Abstract
The rates of obesity and diabetes are increasing worldwide, whereas the age of onset for both obesity and diabetes are decreasing steadily. Obesity and diabetes are associated with multiple factors that contribute to the increased risk of a number of different cancers, including breast cancer. These factors are hyperinsulinemia, elevated IGFs, hyperglycemia, dyslipidemia, adipokines, inflammatory cytokines, and the gut microbiome. In this review, we discuss the current understanding of the complex signaling pathways underlying these multiple factors involved in the obesity/diabetes–breast cancer link, with a focus particularly on the roles of the insulin/IGF system and dyslipidemia in preclinical breast cancer models. We review some of the therapeutic strategies to target these metabolic derangements in cancer. Future research directions and potential therapeutic strategies are also discussed.
Breast cancer is the leading form of cancer in women and the second most common cause of cancer-induced death in the United States and worldwide (1, 2). The World Health Organization statistics show that the obese population, defined as those with a body mass index ≥30 kg/m2, has been growing rapidly worldwide in recent decades (3). In the United States, more than two-thirds of adults are overweight (body mass index ≥25 kg/m2) or obese (4). Obesity is an independent risk factor for a number of diverse cancers including breast cancer. Meta-analyses have reported an ∼30% increased risk of recurrence or death in obese vs normal-weight women diagnosed with breast cancer (5). In the US Cancer and Steroid Hormone study, increasing body size was found to increase the risk of developing premenopausal triple-negative breast cancer (TNBC) by 67% and the risk of premenopausal luminal B breast cancer by 73% compared with women of normal weight (6, 7). Women with diabetes are also at greater risk of developing TNBC, compared with women without diabetes (8). TNBC is a subtype of breast cancer that does not express the estrogen receptor (ER), progesterone receptor, and human epidermal growth factor receptor 2 (HER2). It is the subtype of breast cancer with the worst prognosis, due to the lack of targeted therapy and enhanced metastasis compared with other breast cancer subtypes (5, 9). Luminal B breast cancer is a molecular subtype of breast cancer that is usually ER positive, but has a high grade by histology and a high proliferative index and carries a worse prognosis than ER-positive luminal A breast cancer (10). Type 2 diabetes (T2D) and obesity are also associated with increased risk for postmenopausal breast cancer (11, 12). Obesity and diabetes frequently co-occur in the same individual. Obese individuals frequently have the metabolic syndrome and are therefore at higher risk of developing T2D. Insulin resistance in metabolic organs (skeletal muscle, liver, and adipose tissue) underlies the pathophysiology of the metabolic syndrome and T2D and is frequently observed in obese individuals (13). In this study, we will review the metabolic factors associated with obesity, the metabolic syndrome, and T2D that are potentially involved in breast cancer growth and progression (Fig. 1) (14).
Figure 1.
Potential mechanisms linking obesity/diabetes and cancer. IGFBP, IGF-binding protein; IR, insulin receptor; SHBG, sex hormone–binding globulin. Reproduced with permission from Gallagher EJ, LeRoith D. Obesity and diabetes: the increased risk of cancer and cancer-related mortality. Physiol Rev. 2015;95:727–748 (14).
The Insulin and IGF Family of Ligands and Receptors
Hyperglycemia is the diagnostic hallmark of both type 1 diabetes (T1D) and T2D. Epidemiologic studies have shown a consistent positive correlation between T2D and the risk of developing and dying from certain cancers, including pancreatic cancer, hepatobiliary cancer, endometrial cancer, colorectal cancer, bladder cancer, non-Hodgkin lymphoma, prostate cancer, and breast cancer (11, 15–18). T1D is also associated with an increased risk of certain cancers (16, 19), although the cancers associated with T1D are different from those associated with T2D, suggesting a different mechanism may be involved. Many types of cancer cell take up more glucose than normal nontumor cells. In contrast to normal tissue, cancer cells predominantly rely on aerobic glycolysis rather than mitochondrial oxidative phosphorylation to generate energy, due to their altered metabolism (Warburg effect) (20). The Warburg effect leads to increased glucose uptake by the cancer cells. In vitro cancer cell studies, in vivo animal tumor models, and human studies all show that cancer cells frequently have high glucose uptake; however, in vivo studies suggest that despite the increased uptake of glucose by the tumor, hyperglycemia alone may not increase tumor growth without hyperinsulinemia (21–24). This suggests that although many cancer cells rely on glucose for metabolism, glucose is not the key driver of cancer growth and progression in the setting of obesity, the metabolic syndrome, and diabetes.
In contrast to patients with T1D, who are insulin deficient, individuals with early T2D have hyperinsulinemia secondary to insulin resistance. Elevated levels of circulating insulin or C-peptide (a biomarker of insulin secretion) are strongly associated with breast and colorectal cancer progression, recurrence, and mortality (25, 26). Therefore, hyperinsulinemia, rather than hyperglycemia, in diabetes is suggested to contribute to the increased risk of developing cancer and cancer progression (23). Insulin is a member of the insulin/IGF family, which comprises insulin, IGF-1, IGF-2, IGF-binding proteins (IGFBPs), and their respective receptors (27, 28). Insulin signaling contributes to metabolic signaling pathways, cell survival, and proliferation signaling pathways. The traditional view of insulin and insulin receptor (IR) signaling was that in metabolic tissues, the IR regulates glucose, protein, and lipid metabolism through the phosphatidylinositol 3-kinase (PI3K)/AKT pathway, and in nonmetabolic tissues, it stimulates the RAS/RAF/MAPK kinase/ERK cascade to cause cell proliferation, survival, and migration (29–31). Insulin/IR signaling appears to have an important role in breast cancer. Previous studies have found that breast cancer tissues have significantly higher IR levels (average IR content: 6.15 ± 3.69 ng IR/0.1 mg protein) than normal breast tissues (0.95 ± 0.68 ng IR/0.1 mg protein) (32). Moreover, IR expression in breast cancers is not downregulated in the setting of hyperinsulinemia (33). Therefore, the hyperinsulinemia-IR signaling pathway may play a role in cancer progression, as proposed in breast cancer.
To study the role of hyperinsulinemia in tumor growth and migration in the absence of hyperglycemia, the LeRoith laboratory created a mouse model overexpressing a dominant-negative IGF-1 receptor (IGF-1R) specifically in skeletal muscle [named MKR for the muscle (M) lysine (K) to arginine (R) substitution in the tyrosine kinase motif of the IGF-1R] (34). The male MKR mice exhibit hyperglycemia, hyperinsulinemia, and dyslipidemia. However, the female MKR mice only develop hyperinsulinemia without hyperglycemia or dyslipidemia (34, 35). Therefore, the female MKR mice have served as an animal model for studying the specific effects of hyperinsulinemia on breast cancer development and progression. A number of transgenic and orthotopic breast tumor models were studied in the MKR mice. In each model, tumors grew faster and larger in MKR mice compared with control mice (35, 36). In addition, metastases to the lungs were increased in the MKR mice (36, 37). A β-3 adrenergic receptor agonist (CL-316,243) that reduced circulating insulin levels and a tyrosine kinase inhibitor that inhibited IR and IGF-1R activation reduced the tumor growth in MKR mice (35, 36, 38). Blockade of the PI3K/AKT/mammalian target of rapamycin pathway also attenuated primary tumor growth, which was reduced to the level of the wild-type (WT) mice (39, 40). These results suggested hyperinsulinemia could promote breast cancer growth and migration through the PI3K/AKT signaling pathway. To determine if insulin was acting through the IR or IGF-1R, human cell lines with IR silenced by short hairpin RNA were injected into the MKR mice. A decrease in cancer cell growth and metastasis was found in the MKR mice injected with cells that were transfected with IR short hairpin RNA (37). Furthermore, it was found that silencing the IR suppressed the epithelial-mesenchymal transition (EMT) in cancer cells (37).
Although insulin resistance is associated with impaired IR signaling in metabolic tissues, in tumor cells, there is no evidence that high levels of insulin lead to impaired activation of the IR signaling pathway that promotes cell proliferation (33, 41, 42). Why cancer cells do not demonstrate insulin resistance may in part be explained by the higher relative expression of the IR-A isoform compared with IR-B. The IR has two isoforms, IR-A and IR-B. IR-A lacks 12 amino acids due to the splicing out of exon 11 in the C-terminal of the α-subunit of IR; IR-B contains exon 11. Insulin has slightly higher affinity for IR-A than IR-B, but insulin metabolic signaling through IR-B is more efficient (43). Both IGF-1 and IGF-2 have higher affinity for IR-A than IR-B. Although IGF-1 binding to IR-A is weak, IGF-2 is considered an important ligand for IR-A (44). In normal tissues, IR-A is mainly expressed in fetal tissues, lymphocytes, brain, and spleen, whereas IR-B is predominantly expressed in liver and adipocytes (43). IR-A is frequently overexpressed in various malignant tumors including breast cancer. Higher IR-A/IR-B ratio has been associated with resistance to hormonal therapy in breast cancer (45). Increased high-mobility group A1 (HMGA1) protein, a chromatin-remodeling protein encoded by the HMGA1 oncogene, is a potential cause leading to total IR overexpression in cancer cells (46–49). One of the downstream effects of HMGA1 is to suppress p53, which is a tumor suppressor. p53 suppresses the promoter activity of both IR and IGF-1R (50). Recently, it was reported that discoidin domain receptor 1, which is an IR-A–interacting protein, specifically upregulates IR-A and IGF-1R expression in breast cancer cells (51). Additionally, a number of splicing factors control the IR-A/IR-B ratio. In the hepatocellular carcinoma, upregulation of certain splicing factors (CUGBP1, hnRNPH, hnRNPA1, hnRNPA2B1, and SF2/ASF) occurred in the setting of epidermal growth factor receptor (EGFR) signaling, leading to an increase in the IR-A/IR-B ratio (52). Loss of the splicing factor SRSF3 has also been found to lead to an increase of IR-A and predisposes to murine hepatocellular carcinoma (53). Precisely what splicing factors regulate IR splicing in other tissues and cancers remains to be determined, and whether cancers with higher IR-A/IR-B ratio are more susceptible to the effects of hyperinsulinemia is also unknown.
