U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Red Meat and Processed Meat. Lyon (FR): International Agency for Research on Cancer; 2018. (IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, No. 114.)

Cover of Red Meat and Processed Meat

Red Meat and Processed Meat.

Show details

4. MECHANISTIC AND OTHER RELEVANT DATA

4.1. Digestion and metabolism

The composition of red meat and processed meat, as well as their potential contaminants, is described in detail in Section 1 of this Monograph. Red meat and processed meat are sources of high-quality protein, fat in highly variable amounts, and a range of micronutrients. The impact of the digestion of protein and fat, and the modifications that these macronutrients may undergo in the processing of meat, is addressed in this section. The specific components of red meat and processed meat, including haem iron, lipid oxidation products, heterocyclic aromatic amines (HAAs), polycyclic aromatic hydrocarbons (PAHs), and N-nitroso compounds (NOCs), that are potentially involved in carcinogenesis are discussed in Section 4.5.

After the hydrolytic breakdown of dietary proteins by the activity of proteases, and the absorption of the resultant amino acids and dipeptides in the proximal gut, fermentation of excess proteins may yield toxic compounds. The amount of protein that enters the colon depends on the protein content of the ingested food and the protein digestibility (Windey et al., 2012). Digestibility of dairy and animal proteins exceeds 90%, and is generally higher than the digestibility of plant proteins (70–90%). Storage and processing of meat before consumption may alter its protein digestibility. Cooking of beef affected bovine myofibrillar protein susceptibility to proteases in vitro, with increased or decreased rates depending on the nature of the proteases, and the time and temperature parameters (Santé-Lhoutellier et al., 2008). Similarly, Bax et al. (2012) reported that ageing and mincing had little impact on the in vitro digestion of pig muscle proteins, but heat treatment had temperature-dependent effects. At 70 °C, the proteins underwent denaturation, enhancing the speed of pepsin digestion by increasing enzyme accessibility to protein cleavage sites. At above 100 °C, the proteins underwent oxidation-related aggregation, slowing the speed of pepsin digestion, but improving overall meat protein digestibility. In a study of miniature pigs fed meat from a calf, the true ileal protein digestibility averaged 95%, and was not affected by cooking temperature or by the level of meat intake (Bax et al., 2013). Chemical oxidation of pig myofibrillar proteins has been shown to reduce protein digestibility in vitro (Santé-Lhoutellier et al., 2007). Overall, the impact of thermal denaturation and oxidation of meat proteins during processing and storage on their digestibility, as well as the formation of carcinogenic compounds during digestion, is not well known.

On a normal mixed diet, the amount of protein rather than the source determines the quantity that reaches the colon (Silvester & Cummings, 1995). Hence, high-meat, low-fibre diets may stimulate protein fermentation in the colon, producing short- and branched-chain fatty acids, ammonia, phenolic and indolic compounds, and hydrogen sulfide (O’Keefe, 2008). Bacterial proteases and peptidases are more active when pH is neutral to alkaline. In the proximal colon, pH is more acidic due to the production of short-chain fatty acids, primarily from carbohydrate fermentation, but also from reductive deamination of many amino acids. In more distal parts of the colon, pH is higher and protein fermentation becomes more prominent. In relation to meat intake, ammonia and hydrogen sulfide are the most critical compounds because of their known toxicity (Attene-Ramos et al., 2007; Windey et al., 2012). Meat is rich in sulfur-containing amino acids, possibly leading to higher hydrogen sulfide concentrations in the colon. However, hydrogen sulfide in the gut originates from both the fermentation of sulfur-containing amino acids and dietary sulfate.

A diet high in red meat or processed meat may contain high levels of fat. The digestion of food lipids consists of a series of enzyme-catalysed steps resulting in absorbable components, whereby the release of bile from the gallbladder is essential. It has been suggested that dietary fat promotes the development of cancer of the colorectum (Boyle et al., 1985; Reddy, 1992). Several mechanisms have been postulated to explain this association, including the stimulating effect of high-fat intake on the secretion of secondary bile acids in the gut; this proposed mechanism has received the most attention. These bile acids may promote tumour formation by acting as aggressive surfactants on the mucosa, thus increasing cell loss and proliferation (Bruce, 1987; Owen, 1997; Bernstein et al., 2005). Other proposed mechanisms for the promoting role of dietary fat include an increase in the amount of free fatty acids in the colonic lumen, which may damage the colonic epithelium and induce cell proliferation, and an augmented risk for obesity (Calle & Kaaks, 2004). Dietary fat intake is also associated with peroxidation of unsaturated fatty acids (see Section 4.5.2).

[The Working Group noted that the digestion of red meat and processed meat provides energy and supplies essential nutrients, such as amino acids, iron, other minerals (including zinc), long-chain fatty acids, and various vitamins. At the same time, the digestion of protein and fat yields intrinsically toxic compounds. However, protein and fat are also present in dairy, fish, poultry, and other food products (Demeyer et al., 2015).]

4.2. Mechanisms of carcinogenesis

This section summarizes the evidence for the key characteristics of carcinogens (Smith et al., 2016), concerning whether red and processed meat intake is genotoxic, induces epigenetic effects, induces oxidative stress, and alters cell proliferation, cell death, and nutrient supply. Other mechanistic effects of red and processed meat intake, including whether it induces chronic inflammation and modulates receptor-mediated effects, are also addressed. Potential indirect mediators and studies of hemin and hemin chloride are summarized. Within each topic, studies are presented according to species (human and experimental systems) and test system (in vivo and in vitro), and red meat and processed meat studies are presented separately.

4.2.1. Genetic and related effects

Red meat and processed meat have been tested in studies of DNA damage, gene mutation, chromosomal damage, and epigenetic end-points. These studies are summarized in Table 4.1 to Table 4.6.

(a) Exposed humans

(i) DNA damage and DNA adducts

See Table 4.1.

Table 4.1. Genetic and related effects of red meat or processed meat in exposed humans.

Table 4.1

Genetic and related effects of red meat or processed meat in exposed humans.

Regarding studies of red meat, Lewin et al. (2006) conducted a randomized crossover study in 21 human subjects fed diets that were high in red meat (420 g/day), vegetarian, or high in red meat and fibre for 15 days. Compared with vegetarian diet, red meat intake significantly increased levels of O6-carboxymethyl guanine (O6-CMG), a DNA adduct putatively related to NOCs, in exfoliated colon cells. This adduct was detected by immunohistochemistry using polyclonal antiobodies. [The Working Group noted the lack of specificity of this method for this particular adduct).]

Increased levels of O6-methylguanine (O6-MeG), also a DNA adduct putatively related to NOCs, were shown by immunohistochemistry using monoclonal antibodies in rectal biopsies of human volunteers after an intake period of 4 weeks that was high in red meat (300 g/day) in a randomized crossover study (Le Leu et al., 2015).

No statistically significant associations were found between dietary intake of well-done meat assessed by questionnaire and DNA adducts putatively related to 2-amino-1-methyl-6-phenylimidazo[4, 5-b]pyridine (PhIP) in normal breast tissue (Zhu et al., 2003). The study included 106 women newly diagnosed with cancer of the breast, of which 49 women underwent reduction mammoplasty. PhiP–DNA adducts were assessed by immunohistochemistry using polyclonal antibodies. [The Working Group noted the lack of specificity of this method for these adducts. The type of meat was not specified]

Fried meat [not specified] intake, assessed by food frequency questionnaire (FFQ), was significantly correlated with the presence of bulky, non-specific DNA adducts (32P-postlabelling analysis) in the breast tissue of 44 women undergoing reduction mammoplasty (Rohrmann et al., 2009a).

Chien & Yeh (2010) showed that barbecued pork meat exposure increased oxidative DNA lesions in urine. They gave one meal of barbecued pork meat (reported as 15 or 30 g/kg bw) to eight or five volunteers, respectively. Statistically significant increases in urinary 8-hydroxy-2′-deoxyguanosine (8-OHdG) were observed 2 or 3 days after barbecued pork meat consumption. A correlation was found between PAH metabolites and 8-OHdG in urine (Chien & Yeh, 2010). [The Working Group noted the very high reported intake level of pork meat and of the borderline significance of the results.]

Regarding processed meat, the study by Rohrmann et al. (2009a) previously mentioned reported a significant correlation of intake (assessed by FFQ) with the presence of bulky, non-specific DNA adducts (32P-postlabelling analysis) in the breast tissue of 44 women undergoing reduction mammoplasty.

(ii) Gene mutation

See Table 4.1.

Regarding red meat, no association was found between K-RAS mutation frequency and meat consumption, assessed by FFQ, in colorectal cancer samples from 43 patients (O’ Brien et al., 2000). Similarly, there was no association between tumours with K-RAS mutations and meat consumption in a large cohort study of 448 patients with cancer of the colon and 160 patients with cancer of the rectum from the Netherlands Cohort Study (NLCS) (Brink et al., 2005).

In the randomized crossover study by Lewin et al. (2006) previously mentioned, no K-RAS mutations were present in the exfoliated colon cells of volunteers fed diets that were high in red meat (420 g/day), vegetarian, or high in red meat and fibre for 15 days.

A positive association between haem iron intake and risk of cancer of the colorectum harbouring G→A transitions in K-RAS and APC genes, and TP53 overexpression was found in a prospective study (Gilsing et al., 2013). In the European Prospective Investigation into Cancer and Nutrition (EPIC) study in Norfolk, England, TP53 mutations in cancer of the colorectum were examined in relation to dietary and lifestyle factors (Park et al., 2010). Higher daily total meat and red meat intake (assessed by FFQ) was significantly associated with harbouring TP53 mutations in cancer of the colorectum.

In a case–control study in the Netherlands, Diergaarde et al. (2003) evaluated the association between dietary factors and APC mutations in sporadic colon carcinomas (184 cases, 259 controls). Direct sequencing was used to screen the mutation cluster region of APC in the colon tumours. Red meat intake appeared to be associated with APC mutated tumours: the odds ratios (ORs) for the association between the two highest tertiles (58–87 g/day and ≥ 86 g/day) of red meat intake and APC mutations were 1.5 (95% confidence interval, CI, 0.7–3.0) and 1.7 (95% CI, 0.8–3.6), respectively.

Regarding processed meat, analyses of APC mutations and APC promoter 1A methylation were performed on 185 archival colorectal cancer samples from participants of the EPIC-Norfolk study, with the aim of relating these to a 7-day dietary and lifestyle data collected prospectively (Gay et al., 2012). Truncating APC mutations and APC promoter 1A methylation were identified in 43% and 23% of colorectal cancer samples analysed, respectively. Cases with APC mutations or APC promoter 1A methylation consumed significantly higher levels of processed meat and iron from red meat and red meat products. In a logistic regression model adjusted for age, sex, and cigarette smoking status, each 19 g/day (one standard deviation, SD) increment increase in processed meat consumption was associated with APC mutations with GC→AT transitions (OR, 1.68; 95% CI, 1.03–2.75).

(iii) Faecal water genotoxicity

See Table 4.2

Table 4.2. DNA damage induction by human faecal water following meat consumption.

Table 4.2

DNA damage induction by human faecal water following meat consumption.

Rieger et al. (1999) first reported that a diet high in fat and meat increased faecal water genotoxicity (tested with comet assay in HT-29 cell cultures) in seven healthy volunteers over a period of 12 days. Compared with a diet rich in vegetables and poor in fat and meat, a diet rich in fat (total energy intake, 50%), meat, and sugar, and poor in vegetables and free of wholemeal products [no exact composition was given], significantly increased faecal water genotoxicity. [type of meat was not specified].

Faecal water genotoxicity (tested with comet assay in HT-29 cell cultures) from two randomized controlled studies of red meat (60 or 420 g/day), a vegetarian diet, or haem iron supplements for 15 days in volunteers (n = 21) was evaluated by Cross et al. (2006). Diet had no effect on faecal water genotoxicity (i.e. red meat had no effect). This study was performed under the same conditions as those described by Rieger et al. (1999), but did not confirm those results.

Hughes et al. (2002) studied the effect of vegetables, tea, or soy on faecal water genotoxicity (tested with comet assay in Caco-2 cell cultures) in 11 volunteers fed a high–red meat (420 g/day) diet for 15 days. Low to moderate levels of genotoxicity were observed. [The Working Group noted that the study did not contain a control group consuming a low–red meat diet.]

Faecal water from volunteers (n = 12) fed a red meat (420 g/day, males; 366 g/day, females) or vegetarian diet (Joosen et al., 2009) was tested for genotoxicity by comet assay in Caco-2 cells. Surprisingly, the vegetarian diet produced more DNA strand breaks than the red meat diet. No effect of diet was found in a similar study by the same authors (Joosen et al., 2010) assessing faecal water genotoxicity in volunteers (n = 13) fed a red meat versus fish diet.

More recently, Hebels et al. (2012) showed increased faecal water genotoxicity in a heterogeneous group of inflammatory bowel disease/irritable bowel syndrome patients (n = 12) after 7 days of high–red meat intake (300 g/day), compared with the results obtained before the intervention. In 10 of the subjects, faecal water genotoxicity significantly increased with red meat intake (tested with both standard comet assay and the formamidopyrimidine procedure to measure oxidative damage in Caco-2 cells). Microarray analyses in colon biopsies indicated significant modulation of various signalling pathways (e.g. cytoskeleton remodelling, development, and immune response) (Hebels et al., 2012).

Regarding processed meat, the study by Joosen et al. (2009) previously described found that faecal water from subjects on the vegetarian diet compared with that from subjects on the processed meat diet produced more DNA strand breaks, as assessed by comet assay in Caco-2 cells.

(iv) Mutagenic activity in urine

See Table 4.3

Table 4.3. Bacterial mutagenic activity of human urine following meat consumption.

Table 4.3

Bacterial mutagenic activity of human urine following meat consumption.

The first report of mutagenic activity in the urine of subjects after ingestion of meat was published in 1982 (Baker et al. 1982). Fried pork (150 g) was given to five subjects, and bacterial mutagenicity of urine was determined. Peaks in urinary mutagenicity (in Salmonella typhimurium strains TA98 and TA1538 with S9) were detected 2–4 hours after ingestion.

Dolara et al. (1984) reported a modest increase in mutagenic activity in the urine of subjects fed pork fried in a pan at 200 °C. Mutagenic activity (in S. typhimurium strain TA1538 with S9) was present in 3 of 13 samples analysed and was much lower than that reported by Baker et al. (1982). Hayatsu et al. (1985) also documented mutagenicity (S. typhimurium strain TA98 with S9) in the urine of three volunteers 1.5 hours after consumption of fried ground beef.

Doolittle et al. (1989) studied the effects of different cooking methods on mutagenicity. As a small part of the study, the urinary mutagenicity of different cooking procedures was compared in 12 subjects (6 males, 6 females). Fried meat increased urinary mutagenicity (S. typhimurium strains TA98 and TA100 with S9) compared with boiled or baked meat.

Gabbani et al. (1998) determined urinary mutagenicity 24 hours after ingestion of two pan-fried hamburgers (2 × 100 g) in 32 volunteers. GSTM1 and NAT2 genotypes were also evaluated. Urinary mutagenicity was tested in S. typhimurium strains TA98 and YG1024, with the latter overexpressing O-acetyltransferase. Mutagenicity (a doubling over the spontaneous revertant number) was seen in the YG1024 strain (in 23 of 32 samples), but not in the TA98 strain. Furthermore, NAT2 slow acetylators had higher urinary mutagenicity. Similar results (i.e. increased mutagenic activity after a pan-fried hamburger meal) were shown by Pavanello et al. (2002) in a larger group of subjects (n = 50).

Peters et al. (2004) studied urinary mutagenesis in a group of 60 volunteers who consumed red meat cooked at 100 °C for 7 days followed by red meat cooked at 250 °C for an additional 7 days. Both unhydrolysed and acid-hydrolysed urine samples, containing unmetabolized mutagens and both metabolized and unmetabolized mutagens, respectively, were tested in S. typhimurium strain YG1024. Unhydrolysed and hydrolysed urine samples were 22 and 131 times more mutagenic, respectively, in subjects who consumed red meat cooked at 250 °C compared with those who consumed red meat cooked at 100 °C.

Shaughnessy et al. (2011) reported increased mutagenic activity (in S. typhimurium strains TA98 and YG1024) in the hydrolysed urine and faeces of subjects (n = 8) who consumed red and processed meat cooked at a high temperature of 250 °C (11 minutes/side) for a period of 2 weeks.

Regarding processed meat, the previously mentioned study by Baker et al. (1982) reported increased urinary mutagenicity (in S. typhimurium strains TA98 and TA1538 with S9) in five subjects fed fried bacon (150 g). Similarly, Dolara et al. (1984) reported a modest increase in mutagenic activity in the urine of subjects consuming pan-fried bacon.

(v) Epigenetics

Regarding red meat, microRNA expression in the rectal mucosa of volunteers consuming a high–red meat diet, with or without supplementation with butyrylated high-amylose maize starch (HAMSB), was evaluated by Humphreys et al. (2014). Volunteers received the high–red meat diet for 4 weeks, with washout periods and different orders of treatments. HAMSB significantly lowered a cluster of microRNAs (miR17–92, designated oncomir-1) associated with carcinogenesis. This effect was attributed more to a decrease of these microRNAs by HAMSB than an increase of these microRNAs by red meat.

Tarallo et al. (2014) showed an association between microRNA (miR-92a) levels in the plasma of healthy individuals and consumption of processed meat and other dietary factors (Tarallo et al., 2014).

(b) Human cells in vitro

See Table 4.4

Table 4.4. Genetic and related effects of meat extract in human cells in vitro.

Table 4.4

Genetic and related effects of meat extract in human cells in vitro.

The basic fraction of a beef extract did not induce chromosomal aberrations in human lymphocyte cultures (irrespective of the presence of S9) (Aeschbacher & Ruch, 1989). A small but statistically significant increase in sister-chromatid exchange was seen in the presence of S9.

No study of processed meat in human cells in vitro was available to the Working Group.

(c) Non-human mammals in vivo

See Table 4.5

Table 4.5. Genetic and related effects of red meat or processed meat in non-human mammals in vivo.

Table 4.5

Genetic and related effects of red meat or processed meat in non-human mammals in vivo.

(i) DNA damage

In studies of DNA adducts after red meat consumption, Winter et al. (2011) quantified O6-MeG adducts by immunohistochemistry in CBJ57 mouse colonocytes after mice were fed different diets (containing 15% or 30% protein as casein or red meat, 30% protein with high-amylose maize starch) for 4 weeks. O6-MeG and para-cresol, a protein metabolite with reported genotoxic activity, significantly increased (P < 0.02) with consumption of red meat compared with casein. O6-MeG adducts were present at the apex of the crypts. Starch attenuated the increase in DNA adduct levels.

DNA damage in colonocytes (assessed by comet assay) was measured in Sprague-Dawley rats fed diets containing 25% cooked lean red meat (300 g/kg diet) or casein (15% or 25%), with or without high-amylose maize starch for 4 weeks (Toden et al., 2006). When starch was absent from the diet, red meat caused a significant increase in DNA damage (26%) compared with casein (P < 0.05). When starch was present in the diet, the red meat effect was not significant. The same authors later fed rats diets of 15%, 25%, or 35% cooked beef or chicken, with or without high-amylose maize starch (Toden et al., 2007). DNA single- and double-strand breaks (assessed by comet assay in colonocytes) were significantly higher in the groups fed high levels of both meats compared with those fed low levels of meat. Red meat was more active than chicken, and starch prevented the damage. Apoptotic cells were also increased by red meat (see Section 4.2.3).

The effect of red meat on colonocyte DNA damage was also studied in pigs (Belobrajdic et al., 2012). Ten male animals (Large White strain) were fed diets containing 300 g/kg of cooked red meat or the same diet supplemented with arabinoxylans (arabinoxylan-rich fraction from wheat) for 4 weeks. The comet assay was performed on colonocytes, together with additional determinations (short-chain-fatty-acids (SCFA), phenol, cresol in the feces, bacterial profile). There was a significant decrease in DNA damage in the diet supplemented with arabinoxylans.

Regarding processed meat, 7-methyldeoxyguanosine levels were measured by immuno-slot-blot assay in the colonic DNA of Swiss mice fed hot dogs containing beef or pork (18% of the diet) for 7 days. The levels of this non-mutagenic adduct were similar in control (n = 5) and treated mice (n = 4) (Mirvish et al., 2002).

(ii) Gene mutation

Dolara et al. (1980) reported no increase in the mutagenicity of the urine after administration of a beef extract to Swiss albino mice. Similarly, the beef extract did not have a mutagenic effect as assessed by intrasanguine host-mediated assay.

Regarding processed meat, the mutagenic activity detected by intrasanguine host-mediated assay in NMRI mice given an extract of pan-fried sausage was very low (Gocke et al., 1982). An extract of pan-fried sausage (500 mg/kg bw) fed to pregnant female C57Bl mice did not increase the frequency of coat-coloured spots in the mouse spot test (Gocke et al., 1982).

(iii) Chromosomal aberrations

A basic extract of pan-fried sausage (up to 1000 mg/kg bw) did not increase the frequency of micronucleated erythrocytes in mouse bone marrow (Gocke et al., 1982).

The caecal water from Sprague-Dawley rats fed a high fat, low fibre, and low calcium diet containing barbecued beef as the protein source (equivalent to 17% of the total diet) for 2 weeks significantly increased all the parameters assessed – micronuclei, nucleoplasmic bridges, and nuclear buds –in the WIL2-NS human B-lymphoblastoid cell line. Control rats were fed 17% casein as the protein source in a diet low in fat, and high in fibre and calcium (Benassi et al., 2007).

(d) Non-mammalian experimental systems

See Table 4.6

Table 4.6. Genetic and related effects of red meat or processed meat in non-mammalian experimental systems.

Table 4.6

Genetic and related effects of red meat or processed meat in non-mammalian experimental systems.

(i) Drosophila

No study of red meat in Drosophila was available to the Working Group. Regarding processed meat, an extract of pan-fried sausage did not increase the frequency of sex-linked recessive lethals in Drosophila (Gocke et al., 1982).

(ii) Bacteria

In red meat studies in vitro, extracts of the charred surface of broiled beef meat and fish, together with smoke produced from the broiling of fish, were first demonstrated to be mutagenic by Sugimura and colleagues Nagao et al. (1977). The charred parts of medium-broiled beef were suspended in dimethyl sulfoxide and tested using the Ames test. The dimethyl sulfoxide extract of the charred meat was mutagenic in S. typhimurium strain TA98 with S9 prepared from the liver of rats treated with polychlorinated biphenyl (PCB). This mutagenic activity was much higher than that anticipated from the benzo[a]pyrene (BaP) content of the cooked food.

Commoner et al. (1978) reported that cooked red meat was highly mutagenic (in S. typhimurium strain TA1538). Hamburger meat cooked rare, medium, or well done was extracted with methylene chloride, dried, and dissolved in dimethyl sulfoxide. Mutagenic activity was dependent on the presence of S9. The mutagens formed did not belong to the class of BaP or protein and amino acid pyrolysis products. Mutagens were produced during common cooking procedures, including the use of electrically heated hot plates.

A sharp rise in the frequency of mutations in S. typhimurium strain TA1538 with S9 was detected when beef was boiled for different periods of time and reached temperatures between 140 °C and 180 °C. The mutagenic activity of the beef (hamburger) cooked under different conditions was limited to the surface layer; uncooked meat or microwave-cooked meat did not produce mutagenic activity (Dolara et al., 1979).

Similarly, Pariza et al. (1979) demonstrated that the mutagenic activity of pan-fried hamburger meat was dependent on cooking time and temperature. Mutagenic activity (in S. typhimurium strain TA1538) was not detected in uncooked hamburger or hamburger pan-fried at 143 °C. In contrast, hamburger pan-fried at 191 °C or 210 °C for up to 10 minutes generated considerable mutagenic activity. Higher mutagenic activity was observed when S9 was from Aroclor 1254–treated rats.

Another study with S. typhimurium strain TA98 showed meat cooked only at a high temperature of 250 °C had mutagenic activity (Peters et al., 2004).

Dolara et al. (1980) reported that the mutagenic activity of a beef extract required the presence of S9 fractions from the liver of rats treated with PCB or 3-methylcholanthrene, but no induction was necessary when the liver came from Swiss albino mice. CD-1 mice had intermediate activation capabilities, which increased after the addition of 0.75% butylhydroxyanisole to their diet. S9 from the liver of human donors had low-activation capabilities.

Regarding processed meat, an extract of pan-fried sausage was mutagenic in S. typhimurium strains TA1538 and TA98 in the presence of S9 mix (Gocke et al., 1982).

4.2.2. Oxidative stress

(a) Humans

(i) Red meat

In a randomized crossover study, Pierre et al. (2006) measured the excretion of 1,4-dihydroxynonane mercapturic acid (DHN-MA), the major urinary metabolite of 4-hydroxynonenal (4-HNE). Eight volunteers were fed different diets providing 55, 55, 80, 205, and 110 mg/day of haem for 15 days: red meat (60 g/day) baseline diet, red meat (60 g/day) with non-haem iron diet, red meat (60 g/day) with haem iron in the form of liver pâté diet, red meat (60 g/day) with haem iron in the form of blood sausage diet, or red meat (120 g/day) diet. The blood sausage diet increased urinary DHN-MA by about two-fold (P < 0.001), but mean urinary 8-iso-prostaglandin F was similar in all groups (Pierre et al., 2006).

In contrast, Hodgson et al. reported no elevation in oxidative stress markers with consumption of lean red meat (Hodgson et al., 2007). Sixty participants were randomized to maintain their usual diet for 8 weeks or to partially replace carbohydrate-rich foods with 200 g/day of lean red meat. In terms of the mean between-group difference in comparison to the control diet, the red meat diet increased iron intake (3.2 mg/day; 95% CI, 1.1–5.4), lowered urinary F2-isoprostane excretion (–137 pmol/mmol of creatinine; 95% CI, −264 to −9), and did not change plasma F2-isoprostanes (–12 pmol/L; 95% CI, −122 to 100) or serum γ-glutamyltransferase (–0.8 U/L; 95% CI, −3.2 to 1.5).

Montonen et al. (2013) reported an association between red meat intake and a blood oxidative stress marker. In 2198 participants selected from the EPIC-Potsdam study, higher consumption of red meat was significantly associated with higher levels of γ-glutamyltransferase, even after adjustment for potential confounding factors related to body mass index (BMI), waist circumference, lifestyle, and diet (Montonen et al., 2013).

Lam et al. (2014) identified several genes involved in oxidative stress that were differentially expressed in patients with lung adenocarcinoma who consumed more red meat. Genome-wide expression (HG-U133A) was measured in the tumour tissue and non-involved lung tissue of 64 patients with adenocarcinoma. Gene expression of 232 annotated genes in the tumour tissue significantly distinguished patients who consumed above or below the median intake of fresh red meat. Several genes were involved in lipid metabolism (e.g. NCR1, TNF, UCP3) and oxidative stress (e.g. TPO, SGK2, MTHFR) (Lam et al., 2014).

(ii) Processed meat

As previously noted, Pierre et al. (2006) reported that a blood sausage diet significantly increased DHN-MA by about two-fold. In a later study, Pierre et al. (2013) showed that cured meat intake increased lipid peroxidation and nitroso compounds in human stool. In a single-blind, crossover, randomized controlled trial, 18 volunteers first followed a low-meat diet for a one week control period and were then fed the following diets for four days each, in a random order: cooked, cured pork shoulder meat (similar to air-exposed picnic ham, 180 g/day); cooked, cured pork shoulder meat with a calcium carbonate capsule (1 g/day of calcium); and cooked, cured pork shoulder meat with α-tocopherol (0.05%). Thiobarbituric acid reactive substances (TBARS) and apparent total N-nitroso compounds (ATNC) increased in the faecal water of volunteers given ham compared with control periods. Calcium carbonate normalized both biomarkers, whereas α-tocopherol normalized only lipid peroxidation in the faeces of volunteers (Pierre et al., 2013).

Belinova et al. (2014) showed that consumption of “cooked-pork seasoned meat” was accompanied by increased oxidative stress marker levels in diabetic patients. In a randomized crossover study, 50 type 2 diabetic patients and 50 healthy subjects underwent two 3-hour meal tolerance tests. The acute effects of a processed hamburger meat meal (150 g/meal) were compared with those of a vegan meal (235 g/meal). During the postprandial phase, consumption of the hamburger meat meal was associated with a significant increase in TBARS in the diabetic patients, but not in the healthy subjects, compared with the consumption of the vegan meal. However, superoxide dismutase activity in the healthy subjects was significantly increased after the vegan meal compared with the hamburger meat meal. In the diabetic patients, plasma concentrations of superoxide dismutase, reduced glutathione, or ascorbic acid did not change during the postprandial phase for either meal (Belinova et al., 2014).

No data concerning direct evaluation of red meat or processed meat in human cells in vitro were available to the Working Group.

(b) Rodents

(i) Red meat

Pierre et al. repeatedly showed that diets containing red meat or haemoglobin significantly increased lipid peroxides in faecal water (TBARS) and urinary DHN-MA, a metabolite of the lipid oxidation product 4-HNE, in rats. In a seminal study by Pierre et al. (2004), groups of carcinogen-initiated rats were given one of three low-calcium, meat-based diets containing 60% freeze-dried meat products: raw chicken (low haem), beef (medium haem), or blood sausage (high haem). Two additional groups of rats were given a non-haem control diet supplemented with ferric citrate or a haem control diet supplemented with haemoglobin to match the iron and haem concentrations of the beef diet, respectively. The blood sausage diet increased TBARS in faecal water by 23-fold. The haemoglobin and beef diets increased TBARS in faecal water by two- to four-fold (all P < 0.01), but the chicken diet did not affect TBARS in faecal water compared with the control diets (Pierre et al., 2004). A recent carcinogenesis study confirmed that only diets containing haemoglobin increased faecal and urinary oxidation biomarkers (P < 0.001), independent of dietary HAAs or nitrates and nitrites, and resulting faecal ATNC (Bastide et al., 2015). Other rat studies by the same researchers on beef meat, haemoglobin, or hemin chloride (see Section 4.2.6 for hemin chloride studies) confirmed that dietary haem induced faecal and urinary lipid peroxides (Pierre et al., 2003, 2006, 2008; Guéraud et al., 2015). Additionally, dietary calcium phosphate (31 g/kg) normalized faecal TBARS induced by beef consumption (Pierre et al., 2008). In contrast, dietary antioxidant agents (rutin and butylated hydroxyanisole, 0.05% each) and olive oil (5%) did not reduce faecal TBARS (Pierre et al., 2008).

