Open access peer-reviewed chapter

Body Mass Index and Colorectal Cancer

Written By

Nuri Faruk Aykan, Mehmet Artac and Tahsin Özatli

Submitted: 07 March 2018 Reviewed: 11 May 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.78617

From the Edited Volume

Body-mass Index and Health

Edited by Ayşe Emel Önal

Chapter metrics overview

1,433 Chapter Downloads

View Full Metrics

Abstract

Colorectal cancer (CRC) is one of the most common cancers in the world. Obesity is an established risk factor for colorectal carcinogenesis. Many epidemiological and experimental studies support this link and tumor-promoting effects of obesity. Body mass index (BMI) is a marker of general obesity. Obesity is also a global health problem and is defined by World Health Organization as BMI > 30 kg/m2. In this chapter, we give a general review about the mechanisms of obesity on colorectal carcinogenesis and the effects of obesity on clinical outcomes such as disease-free survival (DFS), progression-free survival (PFS) and overall survival (OS), in adjuvant setting and metastatic disease, respectively.

Keywords

  • colorectal cancer
  • body mass index
  • obesity
  • carcinogenesis

1. Introduction

CRC is third most common cancer in men with age-standardized rate (ASR) 20.6 per 100,000 and second in women (ASR 14.3 per 100,000) in the world [1]. Approximately 1.4 million new cases and 693,900 deaths occurred in 2012. Wide geographical variation in its incidence is observed; the highest incidence is in Australia/New Zealand, the lowest is in Western Africa. Our country, Turkey takes place between them with ASR of 16.6 per 100,000 for both sexes. Environmental factors especially unhealthy lifestyle may increase the burden of CRC and important number of cases can be preventable by changing this lifestyle [2, 3, 4, 5].

Obesity which is characterized by an excess of body fat is an established risk factor for colorectal carcinogenesis [6, 7, 8, 9, 10, 11, 12]. It is defined by World Health Organization (WHO) as BMI > 30 kg/m2 [13]. BMI is the most widely used metric of adiposity in adults and classified by WHO in 1995, 2000 and 2004 (Table 1). Obesity is a global health problem and its prevalence has increased worldwide in all age groups [14, 15]. Overweight individuals account for approximately 30% of the global population (1.9 billion), and more than 650 million people are classified as obese in 2016 [13]. In a large prospective mortality cohort study in the United States, 20% of all cancer deaths in women and 14% in men could be attributed to obesity in 2003 [16]. World Cancer Research Fund (WCRF), American Institute for Cancer Research (AICR) and US National Cancer Institute (NCI) are classified CRC, cancers of kidney, esophagus (adenocarcinoma), gastric cardia, gallbladder, liver, pancreas, thyroid for both sexes and postmenopausal breast cancer, endometrial and ovarian cancers in women as obesity-related cancers [17, 18]. Cohort studies and meta-analyses strengthened the evidence and demonstrate that obesity increases the risk of colon cancer up to 33% as compared to the risk of anybody with a normal BMI [8, 9, 15, 19, 20, 21] (Table 2).

ClassificationBMI (kg/m2)
Underweight<18.50
Normal range18.50–24.99
Overweight≥25.00
Pre-obese25.00–29.99
Obese≥30.00
Obese Class I30.00–34.99
Obese Class II35.00–39.99
Obese Class III (severely obese)≥40.00

Table 1.

The international classification of BMI (Source: Modified from WHO and AICR).

Cancer siteBergström et al. [19]Renehan et al. [21]AICR 2014
RRRelative risk (RR)% link to excess body fat
MenP valueWomenP valueMenWomen
OvarianNR1.03NS5%
Breast (post-menopausal)1.251.12<0.000117%
Endometrium2.521.59<0.000150%
Kidney1.841.24<0.00011.34<0.000120%28%
Gallbladder1.781.09NS1.590.0411%28%
Esophageal adenocarcinomaNR1.52<0.00011.51<0.000132%38%
PancreasNR1.07NS1.120.0117%20%
CRCColon: 1.33Colon: 1.24
Rectum: 1.09
<0.0001
<0.0001
Colon: 1.09
Rectum: 1.02
<0.0001
NS
17%15%
ThyroidNR1.330.021.140.001NRNR

Table 2.

Obesity-related cancers [17, 19, 21].

NS: not significant, NR: not reported

A recent linear dose-response meta-analysis of four prospective studies showed that each 5 kg increase in adult weight gain was associated with an approximately 6% increased risk of colon cancer (RR = 1.06, 95% CI = 1.03–1.10) [8]. Similarly, another meta-analysis of 30 prospective studies reported an increased risk of colon cancer by each 5-unit increase of BMI in both men (RR = 1.30, 95% CI = 1.25–1.35) and women (RR = 1.12, 95% CI = 1.07–1.18), and this association was stronger in men (p < 0.001) [9]. BMI was positively associated with rectal cancer in men, but not in women. In this study, they showed also that increasing waist circumference (per 10 cm increase) and increasing waist-hip ratio (per 0.1 unit increase) were associated with colon cancer risk in both men and women. This study underlines that the variation of the association between obesity and colon and rectal cancer risk is depending on sex and cancer site.

In this chapter, we first overview about the role of obesity in colorectal carcinogenesis and the effect of obesity on clinical outcomes such as disease-free survival (DFS), progression-free survival (PFS) and overall survival (OS), in adjuvant setting and metastatic disease, respectively.

Advertisement

2. The role of obesity in colorectal carcinogenesis

Obesity is a result of energy imbalance which is characterized by an overall rise in caloric intake and reduced physical activity caused by a sedentary lifestyle, besides genetic predisposition [22, 23]. Increased consumption of high carbohydrate food and beverages and dietary fat support total energy intake and priorities to reduce public obesity should focus on these environmental drivers. High BMI greater than 25 kg/m2 reflects high body fatness [24]. The excess energy is stored as fat [23]. Obesity is associated with hyperplasia of the adipocytes which contain abnormally high free fatty acids (FFA) [25]. The exact pathophysiological mechanisms underlying the association between obesity and the risk of colorectal cancer are not fully understood but accumulated evidences coming from preclinical and clinical studies for the tumor-promoting effects of obesity has begun to illuminate many aspects of this connection [25].

2.1. Hyperinsulinemia and the insulin resistance

Adipose tissue excess or obesity, particularly abdominal obesity is linked to hyperinsulinemia, insulin resistance, hyperglycemia, and to the development of type 2 diabetes [9, 26]. High circulating insulin level is a well-established risk factor for cancer [27, 28, 29]. Epidemiological evidences indicate that high serum C-peptide which is a marker of pancreatic insulin secretion, and type 2 diabetes mellitus are associated with a greater risk of colorectal cancer [30, 31, 32, 33, 34, 35].

Hyperinsulinemia is associated with elevated blood levels of free (unbound, bioavailable) insulin-like growth factor-1 (IGF-1) protein [36, 37]. Obesity-associated insulin resistance increases free IGF-1 levels in the postprandial state, unlike the reduction in insulin-sensitive lean people [38]. Inversely, IGF-binding protein-1 (IGFBP-1) concentrations decrease with increasing adiposity which may cause high concentrations of free IGF-1 [39, 40]. IGF-1 concentrations have been shown to be positively associated with colorectal cancer in prospective studies [30, 41, 42, 43, 44].

Insulin is an anabolic hormone which coordinates glucose to either oxidation to provide energy or storage in the body after uptake in insulin-sensitive cells [45]. Insulin receptors (IRs) exist in IR-A, IR-B and IR-related receptor (IRR) isoforms and insulin has been shown also to have tumorigenic effects on preneoplastic cells which have insulin receptors [46, 47]. IRs and IGF receptors have an extracellular ligand-binding domain and an intracellular protein kinase domain. IGF-1 and insulin-like growth factor-2 (IGF-2) are mitogenic growth factors [46]. They have also metabolic functions. IGF receptors include IGF1R and IGF2R [48]. The binding of insulin and/or IGF ligands to cell surface receptors on cancer cells results in cell proliferation and survival [46, 49]. Upregulation of the IR and IGF1R has been demonstrated in human breast cancer and prostate cancer, respectively [50, 51]. IGF receptors are expressed physiologically in the mucosal and muscular layers of the colon and are overexpressed also in colon cancer cells [52, 53].