IGFs are polypeptide hormones that have similar tertiary structure to insulin. IGFs are synthesized in most tissues of the body, although most circulating IGF-1 is made in the liver. The circulating IGF levels are quite stable due to their interactions with IGFBPs, which prevent the degradation of the IGFs (54, 55). GH/GH receptor signaling directly stimulates production of hepatic IGF-1, but has no effect on the expression of IGF-2 (56). Only the free IGFs (unbound to IGFBPs) are biologically available for binding to the IGF-1R (55). As hyperinsulinemia decreases IGFBP-1 and IGFBP-2 levels, insulin may indirectly enhance the IGF–IGF-1R signaling pathway by reducing the expression of certain IGFBPs and increasing bioavailable IGFs. The IR and IGF-1R are transmembrane tyrosine kinase receptors and have well known and common downstream cellular signal pathways, whereas the IGF-2R is the mannose-6-phosphate receptor and has no tyrosine kinase enzymatic activity. Traditionally, IGF-1/IGF-1R signaling has been studied in relation to cancer, though IGF-2 signaling through the IR-A has also been associated with cancer progression (44, 57–59). Some epidemiologic studies have shown that higher IGF-1 levels in the normal population correlate with an increased risk of breast, lung, prostate, and colorectal cancers (58). Furthermore, IGF-1R is overexpressed in several cancers, including liver, colorectal, breast, and prostate. The loss of tumor suppressor genes including BRCA1, p53, and PTEN lead to an increase in IGF-1R expression in tumor (60, 61). In animal studies, exposure to the high levels of IGF-1 was reported to increase tumor growth and metastasis (62, 63). Conversely, both chemically or genetically reducing circulating IGF-1 levels and administration of IGF-1R antibody in cancer cells can reduce tumor growth (61). The IGF-1R signal pathway has been proposed to be responsible for therapeutic resistance in several breast cancer subgroups, although the exact mechanism(s) is still not clear. Signaling through the IGF-1R pathway may play an important role in compensating for therapeutic inhibition of EGFR signaling pathway in breast cancer and may also contribute to breast cancer resistance to chemotherapy and radiotherapy (64). Inhibition of the IGF-1R was found to augment the activity of EGFR in cancer (65). The crosstalk between IGF-1 signaling and ER signaling is considered as a key mechanism of the hormone resistance in ER-positive breast cancer (66, 67). Approximately 20% of human HER2-positive breast cancers express IGF-1R (68). Direct interactions between the IGF-1R and HER2 have been reported and may contribute to the resistance to anti-HER2–targeted therapy (69). In the setting of obesity and diabetes, hyperinsulinemia may therefore directly contribute to tumor growth and progression or indirectly enhance tumorigenesis through IGF-1 signaling.
During the last two decades, many cancer clinical trials have targeted the IGF-1 signaling pathway using various methods in a number of cancers. Unfortunately, in almost all clinical studies, these approaches have had limited success (70, 71). The potential reasons for treatment failure are diverse; however, the possibility that the mitogenic IR pathway compensates for inhibition of the IGF-1 pathway is one of the most striking potential mechanisms. Preclinical studies have supported the hypothesis that silencing the IGF-1R may increase the sensitivity of tumors to signaling through the IR pathway (72, 73). Knockdown or blocking of the IR was reported to inhibit cell proliferation in response to insulin in ER-positive breast cancer cells resistant to endocrine therapies. However, this inhibition was attenuated in the hormone-sensitive ER-positive breast cancer cells, probably due to the IGF-1R/IR hybrid receptors. Although the primary ligand for IGF-1R/IR hybrid receptors is IGF-1, not insulin, insulin can still signal through the IGF-1R/IR hybrid receptors upon IR inhibition (74). Combined targeting of the IR mitogenic pathway and IGF-1R pathway might be valuable therapies for hormone-resistant and TNBC. The other possibilities include the use of monoclonal antibodies toward IGF-1 and IGF-2 ligands simultaneously. Trials using this approach are in fact currently under way (75, 76).
Overall, both IR and IGF-1R are extensively reported to be observed in all breast cancer subtypes (luminal A, luminal B, HER2, and TNBC). The activation of IR and IGF-1R (indicated by phosphorylated IR and IGF-1R) is present in all breast cancer subtypes and is related to poor survival (77). The IR isoforms ratio (IR-A/IR-B) is altered particularly in prognostically unfavorable luminal B cancers and hormone-resistant breast cancers (78). The IGF-1R expression is prognostically favorable in luminal A and B breast cancer but is unfavorable in HER2-positive and TNBC (79, 80). These differences in receptor expression may explain the differences in the sensitivity of certain breast cancers to the direct and indirect tumor-promoting effects of systemic hyperinsulinemia. Additionally, the differences in expression levels may explain why IGF-1R–targeted therapies have had limited success in certain patients. What causes these differences in receptor expression and whether regulation of the expression of the IGF-1R and IR isoforms can be targeted therapeutically remain to be determined.
Dyslipidemia
Dyslipidemia is a feature of the metabolic syndrome in patients with obesity and T2D. Obese patients and patients with diabetes frequently exhibit elevated circulating very-low-density lipoprotein (VLDL) cholesterol, triglycerides, small dense low-density lipoprotein (LDL) cholesterol, and reduced high-density lipoprotein (HDL) cholesterol (81). Elevated total cholesterol, elevated circulating triglycerides, and decreased HDL cholesterol have been associated with an 18%, 15%, and 20% increased risk of cancer, respectively (82). In a recent meta-analysis, it was reported that the dietary cholesterol intake was closely associated with an increased risk of breast cancer occurrence (83). Cholesterol-lowering 3-hydroxy-3-methyl-glutaryl-CoA reductase inhibitors (statin) have proven very effective in lowering circulating cholesterol and are widely used to prevent cardiovascular disease (84). Many studies indicated the link between statin administration and a lower risk of developing specific cancers (85–87). Some studies have suggested that statins reduced recurrence and mortality from prostate cancer and breast cancer (89). However, not all studies have consistently demonstrated these beneficial effects of statins, possibly due to heterogeneous populations or the use of different types of statins (86, 89, 90). Alternatively, the discrepant results on breast cancer outcomes may reflect different breast cancer subtypes having differing sensitivities to circulating cholesterol and/or statin therapy. It is important to consider differences in statins: the type of statin used, when it is used with respect to time of diagnosis of cancer (prediagnosis or postdiagnosis), and duration of treatment. Some statins are lipophilic (simvastatin, lovastatin, and atorvastatin), and others are hydrophilic (pravastatin, rosuvastatin, and fluvastatin) (91). In breast cancer mortality studies, if results were stratified according to statin type, hydrophilic statins had a benefit only when taken postdiagnostically, and lipophilic statins demonstrated survival benefits irrespective of when they were taken. These results suggested cholesterol may not be oncogenic but might promote tumor progression, and lowering circulating cholesterol may improve outcomes (91).
Transgenic animals with dyslipidemia have been used to model and investigate the effect of hyperlipidemia on cancer growth. Several laboratories reported that a high-cholesterol diet promoted tumor growth in mice with ER-positive breast cancer and prostate cancer (92, 93). As mice lack cholesteryl ester transfer protein, and therefore, high-cholesterol diets do not completely reflect human dyslipidemia, genetically modified models of dyslipidemia have also been used (94): the apolipoprotein E knockout (ApoE−/−) mouse has elevated total, VLDL, and LDL cholesterol; the LDL receptor knockout (LDLR−/−) mouse has elevated LDL cholesterol; the transgenic ApoE3 Leiden mouse has elevated VLDL and LDL cholesterol; and the adiponectin knockout mouse has decreased HDL and elevated LDL cholesterol (95–97). Each of these animal models of hypercholesterolemia has been used to study the effects of lipids on breast cancer growth and progression (95). The LeRoith laboratory found that ApoE−/− mice on a high-cholesterol diet had increased growth and metastasis of orthotopic murine ER-negative mammary tumors compared with WT mice (98). Increased Akt phosphorylation has been found in cancer cells in response to cholesterol, and treatment of these mice with a small-molecule inhibitor of PI3K (BKM120) reduced mammary tumor growth, suggesting the PI3K/Akt pathway is partly responsible for the protumorigenic effects of elevated cholesterol (98). LDL receptor (LDLR) is expressed at high levels on certain cancer cells. TNBC cells (MDA-MB-231) have higher levels of expression of the LDLR than ER-positive cells (MCF-7) (99). TNBC and Her2/Neu breast cancer cells with high LDLR expression were found to form larger tumors in ApoE−/− and LDLR−/− mice than in WT mice. Silencing the LDLR in such cancer cells led to decreased tumor growth in both LDLR−/− and ApoE−/− mice (100). Transgenic polyoma virus middle T antigen tumors in adiponectin knockout mice were found to have increased LDLR expression, increased cholesterol content. and accelerated tumor development (101). Similarly, ER-positive breast cancers grew faster in ApoE3-Leiden mice fed a high-fat diet compared with control mice (102).