(ii) Processed meat

Several studies showed that cured meat intake increased lipid peroxidation and nitroso compound (ATNC) formation in rat stool (Pierre et al., 2010, 2013; Santarelli et al., 2010, 2013). For instance, Santarelli et al. (2013) reported increased urinary DHN-MA in rats fed nine different types of purchased cured meats, including hot dogs, sausages, raw and cooked ham, and pâté. Fermented, raw, dry sausages induced 1.8 times more TBARS in faecal water than hot dogs, but only hot dogs promoted preneoplastic lesions in the colon (see Section 4.3). Thus, no association was found between the occurrence of preneoplastic lesions and the biomarkers of lipid oxidation (Santarelli et al., 2013).

No data from non-human mammalian in vitro studies of red meat or processed meat and oxidative stress were available to the Working Group.

4.2.3. Alteration of cell proliferation and cell death

(a) Humans

Regarding red meat, Le Leu et al. (2015) reported an increase in epithelial proliferation in the rectal biopsies of 23 volunteers given cooked lean red meat (300 g/day) for 4 weeks. Proliferating cell nuclear antigen (PCNA) staining revealed a 38% increase in positive cells per crypt (P < 0.001). Caderni et al. (1999) observed that subjects who reported consuming a diet low in red meat had decreased colorectal mucosa proliferation. The labelling index in the upper part of the crypt was increased in subjects at high risk of cancer of the colon. In a study of 69 subjects who previously underwent surgery for at least two sporadic colon adenomas, dietary habit information was collected by FFQ, and proliferation was measured by [3H]thymidine incorporation into colorectal biopsies. Subjects with low–red meat consumption showed decreased proliferation in the upper part of the crypt (mean ± SD: 2.4 ± 2.1, 5.3 ± 4.6, and 5.9 ± 4.8 for low, middle, and high consumption, respectively; P < 0.01) (Caderni et al., 1999).

Humphreys et al. (2014) reported decreased expression of CDKN1A, an inhibitor of cell proliferation, and increased cell proliferation in the rectal cells of volunteers fed a high–red meat diet, with or without supplementation with HAMSB.

In contrast, O’Brien et al. (2000) observed no correlation between red meat consumption and rectal crypt cell proliferation. Crypt cell proliferation was significantly higher in the normal mucosa of patients with left-sided colorectal carcinoma than in that of healthy controls. Meat consumption was assessed by FFQ, and crypt cell proliferation was determined using rectal biopsies obtained before surgery (O’Brien et al., 2000).

Regarding processed meat, Pierre et al. detected no effect of cured meat intake (180 g/day of a model ham for 4 days) in 18 volunteers on faecal water cytotoxicity in two cell lines, nor on genotoxicity (measured by γ-H2AX induction) (Pierre et al., 2013).

No data concerning direct examination of red meat or processed meat on human cells in vitro were available to the Working Group. As described in Section 4.2.6, contrasting effects of hemin chloride on the proliferation of human colon cancer cells were shown in vitro.

(b) Rodents

Regarding red meat, apoptosis (determined by halo assay) increased in a dose-dependent manner in colonocytes isolated from rats fed diets containing 15%, 25%, or 35% cooked beef or chicken for 4 weeks (Toden et al., 2007). In contrast, lean beef meat was without effect on proliferation or apoptosis in the colon in mice fed a standard American Institute of Nutrition (AIN)-76 diet with 15% or 30% protein as casein or cooked, dried lean beef meat for 4 weeks (Winter et al., 2011). Neither the amount nor the type of protein had an effect on cell proliferation (Ki-67), cell mass (crypt height), or rate of apoptosis (terminal deoxynucleotidyl transferase dUTP nick end labelling, TUNEL, assay)

Khil & Gallaher (2004) showed no proliferative effect of dietary beef protein or beef tallow in 1,2-dimethylhydrazine (DMH)-initiated rats given either casein or beef as the protein source and soybean oil or tallow as the fat source in a 2 × 2 factorial design for 9 weeks. However, there was a significantly greater apoptotic labelling index in the distal colonic mucosa of rats fed the beef tallow compared with the soybean oil (Khil & Gallaher, 2004).

Yang et al. (2002) showed that a beef-based diet (24% freeze-dried beef meat vs casein control) reduced caspase-3 activity and neutral ceramidase activity in the colonic mucosa, but had no effect on sphingomyelinase activity in the colonic mucosa.

Pierre et al. repeatedly showed that diets containing red meat or haemoglobin significantly increased faecal water cytotoxicity. In a seminal study, groups of carcinogen-initiated rats were given one of three low-calcium, meat-based diets containing 60% freeze-dried meat products: raw chicken (low haem), beef (medium haem), or blood sausage (high haem). Two additional groups of rats were given a non-haem control diet supplemented with ferric citrate or a haem control diet supplemented with haemoglobin to match the iron and haem concentrations of the beef diet, respectively. The haem control diet was supplemented with haemoglobin to match the haem concentration of the beef diet. The blood sausage diet enhanced erythrocyte cytolysis by more than 50-fold and CMT93 cell toxicity by eight-fold compared with the non-haem control diet. The haemoglobin and beef diets increased CMT93 cell toxicity by four-fold compared with the non-haem control diet; the chicken diet did not increase CMT93 cell toxicity. A correlation was seen between haem intake and faecal water cytotoxicity (r = 0.98), which was correlated with carcinogenesis promotion (r = 0.65; all P < 0.01) (Pierre et al., 2004). Other studies of dietary beef, haemoglobin, or hemin chloride have confirmed that dietary haem can induce faecal water cytotoxicity (Pierre et al., 2003, 2008; Guéraud et al., 2015).

In in vitro studies, faecal water of rats given a diet with red meat or haemoglobin was more cytotoxic to the wild type Apc+/+ murine cells than to premalignant Apc–/+ murine cells (Pierre et al., 2007; Bastide et al., 2015). Trapping of aldehydes from the faecal water of haem-fed rats reduced peroxides by 95% and cytotoxicity by 75%.

Regarding processed meat, several studies by a single research group showed that cured meat intake in rats can increase faecal water cytotoxicity. For instance, faecal water cytotoxicity increased three-fold in rats given a diet with 55% freeze-dried cooked ham for 100 days (Pierre et al., 2010). Santarelli et al. (2010) tested the effect on faecal water cytotoxicity of 16 types of cooked ham diets fed for 2 weeks to rats, with dark or light muscle colour (a proxy for haem level), low or high processing temperature, added nitrite or none, and plastic anaerobic packaging or none, in a 2 × 2 × 2 × 2 design. Faecal water cytotoxicity depended mostly on processing temperature, with cooked ham being more cytotoxic than raw ham, and nitrite, with nitrite being more cytotoxic than no nitrite. [The Working Group noted that both red meat and processed meat were cytotoxic.]

4.2.4. Other mechanisms of carcinogenesis

(a) Chronic inflammation

(i) Humans

Regarding red meat, four observational studies in humans (Azadbakht & Esmaillzadeh, 2009; Montonen et al., 2013; Viscogliosi et al., 2013; Ley et al., 2014) lent little or no support to the hypothesis that red meat intake is directly associated with inflammation markers. Three intervention studies in volunteers found no effect of red meat intake on inflammation markers (Hodgson et al., 2007; Joosen et al., 2010; Maduro et al., 2013).

Regarding processed meat, two studies were identified. In a nested case–control study of 656 women with type 2 diabetes and 694 healthy women from the Nurses’ Health Study (NHS), Schulze et al. (2005) observed that a dietary pattern including processed meat was strongly related to inflammatory markers. This dietary pattern was high in sugar-sweetened soft drinks, refined grains, diet soft drinks, and processed meat, but low in wine, coffee, cruciferous vegetables, and yellow vegetables. Among the inflammatory markers examined, interleukin-6, C-reactive protein, and E-selectin were correlated with processed meat intake. Scores were adjusted for age and BMI, as well as for six other possible confounders (Schulze et al., 2005).

Spehlmann et al. (2012) reported an association between processed meat intake and inflammatory bowel disease in twins. In German monozygotic and dizygotic twins, where at least one sibling had inflammatory bowel disease (n = 512), a high consumption of processed meat, including sausage, was one of the variables most significantly associated with Crohn disease or ulcerative colitis. Likewise, differences in consumption of red meat were also detected in all discordant twin and non-twin Crohn disease groups (Spehlmann et al., 2012).

The hypothesis associating N-glycolylneuraminic acid (Neu5Gc) and chronic inflammation is discussed in Section 4.5.7 (Samraj et al., 2015).

No data from human in vitro studies of red meat or processed meat and inflammation were available to the Working Group.

(ii) Rodents

Regarding studies of red meat, mice fed grain-finished beef for 2 weeks showed enhanced prostaglandin E2 from peritoneal macrophages after inflammatory stimulation. The release of prostaglandin E2 was lowest with diets of range-fed beef, range-fed bison, and elk, and highest in mice fed grain-finished beef (P < 0.05). Prostacyclin release was highest in mice fed elk, intermediate in mice fed feedlot-finished beef or bison, and significantly decreased in mice fed range-fed bison, range-fed beef, or chicken (Broughton et al., 2011). [The Working Group noted that the study design did not include a no-meat control group. Thus, the comparison was done between types of meat, but the effect of meat per se was not assessed.]

Studies that reported on the effect of dietary haem on inflammation markers are described in Section 4.2.6 and Section 4.5.1.

(b) Modulation of receptor-mediated effects (hormones)

Regarding red meat, three observational human studies were found suggesting that red meat intake may be associated with slightly unfavourable insulin-like growth factor-1, sex hormone–binding globulin, or fasting insulin profiles. The associations (expressed as mean or median change across categories of red meat intake) were usually weak, and were often not confirmed by more than one study. In addition, sometimes the trend over categories lost statistical significance when BMI was included in the model (Allen et al., 2000; Brinkman et al., 2010; Ley et al., 2014).

No data from in vitro human studies, or from experimental systems, on hormones or receptor-mediated effects were available to the Working Group.

(c) Telomere length

O’Callaghan et al. (2012) showed that telomere length in colonocytes in Sprague-Dawley rats decreased in proportion to the level of red meat (15%, 25%, and 35% for 4 weeks) in their diet. High-amylose starch attenuated the effect of red meat.

4.2.5. Other relevant data and potential indirect mediators

(a) Dysregulation of the gut microbiota

Diet is a key factor in determining the composition of the human gut microbiota (Graf et al., 2015). A role for microbiota in the development of cancer has been described (Louis et al., 2014; Garrett, 2015), acting via various mechanisms. Bacterial metabolites such as hydrogen sulfide, secondary bile acids, polyamines, and reactive oxygen species (ROS) may provoke inflammation and affect carcinogenesis, while other metabolites such as acetate, propionate, and butyrate may exert protective activities (O’Keefe, 2008). Pathogenic bacteria, in particular, exert proinflammatory effects and might thus increase carcinogenesis (Louis et al., 2014). Specifically, several studies have reported a positive association between the gram-positive Streptococcus gallolyticus (previously named Streptococcus bovis) and cancer of the colorectum (Ellmerich et al., 2000; Tjalsma et al., 2006; Abdulamir et al., 2011).

As discussed in Section 4.5.2(b), Martin et al. (2015) observed reduced faecal TBARS when haemoglobin-fed rats were treated with a cocktail of antibiotics. Some Lactobacillus strains reportedly exert antioxidant behaviour by preventing the Fenton reaction (Sun et al., 2010), while other bacterial species such as Enterococcus faecalis can stimulate extracellular superoxide (Huycke & Moore, 2002). Although the lower faecal TBARS in antibiotic-treated rats in the study by Martin et al. (2015) could be the result of a diminished or altered colonic microbiome, this reduction could be attributed to the direct antioxidant or pro-oxidant effects of the applied antibiotics. For example, metronidazole, which was among the antibiotics administered by Martin et al. (2015), has been described to scavenge ROS in a cell-free environment (Narayanan et al., 2007) and to have an antioxidant effect in colonic tissues (Pélissier et al., 2007).

[The Working Group noted that no direct data on the dysregulation of the gut microbiota by red meat or processed meat were available.]

(b) Type 2 diabetes

A link between high–processed meat intake and diabetes has been hypothesized, and epidemiological meta-analyses have observed a positive association between diabetes and a variety of cancers, including cancer of the liver (El Serag et al., 2006), pancreas (Ben et al., 2011), endometrium (Zhang et al., 2013), colorectum (Larsson et al., 2005), and bladder (Larsson et al., 2006). Various mechanisms have been proposed, such as increased oxidative stress (Ihara et al., 1999; Ceriello & Motz, 2004). Hua et al. (2001) reported lower insulin sensitivity in healthy meat eaters compared with lacto-ovo-vegetarians. Lowering the iron content of the body by phlebotomy improved insulin sensitivity in the meat eaters. The development of insulin resistance increases circulating levels of insulin, triglycerides, and non-esterified fatty acids, which may stimulate colon cell proliferation (Bruce et al., 2000). Other possible mechanisms include the formation of NOCs, advanced glycation end products (AGEPs), trimethylamine N-oxide, branched amino acids, endocrine disruptor chemicals, and inflammation (Azadbakht & Esmaillzadeh, 2009; Tong et al., 2009; Kim et al., 2015). [The Working Group noted that a high–red meat and/or high–processed meat consumption may have an indirect stimulating effect on carcinogenesis by contributing to an increased BMI, which has also been linked to insulin resistance and an increased risk of diabetes.]

4.2.6. Studies of hemin and hemin chloride

In many rodent studies not previously mentioned, a model haem molecule was added to the diet as a surrogate for red meat: hemin chloride. This molecule is a protoporphyrin IX containing a ferric iron ion (haem B) stabilized with a chloride ligand (Deo et al., 2015). It is not present in human diets, is not soluble in water, and quickly induces oxidation of polyunsaturated oils.

(a) Hemin and proliferation

The Van der Meer group repeatedly showed that dietary hemin chloride increased colonic epithelial proliferation and faecal water cytotoxicity in rats (e.g. Sesink et al., 1999, 2000, 2001). For instance, colonic epithelial proliferation increased in rats fed a purified diet supplemented with 1.3 µmol/g of hemin for 14 days. The faecal water of haem-fed rats contained approximately three-fold higher levels of faecal TBARS and was highly cytotoxic compared with that of control rats (Sesink et al., 1999). Cytotoxicity and proliferation were independent of dietary fat content, but were suppressed by dietary calcium phosphate and by dietary chlorophyll both of which bind physically to hemin (Sesink et al., 2000, 2001; de Vogel et al., 2005).

Winter et al. (2014) also showed that dietary hemin chloride increased proliferation in the short term and inhibited apoptosis in the long term in mice fed a high-fat, low-calcium control diet or a high-fat, low-calcium diet with hemin chloride (0.013%). Changes from 1 to 18 months showed increased cell proliferation (P < 0.01) in all groups, but only hemin chloride–fed mice showed reduced apoptosis (P < 0.01) (Winter et al., 2014).

(b) Hemin and inflammation

Several studies investigated the effect of dietary hemin on inflammation markers, showing various effects on myeloperoxidase in the gut mucosa (decreased, increased, or no change). In mice, dietary hemin exacerbated colitis induced by trinitrobenzene sulfonic acid, but decreased myeloperoxidase activity (Schepens et al., 2011). In mice fed a “Western-type” diet with 40% fat (mainly palm oil) and low calcium (30 μmol/g) for 14 days, dietary hemin resulted in a ruffled intestinal epithelium, which was attributed to luminal necrosis. However, there was no indication of local inflammation: no infiltration of neutrophils or macrophages in the lamina propria, no change in the expression of inflammation markers for macrophages (CD14, CD68, CD11b, and F4/80) and for neutrophils (myeloperoxidase, lactoferrin, neutrophil elastase, and EMR4), and no effect on mucins or on gene expression of secreted MUC2 (IJssennagger et al., 2012b). Finally, in rats given a high-fat safflower oil diet, dietary hemin chloride significantly increased colonic myeloperoxidase activity (Guéraud et al., 2015).

(c) Hemin in vitro

Hemin chloride was a potent growth factor in iron-depleted human colon cancer HT-29 cells, but it showed dose-dependent cytotoxic effects on the same cell line (Klenow et al., 2009). It had hyperproliferative effects on Caco-2 cells mediated by haem oxidase and hydrogen peroxide, which was shown using the inhibitors zinc protoporphyrin and catalase (Ishikawa et al., 2010).

[The Working Group noted that dietary hemin chloride markedly increased faecal water cytotoxicity and proliferation of the colonic epithelium in rats and mice. However, the relevance to red meat intake was unclear since the hyperproliferative effect was not reproduced with natural haemoprotein or meat.]

4.3. Precancerous lesions

4.3.1. Precancerous colorectal lesions

(a) Humans

(i) Red meat

Several cohort and case–control studies examined the association between red meat consumption and risk of colorectal adenomas. Of the cohort studies, all showed a positive, but not statistically significant, association between red meat and risk of adenomas (Nagata et al., 2001; Chan et al., 2005a; Wu et al., 2006; Rohrmann et al., 2009b; Tantamango et al., 2011; Ferrucci et al., 2012). However, in a meta-analysis of these studies, the overall association was statistically significant: per 100 g/day increase in intake of red meat, the relative risk (RR) increased by 20% (95% CI, 1.06–1.36) (Aune et al., 2013). The meta-analysis of 10 case–control studies also yielded a positive association (OR, 1.34; 95% CI, 1.12–1.59). Several sensitivity analyses examined potential confounders, also addressed in Section 2.1.5 of this Monograph. These did not appreciably change the risk estimates, such that the associations of the meta-analysis were still statistically significant (Aune et al., 2013). As they are more likely to progress to adenocarcinomas than smaller adenomas, large adenomas were specifically evaluated in some studies, including the EPIC-Heidelberg study (OR, 1.98; 95% CI, 1.09–3.58; top vs bottom quintile) (Rohrmann et al., 2009b) and the Health Professionals Follow-Up Study (HPFS) (OR, 1.95; 95% CI, 0.97–3.91; top vs bottom quintile) (Wu et al., 2006). In single studies, differences in the adenoma characteristics and/or types of red meat were sometimes noted. For example, in the EPIC-Heidelberg study, a high intake of red meat and processed meat (combined) was related to an increased risk of colon adenomas (OR, 1.53; 95% CI, 1.01–2.30) and large adenomas (as noted above), but there was no statistically significant association with adenomas at all sites or small adenomas (Rohrmann et al., 2009b). Ferrucci et al. (2012) did not observe an association between red meat consumption and all types of adenomas. However, they found a statistically significant association between grilled meat (OR, 1.56; 95% CI, 1.04–2.36; top vs bottom quartile), and also well-done or very well-done meat (OR, 1.59; 95% CI, 1.05–2.43), and risk of rectal adenomas. No association was found between these meat types and colon adenomas.

The meta-analysis by Aune et al. (2013) also examined the effects of meat intake by type, and reported statistically significant positive associations between beef and pork intake and risk of adenoma (Aune et al., 2013). A meta-analysis of case–control studies reported a statistically significant increased risk of colorectal adenoma with beef consumption (meta-RR, 1.56; 95% CI, 1.15–2.10; I2, 49.9%) (Carr et al., 2016).

(ii) Processed meat

Fewer studies examined the association between processed meat consumption and risk of colorectal adenomas. In the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial, a non-statistically significant increased risk of colorectal adenomas was observed with high–processed meat consumption (OR, 1.23; 95% CI, 0.99–1.52; top vs bottom quartile) (Ferrucci et al., 2012). In the HPFS, an association was found between high–processed meat consumption and risk of distal colon adenomas (OR, 1.52; 95% CI, 1.12–2.08; top vs bottom quintile), which was stronger than that found for red meat (OR, 1.18; 95% CI, 0.87–1.62; top vs bottom quintile) (Wu et al., 2006). A combined analysis of these two studies revealed a 45% increase in the risk of colorectal adenoma (95% CI, 1.10–1.90) per 50 g increase in consumption of processed meat per day (Aune et al., 2013). The only study that examined adenoma recurrence did not find any statistically significant association between processed meat consumption and risk of recurrence for any adenoma types, advanced adenomas, or multiple adenomas (Mathew et al., 2004). Overall, case–control studies showed positive associations between intake of processed meat and colorectal adenomas, but only a minority of these findings were statistically significant. For example, a 30% increased risk of adenoma (95% CI, 1.1–1.5) was observed in the Tennessee Colorectal Polyp Study (TCPS) with 1881 cases (Fu et al., 2011), but no increased risk of adenoma was found in the PLCO trial with 3696 cases (OR, 1.04; 95% CI, 0.90–1.19) (Sinha et al., 2005b). A meta-analysis of eight case–control studies found a positive association that did not reach statistical significance (OR, 1.23; 95% CI, 0.99–1.52) (Aune et al., 2013).

Several studies reported on specific types of processed meats, including bacon and sausage. Higher intake of bacon and sausage was associated with an increased risk of colorectal adenoma (OR, 1.14; 95% CI, 1.00–1.30) in the PLCO trial (Sinha et al., 2005b). Similarly, in the TCPS, several different types of processed meats (e.g. hot dogs, sausage, or bacon) were associated with an increased risk of adenoma (Fu et al., 2011).

[The Working Group noted that the epidemiological studies supported a positive association between red meat and processed meat consumption and risk of colorectal adenomas. However, results differed with respect to the type of meat, as well as the site, size, number, and histology of the adenomas.]

(b) Experimental systems

(i) Red meat

Parnaud et al. (1998) first studied the effect of meat (beef, chicken, and bacon) on aberrant crypt foci (ACF). Azoxymethane-induced F344 female rats were randomized to 10 different AIN-76–based experimental diets, all high in calcium. Five diets were adjusted to include 14% fat and 23% protein (standard levels), and five other diets were adjusted to include 28% fat and 40% protein (high levels). Fat and protein were supplied by either lard and casein, olive oil and casein, beef, chicken with skin, or bacon. The meat diets contained 30% or 60% freeze-dried, fried meat. The rats were fed ad libitum for 100 days, and ACF multiplicity (the number of crypts forming each focus) was assessed as a parameter related to tumour promotion. ACF multiplicity was similar among the rats, except for the bacon-fed rats.

The same investigators studied the effects of various meats on ACF, using diets high in calcium (Parnaud et al., 2000). Rats fed a diet containing beef, pork, or chicken meat had a lower concentration of faecal NOCs than those fed the control diet (P < 0.01). In the promotion experiment, unprocessed, cooked meat–based diets did not change the number or multiplicity of ACF compared with the control diet.

Hypothesizing that a high level of calcium in the diet may mask the potential carcinogenicity of red meat, subsequent studies administered meat to azoxymethane-induced F344 rats fed a low-calcium diet (0.8%; mimicking the “Western-type” diet). Accordingly, Pierre et al. (2004) formulated the meat-based diets to contain varying concentrations of haem with the addition of raw chicken (low haem), beef (medium haem), or black pudding (blood sausage, high haem). Chicken, beef, and black pudding were administered at 60% of the diet, thus providing a higher intake of protein than the standard nutritional intake of protein (20% of the diet) for rats. Only diets with haem significantly promoted mucin-depleted foci (MDF) formation (P < 0.01), but all meat diets promoted ACF formation. MDF promotion was greater with the high-haem black pudding diet than with the medium-haem beef diet. MDF promotion was also correlated with increased lipid peroxides in faecal water, measured by TBARS, and cytotoxicity in erythrocytes and the mouse epithelial cell line CMT93 (r = 0.65; P < 0.01).

The same group of researchers tested whether calcium and various antioxidants would reduce the promotion of preneoplastic lesions in DMH-induced F344 rats fed red meat (Pierre et al., 2008). Three diets with 60% beef meat were supplemented with calcium phosphate (31 g/kg diet), antioxidant agents (rutin and butylated hydroxyanisole, 0.05% each), and olive oil (5%). The beef meat diet significantly increased the number of ACF (+30%) and MDF (+100%). These results were associated with increased faecal water TBARS (4-fold) and cytotoxicity in CMT93 cells (2-fold), and urinary DHN-MA excretion (15-fold). Calcium fully inhibited beef meat–induced ACF and MDF promotion, and normalized faecal TBARS and cytotoxicity; however, it did not reduce urinary DHN-MA. The antioxidant mix and olive oil did not normalize beef meat promotion or lipid peroxides.

The effects of red meat and whey protein on azoxymethane-induced ACF were studied by Belobrajdic et al. (2003). Wistar rats were fed red meat (barbecued kangaroo muscle meat) or whey protein concentrate to provide 8%, 16%, and 32% protein by body weight in a modified AIN-93 diet with low fibre, low calcium (0.1%), and high polyunsaturated fat. The 32% whey protein group had significantly fewer ACF in the proximal colon than the 16% and 32% red meat groups (P < 0.05). No effect of the diets was observed in the distal colon.

Khil & Gallaher (2004) examined the effects of individual red meat components (beef protein and tallow) on DMH-induced ACF and colon apoptosis and proliferation. DMH-induced Sprague-Dawley rats were fed either casein or beef protein as the protein source, and either soybean oil or tallow as the fat source, for 9 weeks in an AIN-93 standard diet. Rats fed tallow had fewer ACF (only determined in a portion of the distal colon) and significantly higher apoptosis compared with those fed soybean oil. In addition, faecal bile acid concentrations were significantly lower in rats fed tallow than in those fed soybean oil. There were no significant differences in mucosal cell proliferation.

[The Working Group noted that red meat given to carcinogen-initiated animals promoted the growth of preneoplastic lesions in the colon, and that this effect could be modified by factors such as calcium and antioxidants.]

(ii) Processed meat

In the study by Parnaud et al. (1998) previously mentioned, a significant reduction in ACF multiplicity was observed in bacon-fed, carcinogen (azoxymethane)-initiated rats compared with control rats when calcium levels in the diet were high.

Parnaud et al. (2000) also assessed the effect of a high-fat, high-calcium, bacon-based diet on ACF number and multiplicity in the colon of F344 rats (Parnaud et al., 2000). As previously mentioned, other meats tested were pork, chicken, and beef. The faeces of the rats fed the bacon-based diets contained 10–20 times more NOCs than the faeces of the rats fed the casein-based control diet (P < 0.0001). No ACF were detected in the colon of uninitiated, bacon-fed rats. The number of large ACF per rat and ACF multiplicity were consistently reduced by 12% and 20% in rats fed a 30% or 60% high-fat, bacon-based diet and by 17% in rats fed a 30% low-fat, bacon-based diet (all P < 0.01). [The Working Group noted the lack of effect of dietary bacon on rat colon carcinogenesis in the context of a high-calcium diet.]

Using diets containing low levels of calcium (0.8 g/kg diet), Pierre et al. (2010) showed that a freeze-dried, cooked, cured ham diet fed for 100 days to DMH-induced F344 rats significantly increased the number of MDF in the colon. Promotion was associated with cytotoxicity and lipid peroxidation. In a short-term study (14 days) by the same authors, the cytotoxicity (tested in CMT93 cell lines) and lipid peroxidation (TBARS) of faecal water, and the urinary marker of lipid peroxidation (DHN-MA), increased dramatically in ham- and hemin-fed rats; however, this effect was not observed in rats fed the haemoglobin diet or the sodium chloride (NaCl), nitrite, phosphate diet.

This group also demonstrated that experimental cured meat diets (dark cooked pork meat with nitrite, oxidized; dark cooked meat with nitrite, anaerobic; dark cooked meat, oxidized; dark raw meat, anaerobic; with dark meat obtained from supraspinatus and infraspinatus pig muscle, which contained 15–17 mg of haem per 100 g) fed to DMH-induced rats for 100 days significantly increased the number of ACF per colon compared with the no-meat control diet (Santarelli et al., 2010).

In another study, DMH-induced rats were fed a diet containing hot dogs or saucisson (fermented, raw, dry sausage) (40% and 50% on a dry basis) for 100 days (Santarelli et al., 2013). The hot dog diet significantly increased the number of MDF per colon. The saucisson diet increased the number of MDF per colon, but the increase lacked statistical significance compared with the no-meat control diet. The addition of calcium carbonate (150 µmol/g) to the hot dog diet decreased the number of MDF per colon and faecal ATNC compared with the hot dog diet without calcium carbonate.

In DMH-induced F344 rats, the addition of calcium or α-tocopherol to a diet containing cured pork meat (47% meat diet for 100 days) also significantly reduced the number of MDF per colon, but the number of ACF was not affected (Pierre et al., 2013).

[The Working Group noted that results from a single laboratory showed three different kinds of processed meat given to carcinogen-induced animals promoted the growth of preneoplastic lesions in the colon.]

(iii) Haem and other components of red and processed meat

Pierre et al. (2003) showed that haemoglobin or hemin, the ferric porphyrin component of haemoproteins, promoted ACF in azoxymethane-induced rats when dietary calcium was low. This result suggested that myoglobin, the haemoprotein present in red meat, could also promote cancer of the colon when dietary calcium is low.

Santarelli et al. (2010) further evaluated 4 of 16 diets containing cured meat that modified biomarkers of haem-induced carcinogenesis promotion (faecal and urinary fat oxidation and cytotoxicity) in a short-term (14-day) study. The diets differed in muscle colour (a proxy for haem level), processing temperature, nitrite, and packaging. In DMH-induced rats fed for 100 days, only the cooked, nitrite-treated and oxidized, high-haem meat diets significantly increased faecal levels of apparent total N-nitroso compounds (ATNC) and the number of MDF per colon compared with the no-meat control diet. Specifically, the cooked, nitrite-treated and oxidized, high-haem meat diets increased the number of MDF compared with the cooked, non–nitrite-treated meat diet and with the non-oxidized, high-haem meat diet; faecal ATNC levels were 5–15 times higher in the cooked nitrite-treated and oxidized high-haem meat diets than in the other diet groups, but lipid oxidation products (TBARS) in faecal water and urinary DHN-MA were lower in these groups than in the other selected meat diet groups.