There are six high-affinity IGF-binding proteins (from IGFBP1 to IGFBP6); IGFBP1-IGFBP5 have higher affinities for IGF-1, whereas IGFBP6 has a higher affinity for IGF-2 [54]. IGFBPs affect the bioavailability of the IGFs in extracellular fluids [55]. IGFBP/IGF complexes first stabilize IGFs and protect them from degradation, and they inhibit the binding of IGFs to their receptors. Therefore, only released IGFs from IGFBPs by dissociation or protease-mediated cleavage can induce IGF signals [54, 56] (Figure 1).

Figure 1.

Schematic and simplified signaling mechanisms of the “Insulin, IGFs/IR, IGFRs” axis. Insulin and bioavailable IGFs promote carcinogenesis by inhibiting apoptosis and stimulating cell proliferation.

The receptors (IRs, IGFRs) bind the ligands (Insulin, IGF-1, IGF-2) with different affinities (Table 3) [48, 57, 58, 59]. IR-A has a higher IGF-2 affinity than IR-B [59]. IRR is an orphan receptor; its binding ligand is unknown and participates as a heterodimerization partner of the ligand binding family members [60].

ReceptorsAffinity
HighLowVery low
IGF1RIGF-1IGF-2Insulin
IGF2RIGF-2 and other ligands*IGF-1Insulin
IRInsulinIGF-2IGF-1

Table 3.

Insulin, IGF ligands and their receptor affinities [48, 57, 58, 59].

Other ligands such as mannose-6-phosphate.


Insulin/IGFs ligand binding to their receptors activate two major signaling pathways “the phosphatidylinositol 3’-kinase (PI3 kinase)-Akt” and “RAS-RAF-MAPK” [57]. These are classical insulin signaling pathways [61]. In the first pathway, IRs and IGFRs interact with the intracellular insulin receptor substrate 1 (IRS1) after ligand binding and then promotes the PI3K/Akt cascade [62]. This pathway ultimately inhibits apoptosis [63, 64]. In the second pathway, another IRS protein Shc is phosphorylated by IR, and mediate signal transduction through the RAS-MAPK signaling pathway which plays a vital role in cell proliferation [65, 66]. There are several IRS proteins, including IRS1-6, Shc and Gab1 [61]. They are phosphorylated upon IR activation and the tyrosine-phosphorylated IRS sites function by docking with SH2 domain-containing proteins and mediate signal transduction to various downstream factors. Figure 1 illustrates simplified signaling mechanisms of the “Insulin, IGFs/IR, IGFRs” axis. Chronic hyperinsulinemia may reduce the hepatic production of IGFBP resulting in increased level of free IGFs [8, 37]. Finally, insulin and bioavailable IGFs promote carcinogenesis by inhibiting apoptosis and stimulating cell proliferation (mitogenesis) [62, 63, 64].

Activation of the PI3 kinase pathway was also associated pro-invasive phenotype [55]. The role of cell-matrix adhesion molecules (integrins) and cell-cell adhesion molecules (E-cadherin/catenins complex) was investigated in the IGF-1 induced migration. Disruption of the E-cadherin/catenin complex by the activation of IGF1R upon IGF-1 stimulation was shown in human colonic adenocarcinoma cell-line [67].

Cross talks of Insulin/IGF axis to other receptor tyrosine kinase pathways such as epidermal growth factor receptor (EGFR) pathway as well as IGF1R/EGFR heterodimers have also been demonstrated [55, 57]. These interactions may play a critical role in various cellular responses according to the growth factors, the ratio of the receptors and the microenvironment. Regarding mutations, when the mutation threshold in colon cancer was investigated, lower Kras mutations were detected in patients with high BMI [68]. But, in another study that evaluated associations of anthropometric factors with Kras and Braf mutation status in primary CRC, high BMI was found to be associated with the risk of Kras-mutated tumor in men, but not in women [69].

As a conclusion, obesity-related insulin/IGFs signaling pathways may result in evading apoptosis (or resisting cell death), sustaining proliferative signaling and activating invasion and metastasis, which are three of the ten hallmarks of cancer [70, 71].

2.2. Adipocyte-derived factors (leptin, adiponectin)

Adipose tissue is not inert storage depot for lipids, but it is an active endocrine organ [72]. It expresses and secretes a variety of bioactive peptides, known as adipokines. Besides adipocytes, adipose tissue contains connective tissue matrix, stromal and vascular cells, nerve tissue, and immune cells [73]. Adipocytes express and secrete specific endocrine hormones such as leptin and adiponectin; many other proteins are derived from the non-adipocyte fraction of adipose tissue [74].

Leptin is an adipocyte-specific hormone; it is a product of ob gene [75]. Leptin regulates food intake and body weight suppressing appetite and promoting metabolism. Obesity is associated with elevated serum levels of leptin and is often associated with leptin resistance [25]. Epidemiological studies show that high leptin levels are associated with an increased risk of colon cancer [76, 77, 78]. Leptin stimulates cell proliferation by probably MAPK phosphorylation and it is a promoter of cyclin D1 [25, 79, 80]. Leptin also promotes angiogenesis by the activation of PI3K and MAPK pathways [79]. Leptin receptors are expressed on normal epithelial and epithelial-derived tumor cells [81]. A study on obese mice that are genetically deficient in leptin receptors showed significant inhibition of colorectal tumor growth [82]. Leptin levels are correlated with high risk of tubular adenoma and the presence of 3 or more polyps [83, 84].

Other adipocyte-specific peptide hormone adiponectin is secreted mainly from visceral fat adipocytes and is inversely correlated with BMI [21]. It is most abundant adipokine and its circulating levels are higher in women than men. In contrast to leptin, adiponectin levels are significantly lower in obese individuals and a prospective study showed that low adiponectin concentrations was related to an increase in colorectal cancer risk [85, 86]. Physiologically, adiponectin enhances insulin sensitivity and glucose uptake [87]. Induction of insulin sensitivity reduces the circulating levels of insulin and IGF-1. Besides this role in glucose metabolism, it stimulates also fatty acid oxidation [88]. Adiponectin has anti-angiogenic properties; it inhibits Vascular Endothelial Growth Factor-A (VEGF-A) [21, 25]. All of these observations support anti-cancer activity of adiponectin.

Adiponectin exerts its activity via two receptors: AdipoR1 and AdipoR2 [25]. Adiponectin inhibits colorectal cancer cell growth probably by downregulating mammalian target of rapamycin (mTOR) by adenosine monophosphate-activated protein kinase (AMPK) phosphorylation [89].

Inducing angiogenesis is another hallmark of cancer [70, 71].

2.3. Chronic inflammation

Chronic low-grade inflammation is a hallmark of obesity [90]. Increased production of lipids exacerbates inflammation. Increase in visceral adipose tissue (VAT) is accompanied by rising pro-inflammatory adipokines [tumor necrosis factor (TNF) alpha and interleukin (IL)-6 as well as leptin] [25, 81, 91, 92]. In contrary, as mentioned above, there is a decrease in the level of adiponectin which is an anti-inflammatory adipokine [91]. TNF-alpha expression is upregulated in parallel with the increase of BMI and this increase of expression of the TNF-alpha gene in white adipose tissue (WAT) establishes a link between inflammation, insulin resistance and hyperglycemia [93, 94]. Local and systemic chronic inflammation favors tumor initiation and progression [25]. One of the best examples of local chronic inflammatory conditions is inflammatory bowel disease which has an increased risk of colon cancer [95]. Free fatty acids (FFAs) coming from adipocytes are potent activators of macrophages that generate pro-inflammatory cytokines including IL-1beta, IL-6, TNF-alpha [96, 97, 98]. In this microenvironment of an obese host, the activation of nuclear factor (NF)-kappa B via Akt induces cell survival and promotes carcinogenesis [25, 55]. TNF-alpha may function in an autocrine and paracrine mode in the local adipose tissue, but a correlation was also shown between circulating TNF-alpha levels and colorectal adenomas [99, 100]. IL-6 stimulates PI3K/Akt pathway after binding IL-6 receptor (IL-6R), this leads to the expression of the cyclin D1 [101]. The action of IL-6 via JAK2/STAT3 signaling pathway may have a role in carcinogenesis by anti-apoptotic and proliferative mechanisms [102, 103]. Another inflammatory mediator is interferon gamma-inducible protein-10 (IP-10), also known as C-X-C motif chemokine-10 which is a chemo-attractant protein secreted by mature human adipocytes, and enhances local inflammation and tissue damage [104]. Like leptin, high levels of IP-10 are associated with the presence of polyps and tubular adenoma [83]. High serum concentrations of IP-10 are shown to be associated with poor prognosis in patients with CRC [105]. Inflammatory biomarker C Reactive Protein (CRP) and IL-6 were found to be correlated with the presence of CRC [106].

