Targeting obesity-related adipose tissue dysfunction to prevent cancer development and progression

Targeting obesity-related adipose tissue dysfunction to prevent cancer development and progression

Author's Accepted Manuscript Targeting Obesity-Related Adipose Tissue Dysfunction to Prevent Cancer Development and Progression Ayca Gucalp M.D., Nei...

647KB Sizes 0 Downloads 48 Views

Author's Accepted Manuscript

Targeting Obesity-Related Adipose Tissue Dysfunction to Prevent Cancer Development and Progression Ayca Gucalp M.D., Neil M. Iyengar M.D., Clifford A. Hudis M.D., Andrew J. Dannenberg M.D.

www.elsevier.de/endend

PII: DOI: Reference:

S0093-7754(15)00177-3 http://dx.doi.org/10.1053/j.seminoncol.2015.09.012 YSONC51868

To appear in:

Semin Oncol

Cite this article as: Ayca Gucalp M.D., Neil M. Iyengar M.D., Clifford A. Hudis M.D., Andrew J. Dannenberg M.D., Targeting Obesity-Related Adipose Tissue Dysfunction to Prevent Cancer Development and Progression, Semin Oncol, http://dx.doi.org/ 10.1053/j.seminoncol.2015.09.012 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting galley proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Title: Targeting obesity-related adipose tissue dysfunction to prevent cancer development and progression Authors: Ayca Gucalp, Neil M. Iyengar, Clifford A. Hudis, Andrew J. Dannenberg Ayca Gucalp M.D. Assistant Attending, Breast Medicine Service Department of Medicine Memorial Sloan Kettering Cancer Center Instructor of Medicine Weill Cornell Medical College Neil M. Iyengar M.D. Assistant Attending, Breast Medicine Service Department of Medicine Memorial Sloan Kettering Cancer Center Instructor of Medicine Weill Cornell Medical College Associate Attending Physician Rockefeller University Clifford A. Hudis M.D. Chief, Breast Medicine Service Attending, Department of Medicine Memorial Sloan Kettering Cancer Center Professor of Medicine Weill Cornell Medical College Andrew J. Dannenberg M.D. Professor of Medicine Weill Cornell Medical College Grant Support: This work was supported by grants NIH/NCI R01CA154481, the Botwinick-Wolfensohn Foundation (in memory of Mr. and Mrs. Benjamin Botwinick), and the Breast Cancer Research Foundation. Corresponding Author Info: Ayca Gucalp, Department of Medicine, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, NY 10065. Phone: 646-888-4536; Fax: 646-888-4917. E-mail: [email protected]. Conflict of Interest: None

1

Abstract The incidence of obesity, a leading modifiable risk factor for common solid tumors, is increasing. Effective interventions are needed to minimize the public health implications of obesity. Although the mechanisms linking increased adiposity to malignancy are incompletely understood, growing evidence points to complex interactions among multiple systemic and tissue-specific pathways including inflamed white adipose tissue. The metabolic and inflammatory consequences of white adipose tissue dysfunction collectively provide a plausible explanation for the link between overweight/obesity and carcinogenesis. Gaining a better understanding of these underlying molecular pathways and developing risk assessment tools that identify at-risk populations will be critical in implementing effective and novel cancer prevention and management strategies.

I.

Introduction:

Obesity rates continue to rise in the US and in most other industrialized nations. Greater than one third of the US population is obese, defined as a body mass index (BMI) of ≥ 30 kg/m2.1 If the current trends continue, it is estimated that by the year 2030 >50% of the population of 39 states will be obese.2 Epidemiologic studies link obesity with an increased risk of epithelial malignancies.3,4 In 2007, more than 50,000 new cases of cancer in women (7%) and 34,000 new cases in men (4%) were attributable to obesity.5 Accordingly, obesity is projected to

2

replace tobacco as the most common modifiable risk factor for the development of some of the leading preventable causes of death in the US including cardiovascular disease, stroke, and malignancy. In addition to cancer risk, obesity is also associated with poorer cancer-specific outcomes, including higher risk of cancer recurrence and mortality.6-8 Although the mechanisms linking increased adiposity to malignancy remain incompletely understood, growing evidence suggests that complex interactions between multiple pathways regulating steroid hormone synthesis, insulin resistance, adipokine and cytokine production, and chronic local and systemic inflammation may collectively explain the link between overweight/obesity and carcinogenesis.