In DMH-induced F344 rats, various biomarkers (TBARS in faecal water and cytotoxicity of faecal water in CMT93 cell lines, ATNC in faeces and urinary DHN-MA) were all significantly reduced by the addition of calcium to a diet containing cured pork meat (47% meat diet for 100 days), while α-tocopherol decreased only the concentration of haem in faecal water and DHN-MA in urine (Pierre et al., 2013). Within the same report, Pierre et al. (2013) also showed that the consumption of cured meat increased ATNC and lipid peroxidation (TBARS) in the faeces of human volunteers (both P < 0.05). Calcium normalized both biomarkers in the human faeces, whereas α-tocopherol only decreased lipid peroxidation in the human faeces (all P < 0.05).

Bastide et al. (2015) investigated the role of various components present in red meat, including haem iron, HAAs, and endogenous NOCs, in causing promotion of cancer of the colon. The relative contribution of haem iron (1% of the diet), HAAs (PhIP and MeIQx, 50 + 25 μg/kg diet), and NOCs (induced by sodium nitrite and sodium nitrate, 0.17 + 0.23 g/L drinking-water) was determined in chemically (azoxymethane) induced rats and in Min mice (fed a 2.5% haemoglobin diet). Haem iron increased the number of preneoplastic lesions (MDF) in rats, but dietary HAAs and NOCs had no effect. Dietary haemoglobin increased tumour load in the small intestine of the Min mice (ApcMin/+) (control diet, 67 ± 39 mm2; 2.5% haemoglobin diet, 114 ± 47 mm2; P = 0.004). In vitro, faecal water from rats given dietary haemoglobin was rich in aldehydes and was cytotoxic to normal cells (Apc+/+), but not to Apc-deficient cell lines (Apc–/+). The aldehydes 4-HNE and 4-hydroxyhexenal were more toxic to normal cells than mutated cells, and were only genotoxic to normal cells. Genotoxicity (measured by γ-H2AX for DNA double-strand breaks) was also observed in the small intestine of Min mice given haemoglobin.

[The Working Group noted that these studies, coming from a single laboratory, highlighted the contribution of haem iron in the promotion of preneoplastic lesions by red meat. One study also suggested that in cured meat–fed rats, the driving mechanism of promotion was due to NOCs, and not to lipid peroxidation products.]

4.3.2. Other precancerous lesions in exposed humans

(a) Barrett oesophagus

Barrett oesophagus is defined as the replacement of oesophageal squamous epithelium with metaplastic columnar epithelium. Four epidemiological studies, three case–control studies and one cohort study, examined whether the consumption of meat is related to risk of Barrett oesophagus (Kubo et al., 2009; O’Doherty et al., 2011; Jiao et al., 2013; Keszei et al., 2013). Only the USA case–control study observed a statistically significant association between total meat consumption and risk of Barrett oesophagus (multivariate-adjusted OR, 1.91; 95% CI, 1.07–3.38; top vs bottom tertile) (Jiao et al., 2013), but none of the other three studies observed this same risk (Kubo et al., 2009; O’Doherty et al., 2011; Keszei et al., 2013). However, the USA case–control study by Jiao et al. (2013) did not differentiate between the consumption of red, white, and processed meat. In another USA case–control study, there was no association between total meat, well-done meat, or barbecued meat consumption and risk of Barrett oesophagus (Kubo et al., 2009), but an analysis of the same case–control study reported a positive association between a “Western-type” dietary pattern, which is characterized by a high intake of red and processed meat, and risk of Barrett oesophagus (Kubo et al., 2008).

(b) Gastric intestinal metaplasia

Gastric intestinal metaplasia is considered a precursor lesion of cancer of the stomach (Correa et al., 1975). Based on four studies (Nomura et al., 1982; Stemmermann et al., 1990; Fay et al., 1994; Chen et al., 2004), a meta-analysis reported a combined odds ratio of 1.68 (95% CI, 0.98–2.90) for the association between salted/salty meat and intestinal metaplasia, but the heterogeneity between studies was large (I2, 55.4%), which may have been due to their use of different definitions of foods (e.g. all processed meat, cured meat, or bacon only), types of dietary assessment methods, or subgroups of the population (some studies were conducted only among men) (Dias-Neto et al., 2010).

4.4. Cancer susceptibility

4.4.1. Genetic polymorphisms

(a) Humans

(i) Red meat and certain meat components

Several studies have suggested an increased risk of cancer of the colorectum in individuals with NAT2 rapid acetylator status (individuals with two “rapid” alleles), assessed by phenotyping or genotyping. However, meta-analyses of the literature on NAT2 acetylator status (rapid/intermediate vs slow genotype or phenotype) have typically not confirmed such a main effect association (Brockton et al., 2000; Liu et al., 2012; Zhang et al., 2012). This is also true of other cancers, such as those of the lung (Cui et al., 2011), stomach (Zhong et al., 2010), and breast (Ochs-Balcom et al., 2007; Ambrosone et al., 2008), as well as non-Hodgkin lymphoma (Gibson et al., 2013). Unfortunately, adding to the difficulty of interpreting these data, only a few studies, and no meta-analysis or pooled analysis, have reported risk estimates specifically for rapid acetylators – the subset expected to be at the greatest risk. Instead, grouping intermediate with rapid acetylators has been the norm, especially for populations in which the latter phenotype is relatively rare (e.g. Europeans). The inconsistent results for NAT2 in cancer of the colorectum are in sharp contrast to those for NAT2 in cancer of the bladder; the slow NAT2 acetylator status has consistently been associated with an increased risk of cancer of the bladder (except for benzidine (Rothman et al., 1996)), due to the ability of NAT2 to detoxify arylamines, as shown in a meta-analysis and found in a genome-wide association study (Marcus et al., 2000; Figueroa et al., 2014).

A smaller number of studies have explored associations between polymorphisms in other genes involved in the metabolism of HAAs and PAHs (e.g. CYP1B1, GSTM1, GSTT1, SULT1A1, UGT2B17) and cancer risk. The results of these studies have also been inconsistent or have not been replicated (Andersen & Vogel, 2015). Genome-wide association studies have recently shown that common (i.e. allele frequency > 5%) genetic variants have only a small effect on cancer risk. Importantly, few of the risk variants identified in cancer genome-wide association studies have been in metabolic genes, suggesting that stratification of exposure and very large samples sizes are needed to identify such associations. Indeed, it can be expected that polymorphisms in xenobiotic-metabolizing enzymes (XMEs) involved in carcinogen activation or detoxification would only affect cancer risk when there is a high, biologically sufficient level of exposure to a carcinogen. Thus, it is likely important to consider both the exposure and the genetic variants to detect an association with cancer risk.

Studies that have examined the combined effects of exposure (e.g. red meat, well-done meat, or HAA intake) and metabolic genotypes or phenotypes have mainly focused on cancer of the colorectum and its precursor, adenomatous polyps. Interactions were suggested between intake of red meat, well-done meat, or HAAs and NAT2 acetylator status (Welfare et al., 1997; Kampman et al., 1999; Chan et al., 2005b; Lilla et al., 2006; Nöthlings et al., 2009; Voutsinas et al., 2013), NAT1 (Ishibe et al., 2002; Gilsing et al., 2012), AHR (Wang et al., 2011), CYP1B1 (Cotterchio et al., 2008; Wang et al., 2011), CYP1A1 (Turner et al., 2004, Little et al., 2006; Goode et al., 2007), CYP2E1 (Morita et al., 2009), EPHX1 (Cortessis et al., 2001; Ulrich et al., 2001; Goode et al., 2007), NQO1 (Turner et al., 2004), SULT1A1 (Cotterchio et al., 2008; Barbir et al., 2012), and UGTs (Butler et al., 2005; Girard et al., 2008), as well as with a combination of metabolic genes (CYP1A2, CYP2E1, CYP1B1, and CYP2C9) (Küry et al., 2007) and with a polygenic risk score based on variants in AHR, CYP1A2, CYP1B1, NAT2, SULT1A1, UGT1A7, GSTM1, and GSTT1 (Fu et al., 2012) on the risk of colorectal neoplasia. A meta-analysis of three cohort studies (1404 cases, 2186 controls) (Chen et al., 1998, Chan et al., 2005b, Nöthlings et al., 2009) on the modifying effect of NAT2 on the association between red meat and cancer of the colorectum suggested an interaction between NAT2 status and meat intake. High–red meat intake or preference for browned meat was not associated with an increased risk of cancer of the colorectum in carriers of the slow NAT2 phenotype. In contrast, NAT2 fast acetylators with high-meat intake were at increased risk (OR, 1.25; 95% CI, 0.92–2.01) compared with NAT2 slow acetylators with low-meat intake (Pinteraction = 0.07) (Andersen et al., 2013). However, other studies, some with large sample sizes, failed to replicate this interaction between red meat intake and NAT2 acetylator status on risk of cancer of the colorectum or adenoma (Barrett et al., 2003; Murtaugh et al., 2004; Sørensen et al., 2008; Ananthakrishnan et al., 2015; Budhathoki et al., 2015). Of note, the recent pooled analysis conducted by the Genetics and Epidemiology of Colorectal Cancer Consortium (GECCO) (8290 cases, 9115 controls) found no interaction between red meat or processed meat intake and NAT2 acetylator status in case–control and cohort studies (separately and overall) (Ananthakrishnan et al., 2015). High–red meat intake was similarly associated with cancer of the colorectum in subjects with the rapid/intermediate NAT2 genotype (OR, 1.38; 95% CI, 1.20–1.59) and in subjects with the slow NAT2 genotype (OR, 1.43; 95% CI, 1.28–1.61; Pinteraction = 0.9).

Only four studies of adenoma and/or cancer of the colorectum have considered NAT2 jointly with CYP1A2 activity, which, as previously mentioned, shows high inter-individual variability and may account for individual differences in susceptibility to HAAs. Two of the four studies were case–control studies, and both found that rapid NAT2 activity combined with rapid CYP1A2 activity was a risk factor for colorectal neoplasia or cancer of the colorectum in individuals who ate well-done meat (Lang et al., 1994; Le Marchand et al., 2001). However, in one of the case–control studies, this association was limited to smokers (Le Marchand et al., 2001), which is consistent with the inducing effect of smoking on CYP1A2. In the third study, only NAT2 activity, and not CYP1A2 activity, showed an interaction with HAA intake on the risk of adenoma (Voutsinas et al., 2013). The fourth study failed to observe any modifying effect of NAT2 or CYP1A2 activity, also measured by caffeine phenotyping, on the relationship between HAAs and adenoma (Ishibe et al., 2002).

Fewer studies have examined the interaction between meat intake and genetic polymorphisms on the risk of other cancer sites. However, multiple reports have focused on cancer of the breast and NAT2 (Ambrosone et al., 1998; Gertig et al., 1999; Deitz et al., 2000; Delfino et al., 2000; Mignone et al., 2009; Lee et al., 2013), CYP1A2 (Lee et al., 2013), GSTM1 (Zheng et al., 2002), and SULT1A1 (Lee et al., 2012). Similar to the literature on cancer of the colorectum, publications on cancer of the breast have been inconsistent.

Other cancer-related mechanisms, such as DNA repair, have been explored in studies of cancer, genetic variation, and meat intake. For example, a variant in MGMT, a gene involved in the repair of DNA damage caused by alkylating agents, including NOCs from the diet, was found to interact with both red and processed meat intake on the risk of cancer of the colorectum (Loh et al., 2010). Variants in the nucleotide excision repair enzyme gene, xeroderma pigmentosum group D (XPD), have also been found to increase the risk of cancer of the colorectum when combined with a high intake of heavily browned red meat (Joshi et al., 2009).

Thus, a large number of studies have evaluated the role of genetic polymorphisms in an attempt to clarify the association between cancer susceptibility and red meat consumption. Historically, these studies have focused on suspected mechanisms, and mainly on genes involved in the metabolism of carcinogens present in cooked red meat. The results of these candidate gene studies have mostly been inconsistent. Many were underpowered and had multiple testing, publication, and reporting biases. Inconsistencies in the gene–meat interaction studies may also have resulted from differences in the comprehensiveness of the dietary assessments or the lack of consideration for cytochrome P450 (CYP) enzyme inducers (e.g. smoking). The strongest evidence provided by these studies supported an interaction between NAT2, red meat intake, and risk of cancer of the colorectum. However, as previously noted, meta-analyses and pooled analyses (Brockton et al., 2000; Liu et al., 2012; Zhang et al., 2012; Ananthakrishnan et al., 2015) have failed to confirm a main effect or modifying effect of NAT2 or other genes on cancer of the colorectum or other cancers. Insufficient focus has been given to the group expected to be at the highest risk – those with two rapid alleles. Data are lacking for populations in which this genotype is common, and that consume significant amounts of well-done meat and have high rates of cancer of the colorectum (e.g. Japan and Republic of Korea).

In recent years, genome-wide association studies have identified several cancer susceptibility loci, each with a relatively small effect on risk. Statistical methods have been developed to analyse interactions between diet and variants across the entire genome. These analyses, with the currently available sample sizes, have rarely replicated results from candidate gene studies and have not identified interactions with red meat intake (Jiao et al., 2012; Kantor et al., 2014). However, larger sample sizes are needed to detect modest or weak interactions.

(ii) Processed meat

Processed meat has not always been examined separately from red meat in studies of genetic polymorphisms. A population-based case–control study in Hawaii, USA (Le Marchand et al., 2002a) found an increased risk of cancer of the colorectum in individuals who consumed a high amount of red meat or processed meat and who carried a variant in CYP2E1 that had been shown to alter enzyme activity (Lucas et al., 1995; McCarver et al., 1998; Le Marchand et al., 1999). This association was more pronounced for cancer of the rectum and was observed in individuals who consumed salted/dried fish and oriental pickled vegetables, both food sources of NOCs. An association with the same CYP2E1 variant and cancer of the stomach was also observed (Nishimoto et al., 2000; Chen et al., 2004).

Finally, a genome-wide search for diet–gene interactions identified an interaction between processed meat intake and a variant (rs4143094) on 10p14 (near GATA3) on the risk of cancer of the colorectum (Figueiredo et al., 2014). Although the mechanism was unclear, GATA3 was involved in cell maturation, proliferation arrest, and survival. Loss, or silencing, of expression of GATA genes has been described in colorectal tumours.

[The Working Group noted that few studies have explored the role of genetic susceptibility as a potential modifier of the association between processed meat and cancer. These studies have typically been small, and have not allowed for any conclusions to be drawn.]

(b) Experimental systems

Two studies by the same research group showed that mice humanized for CYP1A2 are more susceptible to HAAs (e.g. PhIP) than wildtype mice (Cheung et al., 2011; Li et al., 2012). [The Working Group noted that the doses used in these studies were greater than human exposure levels, and the relative levels of hCYP1A2 expression may have exceeded the range in humans.]

4.4.2. Microflora

Evidence is also available concerning individual differences in intestinal microflora profiles that may affect the carcinogenic effect of red meat. In rodents, the gut microbiota has been shown to facilitate haem-induced hyperproliferation by opening the mucous barrier (Ijssennagger et al., 2015). Similarly, it has been suggested that intestinal microflora play an important role in the bioactivation of HAAs. Kassie et al. (2004) inoculated intestinal flora collected from either vegetarians or meat eaters into germ-free rats. The rats were fed a diet mimicking the donors’ diets, in terms of the origin of the protein and fat (animal or plant). After oral administration of 2-amino-3-methylimidazo[4,5-f]quinoline (IQ), DNA damage in both colon and liver cells, as determined by comet assay, was significantly lower in animals harbouring the flora from vegetarians than in those harbouring the flora of the meat eaters.

The human intestinal microbiota has been shown to selectively convert PhIP to a major metabolite, 7-hydroxy-5-methyl-3-phenyl-6,7,8,9,-tetrahydropyridol[3ʹ,2ʹ:4,5]imidazo[1,2-a]pyrimidin-5-ium chloride (PhIP-M1) (Vanhaecke et al., 2008a). PhIP-M1 was found to cause cell division arrest and induce DNA strand breaks in the human epithelial intestinal colon carcinoma Caco-2 cell line (Vanhaecke et al., 2008b), suggesting that the ability of the colon microbiota to bioactivate PhIP may affect the risk of cancer of the colorectum. [The Working Group could not make any conclusions regarding effect modification due to the microbiota.]

4.5. Meat components potentially involved in carcinogenesis

4.5.1. Haem iron

(a) Iron intake and digestion

One of the defining characteristics of red meat is its haem iron content. Two types of iron occur in foods: haem iron (organic) and non-haem iron (inorganic) (Fonseca-Nunes et al., 2014). Haem, which is made up of an iron atom surrounded by a porphyrin ring, is included in haemoglobin and myoglobin, and is involved in supplying oxygen to the body’s tissues (Bastide et al., 2011). The redness of meat is mainly determined by its concentration of myoglobin, with the oxidation state and sixth ligand of iron determining the specific colour of meat. Haem iron in nitrite-cured meat is mostly nitrosylated (Demeyer et al., 2015).

Iron overload has many adverse health effects, irrespective of the iron source. While the human body maintains homeostatic control of this trace element, the absorption of haem iron in the small intestine is less regulated and more efficient (15–40%) than the absorption of non-haem iron (Layrisse et al., 1969; Carpenter & Mahoney, 1992; Hooda et al., 2014). Red meat is the largest dietary source of haem iron. Non-haem iron, which is present in both animal and vegetable sources, accounts for the majority of total dietary iron intake and has a wide range of absorption (1–40%). The absorption of non-haem iron is influenced by the body’s iron stores, hypoxia, and erythropoietic activity, as well as by the intake of vitamin C, calcium, and haem iron (Layrisse et al., 1969; Carpenter & Mahoney, 1992; Fonseca-Nunes et al., 2014).

Haem in meat may undergo modifications during processing and digestion. Depending on the time and temperature, myoglobin is denatured after cooking, and the haem moiety is liberated. Haem iron can also be converted, to varying degrees, into non-haem iron by heat treatment (Kristensen & Purslow, 2001; Purchas et al., 2006). Purchas et al. (2006) showed an overall loss of iron from cooking of beef, together with a marked shift from soluble haem and non-haem iron to their insoluble forms. However, after simulated stomach and duodenal digestion, solubility was regained to a significant extent. Kristensen & Purslow (2001) reported that NaCl, widely used in meat processing, increased the haem:non-haem ratio in cooked meat by preventing the haem molecule from liberating iron, whereas calcium ions had a negative effect on the haem:non-haem ratio during cooking of meat. Thus, the type of processing and the cooking conditions affected the content and solubility of haem and free iron in meat, determining the absorption of iron in the proximal gut, and thus the amount that entered the distal gut.

(b) Mechanisms of carcinogenesis

Possible mechanisms by which haem iron may promote colon carcinogenesis include its catalytic effect on the formation of NOCs and on the oxidation of polyunsaturated fats. A third potential mechanism involves its direct effect on colon cells (Bastide et al., 2011; Corpet, 2011; Fonseca-Nunes et al., 2014).

A first possible mechanism of tumour promotion by haem iron is related to NOC formation by N-nitrosation of amines and amides by bacterial decarboxylation of amino acids from meat in the presence of a nitrosating agent. A controlled feeding study showed that high–red meat consumption is associated with greater excretion of ATNC (Cross et al., 2003). ATNC is a collective term that encompasses nitrosyl iron, S-nitrosothiols, nitrosamines, and nitrosamides. In another controlled feeding study, the endogenous production of NOCs was further enhanced when the diet contained haem iron from blood sausage compared with red or white meat (Cross et al., 2003; Hammerling et al., 2015). The main ATNC in the faeces of study participants fed a red meat diet was nitrosyl haem, but in those fed cured meat, NOCs predominated (Joosen et al., 2009; Corpet, 2011). Similarly, in animal studies, diets containing red and processed meat increased faecal NOCs (Mirvish et al., 2003; Demeyer et al., 2015). Some NOCs are carcinogenic compounds, inducing multisite tumours in animals (Lijinsky, 1992).

Several mechanisms have been suggested to explain the effect of haem on faecal ATNC content. One hypothesis concerns the combined action of haem and free thiols on NOC formation. Nitrosothiols are readily formed under the acidic conditions of the stomach, a process that is promoted by haem, and may release nitric oxide (NO) once they are exposed to the alkaline and reductive conditions of the small and large intestine, thereby stimulating the nitrosylation of haem iron. Nitrosyl haem is an NO donor and can act as a nitrosating agent in the lower gut (Kuhnle et al., 2007). Although an increase in ATNC after consumption of red and processed meat has been demonstrated, the potential carcinogenicity of the NOCs formed in the gut is unclear (Demeyer et al., 2015); this is addressed further in Section 4.5.5. A second hypothesis that has been proposed is that changes in the microbiota may be related to NOC production (Ijssennagger et al., 2012, 2013, 2015). Ijssennagger et al. (2012) showed a distinctive shift in the colonic microbial composition of mice fed a Westernized diet (40% fat) supplemented with 0.5 µmol/g of haem iron compared with mice fed the same diet without haem iron. After 2 weeks, the colonic contents of the mice given haem iron contained higher amounts of Bacteroidetes (gram-negative) and lower amounts of Firmicutes (gram-positive) than those not given haem iron supplementation. After the haem iron supplementation, Ijssennagger et al. (2012) also observed an increase in the nitrate-reducing capacity of the colonic microflora, while the sulfate-reducing capacity was unchanged. This increase by haem iron in the nitrate-reducing capacity might be important, as considerable inter-individual variation was observed in the ability of different individual porcine and human microbiota to form NOCs and NOC-specific DNA adducts (Engemann et al., 2013; Vanden Bussche et al., 2014). Similarly, Van Hecke et al. (2014b) showed that haem iron had a stimulating effect on O6-CMG production during in vitro fermentation of meat.

A second possible mechanism of tumour promotion by haem iron involves its ability to catalyse the oxidation of polyunsaturated fats (Corpet, 2011). The formation of lipid oxidation products is discussed in Section 4.5.2. Tumour promotion was found to be associated with increased urinary excretion of DHN-MA, a fat peroxidation biomarker, in rats after intake of haem (Pierre et al., 2006). An increase in this biomarker was also observed in humans consuming blood sausage, which is high in haem (Pierre et al., 2006).

A third possible mechanism of tumour promotion by haem iron involves a direct effect of haem or one of its metabolites on colon cells. In vitro studies by Glei et al. (2002) showed that, when haemoglobin was added to a culture medium, it was taken up by human colon cells and participated in the induction of oxidative DNA damage such as DNA breaks and oxidised bases. As reported in Section 4.2.6, the Van der Meer group (e.g. Sesink et al., 1999) showed that supplementing a diet with hemin chloride, which is not present in food, increased epithelial proliferation and enhanced apoptosis in the colonic mucosa, and induced cytotoxicity in faecal water. Cytotoxicity-induced stress, rather than oxidative stress of surface cells, was the determinant of hemin-induced hyperproliferation.

(c) Epidemiological studies

Methods for estimating haem intake in epidemiological studies are varied. The information on haem iron concentrations in meats was sparse, partially due to the lack of appropriate analytical methods, and the variable concentrations across the range of meat types (e.g. beef, chicken, or pork), cuts of meats from the same animal, and methods of preparation (Kongkachuichai et al., 2002; Lombardi-Boccia et al., 2002; Cross et al., 2012). Two methods for estimating haem iron were to use 40% of total iron from meat (Lee et al., 2005) or to use meat-specific proportions (65% for beef; 39% for pork, ham, bacon, pork-based luncheon meats, and veal; and 26% for chicken and fish) (Balder et al., 2006). Recently, a haem iron database and complementary FFQs were developed to estimate haem iron intake from meats prepared by different cooking methods to a range of doneness levels (Cross et al., 2012) for use in etiological studies.

The inconclusive data for an association between haem iron intake and a variety of cancers may be partially explained by inconsistencies in the methods used to measure haem iron intake. Haem iron was positively associated with colorectal adenomas in two cohort studies: the PLCO trial (Ferrucci et al., 2012) and the National Institutes of Health – American Association of Retired Persons (NIH-AARP) Diet and Health Study (Cross et al., 2010). In a meta-analysis of five prospective studies, the summary relative risk for cancer of the colon was 1.18 (95% CI, 1.06–1.32) for those in the highest versus lowest category of haem iron intake (Bastide et al., 2011). In two other meta-analyses of eight studies each, the summary relative risks for cancer of the colorectum were 1.14 (95% CI, 1.04–1.24) for the highest versus the lowest category of haem iron intake (Qiao & Feng, 2013) and 1.08 (95% CI, 1.00–1.17) for an increase of 1 mg/day in the intake of haem iron (Fonseca-Nunes et al., 2014). Although these analyses were suggestive of a significant but modest increased risk, the measurement of haem iron intake differed in each of the studies included.

In the NIH-AARP study, individuals in the highest category of haem iron intake were at increased risk of cancer of the lung (Tasevska et al., 2009) and prostate (Sinha et al., 2009), as well as chronic liver disease mortality (Freedman et al., 2010), but not hepatocellular carcinoma (hazard ratio, HR, 0.95; 95% CI, 0.68 –1.32; top vs bottom quintile) (Freedman et al., 2010), non-Hodgkin lymphoma (Daniel et al., 2012b), or cancer of the breast (Kabat et al., 2010); haem iron intake was also not associated with cancer of the breast in the PLCO trial (Ferrucci et al., 2009). In a meta-analysis of four studies, the summary relative risk for cancer of the breast was 1.03 (95% CI, 0.97–1.09) (Fonseca-Nunes et al., 2014), and the summary relative risk for cancer of the lung was 1.12 (95% CI, 0.98–1.29) for an increase of 1 mg/day in the intake of haem iron (Fonseca-Nunes et al., 2014). Results were heterogeneous for cancer of the stomach (Fonseca-Nunes et al., 2014) and oesophagus (Cross et al., 2011a; Steffen et al., 2012).

Regarding specific gene mutations in colorectal tumours, haem iron intake was positively associated with an increased risk of colorectal tumours with P53 overexpression, but not colorectal tumours without P53 overexpression in the NLCS (Gilsing et al., 2013). Haem iron intake was associated with an increased risk of colorectal tumours harbouring G→A transitions in K-RAS and APC, and overexpression of TP53 (Gilsing et al., 2013).

4.5.2. Lipid oxidation products

(a) Lipid oxidation in meat

The oxidation of unsaturated fatty acids in meat results in the formation of lipid oxidation products, which are in part cytotoxic and genotoxic (Kanner, 2007; Guéraud et al., 2010). Polyunsaturated fatty acids are especially sensitive to oxidation, which proceeds via a free radical chain reaction involving initiation, propagation, and termination steps. Transition metals, especially iron, catalyse this reaction, in the presence of oxygen, producing unstable ROS, including superoxide, hydrogen peroxide, and hydroxyl radicals. The ROS initiates a chain of oxidative reactions, generating lipoperoxyl radicals and lipid hydroperoxides. Lipid hydroperoxides may decompose to several low-molecular-mass breakdown products, such as aldehydes and hydroxyalkenals, or condense to polymers. The main lipid peroxidation by-products are malondialdehyde (MDA) and 4-HNE (Marnett, 2000; Fig. 4.1); both of these lipid oxidation end products are risk factors to human health (Kanner, 2007; Bastide et al., 2011). MDA is most abundant and can reach 300 µM or more in meat products (Kanner, 2007). It is also toxic and binds to DNA and proteins, or undergoes further oxidation to more reactive epoxy derivatives that can be mutagenic in bacterial, mammalian, and human cells (Basu & Marnett, 1983; Esterbauer, 1993; Guéraud et al., 2010; Bastide et al., 2011). In foods, MDA is bound mainly to the lysine residues of proteins, from which it is released in the course of digestion, as N-α-acetyl-ϵ-(2-propenal)lysine (Piche et al., 1988).

Fig. 4.1

Fig. 4.1

Generic scheme of polyunsaturated fatty acid peroxidation

The fat fraction of meat also contains cholesterol, and it is known that dietary fatty acids and dietary cholesterol are co-oxidized (Kanner, 2007). Cholesterol oxidation via lipid free radicals results in the formation of many oxidation by-products, such as oxycholesterol, which has cytotoxic, pro-oxidant, and proinflammatory activities (Lemaire-Ewing et al., 2005). The amount of oxycholesterol in cholesterol-rich food products, including precooked meat and poultry, can reach 10–100 µM (Kanner, 2007).

The degree of lipid oxidation during the manufacturing of processed meat and storage of fresh and processed meat before consumption depends on many factors, such as the iron and polyunsaturated fatty acid content; the presence of endogenous or added antioxidants, and other additives; and the processing and storage conditions (Morrissey et al., 1998). When good storage and processing practices are followed, the levels of lipid oxidation products in meat at the time of consumption are low. Oxidation of myoglobin and other proteins also occurs, which can interfere with lipid oxidation (Faustman et al., 2010).

(b) Lipid oxidation during digestion of meat

The composition of meat and the conditions prevailing in the different compartments of the digestive tract, including interactions with other foods, determine the extent of formation of lipid oxidation products during the digestion of meat. Saliva in the mouth, acidic gastric juice in the stomach, emulsifying pancreatic and bile juice in the small intestine, and anaerobic fermentation by the microbiota in the large intestine all influence lipid oxidation in the gut. Kanner & Lapidot (2001) showed that lipid oxidation in heated muscle tissue was enhanced in the stomach due to the low pH and dissolved oxygen.

Extensive evidence is available indicating that haem iron in red and processed meat is a key factor in promoting lipid oxidation (Carlsen et al., 2005). Feeding rats heated red turkey cutlets, which are high in haem, increased lipid hydroperoxides and MDA in the stomach (Gorelik et al., 2008). As previously noted, urinary excretion of DHN-MA was increased in rats fed haem and in humans fed blood sausage (Pierre et al., 2006). Plasma MDA concentrations were higher in rats fed beef versus chicken (Toden et al., 2010). Higher MDA, 4-HNE, and hexanal concentrations resulted from in vitro duodenal and colonic digests of beef compared with pork, followed by chicken (Van Hecke et al., 2014a).