Adipose tissue contains high concentrations of CD4+ Th1, CD8+ lymphocytes with B cells and dendritic cells. In addition, high levels of anti-inflammatory Th2 and Treg cells exist [25]. In obese individuals, the net balance shifted to a pro-inflammatory state in tissue [107]. In this oncogenic micro-environment, the adipocytes can be seen to be surrounded by syncytium of phagocytic macrophages which is histopathologically described as crownlike structures (CLSs) [108, 109]. These adipocytes are undergoing necrosis, and subsequently, dead adipocytes are surrounded by phagocytic adipose tissue macrophages (ATMs). Proinflammatory cytokine osteopontin (OPN) plays a role in the recruitment and accumulation of ATMs and the development of insulin resistance [110, 111]. Matrix metalloproteinase 9 (MMP9) is expressed and secreted into the circulation by ATMs, and it contributes to adipogenesis and angiogenesis [109]. Elevated leptin levels are also correlated with production of proangiogenic cytokines, such as VEGF, leading to angiogenesis [112, 113]. In fact, inflammation is a complex biological response to cellular stress [90]. Excess fuel or energy leads to cell stress at the level of organelles with increased reactive oxygen species (ROS) in the mitochondria due to FFA oxidation and the unfolded protein response in the endoplasmic reticulum caused by excess demands for protein synthesis for adipose tissue expansion [23, 114].

Glucose intake has also been shown to promote cancer growth via the inflammatory cascade, the 12-lipoxygenase pathway, in mice fed sucrose-enriched diet [115].

Chronic low-grade inflammation developing in adipose tissue during obesity can be transferred to other tissues such as liver, pancreas, skeletal muscle through the appearance of active inflammatory mediators in the bloodstream [116].

As a conclusion, tumor-promoting inflammation, which is one of the enabling characteristics, can contribute to multiple hallmarks of cancer including proliferative signaling, evading apoptosis, angiogenesis, invasion and metastasis [71]. It also supports mutations. This inflammatory biological state is thought to be associated with increased risk of obesity-related colon cancer [117].

2.4. Fat tissue and fatty acid metabolism

Adipose tissue depots are not metabolically equal [23]. In mammals, there are two kinds of adipose tissue: white adipose tissue (WAT) and brown adipose tissue (BAT) [116]. WAT has two compartments: visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) [108, 118]. In adult humans, BAT volume is small, and its deposits are essentially cervical supra-clavicular, supra-adrenal and para-spinal regions [119, 120]. BAT promotes energy expenditure by triggering thermogenesis and suppressing diet-induced weight gain [116, 121]. Decreasing BAT activity in mice was shown to be stimulating hyperglycemia, rising plasma triglyceride levels and insulin resistance [122]. In humans also, BAT activity was found to be inversely related to BMI [123]. TNF-alpha has been shown to induce brown adipocyte apoptosis, and VAT inflammation may be linked to the lower BAT volume [124]. Visceral fat compared to subcutaneous fat is associated with higher insulin resistance [108, 118]. The maximum adipocyte size is lower than in visceral (omental) fat as compared with subcutaneous fat [125]. Therefore, visceral adipocytes are less able to reach greater sizes before death after simple rupture under the increased pressures of their intra-abdominal environment [109]. The rate of death of overproduced fat cells is not increased which is approximately 8–9% per year, in obese individuals [109]. These findings support increased CLSs and necrotic adipocyte death in obese people [109].

BMI is a marker of general obesity, and the markers of abdominal obesity are waist circumference and waist-to-hip ratio [108]. On the other hand, visceral fat area or volume reflects visceral obesity.

Experimental studies on mice suggest that excess fat intake by diet may increase both the number of mammalian intestinal stem cells (ISCs) which are the cell-of-origin for the development of cancer and the proliferation rate of ISCs [126, 127, 128]. Ex vivo treatment of intestinal organoid cultures with fatty acid constituents of the high fat diet may enhance the self-renewal potential of these organoid bodies [127]. High fat diet may also enhance the ability of more differentiated enterocytes (transit-amplifying cells) which are derived from ISCs [127]. These effects appear to be mediated by peroxisome proliferator-activated receptor delta (PPAR-d) activation in colorectal epithelial cells [127, 128]. This activation may induce inflammation-associated colonic carcinogenesis [128].

A recent study suggested a link between obesity and sporadic microsatellite instability (MSI)-high CRC in women [129]. We know that fatty acid synthase (FASN) enzyme is involved in de novo lipogenesis catalyzing the reaction steps in the conversion of acetyl-CoA and malonyl-CoA to long-chain saturated fatty acids [130]. In one study using a large number of samples of CRC, FASN overexpression in CRC was found to be associated with MSI, independent of CpG island methylator phenotype (CIMP) [131]. FASN overexpression is commonly observed in human cancers, including CRC [132, 133, 134]. MSI-high CRC has a deficient mismatch repair system and has been associated with poorly differentiated and mucinous tumors [131, 134, 135, 136]. It is present approximately 15% of CRCs [131]. A possible association between obesity, FASN and MSI in CRC is very interesting. Another study showed that obesity and physical inactivity were associated with elevated risk of MSI-H colon cancer in men, but with non-MSI-H tumors in women [137]. An exact mechanism of these associations awaits further investigations.

Lipid metabolites function as cell signaling molecules directly related to the control of inflammation [90]. One example is sphingolipid metabolite sphingosine-1-phosphate (S1P) which is implicated in adenoma growth and is possibly involved in chronic inflammation and colon cancer via IL-6/STAT3/S1P-receptor 1 positive feedback link [138]. S1P/S1P kinase regulates cyclooxygenase-2 (COX-2), promoting arachidonate cascade which is implicated in colon carcinogenesis [139, 140]. Arachidonic acid (20:4ω6) is a precursor of prostaglandins (PGs) which are key mediators of inflammatory reactions [141]. High level of COX-2 expression is found in cancer cells. PGE2 is a major downstream effector of COX-2, and it inhibits apoptosis, favors invasion, motility and promotes angiogenesis. The efficacy of nonsteroidal anti-inflammatory drugs, especially selective COX-2 inhibitors, was shown in the reduction of colorectal polyps [142].

Feeding high-fat diet activates also the inflammasome and caspase-1 activation which controls adipocyte differentiation and insulin sensitivity [143].

Another new concept is the kynurenine pathway which is implicating in the metabolic syndrome [25]. Most foods contain amino acid tryptophan, and excessive food intake amplifies tryptophan catabolism. There is a clear correlation between plasma levels of tryptophan and its metabolites, leptin and BMI [144]. It seems that the increased tryptophan oxidation is an aspect of fat metabolism. Oxidative enzymes such as indoleamine-2,3-dioxygenase (IDO) and tryptophan-2,3-dioxygenase (TDO) catalyze tryptophan to kynurenine. IDO is activated by interferon-gamma which is the response to immune system stimulation [145]. Kynurenine acts via the Aryl Hydrocarbon Receptor (AHR) to modulate transcription factors and microRNAs. The AHR is known to influence food intake to control body mass [25].

Clustering of clinical findings made up of abdominal obesity (apple-shaped body), hyperglycemia with insulin resistance, dyslipidemia (high triglyceride, low high-density lipoprotein), and hypertension is accepted as metabolic syndrome [23]. It is clear that one of the consequences of metabolic syndrome is the increased risk of carcinogenesis.

Table 4 summarizes obesity-related mechanisms of carcinogenesis.