Several mechanisms potentially contribute to the link between elevated cholesterol and breast cancer growth. As cholesterol is a major component of the plasma membrane, rapidly dividing cells such as cancer cells require large amounts of cholesterol for membrane synthesis; therefore, high levels of circulating cholesterol may provide the substrate for cell proliferation. However, cancer cells may be availing of elevated exogenous cholesterol in other ways. Cholesterol is the precursor of many sex hormones, including progesterone, estrogens, androgens, and their derivatives. The large cohort study in the Women’s Health Initiative indicated the administration of the lipophilic statins independently contributed to a reduction in late-stage breast cancer, especially for the patients with ER-positive breast cancer and androgen receptor–positive breast cancer. The androgen receptor is expressed in some TNBC (103). These results suggest that cholesterol may be used by cancer cells to synthesize sex hormones and thus lead to resistance to systemic hormonal therapies. In addition to the steroid sex hormones, some oxysterol metabolites of cholesterol such as 27-hydroxycholesterol (27HC) and 25-hydroxycholesterol were recently found to play important roles in breast cancer growth (102, 104, 105). 27HC is an endogenous selective ER modulator. It has recently been shown to promote the growth of ER-positive human and murine breast cancers in mice. In functional studies, it was found that 27HC binds to ER to induce a conformational change. The effect of 27HC on the ER is different in various tissues. 27HC works as the ER antagonist in vascular endothelial cells and murine models of cardiovascular disease, whereas it acts as an ER agonist in hepatoma, colorectal, and breast cancer cells. 27HC is also an agonist of liver X receptor (LXR), which induces EMT and subsequent metastasis when activated in breast cancer cells (106). Cholesterol levels are normally tightly controlled by both sterol regulatory element-binding protein-2 and LXR, which suppress cholesterol uptake via the LDLR and increase cholesterol efflux (107, 108). However, in human ER-negative breast cancer cells (MDA-MB-231), no downregulation of the LDLR occurs, and no increase in cholesterol efflux gene transcription is detected. Intracellular cholesterol concentrations are high in this breast cancer cell line (109, 110). The role of LXR in the proliferation of breast cancer is still controversial. Although some studies report that an agonist of LXR promoted both proliferation and lung metastasis of MCF-7 cell xenografts (102). Another group showed that LXR-activated macrophages reduced MCF-7 proliferation and increased apoptosis in vitro (111). Recently, the Nelson laboratory (112) reported that 27HC acts on immune myeloid cells residing at the distal metastatic sites, thus promoting an immune-suppressive environment to facilitate breast cancer metastasis.
In addition to statin treatment, other cholesterol-lowering therapies including the proprotein convertase subtilisin/kexin type 9 inhibitors and inhibitors of 27HC synthesis [sterol 27-hydroxylase (CYP27A1) inhibitors] may also potentially demonstrate benefit in patients with cancer, especially in the statin-resistant or statin-intolerant patients. Studies have not yet been performed with these agents to determine if they will improve responses to therapies, reduce recurrences, or prolong survival in the setting of cancer.
Adipose Tissue
Obese and many patients with T2D have significantly increased total adipose tissue mass compared with the normal-weight population. Large amounts of adipose tissue, including subcutaneous and visceral adipose tissue, surround many organs where cancers, such as the breast cancer, develop (113). Adipose tissue is an important endocrine organ that produces adipokines, inflammatory cytokines, and small amounts of estrogen by aromatization of androgens (114). These adipose tissue factors could affect other organs through both paracrine and endocrine effects. In addition, adipose tissue inflammation may be a favorable environment for the development of cancer. Recently, more studies suggest that adipose tissue factors (cytokines and adipokines) and adipose tissue inflammation also contribute to cancer progression.
Leptin is an important adipokine-regulating appetite and energy balance through its effects on the brain. Many obese individuals acquire leptin resistance and have high levels of circulating leptin (115). Leptin receptor expression has been detected in human breast cancer (116). Leptin binds to the leptin receptor, leading to the activation of several signaling pathways, including Janus kinase/signal transducer and activator of transcription (Stat) signaling, MAPK/ERK, PI3K/Akt, and suppressor of cytokine signaling pathways, promoting cancer cells survival, proliferation, and metastasis (116). A recent study found that enhanced leptin signaling promotes cancer stem cell enrichment and EMT, thus driving obesity-associated TNBC progression in transgenic MMTV-Wnt-1 mice, orthotopic murine E-Wnt and M-Wnt tumors, and human MDA-MB-231 xenografts (117).
Resistin, named for inducing resistance to insulin, is primarily produced by macrophages (118). It mediates insulin resistance by activating the Janus kinase–STAT signaling pathway to activate suppressor of cytokine signaling 3, which binds to endogenous IRS1 and IRS2 and promotes their ubiquitination and subsequent degradation (119). Resistin levels are elevated in patients with both obesity and T2D (120). Some studies suggested the increased resistin in tumor tissues and serum might be a biomarker for the diagnosis of breast cancer (121, 122). Recently, several groups have shown resistin promotes human breast cancer cell growth and metastasis through activation of Stat3 and the ERM family (ezrin, radixin, and moesin) of proteins (123–126).
Adiponectin is considered an anti-inflammatory adipokine. Epidemiological studies have demonstrated that in postmenopausal women with breast cancer, the circulating adiponectin levels are significantly lower than those in women without cancer (127). In vitro studies have reported that adiponectin suppresses the survival and proliferation of T47D, MCF-7, and MDA-MB-231 human breast cancer cells (128). Adiponectin can also inhibit the leptin-mediated migration and invasion of these human breast cancer cells (129, 130). Adiponectin is involved in several signal pathways that may contribute to the inhibition of carcinogenesis. Adiponectin could activate AMP-activated protein kinase, MAPK, and peroxisome proliferator–activated receptor α pathways and block leptin signaling (131–134). Interestingly, as discussed under “Dyslipidemia” in this review, adiponectin deficiency has been found to promote tumor growth, increasing cholesterol content and LDLR expression on breast cancer cells (101).
Gut Microbiome
The human microbiome is a dynamic and functional entity comprising microorganisms including bacteria, viruses, protozoa, and fungi (135). Most of the microbiome is in the gastrointestinal tract and interdependent with the host. The microbiome influences the digestion, metabolism, epithelial homeostasis, local inflammation, cardiovascular, and immune functions of the host (136–139). A study from the Gordon laboratory (140) provided evidence that there are considerable differences in microbiome composition from obese people and lean people. Some studies consecutively exposed the connections between the gut microbiome and obesity. Fasting-induced adipose factor (FIAF), also called angiopoietin-like protein 4, is expressed in the intestine, liver, muscle, and adipose tissue. It is selectively suppressed in the intestinal epithelium. FIAF is an inhibitor of the circulating lipoprotein lipase, which is an enzyme to hydrolyze triglycerides to promote their release from the lipoprotein such as chylomicrons and VLDLs (141, 142). Knocking out FIAF in the obese germ-free mice indicated FIAF is a key regulator on adipose storage for the gut microbiome (143). Another molecular mechanism involved in the microbiome-induced obesogenic progression is lipopolysaccharide (LPS). LPS is a proinflammatory factor existing in the cell wall of gram-negative bacteria. It can be absorbed by the intestine epithelium after the bacteria die. In the intestine, LPS promotes intestinal permeability and induces the production of inflammatory cytokines such as TNF-α and IL-6 (144, 145).
Microbiome perturbations were found to increase the risk of some cancers. The most well-known case is the infection of Helicobacter pylori, which can cause gastric cancer (146). Many studies reported that Fusobacterium nucleatum, which is on the surface of >50% of colorectal adenomas, promotes chronic inflammation in the intestinal tumor microenvironment (147). Studies have also examined links between microbiome alterations and breast cancer. Plottel and Blaser (148) defined a group of human gut organisms as the “estrobolome”: “the aggregate of enteric bacterial genes whose products are capable of metabolizing estrogens.” A high level of the estrobolome could promote the intestinal reabsorption of deconjugated estrogens into the circulation. Epidemiologic studies suggested that some gut microbiome might also affect breast cancer risk through estrogen-independent pathways (149). Another recent study indicated that a bacterial metabolite, lithocholic acid, can limit the proliferation of breast cancer cells both in vitro and in vivo (150). Overall, these studies demonstrate that obesity and diabetes are associated with differences in the intestinal floral composition. Some of these changes may increase cancer risk and development by contributing to inflammation and altering metabolism. Other bacteria may secrete tumor-suppressing factors. Improving our understanding of the composition and characteristics of the microbiome and learning how to manipulate it for our benefit may provide a nontoxic way of improving systemic metabolism and preventing and treating cancer.