A large proportion of ingested haem iron reaches the colon (Pierre et al., 2008), and could thus stimulate oxidation reactions in the colonic contents. However, lower MDA, 4-HNE, and hexanal concentrations resulted from in vitro colonic compared with duodenal digests (Van Hecke et al., 2014a, b; Vanden Bussche et al., 2014). This could be due to the anaerobic conditions in the colon, degradation or metabolism into other compounds. The colonic microbial composition likely also has an influence on oxidation processes (Huycke & Moore, 2002; Sun et al., 2010; Martin et al., 2015).

The high-fat content of many processed meats is likely to result in an increased production of lipid oxidation products. In vitro digestion of a heated pork product containing 5% or 20% pork lard resulted in a higher production of 4-HNE and hexanal compared with heated lean pork containing 1% fat (Van Hecke et al., 2014b). The fatty acid profile is also important. Urinary MDA and DHN-MA in rats increased when haem iron was combined with fish oil (high in n-3 fatty acids) and safflower oil (high in n-6 fatty acids), but not with hydrogenated coconut oil (98% saturated fatty acids) (Guéraud et al., 2015). Similarly, levels of hepatic 4-HNE-histidine protein adducts were higher when haem iron was combined with safflower oil compared with hydrogenated coconut oil.

During the heating of meat, the content of free Fe2+ increases through destruction of the haem-porphyrin moiety, oxymyoglobin releases oxygen with production of hydrogen peroxide, and antioxidant enzymes (e.g. glutathione peroxidase) are inactivated (Kanner, 1994). This stimulates the Fenton reaction, and thus the formation of lipid oxidation products. Rats consuming cooked meat products had increased faecal TBARS and urinary DHN-MA compared with rats consuming raw meats (Santarelli et al., 2010). Similarly, heating compared with not heating a pork product increased the formation of MDA, 4-HNE, and hexanal before and during digestion (Van Hecke et al., 2015).

Nitrite salt is widely used as a curing agent in meat processing. Nitrite has antioxidant properties in processed meat. The formed nitric oxide myoglobin, nitric oxide ferrous complexes, and S-nitrosocysteine have antioxidant properties, and nitric oxide inhibits the Fenton reaction (Kanner, 1994). In acidic conditions, such as in the stomach, nitrous acid generates dinitrogen trioxide and water, which is in equilibrium with nitric oxide (NO) and nitrogen dioxide (NO2) (Honikel, 2008). The balance between NO and ROS has been described as a determinant of the effect of nitrite on oxidant reactions whereby a 1:1 ratio of NO to ROS enhances lipid peroxidation, whereas an excess of NO inhibits oxidation (Darley-Usmar et al., 1995). A study using rats showed that addition of nitrite to meat products reduced TBARS in faecal water (Santarelli et al., 2010). Chenni et al. (2013) found that intake of nitrite through drinking-water (1 g/L) reduced haem-induced lipid peroxidation in the colon of rats by 25%. During in vitro digestion of different nitrite-cured meat products, the formation of lipid oxidation products was markedly inhibited (Van Hecke et al., 2014a, b; 2015). However, this inhibition was less efficient when the fat content of the diet was high (20% fat), and absent when the meat products were subjected to intense heating. The intensely heated meat products, in which nitrite was less efficient at preventing oxidant reactions, contained less residual nitrite.

(c) Absorption, distribution, metabolism, and excretion

A vast amount of literature is available concerning the biotransformation of lipid oxidation products. With respect to toxic aldehydes, the Working Group refers to extensive reviews of Esterbauer et al. (1991) and of Guéraud et al. (2010), and of Poli et al. (2008) for 4-HNE specifically. Most lipid peroxidation–derived aldehydes such as 4-HNE can travel across membranes by passive diffusion. Metabolism occurs in most cells and tissues, and is rapid and complete. As a first and major step is conjugation with glutathione by Michael addition, which may be considered a detoxification reaction, facilitating urinary excretion. Other modifications of the aldehyde function may also occur (e.g. reduction into an alcohol or oxidation into an acid). The liver and the kidneys are the organs primarily involved in the elimination of 4-HNE. DHN-MA appears to be the major urinary metabolite of 4-HNE. MDA is metabolized to carbon dioxide and water via transformation into acetaldehyde, but it is also found unmodified in urine and plasma (Guéraud et al., 2010).

(d) Mechanisms of carcinogenesis

Due to their chemical reactivity, aldehyde breakdown products of lipid oxidation can covalently modify nucleic acids, proteins, and lipids (Guéraud et al., 2010). They also serve as biomarkers of oxidative stress, and are important in cell signalling in both pathological and physiological conditions, mainly in cell cycle regulation. 4-HNE is able to exert cytotoxic, mutagenic, and genotoxic effects. Similarly, MDA has mutagenic and genotoxic properties (Esterbauer, 1993; Guéraud et al., 2010).

After meat consumption, the levels of lipid peroxidation products and their adducts or metabolites increase. For instance, lipid hydroperoxides and MDA accumulation increased more than twofold in the stomach contents of rats fed red turkey cutlets and after pyloric ligation. Postprandial plasma MDA levels increased significantly by 50%. The addition of red wine polyphenols altered these outcomes (Gorelik et al., 2008). In a human study by Brown et al. (1995), urinary MDA increased from 2.1 to 23.1 µmol/day with the consumption of high quantities of cooked meat over a 7-day period. After consumption of red meat by rats and humans, excretion of both MDA and DHN-MA increased in the urine (Pierre et al., 2006, 2008).

4.5.3. Heterocyclic aromatic amines

The following discussion is restricted to studies involving exposure to HAAs as a result of the consumption of red meat or processed meat, together with studies that reported findings directly relevant to the issue of whether such exposure may account for any risk of cancer. The weight accorded to such data depended, among many other considerations, on current knowledge of the carcinogenicity of individual HAAs and of HAAs as a class. See IARC Monographs Volume 56 (IARC, 1993).

Meats cooked at a high temperature contain HAAs (see Section 1.2.3 and Fig. 1.2). HAAs are pyrolysis by-products formed from the reaction between creatine or creatinine found in muscle meats, amino acids, and sugars (Wakabayashi et al., 1992; Sugimura et al., 2004). HAA formation increases with the temperature and duration of cooking, and depends on the type of meat and cooking method (Cross & Sinha, 2004). More than 20 individual HAAs have been identified. After meat consumption of a fried beef meal by human subjects, 24-hour hydrolysed urine contained 2–8.5% PhIP and 13–32% MeIQx of the ingested dose (Reistad et al., 1997).

Most HAAs are potent bacterial mutagens, based on the Ames S. typhimurium test (Ames et al., 1973; Felton et al., 2007). In 1993, the Working Group concluded that HAAs are possibly or probably carcinogenic to humans, including IQ (Group 2A), MeIQ (Group 2B), MeIQx (Group 2B), and PhIP (Group 2B) (IARC, 1993).

(a) Mechanisms of carcinogenesis

Most HAAs are not mutagenic or carcinogenic in their parent form. HAAs acquire the capacity to form DNA adducts and potentially cause DNA damage only after metabolic activation. HAAs undergo rapid and extensive metabolism by phase I and II XMEs (Alexander et al., 1995; Turesky & Le Marchand, 2011), which can lead to either bioactivation or detoxification of the HAAs, as discussed in Section 4.4.

HAA–DNA adduct formation is considered a biomarker for the mutagenic and carcinogenic potential of these xenobiotic compounds (Cheng et al., 2006). Many HAAs have been shown to form DNA adducts in both in vitro and in vivo experiments (Cheng et al., 2006; Turesky & Le Marchand, 2011). The major reaction of the N-hydroxy-HAA derivatives with DNA occurs at deoxyguanosine (dG) to produce dG-C8-HAA adducts, where bond formation occurs between the C8 atom of dG and the activated exocyclic amine group of the HAA (Schut & Snyderwine, 1999; Turesky & Vouros, 2004). For IQ and MeIQx, DNA adducts also form at the N2 group of dG and the C5 atom of the heterocyclic ring structures (Turesky & Vouros, 2004; Turesky & Le Marchand, 2011). While the amount of dG-N2 adducts formed is small relative to the dG-C8 isomers, the dG-N2 adducts can persist in vivo (Turesky & Vouros, 2004; Turesky & Le Marchand, 2011).

In addition to DNA adduct formation, HAAs may exhibit other carcinogenic mechanisms. For example, PhIP may also possess estrogenic activity at very low doses (10−9 to 10−11 M), which can invoke a mitogenic response (Lauber et al., 2004). PhIP at doses as low as 10−11 M had direct effects on a rat pituitary lactotroph model, and induced cell proliferation and the secretion of prolactin. These PhIP-induced effects were suppressed by an estrogen receptor inhibitor. Such hormone-like activities of PhIP provide mechanistic plausibility for carcinogenicity in the breast (Lauber & Gooderham, 2007).

Considerable interspecies differences have been found in the carcinogenicity, mutagenicity and metabolism of HAAs (Hengstler et al., 1999). Carcinogenicity studies have been performed in rats, mice, and monkeys (Ohgaki et al., 1985; Adamson et al., 1990; Hengstler et al., 1999). In rodents, long-term feeding of HAAs induced tumours of the oral cavity, liver, stomach, colon, pancreas, and prostate gland in males and mammary gland in females (Turesky & Le Marchand, 2011). IQ was shown to be a potent hepatocarcinogen in cynomolgus monkeys, but MeIQx failed to induce hepatocellular carcinoma after a 5-year dosing period (Hengstler et al., 1999). Species differences in mutagenicity were most pronounced for MeIQx in S. typhimurium strain TA98 (Ames test) using liver microsomes from cynomolgus monkeys, rats, and humans. Higher mutation rates occurred with human and rat, than with cynomolgus monkey microsomes. DNA adduct levels were highest in male rats, followed by female rats, and were much lower in cynomolgus monkeys after an oral MeIQx dose. Species differences in the bioactivation of PhIP were also observed among in human, rat, and mouse hepatic microsomes, with those of human origin having the highest capacity to catalyse the initial activation step to N-hydroxy-PhIP (Hengstler et al., 1999).

The total dose required to induce tumours formation varied for each HAA, was species-dependent, and could range from 0.1 to 64.6 mg/kg per day in rodents (Turesky & Le Marchand, 2011). Doses of HAAs used in the animal feeding studies exceeded by several orders of magnitude the levels of HAAs found in the human diet (Stavric, 1994). However, several HAA–DNA adducts have been detected in human tissue (Turesky & Le Marchand, 2011). The results reported by Garner et al. (1999) suggest that humans metabolize HAAs differently compared with rats. After low-dose oral administration of MeIQx and PhIP, humans developed higher DNA adduct formation in colonic tissue compared with rats. Similarly, Mauthe et al. (1999) showed that low-dose MeIQx formed DNA adducts in the human colon. This implied that the human colon may be more sensitive to this compound than the mouse or rat colon. Using accelerator mass spectrometry, a tool for measuring isotopes with attomolar sensitivity, Turteltaub et al. (1999) showed that protein and DNA adduct levels in rodents were dose-dependent. The adduct levels in human tissue and blood were generally greater than those in rodents administered equivalent doses. Furthermore, the metabolite profiles for both MeIQx and PhIP differed substantially between humans and rodents, with more N-hydroxylation (bioactivation) and less ring oxidation (detoxification) in humans. There are also important differences between humans and rats in CYP activity and regioselectivity of HAA oxidation, which can affect the toxicological properties of these compounds (Turesky, 2007; Turesky & Le Marchand, 2011).

(b) Epidemiological studies

Estimating HAA exposure in epidemiological studies has been difficult due to the variability of these compounds across the range of meat types, cooking methods and doneness levels. Moreover, there is a lack of gold-standard biomarkers to validate the questionnaires. Surrogate measures of HAA intake, such as cooking methods, meat doneness, and surface browning, have been used to investigate the etiological association between these mutagens and cancer risk. In addition, questionnaires with detailed cooking and doneness information have been linked to an HAA database to estimate individual HAA intake in the USA (Sinha, 2002; Sinha et al., 2005c), Sweden (Augustsson et al., 1997), and Germany (Rohrmann et al., 2009b). The HAA database was created by measuring levels of HAAs in a variety of meats cooked by different high-temperature methods to a range of doneness levels (rare, medium, well done, and very well done) (Sinha et al., 1995, 1998a, b). For example, while grilled, well-done chicken contains high levels of HAAs, roasted chicken contains very low levels of HAAs.

Urinary HAA biomarkers are good indicators of short-term intake, but such one-time measures cannot be used to estimate an individual’s usual exposure level (Cross & Sinha, 2004; Turesky & Le Marchand, 2011; Busquets et al., 2013). Adducts in DNA, haemoglobin, and serum albumin have also been evaluated, but their utility in epidemiological studies at the present time is unclear (Turesky & Le Marchand, 2011). There is enthusiasm for using HAA levels in hair as a long-term measure of exposure, but the use of this measure in epidemiological studies is still being evaluated (Kobayashi et al., 2007; Turesky & Le Marchand, 2011; Kataoka et al., 2013; Iwasaki et al., 2014) (see also Section 1.4.2).

Putative DNA adducts of several HAAs have been detected in human tissues by non-specific 32P-postlabelling (Totsuka et al., 1996) or immunohistochemistry methods (Zhu et al., 2003; Tang et al., 2007), and studies have reported on the analysis of presumed PhIP–DNA adducts after acid hydrolysis of DNA in human lymphocytes or colon DNA samples (Friesen et al., 1994; Magagnotti et al., 2003). However, few studies have unambiguously identified and quantified intact HAA–DNA adducts in human biospecimens by specific tandem mass spectrometry–based methods (Gu et al., 2012).

Using detailed meat cooking questions and linkage to the HAA database, case–control and prospective studies have evaluated the association between HAA intake and cancer risk (Alaejos et al., 2008; Zheng & Lee, 2009; Abid et al., 2014). The results have been mixed, depending on the cancer site and the study population. Results considered here are from large prospective cohort studies. Both MeIQx and DiMeIQx were positively associated with cancer of the colorectum in the NIH-AARP study (Cross et al., 2010), but not with colorectal adenoma incidence in the PLCO trial (Ferrucci et al., 2012). In contrast, MeIQx was associated with colon adenomas in a cohort of men from the USA (Wu et al., 2006). PhIP intake has been linked to colorectal adenomas in the PLCO trial (Ferrucci et al., 2012) and in the EPIC-Heidelberg cohort study (Rohrmann et al., 2009b), but not to cancer of the colorectum in the NIH-AARP study (Cross et al., 2010). PhIP, MeIQx, and DiMeIQx were not associated with cancer of the colorectum in the Multiethnic Cohort Study (Ollberding et al., 2012).

Cancer of the prostate was not associated with PhIP, MeIQx, or DiMeIQx in the EPIC-Heidelberg study, the NIH-AARP study, or the Agricultural Health Study (AHS) (Koutros et al., 2008; Sinha et al., 2009; Sander et al., 2011). In contrast, in the HPFS, intake of PhIP from red meat was associated with advanced cancer of the prostate (Rohrmann et al., 2015). In the PLCO trial, PhIP, but not MeIQx or DiMeIQx, was associated with risk of cancer of the prostate (Cross et al., 2005).

In various prospective studies, none of the HAAs considered were associated with cancer of the breast (Ferrucci et al., 2009; Kabat et al., 2009; Wu et al., 2010). Although DiMeIQx was linked to cancers of the gastric cardia (Cross et al., 2011) and pancreas (Stolzenberg-Solomon et al., 2007; Anderson et al., 2012), no association was found with cancers of the lung (Tasevska et al., 2009, 2011) or liver (Freedman et al., 2010). Similarly, no association between MeIQx intake and cancer of the liver was seen (Freedman et al., 2010). However, MeIQx intake was linked to cancers of the lung (Tasevska et al., 2009) and pancreas (Anderson et al., 2012) in the NIH-AARP study and PLCO trial, respectively. In the NIH-AARP study, both MeIQx and DiMeIQx were associated with a decreased risk of chronic lymphocytic leukaemia and small lymphocytic lymphoma (Daniel et al., 2012b). PhIP was linked to an increased risk of renal cell carcinoma (Daniel et al., 2012a), but not to cancers of the lung (Tasevska et al., 2009, 2011), bladder (Ferrucci et al., 2010b), pancreas (Stolzenberg-Solomon et al., 2007), or liver (Freedman et al., 2010).

(c) HAAs and inter-individual genetic susceptibility

As HAAs can be activated or detoxified by phase I and phase II metabolic reactions, various studies have evaluated single-nucleotide polymorphisms in the genes encoding XMEs. Results were mixed for interactions between XME polymorphisms and HAA consumption for colorectal adenomas or carcinomas (Ishibe et al., 2002; Le Marchand et al., 2002b; Chan et al., 2005b; Lilla et al., 2006; Girard et al., 2008; Shin et al., 2008; Yeh et al., 2009; Ferrucci et al., 2010a; Wang et al., 2011; Fu et al., 2012; Gilsing et al., 2012; Voutsinas et al., 2013). Some studies evaluated XMEs and HAAs for cancer of the breast (Lee et al., 2013), prostate (Nowell et al., 2004), and bladder (Lin et al., 2012). Many of these studies had a small number of cases with inadequate power or examined only a small set of single-nucleotide polymorphisms from a limited number of candidate genes. As the balance of activating and detoxifying enzymes is thought to influence carcinogen metabolism, comprehensive studies including numerous markers across multiple genes involved in xenobiotic metabolism are essential for studying this complex association. Furthermore, the inconsistencies in the data may have resulted partly from the inability of most studies to estimate specific HAAs, due to a lack of information regarding cooking technique and doneness level, or appropriate availability of biomarkers.

4.5.4. Polycyclic aromatic hydrocarbons

The following discussion is restricted to studies involving exposure to PAHs as a result of the consumption of red meat or processed meat, together with studies that reported findings directly relevant to the issue of whether such exposure may account for any risk of cancer. The weight accorded to such data depended, among many other considerations, on current knowledge of the carcinogenicity of individual PAHs and of PAHs as a class. See IARC Monographs Volume 92 (on PAHs) (IARC, 2010) and Volume 100F (on BaP) (IARC, 2012a).

The carcinogenicity of PAHs, specifically BaP (e.g. from active smoking, inhaling second-hand tobacco smoke, or working in coal- and tar-based industries) has prompted scrutiny of other circumstances of PAH exposure (e.g. from air pollution and dietary intake). In non-smoking, non–occupationally exposed populations, diet is frequently the major source of exposure to PAHs (IARC, 2010). Dietary intake of PAHs is often assessed by reference to levels of BaP, which is recognized as a good marker of PAH exposure. When fed to mice, BaP caused multiple tumour types, particularly in the upper gastrointestinal tract (IARC, 2010). PAHs can be formed during the curing and processing of meat, and can be generated during cooking through pyrolysis of fat, particularly if the meat is charred or burned (Phillips, 1999). See Section 1 for further discussion on PAH levels and PAH occurrence in different meat preparations.

(a) Mechanisms of carcinogenesis

The carcinogenic mechanisms of PAHs are extensively reviewed in IARC Monographs Volume 92 and Volume 100F, and include activation and detoxification by phase I and II XMEs.

In a study of 114 subjects (48 women, 66 men), Cocco et al. (2007) reported that frequent intake of grilled meat was a predictor of urinary 1-hydroxypyrene levels of 0.50 μg/g creatine or greater. In the study previously described in Section 4.2 by Chien & Yeh (2010), consumption of barbecued meat (with higher PAH content) resulted in a significant correlation between urinary 8-OHdG concentrations, and 1-hydroxypyrene and 3-hydroxy-BaP concentrations.

A case–control study reported higher PAH–DNA adduct levels in colorectal adenoma cases (median, 1.4 adducts per 108 nucleotides) than in polyp-free controls (median, 1.2 adducts per 108 nucleotides; P = 0.02) (Gunter et al., 2007). The DNA adduct levels were measured by chemiluminescence immunoassay (using an antiserum elicited against DNA modified with (±)-7β,8α-dihydroxy-9α,10α-epoxy-7,8,9,10-tetrahydro-benzo[a]pyrene, which recognizes several PAHs bound to human DNA). Rothman et al. (1990, 1993) found PAHs in urine and DNA adducts in white blood cells. These data support that PAHs are absorbed from the consumption of grilled meat and have a genotoxic effect.

(b) Epidemiological studies

Dietary intake of PAHs, irrespective of the dietary source, has been examined in relation to a range of tumour types, including colorectal adenoma (Sinha et al., 2005a), cancer of the breast (Rundle et al., 2000; Jeffy et al., 2002; Mordukhovich et al., 2010), cancer of the stomach (Liao et al., 2014), and renal cell carcinoma (Daniel et al., 2011). Some positive associations were reported, specifically in relation to colorectal adenoma.

Based on the BaP intake of participants, determined using the Computerized Heterocyclic Amines Resource for Research in Epidemiology of Disease (CHARRED) developed by the National Cancer Institute (NCI), various studies have evaluated PAHs. While studying the association between meat intake and cancer of the colon, a case–control study in North Carolina, USA (Butler et al., 2003), determined levels of BaP intake. Associations with BaP intake, stratified by race, were imprecise, but stronger effects were seen among African Americans than among white Americans. In a large prospective study of meat consumption and risk of cancer of the colorectum, BaP intake was not associated with cancer of the colorectum (Cross et al., 2010). In a study of screening-detected colorectal adenoma (Sinha et al., 2005b) evaluating dietary intake of PAHs, an increased risk of adenoma of the descending colon and sigmoid colon was observed with BaP intake. exposure to BaP from meat consumption was not associated with a risk of cancer of the colorectum in an investigation of the postulated association between high consumption of meat and colorectal carcinoma in a case–control study in Western Australia (Tabatabaei et al., 2010).

No association between PAHs and cancer of the breast was found in a large population-based case–control study that evaluated dietary intake of PAHs from cooked meat, determined by self-administered Block FFQs (Steck et al., 2007). However, this same study did find an association with intake of BaP from meat in postmenopausal women whose tumours were positive for both the estrogen receptor and progesterone receptor.

Daniel et al. (2011) undertook a case–control study that examined exposure to PAHs from meat intake in 1192 newly diagnosed renal cell carcinoma patients and 1175 controls. Risk of malignancy increased with intake of BaP. Risk of renal cell carcinoma was more than two-fold higher in African Americans and current smokers.

In a population-based case–control study, Girard et al. (2008) investigated whether cancer of the colon was associated with genetic variations in UGT1A1 and UGT1A9. The UGT1A1–53 and −3156 genotypes significantly modified the association between dietary BaP and cancer of the colon. The strongest association between dietary BaP exposure was observed in those with less than 7.7 ng/day of BaP exposure and low-activity genotypes. These data support the hypothesis that UDP-glucuronosyltransferases (UGTs) modify the association between meat-derived PAH exposure and cancer of the colon.

4.5.5. N-Nitroso compounds

The following discussion is restricted to studies involving exposure to NOCs as a result of the consumption of red meat or processed meat, together with studies that reported findings directly relevant to the issue of whether such exposure may contribute to any risk of cancer. The weight accorded to such data depended, among many other considerations, on current knowledge of the carcinogenicity of individual NOCs and of NOCs as a class. See IARC Monographs Volume 89 (IARC, 2007) and Volume 100E (IARC, 2012b) for NOCs, and Volume 94 (IARC, 2010) for ingested nitrate and nitrite.

NOCs can be produced during processing, storage, and preparation of foods. They are formed by the reaction of secondary amines (R1NHR2) and N-alkylamides (R1NH·CO·R2) with nitrite in food or in the acidic environment of the stomach (Honikel, 2008). Metabolism of nitrosamines or spontaneous breakdown of nitrosamides can give rise to reactive alkylating intermediates, which can be identified by their reaction with DNA and other macromolecules. Nitrosation of primary amino acids, including glycine and methionine, may also give rise to alkylating intermediates (Issenberg 1976; Mirvish, 1995). NOCs are genotoxic carcinogens associated with particular mutational signatures (Rao, 2013).

The general term NOC covers all substances with N-nitroso groups, including N-nitrosamines and N-nitrosamides. However, the analytical method generally used to analyse NOCs in digestion does not differentiate between N-nitrosamines and other compounds such as S-nitrosothiols, O-nitroso compounds, and iron nitrosyls (Kuhnle & Bingham, 2007). Given this lack of specificity, the term ATNC has been used to describe the substances measured by this technique. Nitrosyl haem and nitrosothiols have been identified as major constituents of both faecal and ileal ATNC, and the formation of these compounds increases significantly after consumption of a diet rich in red meat. Nitrosothiols are readily formed under the acidic conditions of the stomach, a process that is promoted by haem. Haem becomes easily nitrosylated under the anaerobic and reductive conditions of the lower gut to form nitrosyl haem, which is an NO donor and can act as a nitrosating agent. In turn, nitrosothiols can act as NO donors and nitrosating species. Thus, the combined actions of haem and free thiol groups can promote the endogenous formation of NOCs (Kuhnle et al., 2007).

Nitrite used in meat processing is involved in many reactions with myoglobin, proteins, and lipids (Honikel, 2008; Skibsted, 2011; Demeyer et al., 2015; Fig. 4.2). When combined with myoglobin, these reactions result in nitrosomyoglobin in cured meat and nitrosyl haemochromogen after cooking, which are responsible for the characteristic colour of cured meats. Residual nitrite in cured meat proteins may be important as a “hidden NO-generating pool”, a source of nitric oxide for numerous reactions during the storage and cooking of cured meats (Skibsted, 2011).

Fig. 4.2

Fig. 4.2

Nitric oxide formed from nitrite during meat curing can participate in numerous reactions modifying proteins and pigments

The occurrence of nitrate, nitrite, and NOCs in meat is discussed in Section 1. Mirvish et al. (2002) reported that NOC and NOC precursor levels in hot dogs were about 10 and 4 times higher, respectively, than those in fresh meat. The NOC precursors were considered of greater relevance to carcinogenicity, as they are more stable and approximately 1000 times more abundant than NOCs. The main NOC precursors identified were N-glycosyl amino acids and peptides. Dich et al. (1996) described dietary intake of nitrate, nitrite, and N-nitrosodimethylamine (NDMA) in 5304 men and 4750 women who participated in the Finnish Mobile Clinic Health Examination Survey in 1967–1972. Dietary nitrite was mainly provided by meat products (specified as cured meats, cooked sausage, and salami), contributing about 95% of the total intake. The mean daily intake of NDMA was calculated to be 0.052 µg, approximately half of which was derived from meat products.

(a) Absorption, distribution, metabolism, and excretion

Few studies have reported on the absorption, distribution, metabolism, and excretion of NOCs after meat consumption. Human saliva contains nitrate and nitrite due to enterosalivary circulation (Mirvish et al., 2000). Since rats convert a low amount of salivary nitrate into nitrite, it has been argued that the rat may not be a good model for humans (Cockburn et al., 2013). Therefore, Chenni et al. (2013) tested the relevance of this enterosalivary cycle by giving haem iron–fed rats drinking-water containing sodium nitrite, mimicking human salivary nitrite levels. They observed increased faecal ATNC. Phillips et al. (1975) showed that NDMA is absorbed in the rat stomach and the small intestine. Zhou et al. (2014) also reported increased urinary ATNC in rats fed sodium nitrite and/or hot dogs. In a rat model, Santarelli et al. (2010) showed that a combination of nitrite curing, cooking, and oxidation of red meat increased faecal ATNC. In addition, rats fed a diet containing commercially purchased hot dogs or fermented, raw, dry sausages had increased faecal ATNC compared with those fed a control diet without meat (Santarelli et al., 2013). Intake of sodium nitrite (0.17 g/L) and sodium nitrate (0.23 g/L) through drinking-water increased faecal ATNC in rats on a 1% haemoglobin diet (Chenni et al., 2013).

Several mechanisms have been proposed to explain the effect of red meat on the formation of NOCs. Lunn et al. (2007) observed no difference in ATNC levels in the ileal output of ileostomists and in the faecal output of healthy subjects consuming large amounts of red meat. In contrast to the stomach contents, which consisted only of nitrosothiols, nitrosyl iron was present in higher concentrations than nitrosothiols in ileal and faecal samples, with no difference in ATNC composition between both sample types (Kuhnle et al., 2007). Thus nitrosothiols formed in the acidic stomach may release NO once they are exposed to the alkaline and reductive conditions of the small and large intestine, thereby stimulating the nitrosylation of haem iron. However, the consequences of the formation of these products are unclear (Hogg, 2007). On the one hand, nitrosyl haem and nitrosothiols could act as nitrosating agents and promote the formation of NOCs in the intestinal epithelium (Kuhnle et al., 2007). On the other hand, nitrosothiols and nitrosyl iron may act as a protective mechanism by capturing NO and facilitating its excretion, thereby limiting the formation of DNA alkylating agents.

In a series of human intervention studies, Bingham and colleagues demonstrated a dose–response increase in faecal excretion of ATNC with red meat intake. This was not observed with vegetable proteins, white meat, or an Fe2+ supplement, but mimicked by a haem iron supplement (provided by blood sausage) (Bingham et al., 1996; Hughes et al., 2001; Bingham et al., 2002; Cross et al., 2003). Holtrop et al. (2012) conducted three dietary trials in obese men consuming body weight maintenance or weight loss diets, and measured NOCs in faecal samples. The meat-based weight loss diets increased levels of faecal NOCs (P < 0.001). Red meat intake was positively correlated with the faecal log NOC concentrations (r = 0.60; P < 0.001).

The genotoxic effects of faecal ATNC were investigated using different comet assay protocols in individuals consuming high levels of red meat (Cross et al., 2006a). The inter-individual effects were variable, and diet, mean transit time, and weight had no effect on faecal water genotoxicity; see also Lewin et al. (2006), as discussed in Section 4.2. Rats treated with the N-nitrosopeptide N-acetyl--prolyl--nitrosoglycine showed the presence of O6-CMG in the intact small intestine. This was also observed in HT-29 cells treated with diazoacetate (Lewin et al., 2006). Since the analysis of ATNC includes both toxic and non-toxic compounds, the quantification of O6-CMG might offer a more specific insight into the formation of genotoxic NOCs.