Growth factorsPathwaysHallmarks of cancer
Insulin, IGFsPI3K-AktEvading apoptosis (resisting cell death)
Insulin, IGFsRAS-RAF-MAPK-Cyclin D1Sustaining proliferative signaling
IGFsDisruption of E-cadherin/cateninActivating invasion and metastasis
LeptinJAK2-STAT3-MAPKSustaining proliferative signaling
Leptin/VEGFPI3K-MAPKInducing angiogenesis
Leptin, TNF-alpha, IL-6Multiple pathwaysTumor promoting inflammation

Table 4.

Obesity-related mechanisms of carcinogenesis.

Advertisement

3. The effect of obesity on clinical outcomes

3.1. In the adjuvant setting

As we mentioned earlier, several studies have established a link between obesity and colon cancer risk, but there is little information about the effects of obesity on clinical outcomes after diagnosis and surgical treatment. In the first publication, Dignam et al. reported a significant increased risk for recurrence and death from colon cancer in very obese patients (BMI ≥35 kg/m2) who receive adjuvant chemotherapy [146]. They investigated the association of BMI with the outcomes of 4288 patients with Dukes B and C colon cancer. This study cohort was from cooperative group clinical trials. They observed 2074 events in this study population and hazard ratio (HR) for risk of recurrence was 1.38 (95% confidence interval [CI] = 1.10–1.73), and HR for risk of mortality was 1.28 (95% CI = 1.04–1.57) in very obese patients compared with normal weight patients.

The Adjuvant Colon Cancer Endpoints database also compared the outcomes in patients (N = 25,291) with stage II and stage III colon cancer receiving adjuvant chemotherapy, and they showed inferior outcomes both for obese as well as underweight patients [147]. Men with class 2 and 3 obesity (BMI ≥ 35.0 kg/m2) had a statistically significant reduction in DFS (HR: 1.16; 95% CI = 1.01–1.33; p = .0297) compared with normal-weight patients. These worse outcomes appeared to be cancer-related.

On the other hand, Cancer and Leukemia Group B (CALGB) 89803 trial investigators did not find any significant associations between an increased risk of recurrence and mortality and BMI in patients with stage III colon cancer [148]. In this study, they observed 369 events in 1053 patients. The difference between studies should be related to statistical power. Another explanation is related to suboptimal adjuvant therapy (calculating dose according to a maximum body surface area of 2.0 m2) in very obese patients in the study by Dignam et al.

3.2. In metastatic disease

In advanced colorectal cancer, there are more trials than in the adjuvant setting evaluating the role of obesity on clinical outcomes. In a pooled analysis from four large, prospective studies, among 6128 patients with metastatic CRC treated with first-line bevacizumab and chemotherapy, patients with the lowest BMI (<25 kg/m2) experienced the lowest median OS [149]. This study is presented at the World Congress on Gastrointestinal Cancer 2015. This observation concerning the relationship between worse outcome and the lowest BMI does not mean that obesity is an advantage in patients with mCRC [150]. Probably, in patients with mCRC with a lower BMI, the effects of cancer-related cachexia may be more deleterious. These patients may have less tolerance to the treatments. According to another explanation, obesity may promote angiogenesis and bevacizumab, which is an anti-VEGF monoclonal antibody and may have a vital role in obese patients.

There are also other trials with contradictory results [151, 152, 153, 154, 155]. In a trial investigating the influence of BMI on outcomes in advanced CRC patients receiving chemotherapy with or without targeted therapy, BMI was shown as an independent prognostic factor for survival in patients receiving chemotherapy (CT), but not in patients receiving CT plus targeted therapy [151]. In patients receiving only CT, median OS was 19.5 months for BMI category >30 kg/m2 versus 8 months for BMI category <18.5 kg/m2 (p = 0.001). In patients receiving CT plus targeted therapy, median OS was 21.4 months for BMI category >30 kg/m2 versus 16.6 months for BMI category <18.5 kg/m2 (p = 0.8). In another trial, the authors speculate that higher circulating levels of VEGF may confer resistance to bevacizumab [152]. In our retrospective trial, we found better time to progression (TTP) in patients who have BMI < 25 kg/m2 compared to patients who have BMI > 25 kg/m2; median TTP was 11.7 months versus 6 months, respectively (p = 0.004) [153]. All patients had been treated with fluoropyrimidine-based combination CT plus bevacizumab. We think that patients with high BMI may require higher dosages. In another similar trial, the investigators compared OS across the BMI groups for CRC, and they found that the OS was shorter for patients who were underweight and overweight compared to normal in the group of patients receiving CT + targeted therapy. There was no difference in OS for CT alone [154]. In a recent multicentric study, we demonstrated that obesity serves as a prognostic factor for mCRC patients who have been treated with bevacizumab-based regimens. In particular, among Kras wild-type left-sided tumor patients with bevacizumab-based regimens, the prognosis could be worse for obese patients than that for non-obese patients [155].

Advertisement

4. Conclusion

High BMI or obesity is an established risk factor in colorectal carcinogenesis. Adipose tissue excess, particularly abdominal obesity is linked to hyperinsulinemia, insulin resistance, hyperglycemia, and to the development of type 2 diabetes which are the components of metabolic syndrome. Chronic low-grade inflammation is a hallmark of obesity, and obesity supports many hallmarks of carcinogenesis. It is clear that one of the consequences of metabolic syndrome is the increased risk of carcinogenesis. Targeting obesity and metabolic syndrome should be beneficial in the prevention and the treatment of colorectal cancer.

Advertisement

Conflict of interest

Authors declare no conflict of interest.