Conclusions
As obesity and diabetes affect more and more people, it is critical to understand the mechanisms through which they contribute to the development and progression of specific cancers. It is becoming clear that targeting the cancer with specific therapies but ignoring systemic metabolic dysfunction may contribute to resistance to cancer therapy and treatment failure. As our understanding of the mechanisms tying systemic metabolism to cancer grows, it will help to tailor therapies to specific cancer targets altered by metabolic dysfunction or to identify the patients who will benefit from therapies to treat specific metabolic abnormalities.
Acknowledgments
Financial Support: This work was supported by National Cancer Institute Grants R01CA128799 and R01CA200553 to D.L. and K08CA190770 to E.J.G.
Disclosure Summary: The authors have nothing to disclose.
Glossary
Abbreviations:
- 27HC
27-hydroxycholesterol
- ApoE−/−
apolipoprotein E knockout
- EGFR
epidermal growth factor receptor
- EMT
epithelial-mesenchymal transition
- ER
estrogen receptor
- FIAF
fasting-induced adipose factor
- HER2
human epidermal growth factor receptor 2
- HMGA1
high-mobility group A1
- IGF-1R
IGF-1 receptor
- IGFBP
IGF-binding protein
- IR
insulin receptor
- LDL
low-density lipoprotein
- LDLR
low-density lipoprotein receptor
- LDLR−/−
low-density lipoprotein receptor knockout
- LPS
lipopolysaccharide
- LXR
liver X receptor
- PI3K
phosphatidylinositol 3-kinase
- Stat
signal transducer and activator of transcription
- T1D
type 1 diabetes
- T2D
type 2 diabetes
- TNBC
triple-negative breast cancer
- VLDL
very-low-density lipoprotein
- WT
wild-type
References
- 1. DeSantis CE, Ma J, Goding Sauer A, Newman LA, Jemal A. Breast cancer statistics, 2017, racial disparity in mortality by state. CA Cancer J Clin. 2017;67(6):439–448. [DOI] [PubMed] [Google Scholar]
- 2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67(1):7–30. [DOI] [PubMed] [Google Scholar]
- 3. Afshin A, Forouzanfar MH, Reitsma MB, Sur P, Estep K, Lee A, Marczak L, Mokdad AH, Moradi-Lakeh M, Naghavi M, Salama JS, Vos T, Abate KH, Abbafati C, Ahmed MB, Al-Aly Z, Alkerwi A, Al-Raddadi R, Amare AT, Amberbir A, Amegah AK, Amini E, Amrock SM, Anjana RM, Ärnlöv J, Asayesh H, Banerjee A, Barac A, Baye E, Bennett DA, Beyene AS, Biadgilign S, Biryukov S, Bjertness E, Boneya DJ, Campos-Nonato I, Carrero JJ, Cecilio P, Cercy K, Ciobanu LG, Cornaby L, Damtew SA, Dandona L, Dandona R, Dharmaratne SD, Duncan BB, Eshrati B, Esteghamati A, Feigin VL, Fernandes JC, Fürst T, Gebrehiwot TT, Gold A, Gona PN, Goto A, Habtewold TD, Hadush KT, Hafezi-Nejad N, Hay SI, Horino M, Islami F, Kamal R, Kasaeian A, Katikireddi SV, Kengne AP, Kesavachandran CN, Khader YS, Khang YH, Khubchandani J, Kim D, Kim YJ, Kinfu Y, Kosen S, Ku T, Defo BK, Kumar GA, Larson HJ, Leinsalu M, Liang X, Lim SS, Liu P, Lopez AD, Lozano R, Majeed A, Malekzadeh R, Malta DC, Mazidi M, McAlinden C, McGarvey ST, Mengistu DT, Mensah GA, Mensink GBM, Mezgebe HB, Mirrakhimov EM, Mueller UO, Noubiap JJ, Obermeyer CM, Ogbo FA, Owolabi MO, Patton GC, Pourmalek F, Qorbani M, Rafay A, Rai RK, Ranabhat CL, Reinig N, Safiri S, Salomon JA, Sanabria JR, Santos IS, Sartorius B, Sawhney M, Schmidhuber J, Schutte AE, Schmidt MI, Sepanlou SG, Shamsizadeh M, Sheikhbahaei S, Shin MJ, Shiri R, Shiue I, Roba HS, Silva DAS, Silverberg JI, Singh JA, Stranges S, Swaminathan S, Tabarés-Seisdedos R, Tadese F, Tedla BA, Tegegne BS, Terkawi AS, Thakur JS, Tonelli M, Topor-Madry R, Tyrovolas S, Ukwaja KN, Uthman OA, Vaezghasemi M, Vasankari T, Vlassov VV, Vollset SE, Weiderpass E, Werdecker A, Wesana J, Westerman R, Yano Y, Yonemoto N, Yonga G, Zaidi Z, Zenebe ZM, Zipkin B, Murray CJL; GBD 2015 Obesity Collaborators . Health effects of overweight and obesity in 195 countries over 25 years. N Engl J Med. 2017;377(1):13–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Flegal KM, Carroll MD, Kit BK, Ogden CL. Prevalence of obesity and trends in the distribution of body mass index among US adults, 1999-2010. JAMA. 2012;307(5):491–497. [DOI] [PubMed] [Google Scholar]
- 5. Protani M, Coory M, Martin JH. Effect of obesity on survival of women with breast cancer: systematic review and meta-analysis. Breast Cancer Res Treat. 2010;123(3):627–635. [DOI] [PubMed] [Google Scholar]
- 6. Turkoz FP, Solak M, Petekkaya I, Keskin O, Kertmen N, Sarici F, Arik Z, Babacan T, Ozisik Y, Altundag K. Association between common risk factors and molecular subtypes in breast cancer patients. Breast. 2013;22(3):344–350. [DOI] [PubMed] [Google Scholar]
- 7. Gaudet MM, Press MF, Haile RW, Lynch CF, Glaser SL, Schildkraut J, Gammon MD, Douglas Thompson W, Bernstein JL. Risk factors by molecular subtypes of breast cancer across a population-based study of women 56 years or younger. Breast Cancer Res Treat. 2011;130(2):587–597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Bronsveld HK, Jensen V, Vahl P, De Bruin ML, Cornelissen S, Sanders J, Auvinen A, Haukka J, Andersen M, Vestergaard P, Schmidt MK. Diabetes and breast cancer subtypes. PLoS One. 2017;12(1):e0170084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Carey L, Winer E, Viale G, Cameron D, Gianni L. Triple-negative breast cancer: disease entity or title of convenience? Nat Rev Clin Oncol. 2010;7(12):683–692. [DOI] [PubMed] [Google Scholar]
- 10. Tran B, Bedard PL. Luminal-B breast cancer and novel therapeutic targets. Breast Cancer Res. 2011;13(6):221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Larsson SC, Mantzoros CS, Wolk A. Diabetes mellitus and risk of breast cancer: a meta-analysis. Int J Cancer. 2007;121(4):856–862. [DOI] [PubMed] [Google Scholar]
- 12. Neuhouser ML, Aragaki AK, Prentice RL, Manson JE, Chlebowski R, Carty CL, Ochs-Balcom HM, Thomson CA, Caan BJ, Tinker LF, Urrutia RP, Knudtson J, Anderson GL. Overweight, obesity, and postmenopausal invasive breast cancer risk: a secondary analysis of the Women’s Health Initiative randomized clinical trials. JAMA Oncol. 2015;1(5):611–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Gallagher EJ, Leroith D, Karnieli E. The metabolic syndrome--from insulin resistance to obesity and diabetes. Med Clin North Am. 2011;95(5):855–873. [DOI] [PubMed] [Google Scholar]
- 14. Gallagher EJ, LeRoith D. Obesity and diabetes: the increased risk of cancer and cancer-related mortality. Physiol Rev. 2015;95(3):727–748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Campbell PT, Newton CC, Patel AV, Jacobs EJ, Gapstur SM. Diabetes and cause-specific mortality in a prospective cohort of one million U.S. adults. Diabetes Care. 2012;35(9):1835–1844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Harding JL, Shaw JE, Peeters A, Cartensen B, Magliano DJ. Cancer risk among people with type 1 and type 2 diabetes: disentangling true associations, detection bias, and reverse causation. Diabetes Care 2015;38:264-270. Diabetes Care. 2015;38(4):734–735. [DOI] [PubMed] [Google Scholar]
- 17. Ben Q, Xu M, Ning X, Liu J, Hong S, Huang W, Zhang H, Li Z. Diabetes mellitus and risk of pancreatic cancer: A meta-analysis of cohort studies. Eur J Cancer. 2011;47(13):1928–1937. [DOI] [PubMed] [Google Scholar]
- 18. Wang C, Wang X, Gong G, Ben Q, Qiu W, Chen Y, Li G, Wang L. Increased risk of hepatocellular carcinoma in patients with diabetes mellitus: a systematic review and meta-analysis of cohort studies. Int J Cancer. 2012;130(7):1639–1648. [DOI] [PubMed] [Google Scholar]
- 19. Zendehdel K, Nyrén O, Ostenson CG, Adami HO, Ekbom A, Ye W. Cancer incidence in patients with type 1 diabetes mellitus: a population-based cohort study in Sweden. J Natl Cancer Inst. 2003;95(23):1797–1800. [DOI] [PubMed] [Google Scholar]
- 20. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324(5930):1029–1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Muti P, Quattrin T, Grant BJ, Krogh V, Micheli A, Schünemann HJ, Ram M, Freudenheim JL, Sieri S, Trevisan M, Berrino F. Fasting glucose is a risk factor for breast cancer: a prospective study. Cancer Epidemiol Biomarkers Prev. 2002;11(11):1361–1368. [PubMed] [Google Scholar]