Although Lunn et al. (2007) and Kuhnle et al. (2007) observed no difference between ileal and faecal ATNC and its individual components, suggesting no influence of the colonic microbiota on NOC formation, other research has suggested a major facilitating role of the gut microbiota. Using a pig caecum model containing nitrate and amines/amides, Engemann et al. (2013) showed large inter-individual variation in porcine microbiota to form N-nitrosamines N-nitrosomorpholine (NMOR) and N-nitrosopyrrolidine (NPYR), and N-nitrosamides N-methyl-N-nitrosourea and N-ethyl-N-nitrosourea. Moreover, a clear increase in NOCs was observed in time, with the microbiota responsible for the reduction of nitrate to nitrite. In accordance, Vanden Bussche et al. (2014) found considerable inter-individual variation in human microbiota for the in vitro formation of the NOC-specific DNA adduct O6-CMG during fermentation of white and red meat. This large inter-individual variation was also acknowledged by Van Hecke et al. (2014a, b; 2015). However, in vitro formation of O6-CMG was stimulated by a higher haem iron content, a higher fat content, and more intense heating conditions, while nitrite curing was not perceived to be of influence, despite the large impact of the applied microbiota (Van Hecke et al., 2014a, b, 2015).

A broad body of evidence indicates that the formation of NOCs may be inhibited by agents including ascorbic acid and α-tocopherol (Mirvish, 1986). The inhibitory effects of nitrite, l-ascorbic acid, and α-tocopherol on the formation of NOCs in processed meat products were evaluated by comparing samples of sausage with different concentrations of these reductants (Pourazrang et al., 2002). Revertants in the S. typhimurium (TA100) microsome assay were significantly reduced (P < 0.05) by 60% when the reductants were added to the samples. In the study of Hughes et al. (2002) described in Section 4.2 evaluated the effect of soy and other dietary components on faecal NOC excretion with consumption of a high–red meat (420 g/day) diet in 11 male volunteers randomized to 15-day dietary periods. Soy significantly suppressed faecal ATNC concentrations (P = 0.02), but vegetables and tea extract did not affect mean faecal ATNC concentrations or faecal water genotoxicity. However, faecal weight increased and was associated with reduced transit time, decreasing contact between ATNC concentrations, nitrite, and ammonia and the large bowel mucosa.

(b) Mechanisms of carcinogenesis

For decades, experimental animal data have afforded insight into the increased risk of cancer in humans that is attributable to the consumption of different categories of meat, and specifically the possible role of NOCs (Olsen et al., 1984); see also Santarelli et al. (2010), as discussed in Section 4.3.

G→A transitions in K-RAS occur in cancer of the colorectum and are characteristic of the effects of alkylating agents such as NOCs (Bingham et al., 1996). The methylating agent N-methyl-N-nitrosourea produced predominantly (> 80%) transitions (GC→AT), whereas potassium diazoacetate, a stable form of nitrosated glycine, produced transitions (GC→AT) and transversions (GC→TA and AT→TA) in equivalent amounts (Gottschalg et al., 2007). The similarity in the patterns of mutations induced by potassium diazoacetate with those observed in mutated P53 in human gastrointestinal tract tumours suggests that nitrosation of glycine (or glycine derivatives) may contribute to characteristic human P53 mutation profiles.

(c) Epidemiological studies

Studies addressing the association between risk of cancer and dietary intake of nitrate, nitrite, or nitrosamines refer to meat as well as other relevant foods (Loh et al., 2011). [The Working Group noted that dietary intake of nitrate and nitrite does not necessarily reflect NOC intake.]

In the EPIC-Norfolk study (Loh et al., 2011), dietary NDMA intake was significantly associated with an increased risk of cancer of the rectum in women. There was no significant association between cancer risk across quartiles and dietary nitrite and endogenous NOCs. In a case–control study in Canada, NDMA intake was associated with a higher risk of cancer of the colorectum, specifically rectal carcinoma. Risk of cancer of the colorectum also increased with the consumption of NDMA-containing meats (Zhu et al., 2014). Individuals with high-NDMA and low–vitamin E intake had a significantly higher risk than those with both low-NDMA and low–vitamin E intake.

Intake of dietary nitrites and nitrosamines was positively associated with risk of cancer of the lower urinary tract in American men of Japanese ancestry (Wilkens et al., 1996). Consumption of processed meats, in particular bacon, sausage, and ham, was also significantly associated with an increased risk in American men of Japanese ancestry. Three food items accounted for all of the NOC intake: sausage (46%), bacon (33%), and luncheon meats (21%).

The association between nitrate, nitrite, and nitrosamine intake and glioma was examined by Michaud et al. (2009). Risk of glioma was not elevated among individuals in the highest intake category of nitrate, nitrite, or NDMA compared with those in the lowest intake category. In a population-based case–control study of glioma in adults, increased odds ratios were observed in males who consumed high levels of bacon, corned meats, apples, melons, and oil (Giles et al., 1994). Elevated odds ratio in men, but not women, were associated with the intake of NDMA.

4.5.6. Interactions between NOCs, haem iron, and HAAs

Haem in red meat stimulates the endogenous production of NOCs. The effect of red meat and processed meat on endogenous nitrosation, as well as DNA damage (see Section 4.2), was investigated by Joosen et al. (2009). Faecal NOC concentrations in 5 males and 11 females on vegetarian diets were low (2.6 and 3.5 mmol/g, respectively), but significantly increased in those fed meat diets (preserved red meat, 175 ± 19 nmol/g; red meat, 185 ± 22 nmol/g; P = 0.75). The meat diets contained 420 g/day (males) or 366 g/day (females) of meat. The nitrite-cured meat diet had the same effect as the fresh red meat diet on endogenous nitrosation, but increased faecal water–induced oxidative DNA damage.

A high–red meat diet (420 g/day) significantly increased nitrosyl iron and nitrosothiols in ileal and faecal samples compared with a vegetarian diet (Kuhnle et al., 2007). Faecal nitrosyl iron and haem were strongly correlated (r = 0.776; P < 0.0001), suggesting that nitrosyl haem is the main source of nitrosyl iron. Nitrosation of HAAs is depicted in Fig. 4.3 (Lakshmi et al., 2005b). Lakshmi et al. (2005a) demonstrated hemin potentiation of NO-mediated nitrosation using the HAA IQ as a target and by monitoring the formation of 14C-2-nitrosoamino-3-methylimidazo[4,5-f]quinoline (N-NO-IQ) by high-performance liquid chromatography. Faecal NOCs (Mirvish et al., 2003) and urinary nitrite and nitrate were increased in mice with dextran sulfate sodium–induced colitis, which was consistent with increased expression of inducible NO synthase and NO synthesis.

Fig. 4.3

Fig. 4.3

Nitrosation of heterocyclic aromatic amines

IQ and MeIQx can be converted to their corresponding N-nitrosamines, N-NO-IQ and 2-nitrosoamino-3,8-dimethylimidazo[4,5-f]quinoxaline (N-NO-MeIQx) (Zenser et al., 2009). N-NO-IQ and N-NO-MeIQx have been shown to form several putative adducts in common with those formed by 2-hydroxyamino-3-methylimidazo[4,5‑f]quinoline (N-OH-IQ) and 2-hydroxyamino-3,8-dimethylimidazo[4,5-f]quinoxaline (N-OH-MeIQx). These N-nitrosamines might be alternatives to their hydroxylamine analogues, as activated intermediates leading to the initiation of cancer of the colon in individuals with colitis.

4.5.7. Other components

The following subsections address components or contaminants of red meat or processed meat not considered elsewhere in Section 4.5.

(a) Advanced glycation end products

AGEPs form by Maillard reaction after the initial binding of aldehydes with amines or amides in, among other places, heated foods. Within proteins, high molecular-mass AGEPs are formed whereas reactions among small molecules yield low-molecular-mass AGEPs. Some of these compounds interact with specific pro- or anti-inflammatory receptors. In observational studies, dietary AGEPs were strongly associated with late complications in diabetes (e.g. Poulsen et al., 2013).

Levels of representative AGEPs are similar in certain cheeses, fried eggs, cereal products, and broiled steak (Uribarri et al., 2010). Monitoring of representative AGEPs in 19 healthy, overweight individuals who were fed meals of identical ingredients, prepared by either roasting or steaming, indicated that AGEPs may affect postprandial ghrelin, oxidative stress, and glucose responses (Poulsen et al., 2014). In a prospective case–control study of cancer of the colorectum, higher prediagnostic levels of the serum-soluble receptor for AGEPs were associated with a lower risk of cancer of the colorectum in male smokers; no specific relationship with any dietary constituent was reported (Jiao et al., 2011).

(b) N-Glycolylneuraminic acid

Neu5Gc is a predominant sialic acid on most mammalian cells. Humans are genetically deficient in Neu5Gc production, and the compound is metabolically incorporated into human tissue from dietary sources, particularly red meat. Neu5Gc is thus detectable on the surface of human epithelia and endothelia, and in higher amounts in malignant tissues. This xeno-autoantigen can react with circulating anti-Neu5Gc antibodies in humans. The compound has been proposed as a cancer biomarker (Samraj et al., 2014). Among the evidence for its role in tumour progression, Hedlund et al. (2008) reported that murine tumours expressing human-like levels of Neu5Gc showed accelerated growth in syngeneic mice with a human-like Neu5Gc deficiency, which coincided with the induction of anti-Neu5Gc antibodies and increased infiltration of inflammatory cells.

Samraj et al. (2015) employed what was described as an improved method to survey foods for Neu5Gc. They showed that Neu5Gc was highly and selectively enriched in red meat. In the study, Neu5Gc-deficient mice, immunized against Neu5Gc and fed bioavailable Neu5Gc from porcine saliva, developed a much higher incidence of hepatocellular carcinoma than three groups of variously identified control mice.

(c) Proposed oncogenic bovine virus

Noting the cancer incidence in Asian communities known to consume undercooked beef, zur Hausen (2012) hypothesized that the presence of one or more thermoresistant, potentially oncogenic bovine viruses contaminates beef preparations and contributes to development of cancer of the colorectum. The same, or comparable, factors were proposed to be transmitted by the consumption of milk products (zur Hausen & de Villiers, 2015). [The Working Group took note of the lack of supporting evidence for this hypothesis.]