References

  1. 1. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA: a Cancer Journal for Clinicians. 2015;65(2):87-108
  2. 2. Haenszel W, Kurihara M. Studies of Japanese migrants. I. Mortality from cancer and other diseases among Japanese in the United States. Journal of the National Cancer Institute. 1968;40:43-68
  3. 3. Center MM, Jemal A, Ward E. International trends in colorectal cancer incidence rates. Cancer Epidemiology, Biomarkers & Prevention. 2009;18:1688-1694
  4. 4. Durko L, Malecka-Panas E. Lifestyle modifications and colorectal cancer. Current Colorectal Cancer Reports. 2014;10:45-54
  5. 5. Keum N, Giovannucci EL. Epidemiology of colorectal cancer. In: Pathology and Epidemiology of Cancer. Switzerland: Springer; 2017. pp. 391-407
  6. 6. World Cancer Research Fund. Continuous update Project Report: Diet, Nutrition, Physical Activity and Colorectal Cancer: WCRF; 2017. wcrf.org/colorectal-cancer-2017
  7. 7. Lauby-Secretan B, Scoccianti C, Loomis D, Grosse Y, Bianchini F, Straif K. Body fatness and cancer—Viewpoint of the IARC working group. The New England Journal of Medicine. 2016;375:794-798
  8. 8. Keum N, Greenwood DC, Lee DH, et al. Adult weight gain and adiposity-related cancers: A dose-response meta-analysis of prospective observational studies. Journal of the National Cancer Institute. 2015;107:2
  9. 9. Larsson SC, Wolk A. Obesity and colon and rectal cancer risk: A meta-analysis of prospective studies. The American Journal of Clinical Nutrition. 2007;86:556-565
  10. 10. Thygesen LC, Thygesen LC, Grønbaek M, et al. Prospective weight change and colon cancer risk in male US health professionals. International Journal of Cancer. 2008;123:1160-1165
  11. 11. Laake I, Thune I, Selmer R, et al. A prospective study of body mass index, weight change, and risk of cancer in the proximal and distal colon. Cancer Epidemiology Biomarkers & Prevention. 2010;19:1511-1522
  12. 12. Li H, Yang G, Xiang YB, et al. Body weight, fat distribution and colorectal cancer risk: A report from cohort studies of 134,255 Chinese men and women. International Journal of Obesity. 2013;37(6):783-789
  13. 13. WHO. World Health Organisation: Obesity and overweight factsheet. 2018. http://www.who.int/mediacentre/factsheets/fs311/en/ (updated February 2018, Accessed: 14-04-2018)
  14. 14. Wang Y, Beydoun MA. The obesity epidemic in the United States–gender, age, socioeconomic, racial/ethnic, and geographic characteristics: A systematic review and meta-regression analysis. Epidemiologic Reviews. 2007;29:6-28
  15. 15. Renehan AG, Soerjomataram I, Tyson M, et al. Incident cancer burden attributable to excess body mass index in 30 European countries. International Journal of Cancer. 2010;126:692-702
  16. 16. Calle EE, Rodriguez C, Walker-Thurmond K, Thun MJ. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. The New England Journal of Medicine. 2003;348:1625-1638
  17. 17. American Institute for Cancer Research. Updated Estimate on Obesity-Related Cancers. http://www.aicr.org/cancer-research-update/2014/march_19/cru-updated-estimate-on- obesity-related-cancers.html. [Accessed: April 23, 2018]
  18. 18. The U.S. National Cancer Institute. Obesity and Cancer Risk. http://www.cancer.gov/cancertopics/factsheet/Risk/obesity. [Accessed: April 23, 2018]
  19. 19. Bergström A, Pisani P, Tenet V, et al. Overweight as an avoidable cause of cancer in Europe. International Journal of Cancer. 2001;91:421-430
  20. 20. Gilbert CA, Slingerland JM. Cytokines, obesity, and cancer: new insights on mechanisms linking obesity to cancer risk and progression. Annual Review of Medicine. 2013;64:45-57
  21. 21. Renehan AG, Tyson M, Egger M, et al. Body-mass index and incidence of cancer: A systematic review and meta-analysis of prospective observational studies. Lancet. 2008;371:569-578
  22. 22. Swinburn B, Sacks G, Ravussin E. Increased food energy supply is more than sufficient to explain the US epidemic of obesity. The American Journal of Clinical Nutrition. 2009;90:1453-1456
  23. 23. Samson SL, Garber AJ. Metabolic syndrome. Endocrinology and Metabolism Clinics of North America. 2014;43:1-23
  24. 24. Nunez et al. Physical activity, obesity and sedentary behaviour and the risks of colon and rectal cancers in the 45 and up study. BMC Public Health. 2018;18:325
  25. 25. Stone TW, McPherson M, Darlington LG. Obesity and Cancer: Existing and new hypotheses for a causal connection. eBioMedicine. 2018;30:14-28
  26. 26. Kahn BB, Flier JS. Obesity and insulin resistance. The Journal of Clinical Investigation. 2000;106:473-481
  27. 27. Hsing AW, Chua S, Gao YT, et al. Prostate cancer risk and serum levels of insulin and leptin: A population-based study. Journal of the National Cancer Institute. 2001;93:783-789
  28. 28. Goodwin PJ, Ennis M, Pritchard KI, et al. Fasting insulin and outcome in early-stage breast cancer: Results of a prospective cohort study. Journal of Clinical Oncology. 2002;20:42-51
  29. 29. Kim SH, Abbasi F, Reaven GM. Impact of degree of obesity on surrogate estimates of insulin resistance. Diabetes Care. 2004;27:1998-2002
  30. 30. Kaaks R, Toniolo P, Akhmedkhanov A, et al. Serum C-peptide, insulin-like growth factor (IGF)-I, IGF-binding proteins, and colorectal cancer risk in women. Journal of the National Cancer Institute. 2000;92:1592-1600
  31. 31. Wei EK, Ma J, Pollak MN, et al. A prospective study of C-peptide, insulin-like growth factor-I, insulin-like growth factor binding protein-1, and the risk of colorectal cancer in women. Cancer Epidemiology, Biomarkers & Prevention. 2005a;14:850-855
  32. 32. Schoen RE, Tangen CM, Kuller LH, et al. Increased blood glucose and insulin, body size, and incident colorectal cancer. Journal of the National Cancer Institute. 1999;91:1147-1154
  33. 33. Ma J, Giovannucci E, Pollak M, et al. A prospective study of plasma C-peptide and colorectal cancer risk in men. Journal of the National Cancer Institute. 2004;96:546-553
  34. 34. Jenab M, Riboli E, Cleveland RJ, et al. Serum C-peptide, IGFBP-1 and IGFBP-2 and risk of colon and rectal cancers in the European Prospec- tive investigation into Cancer and nutrition. International Journal of Cancer. 2007;121:368-376
  35. 35. Larsson SC, Orsini N, Wolk A. Diabetes mellitus and risk of colorectal cancer: A meta-analysis. Journal of the National Cancer Institute. 2005;97:1679-1687
  36. 36. Frystyk J, Vestbo E, Skjaerbaek C, et al. Free insulin-like growth factors in human obesity. Metabolism. 1995;44:37-44
  37. 37. Nam SY, Lee EJ, Kim KR, et al. Effect of obesity on total and free insulin-like growth factor (IGF)-1, and their relationship to IGF-binding protein (BP)-1, IGFBP-2, IGFBP-3, insulin, and growth hormone. International Journal of Obesity and Related Metabolic Disorders. 1997;21:355-359
  38. 38. Ricart W, Fernández-Real JM. No decrease in free IGF-I with increasing insulin in obesity-related insulin resistance. Obesity Research. 2001;9:631-636
  39. 39. Wolk K, Larsson SC, Vessby B, et al. Metabolic, anthropometric, and nutritional factors as predictors of circulating insulin-like growth factor binding protein-1 levels in middle-aged and elderly men. The Journal of Clinical Endocrinology and Metabolism. 2004;89:1879-1884
  40. 40. Sandhu MS, Dunger DB, Giovannucci EL. Insulin, insulin-like growth factor-I (IGF-I), IGF binding proteins, their biologic interactions, and colorectal cancer. Journal of the National Cancer Institute. 2002;94:972-980
  41. 41. Shimizu N, Nagata C, Shimizu H, et al. Height, weight, and alcohol consumption in relation to the risk of colorectal cancer in Japan: A prospective study. British Journal of Cancer. 2003;88:1038-1043