- 22. Heuson JC, Legros N, Heimann R. Influence of insulin administration on growth of the 7,12-dimethylbenz(a)anthracene-induced mammary carcinoma in intact, oophorectomized, and hypophysectomized rats. Cancer Res. 1972;32(2):233–238. [PubMed] [Google Scholar]
- 23. Giovannucci E, Harlan DM, Archer MC, Bergenstal RM, Gapstur SM, Habel LA, Pollak M, Regensteiner JG, Yee D. Diabetes and cancer: a consensus report. CA Cancer J Clin. 2010;60(4):207–221. [DOI] [PubMed] [Google Scholar]
- 24. Vigneri P, Frasca F, Sciacca L, Pandini G, Vigneri R. Diabetes and cancer. Endocr Relat Cancer. 2009;16(4):1103–1123. [DOI] [PubMed] [Google Scholar]
- 25. Handelsman Y, Leroith D, Bloomgarden ZT, Dagogo-Jack S, Einhorn D, Garber AJ, Grunberger G, Harrell RM, Gagel RF, Lebovitz HE, McGill JB, Hennekens CH. Diabetes and cancer--an AACE/ACE consensus statement. Endocr Pract. 2013;19(4):675–693. [DOI] [PubMed] [Google Scholar]
- 26. Gallagher EJ, LeRoith D. The proliferating role of insulin and insulin-like growth factors in cancer. Trends Endocrinol Metab. 2010;21(10):610–618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Hwa V, Oh Y, Rosenfeld RG. The insulin-like growth factor-binding protein (IGFBP) superfamily. Endocr Rev. 1999;20(6):761–787. [DOI] [PubMed] [Google Scholar]
- 28. Burren CP, Wilson EM, Hwa V, Oh Y, Rosenfeld RG. Binding properties and distribution of insulin-like growth factor binding protein-related protein 3 (IGFBP-rP3/NovH), an additional member of the IGFBP superfamily. J Clin Endocrinol Metab. 1999;84(3):1096–1103. [DOI] [PubMed] [Google Scholar]
- 29. White MF. The IRS-signalling system: a network of docking proteins that mediate insulin action. Mol Cell Biochem. 1998;182(1-2):3–11. [PubMed] [Google Scholar]
- 30. Taniguchi CM, Emanuelli B, Kahn CR. Critical nodes in signalling pathways: insights into insulin action. Nat Rev Mol Cell Biol. 2006;7(2):85–96. [DOI] [PubMed] [Google Scholar]
- 31. Ward CW, Lawrence MC. Ligand-induced activation of the insulin receptor: a multi-step process involving structural changes in both the ligand and the receptor. BioEssays. 2009;31(4):422–434. [DOI] [PubMed] [Google Scholar]
- 32. Papa V, Pezzino V, Costantino A, Belfiore A, Giuffrida D, Frittitta L, Vannelli GB, Brand R, Goldfine ID, Vigneri R. Elevated insulin receptor content in human breast cancer. J Clin Invest. 1990;86(5):1503–1510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Mulligan AM, O’Malley FP, Ennis M, Fantus IG, Goodwin PJ. Insulin receptor is an independent predictor of a favorable outcome in early stage breast cancer. Breast Cancer Res Treat. 2007;106(1):39–47. [DOI] [PubMed] [Google Scholar]
- 34. Fernández AM, Kim JK, Yakar S, Dupont J, Hernandez-Sanchez C, Castle AL, Filmore J, Shulman GI, Le Roith D. Functional inactivation of the IGF-I and insulin receptors in skeletal muscle causes type 2 diabetes. Genes Dev. 2001;15(15):1926–1934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Novosyadlyy R, Lann DE, Vijayakumar A, Rowzee A, Lazzarino DA, Fierz Y, Carboni JM, Gottardis MM, Pennisi PA, Molinolo AA, Kurshan N, Mejia W, Santopietro S, Yakar S, Wood TL, LeRoith D. Insulin-mediated acceleration of breast cancer development and progression in a nonobese model of type 2 diabetes. Cancer Res. 2010;70(2):741–751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Ferguson RD, Novosyadlyy R, Fierz Y, Alikhani N, Sun H, Yakar S, Leroith D. Hyperinsulinemia enhances c-Myc-mediated mammary tumor development and advances metastatic progression to the lung in a mouse model of type 2 diabetes. Breast Cancer Res. 2012;14(1):R8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Zelenko Z, Gallagher EJ, Antoniou IM, Sachdev D, Nayak A, Yee D, LeRoith D. EMT reversal in human cancer cells after IR knockdown in hyperinsulinemic mice. Endocr Relat Cancer. 2016;23(9):747–758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Fierz Y, Novosyadlyy R, Vijayakumar A, Yakar S, LeRoith D. Insulin-sensitizing therapy attenuates type 2 diabetes-mediated mammary tumor progression. Diabetes. 2010;59(3):686–693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Fierz Y, Novosyadlyy R, Vijayakumar A, Yakar S, LeRoith D. Mammalian target of rapamycin inhibition abrogates insulin-mediated mammary tumor progression in type 2 diabetes. Endocr Relat Cancer. 2010;17(4):941–951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Gallagher EJ, Alikhani N, Tobin-Hess A, Blank J, Buffin NJ, Zelenko Z, Tennagels N, Werner U, LeRoith D. Insulin receptor phosphorylation by endogenous insulin or the insulin analog AspB10 promotes mammary tumor growth independent of the IGF-I receptor. Diabetes. 2013;62(10):3553–3560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Belfiore A, Malaguarnera R. Insulin receptor and cancer. Endocr Relat Cancer. 2011;18(4):R125–R147. [DOI] [PubMed] [Google Scholar]
- 42. Kurtzhals P, Schäffer L, Sørensen A, Kristensen C, Jonassen I, Schmid C, Trüb T. Correlations of receptor binding and metabolic and mitogenic potencies of insulin analogs designed for clinical use. Diabetes. 2000;49(6):999–1005. [DOI] [PubMed] [Google Scholar]
- 43. 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. Endocr Rev. 2009;30(6):586–623. [DOI] [PubMed] [Google Scholar]
- 44. Denley A, Bonython ER, Booker GW, Cosgrove LJ, Forbes BE, Ward CW, Wallace JC. Structural determinants for high-affinity binding of insulin-like growth factor II to insulin receptor (IR)-A, the exon 11 minus isoform of the IR. Mol Endocrinol. 2004;18(10):2502–2512. [DOI] [PubMed] [Google Scholar]
- 45. Harrington SC, Weroha SJ, Reynolds C, Suman VJ, Lingle WL, Haluska P. Quantifying insulin receptor isoform expression in FFPE breast tumors. Growth Horm IGF Res. 2012;22(3-4):108–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Chiefari E, Tanyolaç S, Iiritano S, Sciacqua A, Capula C, Arcidiacono B, Nocera A, Possidente K, Baudi F, Ventura V, Brunetti G, Brunetti FS, Vero R, Maio R, Greco M, Pavia M, Hodoglugil U, Durlach V, Pullinger CR, Goldfine ID, Perticone F, Foti D, Brunetti A. A polymorphism of HMGA1 is associated with increased risk of metabolic syndrome and related components. Sci Rep. 2013;3(1):1491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Resar LM. The high mobility group A1 gene: transforming inflammatory signals into cancer? Cancer Res. 2010;70(2):436–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Shah SN, Cope L, Poh W, Belton A, Roy S, Talbot CC Jr, Sukumar S, Huso DL, Resar LM. HMGA1: a master regulator of tumor progression in triple-negative breast cancer cells. PLoS One. 2013;8(5):e63419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Brunetti A, Manfioletti G, Chiefari E, Goldfine ID, Foti D. Transcriptional regulation of human insulin receptor gene by the high-mobility group protein HMGI(Y). FASEB J. 2001;15(2):492–500. [DOI] [PubMed] [Google Scholar]
- 50. Fusco A, Fedele M. Roles of HMGA proteins in cancer. Nat Rev Cancer. 2007;7(12):899–910. [DOI] [PubMed] [Google Scholar]
- 51. Vella V, Malaguarnera R, Nicolosi ML, Palladino C, Spoleti C, Massimino M, Vigneri P, Purrello M, Ragusa M, Morrione A, Belfiore A. Discoidin domain receptor 1 modulates insulin receptor signaling and biological responses in breast cancer cells. Oncotarget. 2017;8(26):43248–43270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Chettouh H, Fartoux L, Aoudjehane L, Wendum D, Clapéron A, Chrétien Y, Rey C, Scatton O, Soubrane O, Conti F, Praz F, Housset C, Rosmorduc O, Desbois-Mouthon C. Mitogenic insulin receptor-A is overexpressed in human hepatocellular carcinoma due to EGFR-mediated dysregulation of RNA splicing factors. Cancer Res. 2013;73(13):3974–3986. [DOI] [PubMed] [Google Scholar]
- 53. Sen S, Langiewicz M, Jumaa H, Webster NJ. Deletion of serine/arginine-rich splicing factor 3 in hepatocytes predisposes to hepatocellular carcinoma in mice. Hepatology. 2015;61(1):171–183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Lammers R, Gray A, Schlessinger J, Ullrich A. Differential signalling potential of insulin- and IGF-1-receptor cytoplasmic domains. EMBO J. 1989;8(5):1369–1375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Firth SM, Baxter RC. Cellular actions of the insulin-like growth factor binding proteins. Endocr Rev. 2002;23(6):824–854. [DOI] [PubMed] [Google Scholar]