References

  • Abdulamir AS, Hafidh RR, Abu Bakar F. The association of Streptococcus bovis/gallolyticus with colorectal tumors: the nature and the underlying mechanisms of its etiological role. J Exp Clin Cancer Res. 2011;30(1):11–11. [PMC free article: PMC3032743] [PubMed: 21247505] [CrossRef]
  • Abid Z, Cross AJ, Sinha R. Meat, dairy, and cancer. Am J Clin Nutr. 2014;100 Suppl 1:386S–93S. [PMC free article: PMC4144110] [PubMed: 24847855] [CrossRef]
  • Adamson RH, Snyderwine EG, Thorgeirsson UP, Schut HA, Turesky RJ, Thorgeirsson SS, et al. Metabolic processing and carcinogenicity of heterocyclic amines in nonhuman primates. Princess Takamatsu Symp. 1990;21:289–301. [PubMed: 2134682]
  • Aeschbacher HU, Ruch E. Effect of heterocyclic amines and beef extract on chromosome aberrations and sister chromatid exchanges in cultured human lymphocytes. Carcinogenesis. 1989;10(3):429–33. [PubMed: 2924390] [CrossRef]
  • Alaejos MS, González V, Afonso AM. Exposure to heterocyclic aromatic amines from the consumption of cooked red meat and its effect on human cancer risk: a review. Food Addit Contam Part A Chem Anal Control Expo Risk Assess. 2008;25(1):2–24. [PubMed: 17952757] [CrossRef]
  • Alexander J, Fossum BH, Reistad R, Holme JA. Metabolism of the food carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) in the rat and other rodents. Princess Takamatsu Symp. 1995;23:113–22. [PubMed: 8844802]
  • Allen NE, Appleby PN, Davey GK, Key TJ. Hormones and diet: low insulin-like growth factor-I but normal bioavailable androgens in vegan men. Br J Cancer. 2000;83(1):95–7. [PMC free article: PMC2374537] [PubMed: 10883675] [CrossRef]
  • Ambrosone CB, Freudenheim JL, Sinha R, Graham S, Marshall JR, Vena JE, et al. Breast cancer risk, meat consumption and N-acetyltransferase (NAT2) genetic polymorphisms. Int J Cancer. 1998;75(6):825–30. [PubMed: 9506525] [CrossRef]
  • Ambrosone CB, Kropp S, Yang J, Yao S, Shields PG, Chang-Claude J. Cigarette smoking, N-acetyltransferase 2 genotypes, and breast cancer risk: pooled analysis and meta-analysis. Cancer Epidemiol Biomarkers Prev. 2008;17(1):15–26. [PubMed: 18187392] [CrossRef]
  • Ames BN, Durston WE, Yamasaki E, Lee FD. Carcinogens are mutagens: a simple test system combining liver homogenates for activation and bacteria for detection. Proc Natl Acad Sci USA. 1973;70(8):2281–5. [PMC free article: PMC433718] [PubMed: 4151811] [CrossRef]
  • Ananthakrishnan AN, Du M, Berndt SI, Brenner H, Caan BJ, Casey G, et al. Red meat intake, NAT2, and risk of colorectal cancer: a pooled analysis of 11 studies. Cancer Epidemiol Biomarkers Prev. 2015;24(1):198–205. [PMC free article: PMC4294960] [PubMed: 25342387] [CrossRef]
  • Andersen V, Holst R, Vogel U. Systematic review: diet-gene interactions and the risk of colorectal cancer. Aliment Pharmacol Ther. 2013;37(4):383–91. [PMC free article: PMC3565452] [PubMed: 23216531] [CrossRef]
  • Andersen V, Vogel U. Interactions between meat intake and genetic variation in relation to colorectal cancer. Genes Nutr. 2015;10(1):448. [PMC free article: PMC4261072] [PubMed: 25491747] [CrossRef]
  • Anderson KE, Mongin SJ, Sinha R, Stolzenberg-Solomon R, Gross MD, Ziegler RG, et al. Pancreatic cancer risk: associations with meat-derived carcinogen intake in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (PLCO) cohort. Mol Carcinog. 2012;51(1):128–37. [PMC free article: PMC3516181] [PubMed: 22162237] [CrossRef]
  • Attene-Ramos MS, Wagner ED, Gaskins HR, Plewa MJ. Hydrogen sulfide induces direct radical-associated DNA damage. Mol Cancer Res. 2007;5(5):455–9. [PubMed: 17475672] [CrossRef]
  • Augustsson K, Skog K, Jägerstad M, Steineck G. Assessment of the human exposure to heterocyclic amines. Carcinogenesis. 1997;18(10):1931–5. [PubMed: 9364002] [CrossRef]
  • Aune D, Chan DS, Vieira AR, Navarro Rosenblatt DA, Vieira R, Greenwood DC, et al. Red and processed meat intake and risk of colorectal adenomas: a systematic review and meta-analysis of epidemiological studies. Cancer Causes Control. 2013;24(4):611–27. [PubMed: 23380943] [CrossRef]
  • Azadbakht L, Esmaillzadeh A. Red meat intake is associated with metabolic syndrome and the plasma C-reactive protein concentration in women. J Nutr. 2009;139(2):335–9. [PubMed: 19074209] [CrossRef]
  • Baker R, Arlauskas A, Bonin A, Angus D. Detection of mutagenic activity in human urine following fried pork or bacon meals. Cancer Lett. 1982;16(1):81–9. [PubMed: 6811131] [CrossRef]
  • Balder HF, Vogel J, Jansen MC, Weijenberg MP, van den Brandt PA, Westenbrink S, et al. Heme and chlorophyll intake and risk of colorectal cancer in the Netherlands cohort study. Cancer Epidemiol Biomarkers Prev. 2006;15(4):717–25. [PubMed: 16614114] [CrossRef]
  • Barbir A, Linseisen J, Hermann S, Kaaks R, Teucher B, Eichholzer M, et al. Effects of phenotypes in heterocyclic aromatic amine (HCA) metabolism-related genes on the association of HCA intake with the risk of colorectal adenomas. Cancer Causes Control. 2012;23(9):1429–42. [PubMed: 22740027] [CrossRef]
  • Barrett JH, Smith G, Waxman R, Gooderham N, Lightfoot T, Garner RC, et al. Colorectal Cancer Study Group. Investigation of interaction between N-acetyltransferase 2 and heterocyclic amines as potential risk factors for colorectal cancer. Carcinogenesis. 2003;24(2):275–82. [PubMed: 12584178] [CrossRef]
  • Bastide NM, Chenni F, Audebert M, Santarelli RL, Taché S, Naud N, et al. A central role for heme iron in colon carcinogenesis associated with red meat intake. Cancer Res. 2015;75(5):870–9. [PubMed: 25592152] [CrossRef]
  • Bastide NM, Pierre FH, Corpet DE. Heme iron from meat and risk of colorectal cancer: a meta-analysis and a review of the mechanisms involved. Cancer Prev Res (Phila). 2011;4(2):177–84. [PubMed: 21209396] [CrossRef]
  • Basu AK, Marnett LJ. Unequivocal demonstration that malondialdehyde is a mutagen. Carcinogenesis. 1983;4(3):331–3. [PubMed: 6339098] [CrossRef]
  • Bax M-L, Aubry L, Ferreira C, Daudin JD, Gatellier P, Rémond D, et al. Cooking temperature is a key determinant of in vitro meat protein digestion rate: investigation of underlying mechanisms. J Agric Food Chem. 2012;60(10):2569–76. [PubMed: 22335241] [CrossRef]
  • Bax M-L, Buffière C, Hafnaoui N, Gaudichon C, Savary-Auzeloux I, Dardevet D, et al. Effects of meat cooking, and of ingested amount, on protein digestion speed and entry of residual proteins into the colon: a study in minipigs. PLoS One. 2013;8(4):e61252. [PMC free article: PMC3625175] [PubMed: 23593443] [CrossRef]
  • Belinova L, Kahleova H, Malinska H, Topolcan O, Vrzalova J, Oliyarnyk O, et al. Differential acute postprandial effects of processed meat and isocaloric vegan meals on the gastrointestinal hormone response in subjects suffering from type 2 diabetes and healthy controls: a randomized crossover study. PLoS One. 2014;9(9):e107561. [PMC free article: PMC4164634] [PubMed: 25222490] [CrossRef]
  • Belobrajdic DP, Bird AR, Conlon MA, Williams BA, Kang S, McSweeney CS, et al. An arabinoxylan-rich fraction from wheat enhances caecal fermentation and protects colonocyte DNA against diet-induced damage in pigs. Br J Nutr. 2012;107(9):1274–82. [PubMed: 22115395] [CrossRef]
  • Belobrajdic DP, McIntosh GH, Owens JA. Whey proteins protect more than red meat against azoxymethane induced ACF in Wistar rats. Cancer Lett. 2003;198(1):43–51. [PubMed: 12893429] [CrossRef]
  • Ben Q, Xu M, Ning X, Liu J, Hong S, Huang W, et al. Diabetes mellitus and risk of pancreatic cancer: A meta-analysis of cohort studies. Eur J Cancer. 2011;47(13):1928–37. [PubMed: 21458985] [CrossRef]
  • Benassi B, Leleu R, Bird T, Clifton P, Fenech M. Cytokinesis-block micronucleus cytome assays for the determination of genotoxicity and cytotoxicity of cecal water in rats and fecal water in humans. Cancer Epidemiol Biomarkers Prev. 2007;16(12):2676–80. [PubMed: 18086773] [CrossRef]
  • Bernstein H, Bernstein C, Payne CM, Dvorakova K, Garewal H. Bile acids as carcinogens in human gastrointestinal cancers. Mutat Res. 2005;589(1):47–65. [PubMed: 15652226] [CrossRef]
  • Bingham SA, Hughes R, Cross AJ. Effect of white versus red meat on endogenous N-nitrosation in the human colon and further evidence of a dose response. J Nutr. 2002;132(11) Suppl:3522S–5S. [PubMed: 12421881] [CrossRef]
  • Bingham SA, Pignatelli B, Pollock JR, Ellul A, Malaveille C, Gross G, et al. Does increased endogenous formation of N-nitroso compounds in the human colon explain the association between red meat and colon cancer? Carcinogenesis. 1996;17(3):515–23. [PubMed: 8631138] [CrossRef]
  • Bocci VA, Zanardi I, Travagli V. Ozone acting on human blood yields a hormetic dose-response relationship. J Transl Med. 2011;9(1):66. [PMC free article: PMC3125221] [PubMed: 21575276] [CrossRef]
  • Boyle P, Zaridze DG, Smans M. Descriptive epidemiology of colorectal cancer. Int J Cancer. 1985;36(1):9–18. [PubMed: 2991145] [CrossRef]
  • Brink M, Weijenberg MP, de Goeij AF, Roemen GM, Lentjes MH, de Bruïne AP, et al. Meat consumption and K-ras mutations in sporadic colon and rectal cancer in The Netherlands Cohort Study. Br J Cancer. 2005;92(7):1310–20. [PMC free article: PMC2361976] [PubMed: 15812479] [CrossRef]
  • Brinkman MT, Baglietto L, Krishnan K, English DR, Severi G, Morris HA, et al. Consumption of animal products, their nutrient components and postmenopausal circulating steroid hormone concentrations. Eur J Clin Nutr. 2010;64(2):176–83. [PubMed: 19904296] [CrossRef]
  • Brockton N, Little J, Sharp L, Cotton SC. N-acetyltransferase polymorphisms and colorectal cancer: a HuGE review. Am J Epidemiol. 2000;151(9):846–61. [PubMed: 10791558] [CrossRef]
  • Broughton KS, Rule DC, Handrich E. Prostaglandin E₂ production in mice is reduced by consumption of range-fed sources of red meat. Nutr Res. 2011;31(12):907–14. [PubMed: 22153516] [CrossRef]
  • Brown ED, Morris VC, Rhodes DG, Sinha R, Levander OA. Urinary malondialdehyde-equivalents during ingestion of meat cooked at high or low temperatures. Lipids. 1995;30(11):1053–6. [PubMed: 8569434] [CrossRef]
  • Bruce WR. Recent hypotheses for the origin of colon cancer. Cancer Res. 1987;47(16):4237–42. [PubMed: 3300962]
  • Bruce WR, Giacca A, Medline A. Possible mechanisms relating diet and risk of colon cancer. Cancer Epidemiol Biomarkers Prev. 2000;9(12):1271–9. [PubMed: 11142411]
  • Budhathoki S, Iwasaki M, Yamaji T, Sasazuki S, Takachi R, Sakamoto H, et al. Dietary heterocyclic amine intake, NAT2 genetic polymorphism, and colorectal adenoma risk: the colorectal adenoma study in Tokyo. Cancer Epidemiol Biomarkers Prev. 2015;24(3):613–20. [PubMed: 25604583] [CrossRef]
  • Busquets R, Frandsen H, Jönsson JA, Puignou L, Galceran MT, Skog K. Biomonitoring of dietary heterocyclic amines and metabolites in urine by liquid phase microextraction: 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), a possible biomarker of exposure to dietary PhIP. Chem Res Toxicol. 2013;26(2):233–40. [PubMed: 23276304] [CrossRef]
  • Butler LM, Sinha R, Millikan RC, Martin CF, Newman B, Gammon MD, et al. Heterocyclic amines, meat intake, and association with colon cancer in a population-based study. Am J Epidemiol. 2003;157(5):434–45. [PubMed: 12615608] [CrossRef]
  • Butler LM, Duguay Y, Millikan RC, Sinha R, Gagné JF, Sandler RS, et al. Joint effects between UDP-glucuronosyltransferase 1A7 genotype and dietary carcinogen exposure on risk of colon cancer. Cancer Epidemiol Biomarkers Prev. 2005;14(7):1626–32. [PubMed: 16030093] [CrossRef]
  • Caderni G, Palli D, Lancioni L, Russo A, Luceri C, Saieva C, et al. Dietary determinants of colorectal proliferation in the normal mucosa of subjects with previous colon adenomas. Cancer Epidemiol Biomarkers Prev. 1999;8(3):219–25. [PubMed: 10090299]
  • Calle EE, Kaaks R. Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat Rev Cancer. 2004;4(8):579–91. [PubMed: 15286738] [CrossRef]
  • Carlsen CU, Moller JK, Skibsted LH. Heme-iron in lipid oxidation. Coord Chem Rev. 2005;249(3-4):485–98. [CrossRef]
  • Carpenter CE, Mahoney AW. Contributions of heme and nonheme iron to human nutrition. Crit Rev Food Sci Nutr. 1992;31(4):333–67. [PubMed: 1581009] [CrossRef]
  • Carr PR, Walter V, Brenner H, Hoffmeister M. Meat subtypes and their association with colorectal cancer: Systematic review and meta-analysis. Int J Cancer. 2016;138(2):293–302. [PubMed: 25583132] [CrossRef]
  • Ceriello A, Motz E. Is oxidative stress the pathogenic mechanism underlying insulin resistance, diabetes, and cardiovascular disease? The common soil hypothesis revisited. Arterioscler Thromb Vasc Biol. 2004;24(5):816–23. [PubMed: 14976002] [CrossRef]
  • Chan AT, Ma J, Tranah GJ, Giovannucci EL, Rifai N, Hunter DJ, et al. Hemochromatosis gene mutations, body iron stores, dietary iron, and risk of colorectal adenoma in women. J Natl Cancer Inst. 2005;97(12):917–26. a. [PubMed: 15956653] [CrossRef]
  • Chan AT, Tranah GJ, Giovannucci EL, Willett WC, Hunter DJ, Fuchs CS. Prospective study of N-acetyltransferase-2 genotypes, meat intake, smoking and risk of colorectal cancer. Int J Cancer. 2005;115(4):648–52. b. [PubMed: 15700302] [CrossRef]
  • Chen J, Stampfer MJ, Hough HL, Garcia-Closas M, Willett WC, Hennekens CH, et al. A prospective study of N-acetyltransferase genotype, red meat intake, and risk of colorectal cancer. Cancer Res. 1998;58(15):3307–11. [PubMed: 9699660]
  • Chen SY, Liu TY, Shun CT, Wu MS, Lu TH, Lin JT, et al. Modification effects of GSTM1, GSTT1 and CYP2E1 polymorphisms on associations between raw salted food and incomplete intestinal metaplasia in a high-risk area of stomach cancer. Int J Cancer. 2004;108(4):606–12. [PubMed: 14696128] [CrossRef]
  • Cheng KW, Chen F, Wang M. Heterocyclic amines: chemistry and health. Mol Nutr Food Res. 2006;50(12):1150–70. [PubMed: 17131456] [CrossRef]
  • Chenni FZ, Taché S, Naud N, Guéraud F, Hobbs DA, Kunhle GG, et al. Heme-induced biomarkers associated with red meat promotion of colon cancer are not modulated by the intake of nitrite. Nutr Cancer. 2013;65(2):227–33. [PMC free article: PMC3733138] [PubMed: 23441609] [CrossRef]
  • Cheung C, Loy S, Li GX, Liu AB, Yang CS. Rapid induction of colon carcinogenesis in CYP1A-humanized mice by 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine and dextran sodium sulfate. Carcinogenesis. 2011;32(2):233–9. [PMC free article: PMC3026842] [PubMed: 21081470] [CrossRef]
  • Chien YC, Yeh CT. Excretion characteristics of urinary 8-hydroxydeoxyguanosine after dietary exposure to polycyclic aromatic hydrocarbons. Environ Mol Mutagen. 2010;51(3):243–50. [PubMed: 19844955]
  • Cocco P, Moore PS, Ennas MG, Tocco MG, Ibba A, Mattuzzi S, et al. Effect of urban traffic, individual habits, and genetic polymorphisms on background urinary 1-hydroxypyrene excretion. Ann Epidemiol. 2007;17(1):1–8. [PubMed: 16406813] [CrossRef]
  • Cockburn A, Brambilla G, Fernández ML, Arcella D, Bordajandi LR, Cottrill B, et al. Nitrite in feed: from animal health to human health. Toxicol Appl Pharmacol. 2013;270(3):209–17. [PubMed: 21095201] [CrossRef]
  • Commoner B, Vithayathil AJ, Dolara P, Nair S, Madyastha P, Cuca GC. Formation of mutagens in beef and beef extract during cooking. Science. 1978;201(4359):913–6. [PubMed: 567374] [CrossRef]
  • Corpet DE. Red meat and colon cancer: should we become vegetarians, or can we make meat safer? Meat Sci. 2011;89(3):310–6. [PubMed: 21558046] [CrossRef]
  • Correa P, Haenszel W, Cuello C, Tannenbaum S, Archer M. A model for gastric cancer epidemiology. Lancet. 1975;2(7924):58–60. [PubMed: 49653] [CrossRef]
  • Cortessis V, Siegmund K, Chen Q, Zhou N, Diep A, Frankl H, et al. A case-control study of microsomal epoxide hydrolase, smoking, meat consumption, glutathione S-transferase M3, and risk of colorectal adenomas. Cancer Res. 2001;61(6):2381–5. [PubMed: 11289100]
  • Cotterchio M, Boucher BA, Manno M, Gallinger S, Okey AB, Harper PA. Red meat intake, doneness, polymorphisms in genes that encode carcinogen-metabolizing enzymes, and colorectal cancer risk. Cancer Epidemiol Biomarkers Prev. 2008;17(11):3098–107. [PMC free article: PMC2751598] [PubMed: 18990750] [CrossRef]
  • Cross AJ, Greetham HL, Pollock JR, Rowland IR, Bingham SA. Variability in fecal water genotoxicity, determined using the Comet assay, is independent of endogenous N-nitroso compound formation attributed to red meat consumption. Environ Mol Mutagen. 2006;47(3):179–84. [PubMed: 16304669] [CrossRef]
  • Cross AJ, Ferrucci LM, Risch A, Graubard BI, Ward MH, Park Y, et al. A large prospective study of meat consumption and colorectal cancer risk: an investigation of potential mechanisms underlying this association. Cancer Res. 2010;70(6):2406–14. [PMC free article: PMC2840051] [PubMed: 20215514] [CrossRef]
  • Cross AJ, Freedman ND, Ren J, Ward MH, Hollenbeck AR, Schatzkin A, et al. Meat consumption and risk of esophageal and gastric cancer in a large prospective study. Am J Gastroenterol. 2011;106(3):432–42. [PMC free article: PMC3039705] [PubMed: 20978481] [CrossRef]
  • Cross AJ,, Ferrucci LM, Risch A, Sinha R. Food Nutr Sci; Developing a heme iron database for meats according to meat type, cooking method and doneness level. 2012; 3(7):905–13. [PMC free article: PMC3583546] [PubMed: 23459329]
  • Cross AJ, Peters U, Kirsh VA, Andriole GL, Reding D, Hayes RB, et al. A prospective study of meat and meat mutagens and prostate cancer risk. Cancer Res. 2005;65(24):11779–84. [PubMed: 16357191] [CrossRef]
  • Cross AJ, Pollock JR, Bingham SA. Haem, not protein or inorganic iron, is responsible for endogenous intestinal N-nitrosation arising from red meat. Cancer Res. 2003;63(10):2358–60. [PubMed: 12750250]
  • Cross AJ, Sinha R. Meat-related mutagens/carcinogens in the etiology of colorectal cancer. Environ Mol Mutagen. 2004;44(1):44–55. [PubMed: 15199546] [CrossRef]
  • Cui D, Wang Z, Zhao E, Ma J, Lu W. NAT2 polymorphism and lung cancer risk: a meta-analysis. Lung Cancer. 2011;73(2):153–7. [PubMed: 21292342] [CrossRef]
  • Daniel CR, Schwartz KL, Colt JS, Dong LM, Ruterbusch JJ, Purdue MP, et al. Meat-cooking mutagens and risk of renal cell carcinoma. Br J Cancer. 2011;105(7):1096–104. [PMC free article: PMC3185955] [PubMed: 21897389] [CrossRef]
  • Daniel CR, Cross AJ, Graubard BI, Park Y, Ward MH, Rothman N, et al. Large prospective investigation of meat intake, related mutagens, and risk of renal cell carcinoma. Am J Clin Nutr. 2012;95(1):155–62. a. [PMC free article: PMC3238458] [PubMed: 22170360] [CrossRef]
  • Daniel CR, Sinha R, Park Y, Graubard BI, Hollenbeck AR, Morton LM, et al. Meat intake is not associated with risk of non-Hodgkin lymphoma in a large prospective cohort of U.S. men and women. J Nutr. 2012;142(6):1074–80. b. [PMC free article: PMC3349978] [PubMed: 22535761] [CrossRef]
  • Darley-Usmar V, Wiseman H, Halliwell B. Nitric oxide and oxygen radicals: a question of balance. FEBS Lett. 1995;369(2-3):131–5. [PubMed: 7649244] [CrossRef]
  • de Vogel J, Jonker-Termont DS, van Lieshout EM, Katan MB, van der Meer R. Green vegetables, red meat and colon cancer: chlorophyll prevents the cytotoxic and hyperproliferative effects of haem in rat colon. Carcinogenesis. 2005;26(2):387–93. [PubMed: 15550456] [CrossRef]
  • Deitz AC, Zheng W, Leff MA, Gross M, Wen WQ, Doll MA, et al. N-Acetyltransferase-2 genetic polymorphism, well-done meat intake, and breast cancer risk among postmenopausal women. Cancer Epidemiol Biomarkers Prev. 2000;9(9):905–10. [PubMed: 11008907]
  • Delfino RJ, Sinha R, Smith C, West J, White E, Lin HJ, et al. Breast cancer, heterocyclic aromatic amines from meat and N-acetyltransferase 2 genotype. Carcinogenesis. 2000;21(4):607–15. [PubMed: 10753193] [CrossRef]
  • Demeyer D, Mertens B, De Smet S, Ulens M. Mechanisms linking colorectal cancer to the consumption of (processed) red meat: a review. Crit Rev Food Sci Nutr. 2015;56(16):2747–66. [PubMed: 25975275] [CrossRef]
  • Deo V, Zhang Y, Soghomonian V, Heremans JJ. Quantum interference measurement of spin interactions in a bio-organic/semiconductor device structure. Sci Rep. 2015;5(1):9487. [PMC free article: PMC4377638] [PubMed: 25820781] [CrossRef]
  • Dias-Neto M, Pintalhao M, Ferreira M, Lunet N. Salt intake and risk of gastric intestinal metaplasia: systematic review and meta-analysis. Nutr Cancer. 2010;62(2):133–47. [PubMed: 20099187] [CrossRef]
  • Dich J, Järvinen R, Knekt P, Penttilä PL. Dietary intakes of nitrate, nitrite and NDMA in the Finnish Mobile Clinic Health Examination Survey. Food Addit Contam. 1996;13(5):541–52. [PubMed: 8799716] [CrossRef]
  • Diergaarde B, van Geloof WL, van Muijen GN, Kok FJ, Kampman E. Dietary factors and the occurrence of truncating APC mutations in sporadic colon carcinomas: a Dutch population-based study. Carcinogenesis. 2003;24(2):283–90. [PubMed: 12584179] [CrossRef]
  • Dolara P, Commoner B, Vithayathil A, Cuca G, Tuley E, Madyastha P, et al. The effect of temperature on the formation of mutagens in heated beef stock and cooked ground beef. Mutat Res. 1979;60(3):231–7. [PubMed: 384210] [CrossRef]
  • Dolara P, Barale R, Mazzoli S, Benetti D. Activation of the mutagens of beef extract in vitro and in vivo. Mutat Res. 1980;79(3):213–21. [PubMed: 7012603] [CrossRef]
  • Dolara P, Caderni G, Salvadori M, Tringale L, Lodovici M. Urinary mutagens in humans after fried pork and bacon meals. Cancer Lett. 1984;22(3):275–80. [PubMed: 6713368] [CrossRef]
  • Doolittle DJ, Rahn CA, Burger GT, Lee CK, Reed B, Riccio E, et al. Effect of cooking methods on the mutagenicity of food and on urinary mutagenicity of human consumers. Food Chem Toxicol. 1989;27(10):657–66. [PubMed: 2606402] [CrossRef]
  • El-Serag HB, Hampel H, Javadi F. The association between diabetes and hepatocellular carcinoma: a systematic review of epidemiologic evidence. Clin Gastroenterol Hepatol. 2006;4(3):369–80. [PubMed: 16527702] [CrossRef]
  • Ellmerich S, Schöller M, Duranton B, Gossé F, Galluser M, Klein JP, et al. Promotion of intestinal carcinogenesis by Streptococcus bovis. Carcinogenesis. 2000;21(4):753–6. [PubMed: 10753212] [CrossRef]
  • Engemann A, Focke C, Humpf HU. Intestinal formation of N-nitroso compounds in the pig cecum model. J Agric Food Chem. 2013;61(4):998–1005. [PubMed: 23297847] [CrossRef]
  • Esterbauer H. Cytotoxicity and genotoxicity of lipid-oxidation products. Am J Clin Nutr. 1993;57(5) Suppl:779S–85S, discussion 785S–6S. [PubMed: 8475896] [CrossRef]
  • Esterbauer H, Schaur RJ, Zollner H. Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radic Biol Med. 1991;11(1):81–128. [PubMed: 1937131] [CrossRef]
  • Faustman C, Sun Q, Mancini R, Suman SP. Myoglobin and lipid oxidation interactions: mechanistic bases and control. Meat Sci. 2010;86(1):86–94. [PubMed: 20554121] [CrossRef]
  • Fay M, Fennerty MB, Emerson J, Larez M. Dietary habits and the risk of stomach cancer: a comparison study of patients with and without intestinal metaplasia. Gastroenterol Nurs. 1994;16(4):158–62. [PubMed: 8110846] [CrossRef]
  • Felton JS, Knize MG, Wu RW, Colvin ME, Hatch FT, Malfatti MA. Mutagenic potency of food-derived heterocyclic amines. Mutat Res. 2007;616(1-2):90–4. [PubMed: 17161439] [CrossRef]
  • Ferrucci LM, Cross AJ, Graubard BI, Brinton LA, McCarty CA, Ziegler RG, et al. Intake of meat, meat mutagens, and iron and the risk of breast cancer in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. Br J Cancer. 2009;101(1):178–84. [PMC free article: PMC2713710] [PubMed: 19513076] [CrossRef]
  • Ferrucci LM, Cross AJ, Gunter MJ, Ahn J, Mayne ST, Ma X, et al. Xenobiotic metabolizing genes, meat-related exposures, and risk of advanced colorectal adenoma. World Rev Nutr Diet. 2010;101:34–45. a. [PMC free article: PMC3051350] [PubMed: 20436251]
  • Ferrucci LM, Sinha R, Ward MH, Graubard BI, Hollenbeck AR, Kilfoy BA, et al. Meat and components of meat and the risk of bladder cancer in the NIH-AARP Diet and Health Study. Cancer. 2010;116(18):4345–53. b. [PMC free article: PMC2936663] [PubMed: 20681011] [CrossRef]
  • Ferrucci LM, Sinha R, Huang WY, Berndt SI, Katki HA, Schoen RE, et al. Meat consumption and the risk of incident distal colon and rectal adenoma. Br J Cancer. 2012;106(3):608–16. [PMC free article: PMC3281548] [PubMed: 22166801] [CrossRef]
  • Figueiredo JC, Hsu L, Hutter CM, Lin Y, Campbell PT, Baron JA, et al. CCFR. GECCO. Genome-wide diet-gene interaction analyses for risk of colorectal cancer. PLoS Genet. 2014;10(4):e1004228. [PMC free article: PMC3990510] [PubMed: 24743840] [CrossRef]
  • Figueroa JD, Han SS, Garcia-Closas M, Baris D, Jacobs EJ, Kogevinas M, et al. Genome-wide interaction study of smoking and bladder cancer risk. Carcinogenesis. 2014;35(8):1737–44. [PMC free article: PMC4123644] [PubMed: 24662972] [CrossRef]
  • Fonseca-Nunes A, Jakszyn P, Agudo A. Iron and cancer risk–a systematic review and meta-analysis of the epidemiological evidence. Cancer Epidemiol Biomarkers Prev. 2014;23(1):12–31. [PubMed: 24243555] [CrossRef]
  • Freedman ND, Cross AJ, McGlynn KA, Abnet CC, Park Y, Hollenbeck AR, et al. Association of meat and fat intake with liver disease and hepatocellular carcinoma in the NIH-AARP cohort. J Natl Cancer Inst. 2010;102(17):1354–65. [PMC free article: PMC2935477] [PubMed: 20729477] [CrossRef]
  • Friesen MD, Kaderlik K, Lin D, Garren L, Bartsch H, Lang NP, et al. Analysis of DNA adducts of 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine in rat and human tissues by alkaline hydrolysis and gas chromatography/electron capture mass spectrometry: validation by comparison with 32P-postlabeling. Chem Res Toxicol. 1994;7(6):733–9. [PubMed: 7696526] [CrossRef]
  • Fu Z, Shrubsole MJ, Smalley WE, Wu H, Chen Z, Shyr Y, et al. Association of meat intake and meat-derived mutagen exposure with the risk of colorectal polyps by histologic type. Cancer Prev Res (Phila). 2011;4(10):1686–97. [PMC free article: PMC3188364] [PubMed: 21803984] [CrossRef]
  • Fu Z, Shrubsole MJ, Li G, Smalley WE, Hein DW, Chen Z, et al. Using gene-environment interaction analyses to clarify the role of well-done meat and heterocyclic amine exposure in the etiology of colorectal polyps. Am J Clin Nutr. 2012;96(5):1119–28. [PMC free article: PMC3471199] [PubMed: 23015320] [CrossRef]
  • Gabbani G, Nardini B, Bordin A, Pavanello S, Janni L, Celotti L, et al. Urinary mutagenicity on TA98 and YG1024 Salmonella typhimurium strains after a hamburger meal: influence of GSTM1 and NAT2 genotypes. Mutagenesis. 1998;13(2):187–91. [PubMed: 9568593] [CrossRef]
  • Garner RC, Lightfoot TJ, Cupid BC, Russell D, Coxhead JM, Kutschera W, et al. Comparative biotransformation studies of MeIQx and PhIP in animal models and humans. Cancer Lett. 1999;143(2):161–5. [PubMed: 10503897] [CrossRef]
  • Garrett WS. Cancer and the microbiota. Science. 2015;348(6230):80–6. [PMC free article: PMC5535753] [PubMed: 25838377] [CrossRef]
  • Gay LJ, Mitrou PN, Keen J, Bowman R, Naguib A, Cooke J, et al. Dietary, lifestyle and clinicopathological factors associated with APC mutations and promoter methylation in colorectal cancers from the EPIC-Norfolk study. J Pathol. 2012;228(3):405–15. [PubMed: 22864938] [CrossRef]
  • Gertig DM, Hankinson SE, Hough H, Spiegelman D, Colditz GA, Willett WC, et al. N-acetyl transferase 2 genotypes, meat intake and breast cancer risk. Int J Cancer. 1999;80(1):13–7. [PubMed: 9935222] [CrossRef]
  • Gibson TM, Smedby KE, Skibola CF, Hein DW, Slager SL, de Sanjosé S, et al. Smoking, variation in N-acetyltransferase 1 (NAT1) and 2 (NAT2), and risk of non-Hodgkin lymphoma: a pooled analysis within the InterLymph consortium. Cancer Causes Control. 2013;24(1):125–34. [PMC free article: PMC3529854] [PubMed: 23160945] [CrossRef]
  • Giles GG, McNeil JJ, Donnan G, Webley C, Staples MP, Ireland PD, et al. Dietary factors and the risk of glioma in adults: results of a case-control study in Melbourne, Australia. Int J Cancer. 1994;59(3):357–62. [PubMed: 7927941] [CrossRef]
  • Gilsing AM, Berndt SI, Ruder EH, Graubard BI, Ferrucci LM, Burdett L, et al. Meat-related mutagen exposure, xenobiotic metabolizing gene polymorphisms and the risk of advanced colorectal adenoma and cancer. Carcinogenesis. 2012;33(7):1332–9. [PMC free article: PMC3499048] [PubMed: 22552404] [CrossRef]
  • Gilsing AM, Fransen F, de Kok TM, Goldbohm AR, Schouten LJ, de Bruïne AP, et al. Dietary heme iron and the risk of colorectal cancer with specific mutations in KRAS and APC. Carcinogenesis. 2013;34(12):2757–66. [PubMed: 23983135] [CrossRef]
  • Girard H, Butler LM, Villeneuve L, Millikan RC, Sinha R, Sandler RS, et al. UGT1A1 and UGT1A9 functional variants, meat intake, and colon cancer, among Caucasians and African-Americans. Mutat Res. 2008;644(1-2):56–63. [PMC free article: PMC2570038] [PubMed: 18675828] [CrossRef]
  • Glei M, Latunde-Dada GO, Klinder A, Becker TW, Hermann U, Voigt K, et al. Iron-overload induces oxidative DNA damage in the human colon carcinoma cell line HT29 clone 19A. Mutat Res. 2002;519(1-2):151–61. [PubMed: 12160900] [CrossRef]
  • Gocke E, Eckhardt K, King MT, Wild D. Mutagenicity study of fried sausages in Salmonella, Drosophila and mammalian cells in vitro and in vivo. Mutat Res. 1982;101(4):293–304. [PubMed: 6810162] [CrossRef]
  • Goode EL, Potter JD, Bamlet WR, Rider DN, Bigler J. Inherited variation in carcinogen-metabolizing enzymes and risk of colorectal polyps. Carcinogenesis. 2007;28(2):328–41. [PubMed: 16926176] [CrossRef]
  • Gorelik S, Ligumsky M, Kohen R, Kanner J. The stomach as a “bioreactor”: when red meat meets red wine. J Agric Food Chem. 2008;56(13):5002–7. [PubMed: 18540628] [CrossRef]
  • Gottschalg E, Scott GB, Burns PA, Shuker DE. Potassium diazoacetate-induced p53 mutations in vitro in relation to formation of O6-carboxymethyl- and O6-methyl-2′-deoxyguanosine DNA adducts: relevance for gastrointestinal cancer. Carcinogenesis. 2007;28(2):356–62. [PubMed: 16926174] [CrossRef]
  • Graf D, Di Cagno R, Fåk F, Flint HJ, Nyman M, Saarela M, et al. Contribution of diet to the composition of the human gut microbiota. Microb Ecol Health Dis. 2015;26:26164. [PMC free article: PMC4318938] [PubMed: 25656825] [CrossRef]