Adipose tissue is increasingly recognized as an active endocrine organ that secretes several adipose tissue-specific hormones, including adipokines. Adipose tissue plays a significant role in the regulation of energy balance and homeostasis.9 Obesity, which is most commonly defined by BMI, is associated with increased adipocyte number, adipocyte hypertrophy, and metabolic dysfunction. Specifically, the metabolic disturbances associated with obesity include changes in steroid hormone and adipokine production, insulin resistance, dyslipidemia, and subclinical chronic inflammation.10,11 Among many other health consequences, these metabolic alterations involve pathways known to contribute to cancer development, progression, and metastasis.11 In addition to

3

being common in the setting of obesity, adipose tissue dysfunction and its consequences including insulin resistance, dyslipidemia and white adipose tissue (WAT) inflammation have also been reported in individuals with normal BMIs.12,13 White adipose tissue is one of two types of fat found in mammals, the other being brown adipose tissue. In contrast to brown adipose tissue which generates heat, WAT stores fat and acts as a thermal insulator, to maintain body temperature. Of course, not all obese individuals develop obesity-related metabolic or cardiovascular disorders and some appear to have preserved normal adipose tissue function.14-16 These metabolically healthy obese individuals are, however, in the minority. Together, these observations highlight the limitations of BMI as a measure of adipose tissue function. Intra-abdominal or visceral adipocytes have the highest rates of metabolic activity17 and truncal obesity has been associated with greater incidence of metabolic derangements.18 The key issue is that BMI alone is not an adequate surrogate of adipose tissue function. Therefore, a clearer understanding of the underlying biology linking obesity and carcinogenesis will be critical for the identification of high-risk individuals and for the development of mechanism-based interventions for the safe and effective prevention and treatment of cancer. It is clear that effective and broadly applicable interventions are needed to decrease the societal impact of this shift in disease causation. Thus, the development of minimally-invasive risk assessment tools represents a critical unmet need. The specific goal is to identify the

4

targetable underlying pathophysiology often but not exclusively associated with obesity. Therefore, this review focuses on the underlying biologic mechanisms associated with impaired adipose tissue function in the context of cancer development. In addition, we will examine potential interventions and strategies to reduce the unfavorable impact of obesity on cancer pathogenesis and progression.

II.

Adipose tissue dysfunction and metabolism

WAT plays a key role in the regulation of energy balance, which is mediated by the production and secretion of metabolically active proteins. Several lines of evidence suggest that obesity-induced tumor development, invasion, and progression are mediated by a complex interplay of systemic alterations at the center of which is dysfunctional adipose tissue.11 In this section, we will review these potentially carcinogenic derangements in WAT including changes in levels of several hormones, adipokines, and proinflammatory mediators.

Insulin resistance, lipid metabolism and the metabolic syndrome Obesity is associated with insulin resistance, characterized by elevated circulating levels of insulin and glucose. Epidemiologic evidence suggests that individuals diagnosed with non-insulin-dependent diabetes mellitus (NIDDM) are at significantly higher risk for several cancers notably breast, endometrial,

5

colorectal, pancreatic, urinary tract, and hepatocellular cancer.19,20 Furthermore, a history of NIDDM has been associated with worse long-term survival outcomes in patients with breast and several other cancers.21-23 There are several possible common mechanisms supporting a biological link between diabetes and cancer.

Insulin has been suggested to stimulate carcinogenesis by inducing the synthesis of insulin-like growth factor-1 (IGF-1) and activating insulin/IGF-1 receptors that can be overexpressed in human cancer cells.21 Downstream activation of the RasRaf-MAPK and PI3K/Akt pathways ultimately leads to increased tumor cell proliferation, and inhibition of apoptosis.20,24,25 Furthermore, IGF-1 and insulin interact with hormone signaling pathways and may impact the growth and progression of hormonally-driven cancers. Moreover, increased insulin levels can lead to decreased hepatic production of steroid hormone binding globulin (SHBG) that normally binds to circulating steroid hormones.4 A decrease in SHBG levels resulting in increased circulating free estradiol and androgens may also contribute to tumorigenesis and progression of hormone-dependent tumors.