- 56. Le Roith D, Bondy C, Yakar S, Liu JL, Butler A. The somatomedin hypothesis: 2001. Endocr Rev. 2001;22(1):53–74. [DOI] [PubMed] [Google Scholar]
- 57. Yakar S, Pennisi P, Zhao H, Zhang Y, LeRoith D.. Circulating IGF-1 and its role in cancer: lessons from the IGF-1 gene deletion (LID) mouse. Novartis Found Symp. 2004;262:3–9; discussion 9–18, 265–268. [PubMed] [Google Scholar]
- 58. Frasca F, Pandini G, Scalia P, Sciacca L, Mineo R, Costantino A, Goldfine ID, Belfiore A, Vigneri R. Insulin receptor isoform A, a newly recognized, high-affinity insulin-like growth factor II receptor in fetal and cancer cells. Mol Cell Biol. 1999;19(5):3278–3288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Sciacca L, Costantino A, Pandini G, Mineo R, Frasca F, Scalia P, Sbraccia P, Goldfine ID, Vigneri R, Belfiore A. Insulin receptor activation by IGF-II in breast cancers: evidence for a new autocrine/paracrine mechanism. Oncogene. 1999;18(15):2471–2479. [DOI] [PubMed] [Google Scholar]
- 60. Hiraga T, Ito S, Nakamura H. Cancer stem-like cell marker CD44 promotes bone metastases by enhancing tumorigenicity, cell motility, and hyaluronan production. Cancer Res. 2013;73(13):4112–4122. [DOI] [PubMed] [Google Scholar]
- 61. Yakar S, Leroith D, Brodt P. The role of the growth hormone/insulin-like growth factor axis in tumor growth and progression: Lessons from animal models. Cytokine Growth Factor Rev. 2005;16(4-5):407–420. [DOI] [PubMed] [Google Scholar]
- 62. Christopoulos PF, Msaouel P, Koutsilieris M. The role of the insulin-like growth factor-1 system in breast cancer. Mol Cancer. 2015;14(1):43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Li ZJ, Ying XJ, Chen HL, Ye PJ, Chen ZL, Li G, Jiang HF, Liu J, Zhou SZ. Insulin-like growth factor-1 induces lymphangiogenesis and facilitates lymphatic metastasis in colorectal cancer. World J Gastroenterol. 2013;19(43):7788–7794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Li P, Veldwijk MR, Zhang Q, Li ZB, Xu WC, Fu S. Co-inhibition of epidermal growth factor receptor and insulin-like growth factor receptor 1 enhances radiosensitivity in human breast cancer cells. BMC Cancer. 2013;13(1):297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Buck E, Eyzaguirre A, Rosenfeld-Franklin M, Thomson S, Mulvihill M, Barr S, Brown E, O’Connor M, Yao Y, Pachter J, Miglarese M, Epstein D, Iwata KK, Haley JD, Gibson NW, Ji QS. Feedback mechanisms promote cooperativity for small molecule inhibitors of epidermal and insulin-like growth factor receptors. Cancer Res. 2008;68(20):8322–8332. [DOI] [PubMed] [Google Scholar]
- 66. Luey BC, May FE. Insulin-like growth factors are essential to prevent anoikis in oestrogen-responsive breast cancer cells: importance of the type I IGF receptor and PI3-kinase/Akt pathway. Mol Cancer. 2016;15(1):8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Motallebnezhad M, Aghebati-Maleki L, Jadidi-Niaragh F, Nickho H, Samadi-Kafil H, Shamsasenjan K, Yousefi M. The insulin-like growth factor-I receptor (IGF-IR) in breast cancer: biology and treatment strategies. Tumour Biol. 2016;37(9):11711–11721. [DOI] [PubMed] [Google Scholar]
- 68. Shin SJ, Gong G, Lee HJ, Kang J, Bae YK, Lee A, Cho EY, Lee JS, Suh KS, Lee DW, Jung WH. Positive expression of insulin-like growth factor-1 receptor is associated with a positive hormone receptor status and a favorable prognosis in breast cancer. J Breast Cancer. 2014;17(2):113–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Menyhárt O, Santarpia L, Győrffy B. A comprehensive outline of trastuzumab resistance biomarkers in HER2 overexpressing breast cancer. Curr Cancer Drug Targets. 2015;15(8):665–683. [DOI] [PubMed] [Google Scholar]
- 70. Buck E, Gokhale PC, Koujak S, Brown E, Eyzaguirre A, Tao N, Rosenfeld-Franklin M, Lerner L, Chiu MI, Wild R, Epstein D, Pachter JA, Miglarese MR. Compensatory insulin receptor (IR) activation on inhibition of insulin-like growth factor-1 receptor (IGF-1R): rationale for cotargeting IGF-1R and IR in cancer. Mol Cancer Ther. 2010;9(10):2652–2664. [DOI] [PubMed] [Google Scholar]
- 71. Ekyalongo RC, Yee D. Revisiting the IGF-1R as a breast cancer target. NPJ Precis Oncol. 2017;1:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Rostoker R, Bitton-Worms K, Caspi A, Shen-Orr Z, LeRoith D. Investigating new therapeutic strategies targeting hyperinsulinemia’s mitogenic effects in a female mouse breast cancer model. Endocrinology. 2013;154(5):1701–1710. [DOI] [PubMed] [Google Scholar]
- 73. Singh P, Alex JM, Bast F. Insulin receptor (IR) and insulin-like growth factor receptor 1 (IGF-1R) signaling systems: novel treatment strategies for cancer. Med Oncol. 2014;31(1):805. [DOI] [PubMed] [Google Scholar]
- 74. Chan JY, LaPara K, Yee D. Disruption of insulin receptor function inhibits proliferation in endocrine-resistant breast cancer cells. Oncogene. 2016;35(32):4235–4243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Salisbury TB, Tomblin JK. Insulin/Insulin-like growth factors in cancer: new roles for the aryl hydrocarbon receptor, tumor resistance mechanisms, and new blocking strategies. Front Endocrinol (Lausanne). 2015;6:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Brahmkhatri VP, Prasanna C, Atreya HS. Insulin-like growth factor system in cancer: novel targeted therapies. BioMed Res Int. 2015;2015:538019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Law JH, Habibi G, Hu K, Masoudi H, Wang MY, Stratford AL, Park E, Gee JM, Finlay P, Jones HE, Nicholson RI, Carboni J, Gottardis M, Pollak M, Dunn SE. Phosphorylated insulin-like growth factor-i/insulin receptor is present in all breast cancer subtypes and is related to poor survival. Cancer Res. 2008;68(24):10238–10246. [DOI] [PubMed] [Google Scholar]
- 78. Huang J, Morehouse C, Streicher K, Higgs BW, Gao J, Czapiga M, Boutrin A, Zhu W, Brohawn P, Chang Y, Viner J, LaVallee T, Richman L, Jallal B, Yao Y. Altered expression of insulin receptor isoforms in breast cancer. PLoS One. 2011;6(10):e26177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Hartog H, Horlings HM, van der Vegt B, Kreike B, Ajouaou A, van de Vijver MJ, Marike Boezen H, de Bock GH, van der Graaf WT, Wesseling J. Divergent effects of insulin-like growth factor-1 receptor expression on prognosis of estrogen receptor positive versus triple negative invasive ductal breast carcinoma. Breast Cancer Res Treat. 2011;129(3):725–736. [DOI] [PubMed] [Google Scholar]
- 80. Mountzios G, Aivazi D, Kostopoulos I, Kourea HP, Kouvatseas G, Timotheadou E, Zebekakis P, Efstratiou I, Gogas H, Vamvouka C, Chrisafi S, Stofas A, Pentheroudakis G, Koutras A, Galani E, Bafaloukos D, Fountzilas G. Differential expression of the insulin-like growth factor receptor among early breast cancer subtypes. PLoS One. 2014;9(3):e91407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Chahil TJ, Ginsberg HN. Diabetic dyslipidemia. Endocrinol Metab Clin North Am. 2006;35(3):491–510, vii–viii. [DOI] [PubMed] [Google Scholar]
- 82. Melvin JC, Holmberg L, Rohrmann S, Loda M, Van Hemelrijck M. Serum lipid profiles and cancer risk in the context of obesity: four meta-analyses. J Cancer Epidemiol. 2013;2013:823849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Li C, Yang L, Zhang D, Jiang W. Systematic review and meta-analysis suggest that dietary cholesterol intake increases risk of breast cancer. Nutr Res. 2016;36(7):627–635. [DOI] [PubMed] [Google Scholar]
- 84. Taylor F, Huffman MD, Macedo AF, Moore TH, Burke M, Davey Smith G, Ward K, Ebrahim S. Statins for the primary prevention of cardiovascular disease. Cochrane Database Syst Rev. 2013;(1):CD004816. [DOI] [PMC free article] [PubMed]
- 85. Geybels MS, Wright JL, Holt SK, Kolb S, Feng Z, Stanford JL. Statin use in relation to prostate cancer outcomes in a population-based patient cohort study. Prostate. 2013;73(11):1214–1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Tan M, Song X, Zhang G, Peng A, Li X, Li M, Liu Y, Wang C. Statins and the risk of lung cancer: a meta-analysis. PLoS One. 2013;8(2):e57349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Singh S, Singh PP, Singh AG, Murad MH, Sanchez W. Statins are associated with a reduced risk of hepatocellular cancer: a systematic review and meta-analysis. Gastroenterology. 2013;144(2):323–332. [DOI] [PubMed] [Google Scholar]