  42. 42. Ma J, Pollak MN, Giovannucci E, et al. Prospective study of colorectal cancer risk in men and plasma levels of insulin-like growth factor (IGF)-I and IGF-binding protein-3. Journal of the National Cancer Institute. 1999;91, 5:620
  43. 43. Giovannucci E, Pollak MN, Platz EA, et al. A prospective study of plasma insulin-like growth factor-1 and binding protein-3 and risk of colorectal neoplasia in women. Cancer Epidemiology, Biomarkers & Prevention. 2000;9:345-349
  44. 44. Palmqvist R, Hallmans G, Rinaldi S, et al. Plasma insulin-like growth factor 1, insulin-like growth factor binding protein 3, and risk of colorectal cancer: A prospective study in northern Sweden. Gut. 2002;50:642-646
  45. 45. Dev R, Bruera E, Dalal S. Insulin resistance and body composition in cancer patients. Annals of Oncology. 2018;29(suppl_2):ii18-ii26
  46. 46. Sipos F, Székely H, Kis ID, et al. Relation of the IGF/IGF1R system to autophagy in colitis and colorectal cancer. World Journal of Gastroenterology. 2017;23(46):8109-8119
  47. 47. Calle EE, Kaaks R. Overweight, obesity and cancer: Epidemiological evidence and proposed mechanisms. Nature Reviews. Cancer. 2004;4(8):579-591
  48. 48. Pandini G, Frasca F, Mineo R, Sciacca L, Vigneri R, Belore A. Insulin/insulin-like growth factor I hybrid receptors have different biological characteristics depending on the insulin receptor isoform involved. The Journal of Biological Chemistry. 2002;277:39684-39695
  49. 49. Hagiwara A, Nishiyama M, Ishizaki S. Branched-chain amino acids prevent insulin-induced hepatic tumor cell proliferation by inducing apoptosis through mTORC1 and mTORC2-dependent mechanisms. Journal of Cellular Physiology. 2012;227(5):2097-2105
  50. 50. Papa V, Pezzino V, Costantino A, Belfiore A, Giuffrida D, Frittitta L, et al. Elevated insulin receptor content in human breast cancer. The Journal of Clinical Investigation. 1990;86:1503-1510
  51. 51. Hellawell GO, Turner GD, Davies DR, Poulsom R, Brewster SF, Macaulay VM. Expression of the type 1 insulin-like growth factor receptor is up-regulated in primary prostate cancer and commonly persists in metastatic disease. Cancer Research. 2002;62:2942-2950
  52. 52. Alemán JO, Eusebi LH, Ricciardiello L, Patidar K, Sanyal AJ, Holt PR. Mechanisms of obesity-induced gastrointestinal neoplasia. Gastroenterology. 2014;146:357-373
  53. 53. Ouban A, Muraca P, Yeatman T, Coppola D. Expression and distribution of insulin-like growth factor-1 receptor in human carcinomas. Human Pathology. 2003;34:803-808
  54. 54. Firth SM, Baxter RC. Cellular actions of the insulin-like growth factor binding proteins. Endocrine Reviews. 2002;23:824-854
  55. 55. LeRoith D, Roberts CT Jr. The insulin-like growth factor system and cancer. Cancer Letters. 2003;195:127-137
  56. 56. Hwa V, Oh Y, Rosenfeld RG. The insulin-like growth factor-binding protein (IGFBP) superfamily. Endocrine Reviews. 1999;20:761-787
  57. 57. Massoner P, Ladurner-Rennau M, Eder IE, Klocker H. Insulin-like growth factors and insulin control a multifunctional signalling network of significant importance in cancer. British Journal of Cancer. 2010 Nov 9;103(10):1479-1484
  58. 58. Ghosh P, Dahms NM, Kornfeld S. Mannose 6-phosphate receptors: New twists in the tale. Nature Reviews. Molecular Cell Biology. 2003;4:202-212
  59. 59. Belfiore A, Frasca F, Pandini G, Sciacca L, Vigneri R. Insulin receptor isoforms and insulin receptor/insulin-like growth factor receptor hybrids in physiology and disease. Endocrine Reviews. 2009;30:586-623
  60. 60. Zhang B, Roth RA. The insulin receptor-related receptor. Tissue expression, ligand binding specificity, and signaling capabilities. The Journal of Biological Chemistry. 1992;267:18320-18328
  61. 61. Du Y, Wei T. Inputs and outputs of insulin receptor. Protein & Cell. 2014;5(3):203-213
  62. 62. Myers MG, Grammer TC, Wang LM, Sun XJ, Pierce JH, Blenis J, et al. Insulin receptor substrate-1 mediates phosphatidylinositol 3′-kinase and p70S6k signaling during insulin, insulin-like growth factor-1, and interleukin-4 stimulation. The Journal of Biological Chemistry. 1994;269:28783-28789
  63. 63. Datta SR, Dudek H, Tao X, Masters S, Fu H, Gotoh Y, Greenberg ME. Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell. 1997;91:231-241
  64. 64. Kulik G, Klippel A, Weber MJ. Antiapoptotic signalling by the insulin-like growth factor I receptor, phosphatidylinositol 3-kinase and Akt. Molecular and Cellular Biology. 1997;17:1595-1606
  65. 65. Parrizas M, Saltiel AR, LeRoith D. Insulin-like growth factor 1 inhibits apoptosis using the phosphatidylinositol 3′-kinase and mitogen-activated protein kinase pathways. The Journal of Biological Chemistry. 1997;272:154-161
  66. 66. Menu E, Kooijman R, Van Valckenborgh E, Asosingh K, Bakkus M, Van Camp B, et al. Specific roles for the PI3K and the MEK–ERK pathway in IGF-1-stimulated che- motaxis, VEGF secretion and proliferation of multiple myeloma cells: Study in the 5T33MM model. British Journal of Cancer. 2004;90:1076-1083
  67. 67. André F, Rigot V, Thimonier J, et al. Integrins and E-cadherin cooperate with IGF-I to induce migration of epithelial colonic cells. International Journal of Cancer. 1999;83:497-505
  68. 68. Bordonaro M, Lazarova D. Hypothesis: Obesity is associated with a lower mutation threshold in colon cancer. Journal of Cancer. 2015 Jul 15;6(9):825-831
  69. 69. Brändstedt J, Wangefjord S, Nodin B, Eberhard J, Sundström M, Manjer J, et al. Associations of anthropometric factors with KRAS and BRAF mutation status of primary colorectal cancer in men and women: A cohort study. PLoS One. 2014;9(6):e98964
  70. 70. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000 Jan 7;100(1):57-70
  71. 71. Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell. 2011 Mar 4;144(5):646-674
  72. 72. Kershaw EE, Flier JS. Adipose tissue as an endocrine organ. The Journal of Clinical Endocrinology and Metabolism. 2004 Jun;89(6):2548-2556
  73. 73. Frayn KN, Karpe F, Fielding BA, Macdonald IA, Coppack SW. Integrative physiology of human adipose tissue. International Journal of Obesity and Related Metabolic Disorders. 2003;27:875-888
  74. 74. Fain JN, Madan AK, Hiler ML, Cheema P, Bahouth SW. Comparison of the release of adipokines by adipose tissue, adipose tissue matrix, and adi- pocytes from visceral and subcutaneous abdominal adipose tissues of obese humans. Endocrinology. 2004;145:2273-2282
  75. 75. Halaas JL, Gajiwala KS, Maffei M, Cohen SL, Chait BT, Rabinowitz D, et al. Weight-reducing effects of the plasma protein encoded by the obese gene. Science. 1995;269:543-546
  76. 76. Stattin P, Palmqvist R, Soderberg S, et al. Plasma leptin and colorectal cancer risk: A prospective study in northern Sweden. Oncology Reports. 2003;10:2015-2021
  77. 77. Stattin P, Lukanova A, Biessy C, Söderberg S, Palmqvist R, Kaaks R, et al. Obesity and colon cancer: Does leptin provide a link? International Journal of Cancer. 2004;109:149-152
  78. 78. Tamakoshi K, Toyoshima H, Wakai K, et al. Leptin is associated with an increased female colorectal cancer risk: A nested case-control study in Japan. Oncology. 2005;68:454-461
  79. 79. Hardwick JC, Van Den Brink GR, Offerhaus GJ, Van Deventer SJ, Peppelenbosch MP. Leptin is a growth factor for colonic epithelial cells. Gastroenterology. 2001;121:79-90
  80. 80. Gonzalez RR, Cherfils S, Escobar M, Yoo JH, Carino C, Styer AK, et al. Leptin signaling promotes the growth of mammary tumors and increases the expression of vascular endothelial growth factor (VEGF) and its receptor type two (VEGF-R2). The Journal of Biological Chemistry. 2006;281:26320-26328
  81. 81. Tandon K, Imam M, Ismail BE, Castro F. Body mass index and colon cancer screening: The road ahead. World Journal of Gastroenterology. 2015 Feb 7;21(5):1371-1376. DOI: 10.3748/wjg.v21.i5.1371