In addition, insulin has critical effects on lipid metabolism. Under normal physiologic conditions, insulin promotes lipid synthesis and inhibits the degradation of lipids. In contrast, obesity-related insulin resistance is associated with increased lipolysis resulting in the release of free fatty acids (FFA).26,27

6

Increased levels of FFA in the circulation can lead to ectopic fat deposition in organs such as the liver, pancreas, skeletal muscle, and heart leading to disruption of metabolic processes, impairment of organ function, and further promotion of insulin resistance, hyperglycemia, dyslipidemia, and hypertension.28 Along with central obesity, these conditions are components of the metabolic syndrome.29 The metabolic syndrome has been associated with development of NIDDM, cardiovascular disease, and more recently several cancers including breast, endometrial, colon, pancreatic, liver, and bladder cancer.30-33 Altered adipokine production Adipokines, which are bioactive proteins synthesized and secreted from adipose tissue, play an important role in lipid metabolism, insulin sensitivity, inflammation,

regulation

of

energy

balance,

angiogenesis,

and

cell

proliferation.11,34 Cross-talk between leptin and adiponectin-induced signaling pathways are thought to maintain metabolic homeostasis and balance cell proliferation and apoptosis. Obesity is commonly associated with altered levels of adipokines particularly increased leptin and decreased adiponectin levels in the circulation.34 Furthermore, adipokine deregulation is implicated in cancer progression and metastasis.35

Adiponectin. Several preclinical and epidemiologic studies suggest an inverse relationship between adiponectin levels and risk for the development and

7

progression of multiple cancers. Adiponectin can inhibit cell proliferation, induce apoptosis, and decrease invasion in cellular models of multiple cancers.36-39 Furthermore, preclinical work in mouse models has shown that lower levels of adiponectin results in accelerated hepatic tumor growth40 and increased colon polyp formation.41 Observational studies suggest a correlation between higher levels of adiponectin and decreased risk of postmenopausal breast cancer, uterine cancer and colorectal cancer.42-46 Variants of the adiponectin (ADIPOQ) and adiponectin receptor 1(ADIPOR1) genes have been associated with increased breast, colorectal and prostate cancer risk.47-49 Adiponectin is thought to affect cell cycle regulation, proliferation and apoptosis through the activation of multiple signaling pathways downstream of the adiponectin receptors, AdipoR1 and AdipoR2, including adenosine monophosphate-activated protein kinase (AMPK), PI3K/mTOR and the transcription factor, nuclear factor-kappaB (NFκB).50-52

Leptin. Leptin is primarily produced in the adipocyte and plays a key role in regulating energy intake and expenditure both centrally, by acting on receptors in the hypothalamus, and peripherally through modulation of glucose and insulin metabolism.53 Obese individuals generally have higher circulating levels of leptin in comparison to individuals with normal BMI.34 Similar to adiponectin, leptin and its transmembrane receptor (ObR) have been implicated in tumor

8

development and metastasis.52,54-57 Leptin is known to activate the JAK/STAT, MAPK/ERK and PI3K/Akt signaling pathways leading to increased cell migration, invasion and cell survival. Additionally, leptin promotes cell proliferation via alteration of cell cycle checkpoints and up-regulation of the cdk2 and cyclin D1 genes, advancing cells from G1 to S phase.50 Finally, leptin has also been reported to have both proinflammatory and angiogenic properties, which may play a role in carcinogenesis (Figure 1). Cancer cell lines exposed to leptin demonstrate increased proliferation, migration, and invasion. In both in vitro and in vivo models of breast cancer, leptin receptor deficiency has been associated with growth suppression and decreased tumor volume. Epidemiologic data regarding leptin and cancer risk remain mixed, however. Several studies have reported a direct association between elevated circulating levels of leptin and increased risk of breast, endometrial, colorectal and prostate cancer, while others have demonstrated no association or an inverse correlation.11,58 Furthermore, increased expression of the leptin receptor has been correlated with decreased survival in ovarian cancer59 and the development of distant metastases in breast cancer.60

More recently, the leptin to adiponectin ratio has been described as a potential marker of cancer risk and outcomes as adiponectin has been shown to counteract the effects of leptin on cellular behavior. In prostate cancer cell lines, leptin can

9

overcome the anti-proliferative effects of adiponectin61 and in in vitro models of hepatocellular carcinoma, adiponectin can block leptin-induced cell proliferation.62 Population-based observational studies have demonstrated a positive correlation between this ratio and increased risk of endometrial, prostate, and breast cancer.63-65 In addition to risk, a direct association between increased leptin to adiponectin ratio and tumor aggressiveness and size has been reported in breast cancer.66 These findings suggest that the balance between these two adipokines may play a central role in risk assessment in the setting of obesityinduced carcinogenesis.