- 88. Nielsen SF, Nordestgaard BG, Bojesen SE. Statin use and reduced cancer-related mortality. N Engl J Med. 2012;367(19):1792–1802. [DOI] [PubMed] [Google Scholar]
- 89. Zhang XL, Liu M, Qian J, Zheng JH, Zhang XP, Guo CC, Geng J, Peng B, Che JP, Wu Y. Statin use and risk of kidney cancer: a meta-analysis of observational studies and randomized trials. Br J Clin Pharmacol. 2014;77(3):458–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Brewer TM, Masuda H, Liu DD, Shen Y, Liu P, Iwamoto T, Kai K, Barnett CM, Woodward WA, Reuben JM, Yang P, Hortobagyi GN, Ueno NT. Statin use in primary inflammatory breast cancer: a cohort study. Br J Cancer. 2013;109(2):318–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Van Wyhe RD, Rahal OM, Woodward WA. Effect of statins on breast cancer recurrence and mortality: a review. Breast Cancer (Dove Med Press). 2017;9:559–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Llaverias G, Danilo C, Mercier I, Daumer K, Capozza F, Williams TM, Sotgia F, Lisanti MP, Frank PG. Role of cholesterol in the development and progression of breast cancer. Am J Pathol. 2011;178(1):402–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Llaverias G, Danilo C, Wang Y, Witkiewicz AK, Daumer K, Lisanti MP, Frank PG. A Western-type diet accelerates tumor progression in an autochthonous mouse model of prostate cancer. Am J Pathol. 2010;177(6):3180–3191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Yin W, Carballo-Jane E, McLaren DG, Mendoza VH, Gagen K, Geoghagen NS, McNamara LA, Gorski JN, Eiermann GJ, Petrov A, Wolff M, Tong X, Wilsie LC, Akiyama TE, Chen J, Thankappan A, Xue J, Ping X, Andrews G, Wickham LA, Gai CL, Trinh T, Kulick AA, Donnelly MJ, Voronin GO, Rosa R, Cumiskey AM, Bekkari K, Mitnaul LJ, Puig O, Chen F, Raubertas R, Wong PH, Hansen BC, Koblan KS, Roddy TP, Hubbard BK, Strack AM. Plasma lipid profiling across species for the identification of optimal animal models of human dyslipidemia. J Lipid Res. 2012;53(1):51–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Dansky HM, Shu P, Donavan M, Montagno J, Nagle DL, Smutko JS, Roy N, Whiteing S, Barrios J, McBride TJ, Smith JD, Duyk G, Breslow JL, Moore KJ. A phenotype-sensitizing Apoe-deficient genetic background reveals novel atherosclerosis predisposition loci in the mouse. Genetics. 2002;160(4):1599–1608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. van den Maagdenberg AM, Hofker MH, Krimpenfort PJ, de Bruijn I, van Vlijmen B, van der Boom H, Havekes LM, Frants RR. Transgenic mice carrying the apolipoprotein E3-Leiden gene exhibit hyperlipoproteinemia. J Biol Chem. 1993;268(14):10540–10545. [PubMed] [Google Scholar]
- 97. Liu Y, Michael MD, Kash S, Bensch WR, Monia BP, Murray SF, Otto KA, Syed SK, Bhanot S, Sloop KW, Sullivan JM, Reifel-Miller A. Deficiency of adiponectin receptor 2 reduces diet-induced insulin resistance but promotes type 2 diabetes. Endocrinology. 2007;148(2):683–692. [DOI] [PubMed] [Google Scholar]
- 98. Alikhani N, Ferguson RD, Novosyadlyy R, Gallagher EJ, Scheinman EJ, Yakar S, LeRoith D. Mammary tumor growth and pulmonary metastasis are enhanced in a hyperlipidemic mouse model. Oncogene. 2013;32(8):961–967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Stranzl A, Schmidt H, Winkler R, Kostner GM. Low-density lipoprotein receptor mRNA in human breast cancer cells: influence by PKC modulators. Breast Cancer Res Treat. 1997;42(3):195–205. [DOI] [PubMed] [Google Scholar]
- 100. Gallagher EJ, Zelenko Z, Neel BA, Antoniou IM, Rajan L, Kase N, LeRoith D. Elevated tumor LDLR expression accelerates LDL cholesterol-mediated breast cancer growth in mouse models of hyperlipidemia. Oncogene. 2017;36(46):6462–6471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Liu J, Xu A, Lam KS, Wong NS, Chen J, Shepherd PR, Wang Y. Cholesterol-induced mammary tumorigenesis is enhanced by adiponectin deficiency: role of LDL receptor upregulation. Oncotarget. 2013;4(10):1804–1818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Nelson ER, Wardell SE, Jasper JS, Park S, Suchindran S, Howe MK, Carver NJ, Pillai RV, Sullivan PM, Sondhi V, Umetani M, Geradts J, McDonnell DP. 27-Hydroxycholesterol links hypercholesterolemia and breast cancer pathophysiology. Science. 2013;342(6162):1094–1098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Desai P, Lehman A, Chlebowski RT, Kwan ML, Arun M, Manson JE, Lavasani S, Wasswertheil-Smoller S, Sarto GE, LeBoff M, Cauley J, Cote M, Beebe-Dimmer J, Jay A, Simon MS. Statins and breast cancer stage and mortality in the Women’s Health Initiative. Cancer Causes Control. 2015;26(4):529–539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Marwarha G, Raza S, Hammer K, Ghribi O. 27-hydroxycholesterol: A novel player in molecular carcinogenesis of breast and prostate cancer. Chem Phys Lipids. 2017;207(Pt B):108–126. [DOI] [PubMed] [Google Scholar]
- 105. Simigdala N, Gao Q, Pancholi S, Roberg-Larsen H, Zvelebil M, Ribas R, Folkerd E, Thompson A, Bhamra A, Dowsett M, Martin LA. Cholesterol biosynthesis pathway as a novel mechanism of resistance to estrogen deprivation in estrogen receptor-positive breast cancer. Breast Cancer Res. 2016;18(1):58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Shen Z, Zhu D, Liu J, Chen J, Liu Y, Hu C, Li Z, Li Y. 27-Hydroxycholesterol induces invasion and migration of breast cancer cells by increasing MMP9 and generating EMT through activation of STAT-3. Environ Toxicol Pharmacol. 2017;51:1–8. [DOI] [PubMed] [Google Scholar]
- 107. Jo Y, Debose-Boyd RA. Control of cholesterol synthesis through regulated ER-associated degradation of HMG CoA reductase. Crit Rev Biochem Mol Biol. 2010;45(3):185–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Martini C, Pallottini V. Cholesterol: from feeding to gene regulation. Genes Nutr. 2007;2(2):181–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Antalis CJ, Arnold T, Rasool T, Lee B, Buhman KK, Siddiqui RA. High ACAT1 expression in estrogen receptor negative basal-like breast cancer cells is associated with LDL-induced proliferation. Breast Cancer Res Treat. 2010;122(3):661–670. [DOI] [PubMed] [Google Scholar]
- 110. Zelcer N, Hong C, Boyadjian R, Tontonoz P. LXR regulates cholesterol uptake through Idol-dependent ubiquitination of the LDL receptor. Science. 2009;325(5936):100–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. El Roz A, Bard JM, Valin S, Huvelin JM, Nazih H. Macrophage apolipoprotein E and proliferation of MCF-7 breast cancer cells: role of LXR. Anticancer Res. 2013;33(9):3783–3789. [PubMed] [Google Scholar]
- 112. Baek AE, Yu YA, He S, Wardell SE, Chang CY, Kwon S, Pillai RV, McDowell HB, Thompson JW, Dubois LG, Sullivan PM, Kemper JK, Gunn MD, McDonnell DP, Nelson ER. The cholesterol metabolite 27 hydroxycholesterol facilitates breast cancer metastasis through its actions on immune cells. Nat Commun. 2017;8(1):864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Reeves GK, Pirie K, Beral V, Green J, Spencer E, Bull D; Million Women Study Collaboration . Cancer incidence and mortality in relation to body mass index in the Million Women Study: cohort study. BMJ. 2007;335(7630):1134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Goodwin PJ, Stambolic V. Impact of the obesity epidemic on cancer. Annu Rev Med. 2015;66(1):281–296. [DOI] [PubMed] [Google Scholar]
- 115. Ouchi N, Parker JL, Lugus JJ, Walsh K. Adipokines in inflammation and metabolic disease. Nat Rev Immunol. 2011;11(2):85–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Surmacz E. Leptin and adiponectin: emerging therapeutic targets in breast cancer. J Mammary Gland Biol Neoplasia. 2013;18(3-4):321–332. [DOI] [PubMed] [Google Scholar]