  82. 82. Endo H, Hosono K, Uchiyama T, et al. Leptin acts as a growth factor for colorectal tumours at stages subsequent to tumour initiation in murine colon carcinogenesis. Gut. 2011;60:1363-1371 [PMID: 21406387]. DOI: 10.1136/gut.2010.235754
  83. 83. Comstock SS, Hortos K, Kovan B, McCaskey S, Pathak DR, Fenton JI. Adipokines and obesity are associated with colorectal polyps in adult males: A cross-sectional study. PLoS One. 2014;9:e85939 [PMID: 24465801]. DOI: 10.1371/journal.pone.0085939
  84. 84. Chia VM, Newcomb PA, Lampe JW, White E, Mandelson MT, McTiernan A, Potter JD. Leptin concentrations, leptin receptor polymorphisms, and colorectal adenoma risk. Cancer Epidemiology, Biomarkers & Prevention. 2007;16:2697-2703 [PMID: 18086776]. DOI: 10.1158/1055-9965.EPI-07-0467
  85. 85. Arita Y, Kihara S, Ouchi N, Takahashi M, Maeda K, Miyagawa J, et al. Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochemical and Biophysical Research Communications. 1999;257:79-83
  86. 86. Wei EK, Giovannucci E, Fuchs CS, Willett WC, Mantzoros CS. Low plasma adiponectin levels and risk of colorectal cancer in men: A prospective study. Journal of the National Cancer Institute. 2005;97:1688-1694
  87. 87. Berg AH, Combs TP, Du X, Brownlee M, Scherer PE. The adipocyte-secreted protein Acrp30 enhances hepatic insulin action. Nature Medicine. 2001;7:947-953
  88. 88. Yamauchi T, Kamon J, Minokoshi YA, Ito Y, Waki H, Uchida S, et al. Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nature Medicine. 2002;8:1288-1295
  89. 89. Sugiyama M, Takahashi H, Hosono K, Endo H, Kato S, Yoneda K, et al. Adiponectin inhibits colorectal cancer cell growth through the AMPK/mTOR pathway. Int. Journal of Oncology. 2009;34:339-344
  90. 90. Jain R, Austin Pickens C, Fenton JI. The role of the lipidome in obesity-mediated colon cancer risk. The Journal of Nutritional Biochemistry. 2018;59:1-9
  91. 91. Lyon CJ, Law RE, Hsueh WA. Minireview: Adiposity, inflammation, and atherogenesis. Endocrinology. 2003;144:2195-2200 [PMID: 12746274]. DOI: 10.1210/en.2003-0285
  92. 92. Grivennikov S, Karin E, Terzic J, Mucida D, Yu GY, Vallabhapurapu S, et al. IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell. 2009;15:103-113
  93. 93. Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. The Journal of Clinical Investigation. 1995;95:2409-2415
  94. 94. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: Direct role in obesity-linked insulin resistance. Science. 1993;259:87-91
  95. 95. Bernstein CN, Blanchard JF, Kliewer E, Wajda A. Cancer risk in patients with inflammatory bowel disease. Cancer. 2001;91:854-862
  96. 96. Howe LR, Subbaramaiah K, Hudis CA, Dannenberg AJ. Molecular pathways: Adipose inflammation as a mediator of obesity-associated cancer. Clinical Cancer Research. 2013;19:6074-6083
  97. 97. Iyengar NM, Hudis CA, Dannenberg AJ. Obesity and cancer: Local and systemic mechanisms. Annual Review of Medicine. 2015;66:297-309
  98. 98. Morris PG, Zhou XK, Milne GL, Goldstein D, Hawks LC, Dang CT, et al. Increased levels of urinary PGE-M, a biomarker of inflammation, occur in association with obesity, aging, and lung metastases in patients with breast cancer. Cancer Prevention Research. 2013;6:428-436
  99. 99. Mohamed-Ali V, Goodrick S, Rawesh A, Katz DR, Miles JM, Yudkin JS, et al. Subcutaneous adipose tissue releases interleukin-6, but not tumor necrosis factor-α, in vivo 1. The Journal of Clinical Endocrinology and Metabolism. 1997;82:4196-4200
  100. 100. Kim S, Keku TO, Martin C, Galanko J, Woosley JT, Schroeder JC, et al. Circulating levels of inflammatory cytokines and risk of colorectal adenomas. Cancer Research. 2008;68:323-328
  101. 101. Wegiel B, Bjartell A, Culig Z, Persson JL. Interleukin-6 activates PI3K/Akt path- way and regulates cyclin A1 to promote prostate cancer cell survival. International Journal of Cancer. 2008;122:1521-1529
  102. 102. Loffler D, Brocke-Heidrich K, Pfeifer G, et al. Interleukin-6-dependent survival of multiple myeloma cells involves the Stat3-mediated induction of microRNA-21 through a highly conserved enhancer. Blood. 2007;110:1330-1333
  103. 103. Wang Y, van Boxel-Dezaire AH, Cheon H, Yang J, Stark GR, et al. STAT3 acti- vation in response to IL-6 is prolonged by the binding of IL-6 receptor to EGF recep- tor. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:16975-16980
  104. 104. Lee EY, Lee ZH, Song YW. CXCL10 and autoimmune diseases. Autoimmunity Reviews. 2009;8:379-383 [PMID: 19105984]. DOI: 10.1016/j.autrev.2008.12.002
  105. 105. Toiyama Y, Fujikawa H, Kawamura M, Matsushita K, Saigusa S, Tanaka K, Inoue Y, Uchida K, Mohri Y, Kusunoki M. Evaluation of CXCL10 as a novel serum marker for predicting liver metastasis and prognosis in colorectal cancer. International Journal of Oncology. 2012;40:560-566. DOI: 10.3892/ijo.2011.1247]
  106. 106. Il'yasova D, Colbert LH. Harris, et al. circulating levels of inflammatory markers and cancer risk in the health aging and body composition cohort. Cancer Epidemiol. Biomark. Prevention. 2005;14:2413-2418
  107. 107. Lee BC, Lee J. Cellular and molecular players in adipose tissue inflammation in the development of obesity-induced insulin resistance. Biochimica et Biophysica Acta. 2014;1842:446-462
  108. 108. Oda E. The metabolic syndrome as a concept of adipose tissue disease. Hypertension Research. 2008;31:1283-1291
  109. 109. West M. Dead adipocytes and metabolic dysfunction: Recent progress. Current Opinion in Endocrinology, Diabetes, and Obesity. 2009;16:178-182
  110. 110. Nomiyama T, Perez-Tilve D, Ogawa D, et al. Osteopontin mediates obesity- induced adipose tissue macrophage infiltration and insulin resistance in mice. The Journal of Clinical Investigation. 2007;117:2877-2888
  111. 111. Bertola A, Deveaux V, Bonnafous S, et al. Elevated expression of osteopontin may be related to adipose tissue macrophage accumulation and liver steatosis in morbid obesity. Diabetes. 2009;58:125-133
  112. 112. Miyazawa-Hoshimoto S, Takahashi K, Bujo H, Hashimoto N, Saito Y. Elevated serum vascular endothelial growth factor is associated with visceral fat accumulation in human obese subjects. Diabetologia. 2003;46:1483-1488
  113. 113. Birmingham JM, Busik JV, Hansen-Smith FM, Fenton JI. Novel mechanism for obesity- induced colon cancer progression. Carcinogenesis. 2009;30:690-697
  114. 114. Lusis AJ, Attie AD, Reue K. Metabolic syndrome: From epidemiology to systems biology. Nature Reviews. Genetics. 2008;9(11):819-830
  115. 115. Jiang Y, Pan Y, Rhea PR, et al. A sucrose-enriched diet promotes tumorigenesis in mammary gland in part through the 12-lipoxygenase pathway. Cancer Research. 2016;76:24-29
  116. 116. Rogero MM, Calder PC. Obesity, inflammation, toll-like receptor 4 and fatty acids. Nutrients. 2018;10(4)
  117. 117. Ramos-Nino ME. The role of chronic inflammation in obesity-associated cancers. ISRN Oncology. 2013;2013:697521
  118. 118. Fox CS, Massaro JM, Hoffmann U, et al. Abdominal visceral and subcutaneous adipose tissue compartments: Association with metabolic risk factors in the Framingham heart study. Circulation. 2007 Jul 3;116(1):39-48
  119. 119. Cannon B, Nedergaard J. Brown adipose tissue: Function and physiological significance. Physiological Reviews. 2004;84:277-359
  120. 120. Nedergaard J, Bengtsson T, Cannon B. Unexpected evidence for active brown adipose tissue in adult humans. American Journal of Physiology. Endocrinology and Metabolism. 2007;293:E444-E452
  121. 121. Cao L, Choi EY, Liu X, Martin A, Wang C, Xu X, During MJ. White to brown fat phenotypic switch induced by genetic and environmental activation of a hypothalamic-adipocyte axis. Cell Metabolism. 2011;14:324-338