Steroid hormone synthesis Estrogen. Somewhat paradoxically, epidemiologic data demonstrate an increased incidence of hormonally-driven cancers among postmenopausal women. Increasing BMI is one known risk factor for the development of postmenopausal hormone receptor-positive breast cancer as it is for endometrial cancer.67,68 The effect of overweight and obesity on cancer risk in premenopausal women is less clearly defined. While obesity among premenopausal women has been associated with an increased risk of colorectal cancer and possibly malignant melanoma,68 it has been reported in some, but not all studies to have an inverse association in terms of breast cancer risk. Among premenopausal women, estrogen is produced primarily in the ovaries. The transition into menopause is associated with a

10

decline of ovarian function, decreased estrogen production, and a relative shift in estrogen production to extragonadal sites such as adipose tissue. Peripheral conversion of androgens to estrogens is catalyzed by the enzyme aromatase, encoded by the CYP19 gene. Notably, the incidence of hormone receptor-positive breast cancer increases with age in spite of the decrease in circulating estrogen levels with menopause.69 Weight gain and central adiposity associated with menopause are thought to contribute to this seemingly paradoxical phenomenon as circulating estrogen levels are higher among overweight and obese versus normal weight postmenopausal women.70

Hormone receptor-positive breast cancer development and growth are believed to be dependent on locally produced estrogens, and, possibly, ligand-independent activation of estrogen receptor-α.71 Estrogens can directly stimulate cell division and proliferation, inhibit apoptosis, and induce angiogenesis.42,43 Multiple interactive signaling pathways involving estrogen and its receptors, insulin and IGF-1, and adipokines are also implicated in breast carcinogenesis. Furthermore, complex interactions between adipocytes and immune cells, such as macrophages, have been shown to result in local WAT inflammation (WATi) which appears to be associated with enhanced local estrogen receptor-alpha signaling.

11

Androgens.

The relationships among obesity, androgens, and tumorigenesis

represent an area of active investigation. While epidemiologic studies regarding obesity and risk of prostate cancer development have yielded mixed results, obesity has consistently been associated with poorer outcomes and higher grade disease.72-74 The underlying mechanisms for these observations remain incompletely understood. Inflammatory cytokines, e.g., IL-6, and IGF-1 have been implicated in the modulation of androgen receptor (AR) signaling in prostate cancer cell lines.75 Furthermore, in obese men, circulating levels of total testosterone are decreased76 suggesting potential ligand independent AR activation by cytokines or growth factors as a mechanism underlying the link between obesity and the development of high grade prostate cancer. Furthermore, obesity-induced WATi is associated with increased levels of proinflammatory mediators which may contribute to tumor formation and progression via either local or systemic interactions with the AR-signaling pathway.

Local and systemic inflammation Emerging data point to chronic inflammation as a potentially targetable risk factor for the development and progression of multiple malignancies.4,77,78 Obesity has been identified as a cause of both local and systemic inflammation. In this section, we will explore the complex local and systemic effects of obesity-induced

12

inflammation and how these processes promote malignant transformation and tumor progression.

The tumor microenvironment is comprised of stromal components including fibroblasts, blood and lymphatic vessels, and immune cells, e.g., macrophages and lymphocytes. Adipocytes are also present in the microenvironment of some tumor types. Similar to a wound, the microenvironment surrounding tumor cells is characterized by aggregation of immune cells and production of inflammatory cytokines.79 Infiltration of non-tumorous WAT by macrophages and lymphocytes has also been reported among obese individuals26 and may provide a proneoplastic microenvironment.80

As discussed earlier, WATi is well described as a driver of several obesityassociated diseases including NIDDM, dyslipidemia, and cardiovascular disease.26 Recently, there has been increased focus on the complex interactions between adipocytes and the immune cell component of the stromal-vascular fraction. Macrophages comprise up to 40% of the cells in obese WAT and produce several proinflammatory cytokines that contribute to obesity-related insulin resistance.81 Emerging data suggest a central role for the macrophageadipocyte interaction in connecting WATi and cancer promotion.

13

Increased BMI is associated with adipocyte hypertrophy and death. Adipocyte death is associated with increased production of chemokines and recruitment of macrophages.82 Infiltrating macrophages surround the dead or dying adipocyte and form a crown-like structure (CLS).83 This histologic structure was first described in the setting of metabolic disorders, such as diabetes, and more recently has been identified in both the mammary gland of obese mice and in human breast tissue (termed CLS-B).83,84 The presence of these inflammatory foci is associated with activation of NF-κB and increased levels of proinflammatory mediators including tumor necrosis factor α (TNFα), interleukin 1β (IL-1β), interleukin-6 (IL-6) and cyclooxygenase-2 (COX-2)-derived prostaglandin E2 (PGE2) (Figure 1). Importantly, increased circulating levels of proinflammatory mediators are commonly found in obese women and have been associated with breast cancer development and progression.85-87 Furthermore, gene expression profiling of breast tissue from obese women has demonstrated an excess of monocyte and macrophage-associated genes.88