- 117. Bowers LW, Rossi EL, McDonell SB, Doerstling SS, Khatib SA, Lineberger CG, Albright JE, Tang X, deGraffenried LA, Hursting SD. Leptin signaling mediates obesity-associated CSC enrichment and EMT in preclinical TNBC models. Mol Cancer Res. 2018;16(5):869–879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. McTernan PG, Kusminski CM, Kumar S. Resistin. Curr Opin Lipidol. 2006;17(2):170–175. [DOI] [PubMed] [Google Scholar]
- 119. Rui L, Yuan M, Frantz D, Shoelson S, White MF. SOCS-1 and SOCS-3 block insulin signaling by ubiquitin-mediated degradation of IRS1 and IRS2. J Biol Chem. 2002;277(44):42394–42398. [DOI] [PubMed] [Google Scholar]
- 120. Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, Patel HR, Ahima RS, Lazar MA. The hormone resistin links obesity to diabetes. Nature. 2001;409(6818):307–312. [DOI] [PubMed] [Google Scholar]
- 121. Dalamaga M, Sotiropoulos G, Karmaniolas K, Pelekanos N, Papadavid E, Lekka A. Serum resistin: a biomarker of breast cancer in postmenopausal women? Association with clinicopathological characteristics, tumor markers, inflammatory and metabolic parameters. Clin Biochem. 2013;46(7-8):584–590. [DOI] [PubMed] [Google Scholar]
- 122. Lee YC, Chen YJ, Wu CC, Lo S, Hou MF, Yuan SS. Resistin expression in breast cancer tissue as a marker of prognosis and hormone therapy stratification. Gynecol Oncol. 2012;125(3):742–750. [DOI] [PubMed] [Google Scholar]
- 123. Barbieri I, Pensa S, Pannellini T, Quaglino E, Maritano D, Demaria M, Voster A, Turkson J, Cavallo F, Watson CJ, Provero P, Musiani P, Poli V. Constitutively active Stat3 enhances neu-mediated migration and metastasis in mammary tumors via upregulation of Cten. Cancer Res. 2010;70(6):2558–2567. [DOI] [PubMed] [Google Scholar]
- 124. Yu H, Lee H, Herrmann A, Buettner R, Jove R. Revisiting STAT3 signalling in cancer: new and unexpected biological functions. Nat Rev Cancer. 2014;14(11):736–746. [DOI] [PubMed] [Google Scholar]
- 125. Wang CH, Wang PJ, Hsieh YC, Lo S, Lee YC, Chen YC, Tsai CH, Chiu WC, Chu-Sung Hu S, Lu CW, Yang YF, Chiu CC, Ou-Yang F, Wang YM, Hou MF, Yuan SS. Resistin facilitates breast cancer progression via TLR4-mediated induction of mesenchymal phenotypes and stemness properties. Oncogene. 2018;37(5):589–600. [DOI] [PubMed] [Google Scholar]
- 126. Lee JO, Kim N, Lee HJ, Lee YW, Kim SJ, Park SH, Kim HS. Resistin, a fat-derived secretory factor, promotes metastasis of MDA-MB-231 human breast cancer cells through ERM activation. Sci Rep. 2016;6(1):18923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Gunter MJ, Wang T, Cushman M, Xue X, Wassertheil-Smoller S, Strickler HD, Rohan TE, Manson JE, McTiernan A, Kaplan RC, Scherer PE, Chlebowski RT, Snetselaar L, Wang D, Ho GY. Circulating adipokines and inflammatory markers and postmenopausal breast cancer risk. J Natl Cancer Inst. 2015;107(9):djv169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Grossmann ME, Nkhata KJ, Mizuno NK, Ray A, Cleary MP. Effects of adiponectin on breast cancer cell growth and signaling. Br J Cancer. 2008;98(2):370–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Taliaferro-Smith L, Nagalingam A, Zhong D, Zhou W, Saxena NK, Sharma D. LKB1 is required for adiponectin-mediated modulation of AMPK-S6K axis and inhibition of migration and invasion of breast cancer cells. Oncogene. 2009;28(29):2621–2633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Taliaferro-Smith L, Nagalingam A, Knight BB, Oberlick E, Saxena NK, Sharma D. Integral role of PTP1B in adiponectin-mediated inhibition of oncogenic actions of leptin in breast carcinogenesis. Neoplasia. 2013;15(1):23–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Thompson HJ, Sedlacek SM, Wolfe P, Paul D, Lakoski SG, Playdon MC, McGinley JN, Matthews SB. Impact of weight loss on plasma leptin and adiponectin in overweight-to-obese post menopausal breast cancer survivors. Nutrients. 2015;7(7):5156–5176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Panno ML, Naimo GD, Spina E, Andò S, Mauro L. Different molecular signaling sustaining adiponectin action in breast cancer. Curr Opin Pharmacol. 2016;31:1–7. [DOI] [PubMed] [Google Scholar]
- 133. Tahergorabi Z, Khazaei M, Moodi M, Chamani E. From obesity to cancer: a review on proposed mechanisms. Cell Biochem Funct. 2016;34(8):533–545. [DOI] [PubMed] [Google Scholar]
- 134. Libby EF, Frost AR, Demark-Wahnefried W, Hurst DR. Linking adiponectin and autophagy in the regulation of breast cancer metastasis. J Mol Med (Berl). 2014;92(10):1015–1023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol. 2016;14(8):e1002533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Francescone R, Hou V, Grivennikov SI. Microbiome, inflammation, and cancer. Cancer J. 2014;20(3):181–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Yurkovetskiy LA, Pickard JM, Chervonsky AV. Microbiota and autoimmunity: exploring new avenues. Cell Host Microbe. 2015;17(5):548–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Reijnders D, Goossens GH, Hermes GD, Neis EP, van der Beek CM, Most J, Holst JJ, Lenaerts K, Kootte RS, Nieuwdorp M, Groen AK, Olde Damink SW, Boekschoten MV, Smidt H, Zoetendal EG, Dejong CH, Blaak EE. Effects of gut microbiota manipulation by antibiotics on host metabolism in obese humans: a randomized double-blind placebo-controlled trial. Cell Metab. 2016;24(2):341. [DOI] [PubMed] [Google Scholar]
- 139. Krishnan S, Alden N, Lee K. Pathways and functions of gut microbiota metabolism impacting host physiology. Curr Opin Biotechnol. 2015;36:137–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444(7122):1022–1023. [DOI] [PubMed] [Google Scholar]
- 141. Kersten S, Mandard S, Tan NS, Escher P, Metzger D, Chambon P, Gonzalez FJ, Desvergne B, Wahli W. Characterization of the fasting-induced adipose factor FIAF, a novel peroxisome proliferator-activated receptor target gene. J Biol Chem. 2000;275(37):28488–28493. [DOI] [PubMed] [Google Scholar]
- 142. Cushing EM, Chi X, Sylvers KL, Shetty SK, Potthoff MJ, Davies BSJ. Angiopoietin-like 4 directs uptake of dietary fat away from adipose during fasting. Mol Metab. 2017;6(8):809–818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, Gordon JI. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci USA. 2004;101(44):15718–15723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Boutagy NE, McMillan RP, Frisard MI, Hulver MW. Metabolic endotoxemia with obesity: is it real and is it relevant? Biochimie. 2016;124:11–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Ghanim H, Abuaysheh S, Sia CL, Korzeniewski K, Chaudhuri A, Fernandez-Real JM, Dandona P. Increase in plasma endotoxin concentrations and the expression of Toll-like receptors and suppressor of cytokine signaling-3 in mononuclear cells after a high-fat, high-carbohydrate meal: implications for insulin resistance. Diabetes Care. 2009;32(12):2281–2287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Amieva M, Peek RM Jr. Pathobiology of Helicobacter pylori-induced gastric cancer. Gastroenterology. 2016;150(1):64–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Arthur JC, Perez-Chanona E, Mühlbauer M, Tomkovich S, Uronis JM, Fan TJ, Campbell BJ, Abujamel T, Dogan B, Rogers AB, Rhodes JM, Stintzi A, Simpson KW, Hansen JJ, Keku TO, Fodor AA, Jobin C. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science. 2012;338(6103):120–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Plottel CS, Blaser MJ. Microbiome and malignancy. Cell Host Microbe. 2011;10(4):324–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Goedert JJ, Jones G, Hua X, Xu X, Yu G, Flores R, Falk RT, Gail MH, Shi J, Ravel J, Feigelson HS. Investigation of the association between the fecal microbiota and breast cancer in postmenopausal women: a population-based case-control pilot study. J Natl Cancer Inst. 2015;107(8):107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Mikó E, Vida A, Kovács T, Ujlaki G, Trencsényi G, Márton J, Sári Z, Kovács P, Boratkó A, Hujber Z, Csonka T, Antal-Szalmás P, Watanabe M, Gombos I, Csoka B, Kiss B, Vígh L, Szabó J, Méhes G, Sebestyén A, Goedert JJ, Bai P. Lithocholic acid, a bacterial metabolite reduces breast cancer cell proliferation and aggressiveness. Biochim Biophys Acta. 2018;1859(9):958–974. [DOI] [PubMed] [Google Scholar]