  122. 122. Connolly E, Morrisey RD, Carnie JA. The effect of interscapular brown adipose tissue removal on body-weight and cold response in the mouse. The British Journal of Nutrition. 1982;47:653-658
  123. 123. van Marken Lichtenbelt WD, Vanhommerig JW, Smulders NM, Drossaerts JM, Kemerink GJ, Bouvy ND, Schrauwen P, Teule GJ. Cold-activated brown adipose tissue in healthy men. The New England Journal of Medicine. 2009;360:1500-1508
  124. 124. Nisoli E, Briscini L, Giordano A, Tonello C, Wiesbrock SM, Uysal KT, Cinti S, Carruba MO, Hotamisligil GS. Tumor necrosis factor alpha mediates apoptosis of brown adipocytes and defective brown adipocyte function in obesity. Proceedings of the National Academy of Sciences of the United States of America. 2000;97:8033-8038
  125. 125. Murano I, Barbatelli G, Parisani V, et al. Dead adipocytes, detected as crown-like structures, are prevalent in visceral fat depots of genetically obese mice. Journal of Lipid Research. 2008;49:1562-1568
  126. 126. Hanyuda A, Cao Y, Hamada T, et al. Body mass index and risk of colorectal carcinoma subtypes classified by tumor differentiation status. European Journal of Epidemiology. 2017;32:393-407. DOI: 10.1007/s10654-017-0254-y
  127. 127. Beyaz S, Mana MD, Roper J, et al. High-fat diet enhances stemness and tumorigenicity of intestinal progenitors. Nature. 2016;531(7592):53-58
  128. 128. Wang D, Fu L, Ning W, Guo L, Sun X, Dey SK, Chaturvedi R, Wilson KT, DuBois RN. Peroxisome proliferator-activated receptor d promotes colonic inflammation and tumor growth. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(19):7084-7089
  129. 129. Hoffmeister M, Bläker H, Kloor M, Roth W, Toth C, Herpel E, et al. Body mass index and microsatellite instability in colorectal cancer: A population-based study. Cancer Epidemiology, Biomarkers & Prevention. 2013;22(12):2303-2311. DOI: 10.1158/1055-9965.EPI-13-0239. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24127414
  130. 130. Semenkovich CF. Regulation of fatty acid synthase (FAS). Progress in Lipid Research. 1997;36:43-53
  131. 131. Ogino S, Kawasaki T, Ogawa A, Kirkner GJ, Loda M, Fuchs CS. Fatty acid synthase overexpression in colorectal cancer is associated with microsatellite instability, independent of CpG island methylator phenotype. Human Pathology. 2007;38(6):842-849
  132. 132. Rashid A, Pizer ES, Moga M, et al. Elevated expression of fatty acid synthase and fatty acid synthetic activity in colorectal neoplasia. The American Journal of Pathology. 1997;150:201-208
  133. 133. Visca P, Alo PL, Del Nonno F, et al. Immunohistochemical expression of fatty acid synthase, apoptotic-regulating genes, proliferating factors, and ras protein product in colorectal adenomas, carcinomas, and adjacent nonneoplastic mucosa. Clinical Cancer Research. 1999;5:4111-4118
  134. 134. Ogino S, Brahmandam M, Cantor M, et al. Distinct molecular features of colorectal carcinoma with signet ring cell component and colorectal carcinoma with mucinous component. Modern Pathology. 2006;19:59-68
  135. 135. Ogino S, Odze RD, Kawasaki T, Brahmandam M, Kirkner GJ, Laird PW, Loda M, Fuchs CS. Correlation of pathologic features with CpG island methylator phenotype (CIMP) by quantitative DNA methylation analysis in colorectal carcinoma. The American Journal of Surgical Pathology. 2006;30(9):1175-1183
  136. 136. Haydon AMM, Jass JR. Emerging pathways in colorectal-cancer development. The Lancet Oncology. 2002;3(2):83-88
  137. 137. Slattery ML, Potter JD, Curtin K, et al. Estrogens reduce and withdrawal of estrogens increase risk of microsatellite instability-Positive colon cancer. Cancer Research. 2001;61:126-130
  138. 138. Pyne NJ, Pyne S. Sphingosine 1-phosphate is a missing link between chronic inflammation and colon cancer. Cancer Cell. 2013;23:5-7
  139. 139. Kawamori T, Osta W, Johnson KR, Pettus BJ, Bielawski J, Tanaka T, et al. Sphingosine kinase 1 is up-regulated in colon carcinogenesis. FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology. 2006;20:386-388
  140. 140. Kawamori T, Kaneshiro T, Okumura M, Maalouf S, Uflacker A, Bielawski J, et al. Role for sphingosine kinase 1 in colon carcinogenesis. FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology. 2009;23:405-414
  141. 141. Phinney SD. Metabolism of exogenous and endogenous arachidonic acid in cancer. In: Heber, Kritchevsky, editors. Dietary Fats, Lipids, Hormones, and Tumorigenesis. New York: Plenum Press; 1996. pp. 87-94
  142. 142. Ghosh N, Chaki R, Mandal V, Mandal SC. COX-2 as a target for cancer chemotherapy. Pharmacological Reports. 2010;62:233-244
  143. 143. Stienstra R, Joosten LA, Koenen T, van Tits B, van Diepen JA, van den Berg SA, et al. The inflammasome-mediated caspase-1 activation controls adipocyte differentiation and insulin sensitivity. Cell Metabolism. 2010;12:593-605
  144. 144. Samad N, Yasmin F, Naheed S, Bari AZ, Ayaz MM, Zaman A, et al. Serum levels of leptin, zinc and tryptophan in obese subjects with sleep deficits. Pakistan Journal of Pharmaceutical Sciences. 2017;30:1431-1438
  145. 145. Prendergast GC, Smith C, Thomas S, et al. Indoleamine 2,3-dioxygenase pathways of pathogenic inflammation and immune escape in cancer. Cancer Immunology, Immunotherapy. 2014;63:721-735
  146. 146. Dignam JJ, Polite BN, Yothers G, et al. Body mass index and outcomes in patients who receive adjuvant chemotherapy for colon cancer. Journal of the National Cancer Institute. 2006;98:1647-1654
  147. 147. Sinicrope FA, Foster NR, Yothers G, et al. Body mass index at diagnosis and survival among colon cancer patients enrolled in clinical trials of adjuvant chemotherapy. Cancer. 2013;119:1528-1536
  148. 148. Meyerhardt JA, Niedzwiecki D, Hollis D, et al. Impact of body mass index and weight change after treatment on cancer recurrence and survival in patients with stage III colon cancer: Findings from Cancer and leukemia group B 89803. Journal of Clinical Oncology. 2008;26:4109-4115. DOI: 10.1200/JCO.2007.15.6687
  149. 149. Zafar Y, Hubbard J, Van Cutsem E, et al. Survival Outcomes According to Body Mass Index (BMI): Results from a Pooled Analysis of 5 Observational or Phase IV Studies of Bevacizumab in Metastatic Colorectal Cancer (mCRC). Barcelona: ESMO World Congress on Gastrointestinal Cancer; 2015
  150. 150. Kasi PM, Zafar SY, Grothey A. Is obesity an advantage in patients with colorectal cancer? Expert Review of Gastroenterology & Hepatology. 2015;9:1339-1342. DOI: 10.1586/17474124.2015.1089170
  151. 151. Simkens LH, Koopman M, Mol L, et al. Influence of body mass index on outcome in advanced colorectal cancer patients receiving chemotherapy with or without targeted therapy. European Journal of Cancer. 2011;47:2560-2567
  152. 152. Guiu B, Petit JM, Bonnetain F, et al. Visceral fat area is an independent predictive biomarker of outcome after first-line bevacizumab-based treatment in metastatic colorectal cancer. Gut. 2010;59:341-347
  153. 153. Aykan NF, Yildiz I, Sen F, et al. Effect of increased body mass index (BMI) on time to tumour progression (TTP) in unresectable metastatic colorectal cancer (mCRC) patients treated with bevacizumab-based therapy. Medical Oncology. 2013;30(3):679. DOI: 10.1007/s12032-013-0679-4
  154. 154. Patel GS, Ullah S, Beeke C, et al. Association of BMI with overall survival in patients with mCRC who received chemotherapy versus EGFR and VEGF-targeted therapies. Cancer Medicine. 2015;4(10):1461-1471. DOI: 10.1002/cam4.490
  155. 155. Artac M, Korkmaz L, Coskun HS, et al. Bevacuzimab may be less effective in obese metastatic colorectal Cancer patients. Journal of Gastrointestinal Cancer. 2018 Jan 4. DOI: 10.1007/s12029-017-0047-2

Written By

Nuri Faruk Aykan, Mehmet Artac and Tahsin Özatli

Submitted: 07 March 2018 Reviewed: 11 May 2018 Published: 05 November 2018