  • Gu D, Turesky RJ, Tao Y, Langouët SA, Nauwelaërs GC, Yuan JM, et al. DNA adducts of 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine and 4-aminobiphenyl are infrequently detected in human mammary tissue by liquid chromatography/tandem mass spectrometry. Carcinogenesis. 2012;33(1):124–30. [PMC free article: PMC3276332] [PubMed: 22072616] [CrossRef]
  • Guéraud F, Atalay M, Bresgen N, Cipak A, Eckl PM, Huc L, et al. Chemistry and biochemistry of lipid peroxidation products. Free Radic Res. 2010;44(10):1098–124. [PubMed: 20836659] [CrossRef]
  • Guéraud F, Taché S, Steghens JP, Milkovic L, Borovic-Sunjic S, Zarkovic N, et al. Dietary polyunsaturated fatty acids and heme iron induce oxidative stress biomarkers and a cancer promoting environment in the colon of rats. Free Radic Biol Med. 2015;83:192–200. [PubMed: 25744414] [CrossRef]
  • Gunter MJ, Divi RL, Kulldorff M, Vermeulen R, Haverkos KJ, Kuo MM, et al. Leukocyte polycyclic aromatic hydrocarbon-DNA adduct formation and colorectal adenoma. Carcinogenesis. 2007;28(7):1426–9. [PubMed: 17277232] [CrossRef]
  • Hammerling U, Laurila JB, Grafström R, Ilbäck NG. Consumption of red/processed meat and colorectal carcinoma: possible mechanisms underlying the significant association. Crit Rev Food Sci Nutr. 2015;56(4)::614–34. [PubMed: 25849747] [CrossRef]
  • Hayatsu H, Hayatsu T, Ohara Y. Mutagenicity of human urine caused by ingestion of fried ground beef. Jpn J Cancer Res. 1985;76(6):445–8. [PubMed: 3926577]
  • Hebels DG, Sveje KM, de Kok MC, van Herwijnen MH, Kuhnle GG, Engels LG, et al. Red meat intake-induced increases in fecal water genotoxicity correlate with pro-carcinogenic gene expression changes in the human colon. Food Chem Toxicol. 2012;50(2):95–103. [PubMed: 22019696] [CrossRef]
  • Hedlund M, Padler-Karavani V, Varki NM, Varki A. Evidence for a human-specific mechanism for diet and antibody-mediated inflammation in carcinoma progression. Proc Natl Acad Sci USA. 2008;105(48):18936–41. [PMC free article: PMC2596253] [PubMed: 19017806] [CrossRef]
  • Hengstler JG, Van der Burg B, Steinberg P, Oesch F. Interspecies differences in cancer susceptibility and toxicity. Drug Metab Rev. 1999;31(4):917–70. [PubMed: 10575555] [CrossRef]
  • Hodgson JM, Ward NC, Burke V, Beilin LJ, Puddey IB. Increased lean red meat intake does not elevate markers of oxidative stress and inflammation in humans. J Nutr. 2007;137(2):363–7. [PubMed: 17237312] [CrossRef]
  • Hogg N. Red meat and colon cancer: heme proteins and nitrite in the gut. A commentary on “diet-induced endogenous formation of nitroso compounds in the GI tract”. Free Radic Biol Med. 2007;43(7):1037–9. [PubMed: 17761299] [CrossRef]
  • Holtrop G, Johnstone AM, Fyfe C, Gratz SW. Diet composition is associated with endogenous formation of N-nitroso compounds in obese men. J Nutr. 2012;142(9):1652–8. [PubMed: 22833653] [CrossRef]
  • Honikel KO. The use and control of nitrate and nitrite for the processing of meat products. Meat Sci. 2008;78(1-2):68–76. [PubMed: 22062097] [CrossRef]
  • Hooda J, Shah A, Zhang L. Heme, an essential nutrient from dietary proteins, critically impacts diverse physiological and pathological processes. Nutrients. 2014;6(3):1080–102. [PMC free article: PMC3967179] [PubMed: 24633395] [CrossRef]
  • Hua NW, Stoohs RA, Facchini FS. Low iron status and enhanced insulin sensitivity in lacto-ovo vegetarians. Br J Nutr. 2001;86(4):515–9. [PubMed: 11591239] [CrossRef]
  • Hughes R, Cross AJ, Pollock JR, Bingham S. Dose-dependent effect of dietary meat on endogenous colonic N-nitrosation. Carcinogenesis. 2001;22(1):199–202. [PubMed: 11159760] [CrossRef]
  • Hughes R, Pollock JR, Bingham S. Effect of vegetables, tea, and soy on endogenous N-nitrosation, fecal ammonia, and fecal water genotoxicity during a high red meat diet in humans. Nutr Cancer. 2002;42(1):70–7. [PubMed: 12235653] [CrossRef]
  • Humphreys KJ, Conlon MA, Young GP, Topping DL, Hu Y, Winter JM, et al. Dietary manipulation of oncogenic microRNA expression in human rectal mucosa: a randomized trial. Cancer Prev Res (Phila). 2014;7(8):786–95. [PubMed: 25092886] [CrossRef]
  • Huycke MM, Moore DR. In vivo production of hydroxyl radical by Enterococcus faecalis colonizing the intestinal tract using aromatic hydroxylation. Free Radic Biol Med. 2002;33(6):818–26. [PubMed: 12208369] [CrossRef]
  • IARC (1993). Some naturally occurring substances: food items and constituents, heterocyclic aromatic amines and mycotoxins. IARC Monogr Eval Carcinog Risks Hum, 56:1–599. Available from: http://monographs​.iarc​.fr/ENG/Monographs/vol56/index.php.
  • IARC (2007). Smokeless tobacco and some tobacco-specific N-nitrosamines. IARC Monogr Eval Carcinog Risks Hum, 89:1–592. Available from: http://monographs​.iarc​.fr/ENG/Monographs/vol89/index.php. [PMC free article: PMC4781254] [PubMed: 18335640]
  • IARC (2010). Some non-heterocyclic polycyclic aromatic hydrocarbons and some related exposures. IARC Monogr Eval Carcinog Risks Hum, 92:1–853. Available from: http://monographs​.iarc​.fr/ENG/Monographs/vol92/index.php. [PMC free article: PMC4781319] [PubMed: 21141735]
  • IARC (2012a). Chemical agents and related occupations. IARC Monogr Eval Carcinog Risks Hum, 100F:1–599. . Available from: http://monographs​.iarc​.fr/ENG/Monographs/vol100F/index.php. [PMC free article: PMC4781612] [PubMed: 23189753]
  • IARC (2012b). Personal habits and indoor combustions. IARC Monogr Eval Carcinog Risks Hum, 100E:1–575. . Available from: http://monographs​.iarc​.fr/ENG/Monographs/vol100E/index.php. [PMC free article: PMC4781577] [PubMed: 23193840]
  • Ihara Y, Toyokuni S, Uchida K, Odaka H, Tanaka T, Ikeda H, et al. Hyperglycemia causes oxidative stress in pancreatic beta-cells of GK rats, a model of type 2 diabetes. Diabetes. 1999;48(4):927–32. [PubMed: 10102716] [CrossRef]
  • Ijssennagger N, de Wit N, Müller M, van der Meer R. Dietary heme-mediated PPARα activation does not affect the heme-induced epithelial hyperproliferation and hyperplasia in mouse colon. PLoS One. 2012;7(8):e43260. [PMC free article: PMC3419209] [PubMed: 22905243] [CrossRef]
  • Ijssennagger N, Rijnierse A, de Wit NJ, Boekschoten MV, Dekker J, Schonewille A, et al. Dietary heme induces acute oxidative stress, but delayed cytotoxicity and compensatory hyperproliferation in mouse colon. Carcinogenesis. 2013;34(7):1628–35. [PubMed: 23455377] [CrossRef]
  • Ijssennagger N, Belzer C, Hooiveld GJ, Dekker J, van Mil SW, Müller M, et al. Gut microbiota facilitates dietary heme-induced epithelial hyperproliferation by opening the mucus barrier in colon. Proc Natl Acad Sci USA. 2015;112(32):10038–43. [PMC free article: PMC4538683] [PubMed: 26216954] [CrossRef]
  • Ishibe N, Sinha R, Hein DW, Kulldorff M, Strickland P, Fretland AJ, et al. Genetic polymorphisms in heterocyclic amine metabolism and risk of colorectal adenomas. Pharmacogenetics. 2002;12(2):145–50. [PubMed: 11875368] [CrossRef]
  • Ishikawa S, Tamaki S, Ohata M, Arihara K, Itoh M. Heme induces DNA damage and hyperproliferation of colonic epithelial cells via hydrogen peroxide produced by heme oxygenase: a possible mechanism of heme-induced colon cancer. Mol Nutr Food Res. 2010;54(8):1182–91. [PubMed: 20112302]
  • Issenberg P. Nitrite, nitrosamines, and cancer. Fed Proc. 1976;35(6):1322–6. [PubMed: 4342]
  • Iwasaki M, Mukai T, Takachi R, Ishihara J, Totsuka Y, Tsugane S. Validity of a self-administered food frequency questionnaire in the estimation of heterocyclic aromatic amines. Cancer Causes Control. 2014;25(8):1015–28. [PubMed: 24890804] [CrossRef]
  • Jeffy BD, Chirnomas RB, Romagnolo DF. Epigenetics of breast cancer: polycyclic aromatic hydrocarbons as risk factors. Environ Mol Mutagen. 2002;39(2-3):235–44. [PubMed: 11921194] [CrossRef]
  • Jiao L, Taylor PR, Weinstein SJ, Graubard BI, Virtamo J, Albanes D, et al. Advanced glycation end products, soluble receptor for advanced glycation end products, and risk of colorectal cancer. Cancer Epidemiol Biomarkers Prev. 2011;20(7):1430–8. [PMC free article: PMC3132292] [PubMed: 21527578] [CrossRef]
  • Jiao S, Hsu L, Berndt S, Bézieau S, Brenner H, Buchanan D, et al. Genome-wide search for gene-gene interactions in colorectal cancer. PLoS One. 2012;7(12):e52535. [PMC free article: PMC3530500] [PubMed: 23300701] [CrossRef]
  • Jiao L, Kramer JR, Chen L, Rugge M, Parente P, Verstovsek G, et al. Dietary consumption of meat, fat, animal products and advanced glycation end-products and the risk of Barrett’s oesophagus. Aliment Pharmacol Ther. 2013;38(7):817–24. [PMC free article: PMC3811083] [PubMed: 23957669] [CrossRef]
  • Joosen AM, Kuhnle GG, Aspinall SM, Barrow TM, Lecommandeur E, Azqueta A, et al. Effect of processed and red meat on endogenous nitrosation and DNA damage. Carcinogenesis. 2009;30(8):1402–7. [PMC free article: PMC2718076] [PubMed: 19498009] [CrossRef]
  • Joosen AM, Lecommandeur E, Kuhnle GG, Aspinall SM, Kap L, Rodwell SA. Effect of dietary meat and fish on endogenous nitrosation, inflammation and genotoxicity of faecal water. Mutagenesis. 2010;25(3):243–7. [PubMed: 20106932] [CrossRef]
  • Joshi AD, Corral R, Siegmund KD, Haile RW, Le Marchand L, Martínez ME, et al. Red meat and poultry intake, polymorphisms in the nucleotide excision repair and mismatch repair pathways and colorectal cancer risk. Carcinogenesis. 2009;30(3):472–9. [PMC free article: PMC2722151] [PubMed: 19029193] [CrossRef]
  • Kabat GC, Cross AJ, Park Y, Schatzkin A, Hollenbeck AR, Rohan TE, et al. Meat intake and meat preparation in relation to risk of postmenopausal breast cancer in the NIH-AARP diet and health study. Int J Cancer. 2009;124(10):2430–5. [PMC free article: PMC3491884] [PubMed: 19165862] [CrossRef]
  • Kabat GC, Cross AJ, Park Y, Schatzkin A, Hollenbeck AR, Rohan TE, et al. Intakes of dietary iron and heme-iron and risk of postmenopausal breast cancer in the National Institutes of Health-AARP Diet and Health Study. Am J Clin Nutr. 2010;92(6):1478–83. [PMC free article: PMC3478325] [PubMed: 20962158] [CrossRef]
  • Kampman E, Slattery ML, Bigler J, Leppert M, Samowitz W, Caan BJ, et al. Meat consumption, genetic susceptibility, and colon cancer risk: a United States multicenter case-control study. Cancer Epidemiol Biomarkers Prev. 1999;8(1):15–24. [PubMed: 9950235]
  • Kanner J. Oxidative processes in meat and meat products: Quality implications. Meat Sci. 1994;36(1-2):169–89. [PubMed: 22061459] [CrossRef]
  • Kanner J. Dietary advanced lipid oxidation endproducts are risk factors to human health. Mol Nutr Food Res. 2007;51(9):1094–101. [PubMed: 17854006] [CrossRef]
  • Kanner J, Lapidot T. The stomach as a bioreactor: dietary lipid peroxidation in the gastric fluid and the effects of plant-derived antioxidants. Free Radic Biol Med. 2001;31(11):1388–95. [PubMed: 11728810] [CrossRef]
  • Kantor ED, Hutter CM, Minnier J, Berndt SI, Brenner H, Caan BJ, et al. Gene-environment interaction involving recently identified colorectal cancer susceptibility Loci. Cancer Epidemiol Biomarkers Prev. 2014;23(9):1824–33. [PMC free article: PMC4209726] [PubMed: 24994789] [CrossRef]
  • Kassie F, Lhoste EF, Bruneau A, Zsivkovits M, Ferk F, Uhl M, et al. Effect of intestinal microfloras from vegetarians and meat eaters on the genotoxicity of 2-amino-3-methylimidazo[4,5-f]quinoline, a carcinogenic heterocyclic amine. J Chromatogr B Analyt Technol Biomed Life Sci. 2004;802(1):211–5. [PubMed: 15036013] [CrossRef]
  • Kataoka H, Inoue T, Saito K, Kato H, Masuda K. Analysis of heterocyclic amines in hair by on-line in-tube solid-phase microextraction coupled with liquid chromatography-tandem mass spectrometry. Anal Chim Acta. 2013;786:54–60. [PubMed: 23790292] [CrossRef]
  • Keszei AP, Schouten LJ, Driessen AL, Huysentruyt CJ, Keulemans YC, van den Brandt PA. Meat consumption and the risk of Barrett’s esophagus in a large Dutch cohort. Cancer Epidemiol Biomarkers Prev. 2013;22(6):1162–6. [PubMed: 23580699] [CrossRef]
  • Khil J, Gallaher DD. Beef tallow increases apoptosis and decreases aberrant crypt foci formation relative to soybean oil in rat colon. Nutr Cancer. 2004;50(1):55–62. [PubMed: 15572298] [CrossRef]
  • Kim Y, Keogh J, Clifton P. A review of potential metabolic etiologies of the observed association between red meat consumption and development of type 2 diabetes mellitus. Metabolism. 2015;64(7):768–79. [PubMed: 25838035] [CrossRef]
  • Klenow S, Pool-Zobel BL, Glei M. Influence of inorganic and organic iron compounds on parameters of cell growth and survival in human colon cells. Toxicol In Vitro. 2009;23(3):400–7. [PubMed: 19444923] [CrossRef]
  • Kobayashi M, Hanaoka T, Tsugane S. Validity of a self-administered food frequency questionnaire in the assessment of heterocyclic amine intake using 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) levels in hair. Mutat Res. 2007;630(1-2):14–9. [PubMed: 17392018] [CrossRef]
  • Kongkachuichai R, Napatthalung P, Charoensiri R. Heme and nonheme iron content of animal products commonly consumed in Thailand. J Food Compos Anal. 2002;15(4):389–98. [CrossRef]
  • Koutros S, Cross AJ, Sandler DP, Hoppin JA, Ma X, Zheng T, et al. Meat and meat mutagens and risk of prostate cancer in the Agricultural Health Study. Cancer Epidemiol Biomarkers Prev. 2008;17(1):80–7. [PMC free article: PMC2935655] [PubMed: 18199713] [CrossRef]
  • Kristensen L, Purslow PP. The effect of processing temperature and addition of mono- and di-valent salts on the heme- nonheme-iron ratio in meat. Food Chem. 2001;73(4):433–9. [CrossRef]
  • Kubo A, Levin TR, Block G, Rumore GJ, Quesenberry CP Jr, Buffler P, et al. Dietary patterns and the risk of Barrett’s esophagus. Am J Epidemiol. 2008;167(7):839–46. [PMC free article: PMC5630266] [PubMed: 18218607] [CrossRef]
  • Kubo A, Block G, Quesenberry CP Jr, Buffler P, Corley DA. Effects of dietary fiber, fats, and meat intakes on the risk of Barrett’s esophagus. Nutr Cancer. 2009;61(5):607–16. [PMC free article: PMC2765669] [PubMed: 19838934] [CrossRef]
  • Kuhnle GG, Bingham SA. Dietary meat, endogenous nitrosation and colorectal cancer. Biochem Soc Trans. 2007;35(Pt 5):1355–7. [PubMed: 17956350] [CrossRef]
  • Kuhnle GG, Story GW, Reda T, Mani AR, Moore KP, Lunn JC, et al. Diet-induced endogenous formation of nitroso compounds in the GI tract. Free Radic Biol Med. 2007;43(7):1040–7. [PubMed: 17761300] [CrossRef]
  • Küry S, Buecher B, Robiou-du-Pont S, Scoul C, Sébille V, Colman H, et al. Combinations of cytochrome P450 gene polymorphisms enhancing the risk for sporadic colorectal cancer related to red meat consumption. Cancer Epidemiol Biomarkers Prev. 2007;16(7):1460–7. [PubMed: 17627011] [CrossRef]
  • Lakshmi VM, Clapper ML, Chang WC, Zenser TV. Hemin potentiates nitric oxide-mediated nitrosation of 2-amino-3-methylimidazo[4,5-f]quinoline (IQ) to 2-nitrosoamino-3-methylimidazo[4,5-f]quinoline. Chem Res Toxicol. 2005;18(3):528–35. a. [PubMed: 15777093] [CrossRef]
  • Lakshmi VM, Hsu FF, Zenser TV. Nitric oxide-mediated nitrosation of 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline potentiated by hemin and myeloperoxidase. Chem Res Toxicol. 2005;18(6):1038–47. b. [PubMed: 15962939] [CrossRef]
  • Lam TK, Rotunno M, Ryan BM, Pesatori AC, Bertazzi PA, Spitz M, et al. Heme-related gene expression signatures of meat intakes in lung cancer tissues. Mol Carcinog. 2014;53(7):548–56. [PMC free article: PMC4152901] [PubMed: 23681825] [CrossRef]
  • Lang NP, Butler MA, Massengill J, Lawson M, Stotts RC, Hauer-Jensen M, et al. Rapid metabolic phenotypes for acetyltransferase and cytochrome P4501A2 and putative exposure to food-borne heterocyclic amines increase the risk for colorectal cancer or polyps. Cancer Epidemiol Biomarkers Prev. 1994;3(8):675–82. [PubMed: 7881341]
  • Larsson SC, Orsini N, Wolk A. Diabetes mellitus and risk of colorectal cancer: a meta-analysis. J Natl Cancer Inst. 2005;97(22):1679–87. [PubMed: 16288121] [CrossRef]
  • Larsson SC, Orsini N, Brismar K, Wolk A. Diabetes mellitus and risk of bladder cancer: a meta-analysis. Diabetologia. 2006;49(12):2819–23. [PubMed: 17021919] [CrossRef]
  • Lauber SN, Ali S, Gooderham NJ. The cooked food derived carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b] pyridine is a potent oestrogen: a mechanistic basis for its tissue-specific carcinogenicity. Carcinogenesis. 2004;25(12):2509–17. [PubMed: 15319301] [CrossRef]
  • Lauber SN, Gooderham NJ. The cooked meat derived genotoxic carcinogen 2-amino-3-methylimidazo[4,5-b]pyridine has potent hormone-like activity: mechanistic support for a role in breast cancer. Cancer Res. 2007;67(19):9597–602. [PubMed: 17909072] [CrossRef]
  • Layrisse M, Cook JD, Martinez C, Roche M, Kuhn IN, Walker RB, et al. Food iron absorption: a comparison of vegetable and animal foods. Blood. 1969;33(3):430–43. [PubMed: 5766781]
  • Le Leu RK, Winter JM, Christophersen CT, Young GP, Humphreys KJ, Hu Y, et al. Butyrylated starch intake can prevent red meat-induced O6-methyl-2-deoxyguanosine adducts in human rectal tissue: a randomised clinical trial. Br J Nutr. 2015;114(2):220–30. [PMC free article: PMC4531472] [PubMed: 26084032] [CrossRef]
  • LeMarchand L, Wilkinson GR, Wilkens LR. 1999Genetic and dietary predictors of CYP2E1 activity: a phenotyping study in Hawaii Japanese using chlorzoxazone. Cancer Epidemiol Biomarkers Prev 8(6):495–500. [PubMed: 10385138]
  • Le Marchand L, Donlon T, Seifried A, Wilkens LR. Red meat intake, CYP2E1 genetic polymorphisms, and colorectal cancer risk. Cancer Epidemiol Biomarkers Prev. 2002;11(10 Pt 1):1019–24. a. [PubMed: 12376502]
  • Le Marchand L, Hankin JH, Pierce LM, Sinha R, Nerurkar PV, Franke AA, et al. Well-done red meat, metabolic phenotypes and colorectal cancer in Hawaii. Mutat Res. 2002;506-507:205–14. b. [PubMed: 12351160] [CrossRef]
  • Le Marchand L, Hankin JH, Wilkens LR, Pierce LM, Franke A, Kolonel LN, et al. Combined effects of well-done red meat, smoking, and rapid N-acetyltransferase 2 and CYP1A2 phenotypes in increasing colorectal cancer risk. Cancer Epidemiol Biomarkers Prev. 2001;10(12):1259–66. [PubMed: 11751443]
  • Lee DH, Anderson KE, Folsom AR, Jacobs DR Jr. Heme iron, zinc and upper digestive tract cancer: the Iowa Women’s Health Study. Int J Cancer. 2005;117(4):643–7. [PubMed: 15929082] [CrossRef]
  • Lee H, Wang Q, Yang F, Tao P, Li H, Huang Y, et al. SULT1A1 Arg213His polymorphism, smoked meat, and breast cancer risk: a case-control study and meta-analysis. DNA Cell Biol. 2012;31(5):688–99. [PubMed: 22011087] [CrossRef]
  • Lee HJ, Wu K, Cox DG, Hunter D, Hankinson SE, Willett WC, et al. Polymorphisms in xenobiotic metabolizing genes, intakes of heterocyclic amines and red meat, and postmenopausal breast cancer. Nutr Cancer. 2013;65(8):1122–31. [PMC free article: PMC3830653] [PubMed: 24099317] [CrossRef]
  • Lemaire-Ewing S, Prunet C, Montange T, Vejux A, Berthier A, Bessède G, et al. Comparison of the cytotoxic, pro-oxidant and pro-inflammatory characteristics of different oxysterols. Cell Biol Toxicol. 2005;21(2):97–114. [PubMed: 16142584] [CrossRef]
  • Lewin MH, Bailey N, Bandaletova T, Bowman R, Cross AJ, Pollock J, et al. Red meat enhances the colonic formation of the DNA adduct O6-carboxymethyl guanine: implications for colorectal cancer risk. Cancer Res. 2006;66(3):1859–65. [PubMed: 16452248] [CrossRef]
  • Ley SH, Sun Q, Willett WC, Eliassen AH, Wu K, Pan A, et al. Associations between red meat intake and biomarkers of inflammation and glucose metabolism in women. Am J Clin Nutr. 2014;99(2):352–60. [PMC free article: PMC3893727] [PubMed: 24284436] [CrossRef]
  • Li G, Wang H, Liu AB, Cheung C, Reuhl KR, Bosland MC, et al. Dietary carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine-induced prostate carcinogenesis in CYP1A-humanized mice. Cancer Prev Res (Phila). 2012;5(7):963–72. [PMC free article: PMC4147397] [PubMed: 22581815] [CrossRef]
  • Liao LM, Hofmann JN, Kamangar F, Strickland PT, Ji BT, Yang G, et al. Polycyclic aromatic hydrocarbons and risk of gastric cancer in the Shanghai Women’s Health Study. Int J Mol Epidemiol Genet. 2014;5(3):140–4. [PMC free article: PMC4214261] [PubMed: 25379133]
  • Lijinsky W. 1992). Chemistry and biology of N-nitroso compounds. Cambridge, United Kingdom: Cambridge University Press.
  • Lilla C, Verla-Tebit E, Risch A, Jäger B, Hoffmeister M, Brenner H, et al. Effect of NAT1 and NAT2 genetic polymorphisms on colorectal cancer risk associated with exposure to tobacco smoke and meat consumption. Cancer Epidemiol Biomarkers Prev. 2006;15(1):99–107. [PubMed: 16434594] [CrossRef]
  • Lin J, Forman MR, Wang J, Grossman HB, Chen M, Dinney CP, et al. Intake of red meat and heterocyclic amines, metabolic pathway genes and bladder cancer risk. Int J Cancer. 2012;131(8):1892–903. [PMC free article: PMC3415602] [PubMed: 22261697] [CrossRef]
  • Little J, Sharp L, Masson LF, Brockton NT, Cotton SC, Haites NE, et al. Colorectal cancer and genetic polymorphisms of CYP1A1, GSTM1 and GSTT1: a case-control study in the Grampian region of Scotland. Int J Cancer. 2006;119(9):2155–64. [PubMed: 16823842] [CrossRef]
  • Liu J, Ding D, Wang X, Chen Y, Li R, Zhang Y, et al. N-acetyltransferase polymorphism and risk of colorectal adenoma and cancer: a pooled analysis of variations from 59 studies. PLoS One. 2012;7(8):e42797. [PMC free article: PMC3419224] [PubMed: 22905173] [CrossRef]
  • Loh YH, Mitrou PN, Bowman R, Wood A, Jeffery H, Luben RN, et al. MGMT Ile143Val polymorphism, dietary factors and the risk of breast, colorectal and prostate cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC)-Norfolk study. DNA Repair (Amst). 2010;9(4):421–8. [PubMed: 20096652] [CrossRef]
  • Loh YH, Jakszyn P, Luben RN, Mulligan AA, Mitrou PN, Khaw KT. N-Nitroso compounds and cancer incidence: the European Prospective Investigation into Cancer and Nutrition (EPIC)-Norfolk Study. Am J Clin Nutr. 2011;93(5):1053–61. [PubMed: 21430112] [CrossRef]
  • Lombardi-Boccia G, Martinez-Dominguez B, Aguzzi A. Total heme and non-heme iron in raw and cooked meats. Food Chem Toxicol. 2002;67:1738–41.
  • Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nat Rev Microbiol. 2014;12(10):661–72. [PubMed: 25198138] [CrossRef]
  • Lucas D, Ménez C, Girre C, Berthou F, Bodénez P, Joannet I, et al. Cytochrome P450 2E1 genotype and chlorzoxazone metabolism in healthy and alcoholic Caucasian subjects. Pharmacogenetics. 1995;5(5):298–304. [PubMed: 8563770] [CrossRef]
  • Lunn JC, Kuhnle G, Mai V, Frankenfeld C, Shuker DE, Glen RC, et al. The effect of haem in red and processed meat on the endogenous formation of N-nitroso compounds in the upper gastrointestinal tract. Carcinogenesis. 2007;28(3):685–90. [PubMed: 17052997] [CrossRef]
  • Maduro IP, Nonino CB, Sakamoto LM, Meirelles MG, Cardeal Da Costa JA, Marchini JS. Red meat snacks for chronic hemodialysis patients: effect on inflammatory activity (a pilot study). Ren Fail. 2013;35(6):830–4. [PubMed: 23713604] [CrossRef]
  • Magagnotti C, Pastorelli R, Pozzi S, Andreoni B, Fanelli R, Airoldi L. Genetic polymorphisms and modulation of 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP)-DNA adducts in human lymphocytes. Int J Cancer. 2003;107(6):878–84. [PubMed: 14601045] [CrossRef]
  • Marcus PM, Vineis P, Rothman N. NAT2 slow acetylation and bladder cancer risk: a meta-analysis of 22 case-control studies conducted in the general population. Pharmacogenetics. 2000;10(2):115–22. [PubMed: 10761999] [CrossRef]
  • Marnett LJ. Oxyradicals and DNA damage. Carcinogenesis. 2000;21(3):361–70. [PubMed: 10688856] [CrossRef]
  • Martin OCB, Lin C, Naud N, Tache S, Raymond-Letron I, Corpet DE, et al. Antibiotic suppression of intestinal microbiota reduces heme-induced lipoperoxidation associated with colon carcinogenesis in rats. Nutr Cancer. 2015;67(1):119–25. [PubMed: 25514759] [CrossRef]
  • Mathew A, Sinha R, Burt R, Caan B, Paskett E, Iber F, et al. Polyp Prevention Study Group. Meat intake and the recurrence of colorectal adenomas. Eur J Cancer Prev. 2004;13(3):159–64. [PubMed: 15167213] [CrossRef]
  • Mauthe RJ, Dingley KH, Leveson SH, Freeman SP, Turesky RJ, Garner RC, et al. Comparison of DNA-adduct and tissue-available dose levels of MeIQx in human and rodent colon following administration of a very low dose. Int J Cancer. 1999;80(4):539–45. [PubMed: 9935154] [CrossRef]
  • McCarver DG, Byun R, Hines RN, Hichme M, Wegenek W. A genetic polymorphism in the regulatory sequences of human CYP2E1: association with increased chlorzoxazone hydroxylation in the presence of obesity and ethanol intake. Toxicol Appl Pharmacol. 1998;152(1):276–81. [PubMed: 9772223] [CrossRef]
  • Michaud DS, Holick CN, Batchelor TT, Giovannucci E, Hunter DJ. Prospective study of meat intake and dietary nitrates, nitrites, and nitrosamines and risk of adult glioma. Am J Clin Nutr. 2009;90(3):570–7. [PMC free article: PMC2728643] [PubMed: 19587083] [CrossRef]
  • Mignone LI, Giovannucci E, Newcomb PA, Titus-Ernstoff L, Trentham-Dietz A, Hampton JM, et al. Meat consumption, heterocyclic amines, NAT2, and the risk of breast cancer. Nutr Cancer. 2009;61(1):36–46. [PMC free article: PMC4902008] [PubMed: 19116874] [CrossRef]
  • Mirvish SS. Effects of vitamins C and E on N-nitroso compound formation, carcinogenesis, and cancer. Cancer. 1986;58(8) Suppl:1842–50. [PubMed: 3756808] [CrossRef]
  • Mirvish SS. Role of N-nitroso compounds (NOC) and N-nitrosation in etiology of gastric, esophageal, nasopharyngeal and bladder cancer and contribution to cancer of known exposures to NOC. Cancer Lett. 1995;93(1):17–48. [PubMed: 7600541] [CrossRef]
  • Mirvish SS, Haorah J, Zhou L, Clapper ML, Harrison KL, Povey AC. Total N-nitroso compounds and their precursors in hot dogs and in the gastrointestinal tract and feces of rats and mice: possible etiologic agents for colon cancer. J Nutr. 2002;132(11) Suppl:3526S–9S. [PubMed: 12421882] [CrossRef]
  • Mirvish SS, Haorah J, Zhou L, Hartman M, Morris CR, Clapper ML. N-nitroso compounds in the gastrointestinal tract of rats and in the feces of mice with induced colitis or fed hot dogs or beef. Carcinogenesis. 2003;24(3):595–603. [PubMed: 12663523] [CrossRef]
  • Mirvish SS, Reimers KJ, Kutler B, Chen SC, Haorah J, Morris CR, et al. Nitrate and nitrite concentrations in human saliva for men and women at different ages and times of the day and their consistency over time. Eur J Cancer Prev. 2000;9(5):335–42. [PubMed: 11075887] [CrossRef]
  • Montonen J, Boeing H, Fritsche A, Schleicher E, Joost HG, Schulze MB, et al. Consumption of red meat and whole-grain bread in relation to biomarkers of obesity, inflammation, glucose metabolism and oxidative stress. Eur J Nutr. 2013;52(1):337–45. [PMC free article: PMC3549403] [PubMed: 22426755] [CrossRef]
  • Mordukhovich I, Rossner P Jr, Terry MB, Santella R, Zhang YJ, Hibshoosh H, et al. Associations between polycyclic aromatic hydrocarbon-related exposures and p53 mutations in breast tumors. Environ Health Perspect. 2010;118(4):511–8. [PMC free article: PMC2854728] [PubMed: 20064791] [CrossRef]
  • Morita M, Le Marchand L, Kono S, Yin G, Toyomura K, Nagano J, et al. Genetic polymorphisms of CYP2E1 and risk of colorectal cancer: the Fukuoka Colorectal Cancer Study. Cancer Epidemiol Biomarkers Prev. 2009;18(1):235–41. [PubMed: 19124503] [CrossRef]
  • Morrissey PA, Sheehy PJ, Galvin K, Kerry JP, Buckley DJ. Lipid stability in meat and meat products. Meat Sci. 1998;49S1 S1:S73–86. [PubMed: 22060722] [CrossRef]
  • Murtaugh MA, Ma KN, Sweeney C, Caan BJ, Slattery ML. Meat consumption patterns and preparation, genetic variants of metabolic enzymes, and their association with rectal cancer in men and women. J Nutr. 2004;134(4):776–84. [PubMed: 15051825] [CrossRef]
  • Nagao M, Honda M, Seino Y, Yahagi T, Sugimura T. Mutagenicities of smoke condensates and the charred surface of fish and meat. Cancer Lett. 1977;2(4-5):221–6. [PubMed: 45723] [CrossRef]
  • Nagata C, Shimizu H, Kametani M, Takeyama N, Ohnuma T, Matsushita S. Diet and colorectal adenoma in Japanese males and females. Dis Colon Rectum. 2001;44(1):105–11. [PubMed: 11805576] [CrossRef]
  • Narayanan S, Hünerbein A, Getie M, Jäckel A, Neubert RH. Scavenging properties of metronidazole on free oxygen radicals in a skin lipid model system. J Pharm Pharmacol. 2007;59(8):1125–30. [PubMed: 17725855] [CrossRef]
  • Nishimoto IN, Hanaoka T, Sugimura H, Nagura K, Ihara M, Li XJ, et al. Cytochrome P450 2E1 polymorphism in gastric cancer in Brazil: case-control studies of Japanese Brazilians and non-Japanese Brazilians. Cancer Epidemiol Biomarkers Prev. 2000;9(7):675–80. [PubMed: 10919737]
  • Nomura A, Yamakawa H, Ishidate T, Kamiyama S, Masuda H, Stemmermann GN, et al. Intestinal metaplasia in Japan: association with diet. J Natl Cancer Inst. 1982;68(3):401–5. [PubMed: 6950167]
  • Nöthlings U, Yamamoto JF, Wilkens LR, Murphy SP, Park SY, Henderson BE, et al. Meat and heterocyclic amine intake, smoking, NAT1 and NAT2 polymorphisms, and colorectal cancer risk in the multiethnic cohort study. Cancer Epidemiol Biomarkers Prev. 2009;18(7):2098–106. [PMC free article: PMC2771770] [PubMed: 19549810] [CrossRef]
  • Nowell S, Ratnasinghe DL, Ambrosone CB, Williams S, Teague-Ross T, Trimble L, et al. Association of SULT1A1 phenotype and genotype with prostate cancer risk in African-Americans and Caucasians. Cancer Epidemiol Biomarkers Prev. 2004;13(2):270–6. [PubMed: 14973106] [CrossRef]
  • O’Brien H, Matthew JA, Gee JM, Watson M, Rhodes M, Speakman CT, et al. K-ras mutations, rectal crypt cells proliferation, and meat consumption in patients with left-sided colorectal carcinoma. Eur J Cancer Prev. 2000;9(1):41–7. [PubMed: 10777009] [CrossRef]
  • O’Callaghan NJ, Toden S, Bird AR, Topping DL, Fenech M, Conlon MA. Colonocyte telomere shortening is greater with dietary red meat than white meat and is attenuated by resistant starch. Clin Nutr. 2012;31(1):60–4. [PubMed: 21963168] [CrossRef]
  • O’Doherty MG, Cantwell MM, Murray LJ, Anderson LA, Abnet CC., FINBAR Study Group. Dietary fat and meat intakes and risk of reflux esophagitis, Barrett’s esophagus and esophageal adenocarcinoma. Int J Cancer. 2011;129(6):1493–502. [PMC free article: PMC3144995] [PubMed: 21455992] [CrossRef]
  • O’Keefe SJ. Nutrition and colonic health: the critical role of the microbiota. Curr Opin Gastroenterol. 2008;24(1):51–8. [PubMed: 18043233] [CrossRef]
  • Ochs-Balcom HM, Wiesner G, Elston RC. A meta-analysis of the association of N-acetyltransferase 2 gene (NAT2) variants with breast cancer. Am J Epidemiol. 2007;166(3):246–54. [PubMed: 17535831] [CrossRef]
  • Ohgaki H, Hasegawa H, Kato T, Suenaga M, Sato S, Takayama S, et al. Carcinogenicities in mice and rats of IQ, MeIQ, and MeIQx. Princess Takamatsu Symp. 1985;16:97–105. [PubMed: 3842704]
  • Ollberding NJ, Wilkens LR, Henderson BE, Kolonel LN, Le Marchand L. Meat consumption, heterocyclic amines and colorectal cancer risk: the Multiethnic Cohort Study. Int J Cancer. 2012;131(7):E1125–33. [PMC free article: PMC3553660] [PubMed: 22438055] [CrossRef]
  • Olsen P, Gry J, Knudsen I, Meyer O, Poulsen E. Animal feeding study with nitrite-treated meat. IARC Sci Publ. 1984;57(57):667–75. [PubMed: 6533058]
  • Owen RW. Faecal steroids and colorectal carcinogenesis. Scand J Gastroenterol Suppl. 1997;222 sup222:76–82. [PubMed: 9145454] [CrossRef]
  • Pariza MW, Ashoor SH, Chu FS, Lund DB. Effects of temperature and time on mutagen formation in pan-fried hamburger. Cancer Lett. 1979;7(2-3):63–9. [PubMed: 476611] [CrossRef]
  • Park JY, Mitrou PN, Keen J, Dahm CC, Gay LJ, Luben RN, et al. Lifestyle factors and p53 mutation patterns in colorectal cancer patients in the EPIC-Norfolk study. Mutagenesis. 2010;25(4):351–8. [PubMed: 20228093] [CrossRef]
  • Parnaud G, Peiffer G, Taché S, Corpet DE. Effect of meat (beef, chicken, and bacon) on rat colon carcinogenesis. Nutr Cancer. 1998;32(3):165–73. [PMC free article: PMC2527479] [PubMed: 10050267] [CrossRef]
  • Parnaud G, Pignatelli B, Peiffer G, Taché S, Corpet DE. Endogenous N-nitroso compounds, and their precursors, present in bacon, do not initiate or promote aberrant crypt foci in the colon of rats. Nutr Cancer. 2000;38(1):74–80. [PMC free article: PMC2638100] [PubMed: 11341048] [CrossRef]
  • Pavanello S, Simioli P, Mastrangelo G, Lupi S, Gabbani G, Gregorio P, et al. Role of metabolic polymorphisms NAT2 and CYP1A2 on urinary mutagenicity after a pan-fried hamburger meal. Food Chem Toxicol. 2002;40(8):1139–44. [PubMed: 12067576] [CrossRef]
  • Pélissier MA, Marteau P, Pochart P. Antioxidant effects of metronidazole in colonic tissue. Dig Dis Sci. 2007;52(1):40–4. [PubMed: 17151808] [CrossRef]
  • Peters U, Sinha R, Bell DA, Rothman N, Grant DJ, Watson MA, et al. Urinary mutagenesis and fried red meat intake: influence of cooking temperature, phenotype, and genotype of metabolizing enzymes in a controlled feeding study. Environ Mol Mutagen. 2004;43(1):53–74. [PubMed: 14743346] [CrossRef]
  • Phillips DH. Polycyclic aromatic hydrocarbons in the diet. Mutat Res. 1999;443(1-2):139–47. [PubMed: 10415437] [CrossRef]
  • Phillips JC, Lake BG, Heading CE, Gangolli SD, Lloyd AG. Studies on the metabolism of dimethylnitrosamine in the rat I. Effect of dose, route of administration and sex. Food Cosmet Toxicol. 1975;13(2):203–9. [PubMed: 1132850] [CrossRef]
  • Piche LA, Cole PD, Hadley M, van den Bergh R, Draper HH. Identification of N-epsilon-(2-propenal)lysine as the main form of malondialdehyde in food digesta. Carcinogenesis. 1988;9(3):473–7. [PubMed: 3125998] [CrossRef]
  • Pierre F, Taché S, Petit CR, Van der Meer R, Corpet DE. Meat and cancer: haemoglobin and haemin in a low-calcium diet promote colorectal carcinogenesis at the aberrant crypt stage in rats. Carcinogenesis. 2003;24(10):1683–90. [PMC free article: PMC2754080] [PubMed: 12896910] [CrossRef]
  • Pierre F, Freeman A, Taché S, Van der Meer R, Corpet DE. Beef meat and blood sausage promote the formation of azoxymethane-induced mucin-depleted foci and aberrant crypt foci in rat colons. J Nutr. 2004;134(10):2711–6. [PubMed: 15465771] [CrossRef]
  • Pierre F, Peiro G, Taché S, Cross AJ, Bingham SA, Gasc N, et al. New marker of colon cancer risk associated with heme intake: 1,4-dihydroxynonane mercapturic acid. Cancer Epidemiol Biomarkers Prev. 2006;15(11):2274–9. [PubMed: 17119057] [CrossRef]
  • Pierre F, Tache S, Guéraud F, Rerole AL, Jourdan ML, Petit C. Apc mutation induces resistance of colonic cells to lipoperoxide-triggered apoptosis induced by faecal water from haem-fed rats. Carcinogenesis. 2007;28(2):321–7. [PubMed: 16885197] [CrossRef]
  • Pierre F, Santarelli R, Taché S, Guéraud F, Corpet DE. Beef meat promotion of dimethylhydrazine-induced colorectal carcinogenesis biomarkers is suppressed by dietary calcium. Br J Nutr. 2008;99(5):1000–6. [PMC free article: PMC2666370] [PubMed: 17953789] [CrossRef]
  • Pierre FH, Santarelli RL, Allam O, Tache S, Naud N, Gueraud F, et al. Freeze-dried ham promotes azoxymethane-induced mucin-depleted foci and aberrant crypt foci in rat colon. Nutr Cancer. 2010;62(5):567–73. [PMC free article: PMC2936166] [PubMed: 20574917] [CrossRef]
  • Pierre FH, Martin OC, Santarelli RL, Taché S, Naud N, Guéraud F, et al. Calcium and α-tocopherol suppress cured-meat promotion of chemically induced colon carcinogenesis in rats and reduce associated biomarkers in human volunteers. Am J Clin Nutr. 2013;98(5):1255–62. [PMC free article: PMC3798078] [PubMed: 24025632] [CrossRef]
  • Poli G, Schaur RJ, Siems WG, Leonarduzzi G. 4-hydroxynonenal: a membrane lipid oxidation product of medicinal interest. Med Res Rev. 2008;28(4):569–631. [PubMed: 18058921] [CrossRef]
  • Poulsen MW, Hedegaard RV, Andersen JM, de Courten B, Bügel S, Nielsen J, et al. Advanced glycation endproducts in food and their effects on health. Food Chem Toxicol. 2013;60:10–37. [PubMed: 23867544] [CrossRef]
  • Poulsen MW, Bak MJ, Andersen JM, Monošík R, Giraudi-Futin AC, Holst JJ, et al. Effect of dietary advanced glycation end products on postprandial appetite, inflammation, and endothelial activation in healthy overweight individuals. Eur J Nutr. 2014;53(2):661–72. [PubMed: 23929260] [CrossRef]
  • Pourazrang H, Moazzami AA, Bazzaz BS. Inhibition of mutagenic N-nitroso compound formation in sausage samples by using l-ascorbic acid and α-tocopherol. Meat Sci. 2002;62(4):479–83. [PubMed: 22061756] [CrossRef]
  • Purchas RW, Busboom JR, Wilkinson BH. Changes in the forms of iron and in concentrations of taurine, carnosine, coenzyme Q(10), and creatine in beef longissimus muscle with cooking and simulated stomach and duodenal digestion. Meat Sci. 2006;74(3):443–9. [PubMed: 22063048] [CrossRef]
  • Qiao L, Feng Y. Intakes of heme iron and zinc and colorectal cancer incidence: a meta-analysis of prospective studies. Cancer Causes Control. 2013;24(6):1175–83. [PubMed: 23568532] [CrossRef]
  • Rao T, editor. (2013). Genotoxicity of N-nitroso compounds. Berlin, Germany: Springer Science and Business Media.
  • Reddy BS. Dietary fat and colon cancer: animal model studies. Lipids. 1992;27(10):807–13. [PubMed: 1435100] [CrossRef]
  • Reistad R, Rossland OJ, Latva-Kala KJ, Rasmussen T, Vikse R, Becher G, et al. Heterocyclic aromatic amines in human urine following a fried meat meal. Food Chem Toxicol. 1997;35(10-11):945–55. [PubMed: 9463528] [CrossRef]
  • Rieger MA, Parlesak A, Pool-Zobel BL, Rechkemmer G, Bode C. A diet high in fat and meat but low in dietary fibre increases the genotoxic potential of ‘faecal water’. Carcinogenesis. 1999;20(12):2311–6. [PubMed: 10590225] [CrossRef]
  • Rohrmann S, Lukas Jung SU, Linseisen J, Pfau W. Dietary intake of meat and meat-derived heterocyclic aromatic amines and their correlation with DNA adducts in female breast tissue. Mutagenesis. 2009;24(2):127–32. a. [PubMed: 18980957] [CrossRef]
  • Rohrmann S, Hermann S, Linseisen J. Heterocyclic aromatic amine intake increases colorectal adenoma risk: findings from a prospective European cohort study. Am J Clin Nutr. 2009;89(5):1418–24. b. [PubMed: 19261727] [CrossRef]
  • Rohrmann S, Nimptsch K, Sinha R, Willett WC, Giovannucci EL, Platz EA, et al. Intake of meat mutagens and risk of prostate cancer in a cohort of U.S. health professionals. Cancer Epidemiol Biomarkers Prev. 2015;24(10):1557–63. [PMC free article: PMC4592459] [PubMed: 26224797] [CrossRef]
  • Rothman N, Bhatnagar VK, Hayes RB, Zenser TV, Kashyap SK, Butler MA, et al. The impact of interindividual variation in NAT2 activity on benzidine urinary metabolites and urothelial DNA adducts in exposed workers. Proc Natl Acad Sci USA. 1996;93(10):5084–9. [PMC free article: PMC39410] [PubMed: 8643532] [CrossRef]
  • Rothman N, Poirier MC, Baser ME, Hansen JA, Gentile C, Bowman ED, et al. Formation of polycyclic aromatic hydrocarbon-DNA adducts in peripheral white blood cells during consumption of charcoal-broiled beef. Carcinogenesis. 1990;11(7):1241–3. [PubMed: 2372884] [CrossRef]
  • Rothman N, Poirier MC, Haas RA, Correa-Villasenor A, Ford P, Hansen JA, et al. Association of PAH-DNA adducts in peripheral white blood cells with dietary exposure to polyaromatic hydrocarbons. Environ Health Perspect. 1993;99:265–7. [PMC free article: PMC1567054] [PubMed: 8319640] [CrossRef]
  • Rundle A, Tang D, Hibshoosh H, Estabrook A, Schnabel F, Cao W, et al. The relationship between genetic damage from polycyclic aromatic hydrocarbons in breast tissue and breast cancer. Carcinogenesis. 2000;21(7):1281–9. [PubMed: 10874004] [CrossRef]
  • Samraj AN, Läubli H, Varki N, Varki A. Involvement of a non-human sialic Acid in human cancer. Front Oncol. 2014;4:33. [PMC free article: PMC3928833] [PubMed: 24600589]
  • Samraj AN, Pearce OM, Läubli H, Crittenden AN, Bergfeld AK, Banda K, et al. A red meat-derived glycan promotes inflammation and cancer progression. Proc Natl Acad Sci USA. 2015;112(2):542–7. [PMC free article: PMC4299224] [PubMed: 25548184] [CrossRef]
  • Sander A, Linseisen J, Rohrmann S. Intake of heterocyclic aromatic amines and the risk of prostate cancer in the EPIC-Heidelberg cohort. Cancer Causes Control. 2011;22(1):109–14. [PubMed: 21103922] [CrossRef]
  • Santarelli RL, Vendeuvre JL, Naud N, Taché S, Guéraud F, Viau M, et al. Meat processing and colon carcinogenesis: cooked, nitrite-treated, and oxidized high-heme cured meat promotes mucin-depleted foci in rats. Cancer Prev Res (Phila). 2010;3(7):852–64. [PMC free article: PMC2931773] [PubMed: 20530708] [CrossRef]
  • Santé-Lhoutellier V, Astruc T, Marinova P, Greve E, Gatellier P. Effect of meat cooking on physicochemical state and in vitro digestibility of myofibrillar proteins. J Agric Food Chem. 2008;56(4):1488–94. [PubMed: 18237130] [CrossRef]
  • Santarelli RL, Naud N, Taché S, Guéraud F, Vendeuvre JL, Zhou L, et al. Calcium inhibits promotion by hot dog of 1,2-dimethylhydrazine-induced mucin-depleted foci in rat colon. Int J Cancer. 2013;133(11):2533–41. [PMC free article: PMC3788046] [PubMed: 23712585]
  • Santé-Lhoutellier V, Aubry L, Gatellier P. Effect of oxidation on in vitro digestibility of skeletal muscle myofibrillar proteins. J Agric Food Chem. 2007;55(13):5343–8. [PubMed: 17530859] [CrossRef]
  • Schepens MA, Vink C, Schonewille AJ, Dijkstra G, van der Meer R, Bovee-Oudenhoven IM. Dietary heme adversely affects experimental colitis in rats, despite heat-shock protein induction. Nutrition. 2011;27(5):590–7. [PubMed: 20705428] [CrossRef]
  • Schulze MB, Hoffmann K, Manson JE, Willett WC, Meigs JB, Weikert C, et al. Dietary pattern, inflammation, and incidence of type 2 diabetes in women. Am J Clin Nutr. 2005;82(3):675–84, quiz 714–5. [PMC free article: PMC2563043] [PubMed: 16155283] [CrossRef]
  • Schut HA, Snyderwine EG. DNA adducts of heterocyclic amine food mutagens: implications for mutagenesis and carcinogenesis. Carcinogenesis. 1999;20(3):353–68. [PubMed: 10190547] [CrossRef]
  • Sesink AL, Termont DS, Kleibeuker JH, Van der Meer R. Red meat and colon cancer: the cytotoxic and hyperproliferative effects of dietary heme. Cancer Res. 1999;59(22):5704–9. [PubMed: 10582688]
  • Sesink AL, Termont DS, Kleibeuker JH, Van Der Meer R. Red meat and colon cancer: dietary haem, but not fat, has cytotoxic and hyperproliferative effects on rat colonic epithelium. Carcinogenesis. 2000;21(10):1909–15. [PubMed: 11023550] [CrossRef]
  • Sesink AL, Termont DS, Kleibeuker JH, Van der Meer R. Red meat and colon cancer: dietary haem-induced colonic cytotoxicity and epithelial hyperproliferation are inhibited by calcium. Carcinogenesis. 2001;22(10):1653–9. [PubMed: 11577005] [CrossRef]
  • Shaughnessy DT, Gangarosa LM, Schliebe B, Umbach DM, Xu Z, MacIntosh B, et al. Inhibition of fried meat-induced colorectal DNA damage and altered systemic genotoxicity in humans by crucifera, chlorophyllin, and yogurt. PLoS One. 2011;6(4):e18707. [PMC free article: PMC3081825] [PubMed: 21541030] [CrossRef]
  • Shin A, Shrubsole MJ, Rice JM, Cai Q, Doll MA, Long J, et al. Meat intake, heterocyclic amine exposure, and metabolizing enzyme polymorphisms in relation to colorectal polyp risk. Cancer Epidemiol Biomarkers Prev. 2008;17(2):320–9. [PMC free article: PMC2572782] [PubMed: 18268115] [CrossRef]
  • Silvester KR, Cummings JH. Does digestibility of meat protein help explain large bowel cancer risk? Nutr Cancer. 1995;24(3):279–88. [PubMed: 8610047] [CrossRef]
  • Sinha R, Rothman N, Brown ED, Salmon CP, Knize MG, Swanson CA, et al. High concentrations of the carcinogen 2-amino-1-methyl-6-phenylimidazo- [4,5-b]pyridine (PhIP) occur in chicken but are dependent on the cooking method. Cancer Res. 1995;55(20):4516–9. [PubMed: 7553619]
  • Sinha R, Knize MG, Salmon CP, Brown ED, Rhodes D, Felton JS, et al. Heterocyclic amine content of pork products cooked by different methods and to varying degrees of doneness. Food Chem Toxicol. 1998;36(4):289–97. a. [PubMed: 9651045] [CrossRef]
  • Sinha R, Rothman N, Salmon CP, Knize MG, Brown ED, Swanson CA, et al. Heterocyclic amine content in beef cooked by different methods to varying degrees of doneness and gravy made from meat drippings. Food Chem Toxicol. 1998;36(4):279–87. b. [PubMed: 9651044] [CrossRef]
  • Sinha R. An epidemiologic approach to studying heterocyclic amines. Mutat Res. 2002;506-507:197–204. [PubMed: 12351159] [CrossRef]
  • Sinha R, Kulldorff M, Gunter MJ, Strickland P, Rothman N. Dietary benzo[a]pyrene intake and risk of colorectal adenoma. Cancer Epidemiol Biomarkers Prev. 2005;14(8):2030–4. a. [PubMed: 16103456] [CrossRef]
  • Sinha R, Peters U, Cross AJ, Kulldorff M, Weissfeld JL, Pinsky PF, et al. Meat, meat cooking methods and preservation, and risk for colorectal adenoma. Cancer Res. 2005;65(17):8034–41. b. [PubMed: 16140978] [CrossRef]
  • Sinha R, Cross A, Curtin J, Zimmerman T, McNutt S, Risch A, et al. Development of a food frequency questionnaire module and databases for compounds in cooked and processed meats. Mol Nutr Food Res. 2005;49(7):648–55. c. [PubMed: 15986387] [CrossRef]
  • Sinha R, Park Y, Graubard BI, Leitzmann MF, Hollenbeck A, Schatzkin A, et al. Meat and meat-related compounds and risk of prostate cancer in a large prospective cohort study in the United States. Am J Epidemiol. 2009;170(9):1165–77. [PMC free article: PMC2781742] [PubMed: 19808637] [CrossRef]
  • Skibsted LH. Nitric oxide and quality and safety of muscle based foods. Nitric Oxide. 2011;24(4):176–83. [PubMed: 21605822] [CrossRef]
  • Smith MT, Guyton KZ, Gibbons CF, Fritz JM, Portier CJ, Rusyn I, et al. Key characteristics of carcinogens as a basis for organizing data on mechanisms of carcinogenesis. Environ Health Perspect. 2016;124(6):713–21. [PMC free article: PMC4892922] [PubMed: 26600562]
  • Sørensen M, Autrup H, Olsen A, Tjønneland A, Overvad K, Raaschou-Nielsen O. Prospective study of NAT1 and NAT2 polymorphisms, tobacco smoking and meat consumption and risk of colorectal cancer. Cancer Lett. 2008;266(2):186–93. [PubMed: 18372103] [CrossRef]
  • Spehlmann ME, Begun AZ, Saroglou E, Hinrichs F, Tiemann U, Raedler A, et al. Risk factors in German twins with inflammatory bowel disease: results of a questionnaire-based survey. J Crohn’s Colitis. 2012;6(1):29–42. [PubMed: 22261525] [CrossRef]
  • Stavric B. Biological significance of trace levels of mutagenic heterocyclic aromatic amines in human diet: a critical review. Food Chem Toxicol. 1994;32(10):977–94. [PubMed: 7959450] [CrossRef]
  • Steck SE, Gaudet MM, Eng SM, Britton JA, Teitelbaum SL, Neugut AI, et al. Cooked meat and risk of breast cancer–lifetime versus recent dietary intake. Epidemiology. 2007;18(3):373–82. [PubMed: 17435448] [CrossRef]
  • Steffen A, Bergmann MM, Sánchez MJ, Chirlaque MD, Jakszyn P, Amiano P, et al. Meat and heme iron intake and risk of squamous cell carcinoma of the upper aero-digestive tract in the European Prospective Investigation into Cancer and Nutrition (EPIC). Cancer Epidemiol Biomarkers Prev. 2012;21(12):2138–48. [PMC free article: PMC3519922] [PubMed: 23033453] [CrossRef]
  • Stemmermann GN, Nomura AM, Chyou PH, Hankin J. Impact of diet and smoking on risk of developing intestinal metaplasia of the stomach. Dig Dis Sci. 1990;35(4):433–8. [PubMed: 2318088] [CrossRef]
  • Stolzenberg-Solomon RZ, Cross AJ, Silverman DT, Schairer C, Thompson FE, Kipnis V, et al. Meat and meat-mutagen intake and pancreatic cancer risk in the NIH-AARP cohort. Cancer Epidemiol Biomarkers Prev. 2007;16(12):2664–75. [PubMed: 18086772] [CrossRef]
  • Sugimura T, Wakabayashi K, Nakagama H, Nagao M. Heterocyclic amines: Mutagens/carcinogens produced during cooking of meat and fish. Cancer Sci. 2004;95(4):290–9. [PubMed: 15072585] [CrossRef]
  • Sun J, Hu XL, Le GW, Shi YH. Lactobacilli prevent hydroxy radical production and inhibit Escherichia coli and Enterococcus growth in system mimicking colon fermentation. Lett Appl Microbiol. 2010;50(3):264–9. [PubMed: 20059670] [CrossRef]
  • Tabatabaei SM, Heyworth JS, Knuiman MW, Fritschi L. Dietary benzo[a]pyrene intake from meat and the risk of colorectal cancer. Cancer Epidemiol Biomarkers Prev. 2010;19(12):3182–4. [PubMed: 20962298] [CrossRef]
  • Tang D, Liu JJ, Rundle A, Neslund-Dudas C, Savera AT, Bock CH, et al. Grilled meat consumption and PhIP-DNA adducts in prostate carcinogenesis. Cancer Epidemiol Biomarkers Prev. 2007;16(4):803–8. [PMC free article: PMC2132437] [PubMed: 17416774] [CrossRef]
  • Tantamango YM, Knutsen SF, Beeson WL, Fraser G, Sabate J. Foods and food groups associated with the incidence of colorectal polyps: the Adventist Health Study. Nutr Cancer. 2011;63(4):565–72. [PMC free article: PMC3427008] [PubMed: 21547850] [CrossRef]
  • Tarallo S, Pardini B, Mancuso G, Rosa F, Di Gaetano C, Rosina F, et al. MicroRNA expression in relation to different dietary habits: a comparison in stool and plasma samples. Mutagenesis. 2014;29(5):385–91. [PubMed: 25150024] [CrossRef]
  • Tasevska N, Sinha R, Kipnis V, Subar AF, Leitzmann MF, Hollenbeck AR, et al. A prospective study of meat, cooking methods, meat mutagens, heme iron, and lung cancer risks. Am J Clin Nutr. 2009;89(6):1884–94. [PMC free article: PMC2683000] [PubMed: 19369370] [CrossRef]
  • Tasevska N, Cross AJ, Dodd KW, Ziegler RG, Caporaso NE, Sinha R. No effect of meat, meat cooking preferences, meat mutagens or heme iron on lung cancer risk in the prostate, lung, colorectal and ovarian cancer screening trial. Int J Cancer. 2011;128(2):402–11. [PMC free article: PMC2970721] [PubMed: 20232386] [CrossRef]
  • Tjalsma H, Schöller-Guinard M, Lasonder E, Ruers TJ, Willems HL, Swinkels DW. Profiling the humoral immune response in colon cancer patients: diagnostic antigens from Streptococcus bovis. Int J Cancer. 2006;119(9):2127–35. [PubMed: 16841330] [CrossRef]
  • Toden S, Bird AR, Topping DL, Conlon MA. Resistant starch prevents colonic DNA damage induced by high dietary cooked red meat or casein in rats. Cancer Biol Ther. 2006;5(3):267–72. [PubMed: 16410726] [CrossRef]
  • Toden S, Bird AR, Topping DL, Conlon MA. High red meat diets induce greater numbers of colonic DNA double-strand breaks than white meat in rats: attenuation by high-amylose maize starch. Carcinogenesis. 2007;28(11):2355–62. [PubMed: 17916911] [CrossRef]
  • Toden S, Belobrajdic DP, Bird AR, Topping DL, Conlon MA. Effects of dietary beef and chicken with and without high amylose maize starch on blood malondialdehyde, interleukins, IGF-I, insulin, leptin, MMP-2, and TIMP-2 concentrations in rats. Nutr Cancer. 2010;62(4):454–65. [PubMed: 20432166] [CrossRef]
  • Tong M, Neusner A, Longato L, Lawton M, Wands JR, de la Monte SM. Nitrosamine exposure causes insulin resistance diseases: relevance to type 2 diabetes mellitus, non-alcoholic steatohepatitis, and Alzheimer’s disease. J Alzheimers Dis. 2009;17(4):827–44. [PMC free article: PMC2952429] [PubMed: 20387270]
  • Totsuka Y, Fukutome K, Takahashi M, Takahashi S, Tada A, Sugimura T, et al. Presence of N2-(deoxyguanosin-8-yl)-2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline (dG-C8-MeIQx) in human tissues. Carcinogenesis. 1996;17(5):1029–34. [PubMed: 8640908] [CrossRef]
  • Turesky RJ, Vouros P. Formation and analysis of heterocyclic aromatic amine-DNA adducts in vitro and in vivo. J Chromatogr B Analyt Technol Biomed Life Sci. 2004;802(1):155–66. [PubMed: 15036007] [CrossRef]
  • Turesky RJ. Formation and biochemistry of carcinogenic heterocyclic aromatic amines in cooked meats. Toxicol Lett. 2007;168(3):219–27. [PubMed: 17174486] [CrossRef]
  • Turesky RJ, Le Marchand L. Metabolism and biomarkers of heterocyclic aromatic amines in molecular epidemiology studies: lessons learned from aromatic amines. Chem Res Toxicol. 2011;24(8):1169–214. [PMC free article: PMC3156293] [PubMed: 21688801] [CrossRef]
  • Turner F, Smith G, Sachse C, Lightfoot T, Garner RC, Wolf CR, et al. Vegetable, fruit and meat consumption and potential risk modifying genes in relation to colorectal cancer. Int J Cancer. 2004;112(2):259–64. [PubMed: 15352038] [CrossRef]
  • Turteltaub KW, Dingley KH, Curtis KD, Malfatti MA, Turesky RJ, Garner RC, et al. Macromolecular adduct formation and metabolism of heterocyclic amines in humans and rodents at low doses. Cancer Lett. 1999;143(2):149–55. [PubMed: 10503895] [CrossRef]
  • Ulrich CM, Bigler J, Whitton JA, Bostick R, Fosdick L, Potter JD. Epoxide hydrolase Tyr113His polymorphism is associated with elevated risk of colorectal polyps in the presence of smoking and high meat intake. Cancer Epidemiol Biomarkers Prev. 2001;10(8):875–82. [PubMed: 11489754]
  • Uribarri J, Woodruff S, Goodman S, Cai W, Chen X, Pyzik R, et al. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J Am Diet Assoc. 2010;110(6):911–16.e12, e12. [PMC free article: PMC3704564] [PubMed: 20497781] [CrossRef]
  • Van Hecke T, Vanden Bussche J, Vanhaecke L, Vossen E, Van Camp J, De Smet S. Nitrite curing of chicken, pork, and beef inhibits oxidation but does not affect N-nitroso compound (NOC)-specific DNA adduct formation during in vitro digestion. J Agric Food Chem. 2014;62(8):1980–8. a. [PubMed: 24499368] [CrossRef]
  • Van Hecke T, Vossen E, Vanden Bussche J, Raes K, Vanhaecke L, De Smet S. Fat content and nitrite-curing influence the formation of oxidation products and NOC-specific DNA adducts during in vitro digestion of meat. PLoS One. 2014;9(6):e101122. b. [PMC free article: PMC4076193] [PubMed: 24978825] [CrossRef]
  • Van Hecke T, Vossen E, Hemeryck LY, Vanden Bussche J, Vanhaecke L, De Smet S. Increased oxidative and nitrosative reactions during digestion could contribute to the association between well-done red meat consumption and colorectal cancer. Food Chem. 2015;187:29–36. [PubMed: 25976994] [CrossRef]
  • Vanden Bussche J, Hemeryck LY, Van Hecke T, Kuhnle GG, Pasmans F, Moore SA, et al. O⁶-carboxymethylguanine DNA adduct formation and lipid peroxidation upon in vitro gastrointestinal digestion of haem-rich meat. Mol Nutr Food Res. 2014;58(9):1883–96. [PubMed: 24990219] [CrossRef]
  • Vanhaecke L, Knize MG, Noppe H, De Brabander H, Verstraete W, Van de Wiele T. Intestinal bacteria metabolize the dietary carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine following consumption of a single cooked chicken meal in humans. Food Chem Toxicol. 2008;46(1):140–8. a. [PubMed: 17766021] [CrossRef]
  • Vanhaecke L, Derycke L, Le Curieux F, Lust S, Marzin D, Verstraete W, et al. The microbial PhIP metabolite 7-hydroxy-5-methyl-3-phenyl-6,7,8,9-tetrahydropyrido[3′,2′:4,5]imidazo[1,2-a]pyrimidin-5-ium chloride (PhIP-M1) induces DNA damage, apoptosis and cell cycle arrest towards Caco-2 cells. Toxicol Lett. 2008;178(1):61–9. b. [PubMed: 18375078] [CrossRef]
  • Viscogliosi G, Cipriani E, Liguori ML, Marigliano B, Saliola M, Ettorre E, et al. Mediterranean dietary pattern adherence: associations with prediabetes, metabolic syndrome, and related microinflammation. Metab Syndr Relat Disord. 2013;11(3):210–6. [PMC free article: PMC3696914] [PubMed: 23451814] [CrossRef]
  • Voutsinas J, Wilkens LR, Franke A, Vogt TM, Yokochi LA, Decker R, et al. Heterocyclic amine intake, smoking, cytochrome P450 1A2 and N-acetylation phenotypes, and risk of colorectal adenoma in a multiethnic population. Gut. 2013;62(3):416–22. [PMC free article: PMC4491437] [PubMed: 22628494] [CrossRef]
  • Wakabayashi K, Nagao M, Esumi H, Sugimura T. Food-derived mutagens and carcinogens. Cancer Res. 1992;52(7) Suppl:2092s–8s. [PubMed: 1544146]
  • Wang H, Yamamoto JF, Caberto C, Saltzman B, Decker R, Vogt TM, et al. Genetic variation in the bioactivation pathway for polycyclic hydrocarbons and heterocyclic amines in relation to risk of colorectal neoplasia. Carcinogenesis. 2011;32(2):203–9. [PMC free article: PMC3026844] [PubMed: 21081473] [CrossRef]
  • Welfare MR, Cooper J, Bassendine MF, Daly AK. Relationship between acetylator status, smoking, and diet and colorectal cancer risk in the north-east of England. Carcinogenesis. 1997;18(7):1351–4. [PubMed: 9230278] [CrossRef]
  • Wilkens LR, Kadir MM, Kolonel LN, Nomura AM, Hankin JH. Risk factors for lower urinary tract cancer: the role of total fluid consumption, nitrites and nitrosamines, and selected foods. Cancer Epidemiol Biomarkers Prev. 1996;5(3):161–6. [PubMed: 8833615]
  • Windey K, De Preter V, Verbeke K. Relevance of protein fermentation to gut health. Mol Nutr Food Res. 2012;56(1):184–96. [PubMed: 22121108] [CrossRef]
  • Winter J, Nyskohus L, Young GP, Hu Y, Conlon MA, Bird AR, et al. Inhibition by resistant starch of red meat-induced promutagenic adducts in mouse colon. Cancer Prev Res (Phila). 2011;4(11):1920–8. [PubMed: 21885815] [CrossRef]
  • Winter J, Young GP, Hu Y, Gratz SW, Conlon MA, Le Leu RK. Accumulation of promutagenic DNA adducts in the mouse distal colon after consumption of heme does not induce colonic neoplasms in the western diet model of spontaneous colorectal cancer. Mol Nutr Food Res. 2014;58(3):550–8. [PubMed: 24115497] [CrossRef]
  • Wu K, Giovannucci E, Byrne C, Platz EA, Fuchs C, Willett WC, et al. Meat mutagens and risk of distal colon adenoma in a cohort of U.S. men. Cancer Epidemiol Biomarkers Prev. 2006;15(6):1120–5. [PubMed: 16775169] [CrossRef]
  • Wu K, Sinha R, Holmes MD, Giovannucci E, Willett W, Cho E. Meat mutagens and breast cancer in postmenopausal women–a cohort analysis. Cancer Epidemiol Biomarkers Prev. 2010;19(5):1301–10. [PMC free article: PMC3065926] [PubMed: 20447922] [CrossRef]
  • Yang L, Mutanen M, Cheng Y, Duan R. Effects of red meat and fiber in high fat diet on activities of sphingomyelinase, ceramidase and caspase-3 in rat colonic mucosa. J Nutr Biochem. 2002;13(8):499–504. [PubMed: 12165363] [CrossRef]
  • Yeh CC, Sung FC, Tang R, Chang-Chieh CR, Hsieh LL. Polymorphisms of cytochrome P450 1A2 and N-acetyltransferase genes, meat consumption, and risk of colorectal cancer. Dis Colon Rectum. 2009;52(1):104–11. [PubMed: 19273964] [CrossRef]
  • Zenser TV, Lakshmi VM, Schut HA, Zhou HJ, Josephy P. Activation of aminoimidazole carcinogens by nitrosation: mutagenicity and nucleotide adducts. Mutat Res. 2009;673(2):109–15. [PMC free article: PMC2775548] [PubMed: 19449459] [CrossRef]
  • Zhang L, Zhou J, Wang J, Liang G, Li J, Zhu Y, et al. Absence of association between N-acetyltransferase 2 acetylator status and colorectal cancer susceptibility: based on evidence from 40 studies. PLoS One. 2012;7(3):e32425. [PMC free article: PMC3293792] [PubMed: 22403658] [CrossRef]
  • Zhang ZH, Su PY, Hao JH, Sun YH. The role of preexisting diabetes mellitus on incidence and mortality of endometrial cancer: a meta-analysis of prospective cohort studies. Int J Gynecol Cancer. 2013;23(2):294–303. [PubMed: 23287960] [CrossRef]
  • Zheng W, Wen WQ, Gustafson DR, Gross M, Cerhan JR, Folsom AR. GSTM1 and GSTT1 polymorphisms and postmenopausal breast cancer risk. Breast Cancer Res Treat. 2002;74(1):9–16. [PubMed: 12150456] [CrossRef]
  • Zheng W, Lee SA. Well-done meat intake, heterocyclic amine exposure, and cancer risk. Nutr Cancer. 2009;61(4):437–46. [PMC free article: PMC2769029] [PubMed: 19838915] [CrossRef]
  • Zhong X, Hui C, Xiao-Ling W, Yan L, Na L. NAT2 polymorphism and gastric cancer susceptibility: a meta-analysis. Arch Med Res. 2010;41(4):275–80. [PubMed: 20637371] [CrossRef]
  • Zhou L, Anwar MM, Zahid M, Shostrom V, Mirvish SS. Urinary excretion of N-nitroso compounds in rats fed sodium nitrite and/or hot dogs. Chem Res Toxicol. 2014;27(10):1669–74. [PMC free article: PMC4203384] [PubMed: 25183213] [CrossRef]
  • Zhu J, Chang P, Bondy ML, Sahin AA, Singletary SE, Takahashi S, et al. Detection of 2-amino-1-methyl-6-phenylimidazo[4,5-b]-pyridine-DNA adducts in normal breast tissues and risk of breast cancer. Cancer Epidemiol Biomarkers Prev. 2003;12(9):830–7. [PubMed: 14504191]
  • Zhu Y, Wang PP, Zhao J, Green R, Sun Z, Roebothan B, et al. Dietary N-nitroso compounds and risk of colorectal cancer: a case-control study in Newfoundland and Labrador and Ontario, Canada. Br J Nutr. 2014;111(6):1109–17. [PMC free article: PMC4339287] [PubMed: 24160559] [CrossRef]
  • zur Hausen H. Red meat consumption and cancer: reasons to suspect involvement of bovine infectious factors in colorectal cancer. Int J Cancer. 2012;130(11):2475–83. [PubMed: 22212999] [CrossRef]
  • zur Hausen H, de Villiers EM. Dairy cattle serum and milk factors contributing to the risk of colon and breast cancers. Int J Cancer. 2015;137(4):959–67. [PubMed: 25648405] [CrossRef]
© International Agency for Research on Cancer, 2018. For more information contact publications@iarc.fr.
Bookshelf ID: NBK507976

Views

  • PubReader
  • Print View
  • Cite this Page
  • PDF version of this title (7.0M)

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...