The presence of CLS and the associated increase in tissue levels of proinflammatory mediators, are believed to stimulate transcription of CYP19 leading to increased expression of aromatase, the rate-limiting enzyme responsible for estrogen synthesis.89 This finding is consistent with prior evidence that proinflammatory mediators associated with WATi, including TNF-

14

α, COX-2, and IL-1β can induce aromatase.89-91 Through these pathways, WATi is believed to be associated with increased local production of estrogen thereby linking obesity with the development of hormone-sensitive breast cancers. Additionally, the postmenopausal state was recently shown to be associated with WATi, independently of BMI, which further supports the role of adipose inflammation in the development of postmenopausal hormone-sensitive breast cancers.92 Adipose inflammation may also be associated with ligand-independent activation of ER-signaling.

Collectively, these findings provide evidence that WATi, manifest as CLS-B, is likely to be a driver of estrogen-signaling in the breast. This highlights a potential role of WATi in breast cancer pathogenesis and may partially explain the seemingly paradoxical phenomenon of the increased incidence of estrogensensitive breast cancer after menopause.93,94

While both the presence and severity of WATi are associated with increased BMI,82,84,95 WATi also occurs in a minority of women (and men) with normal BMI.92 Furthermore, aromatase activity correlates more strongly with the severity of WATi than with BMI.84 These findings underscore the central role of inflammation, rather than obesity alone, as a driver of aromatase activity in the breast and highlight CLS as a potential actionable target in this setting. In

15

addition, the observation that WATi is present in some normal BMI individuals and is absent in a minority of obese individuals, is consistent with reports of obese individuals, albeit a minority, who are metabolically healthy.14-16 This point emphasizes the clinical significance of developing biomarkers of adipose inflammation that accurately select for at-risk individuals as we strive to develop effective prevention and management strategies.

Taken together, these findings support the existence of an obesity-inflammationaromatase axis active in the breasts of a majority of overweight and obese women and a smaller subset of normal weight women. However, the proinflammatory mediators that are enriched in inflamed WAT may also drive the development and progression of other subtypes of breast cancer and other tumor types. Reversing chronic inflammation associated with adipose tissue dysfunction may become a clearer goal as we gain a more comprehensive understanding of the complex signaling pathways underlying this process.

III.

Diagnostic and interventional strategies

Key to the development of effective risk reduction and therapeutic strategies will be the ability to accurately identify at-risk populations who harbor the detrimental biology discussed in the preceding section. Potential intervention strategies in this setting can be broadly grouped into the following categories: 1) preventing or

16

reversing increased adiposity; 2) targeting metabolic alterations associated with adipose tissue dysfunction; and 3) targeting white adipose tissue inflammation. A key challenge is identifying subsets of the population that can be both selected and possibly tracked to determine if a putative intervention is actually beneficial. Reliable surrogates that serve as early indicators of both risk and effective risk reduction are needed. Until we have such surrogates, we are forced to rely on broader population-based approaches.

Weight reduction is one potential strategy to suppress inflammation and consequently reduce cancer risk and improve prognosis. Weight loss achieved with a very low calorie diet has been shown to significantly improve the inflammatory gene expression profiles of obese subjects.96 Weight loss or maintenance of a normal BMI, achieved by dietary modification and/or physical activity, has been associated with improved outcomes after breast cancer diagnosis.97-105 Women randomized to the low fat diet group in the Women’s Intervention Nutrition Study lost an average of 2.77 kg and had a 24% reduction in the risk of breast cancer relapse.105 Preclinical studies have also demonstrated that calorie restriction halts weight gain in a mouse model of diet-induced obesity and is associated with a reduction in mammary gland inflammation, normalization of levels of proinflammatory mediators, and aromatase.106 Furthermore, several studies demonstrate significant associations between weight loss and

17

improvements in circulating levels of insulin, estrogens, and proinflammatory mediators.99,107-110 Bariatric surgery can result in significant and sustained weight loss and is associated with post-operative decreases in adipose tissue macrophage density, reduced tissue levels of chemokines involved in immune cell recruitment, and decreased cancer incidence and improved outcomes among female patients when compared to matched controls.111-113 Collectively, these findings suggest the potential utility of improved energy balance by limiting caloric intake as a strategy to reverse obesity-associated inflammation and potentially reduce cancer development or related mortality. However, achieving and maintaining an improved state of energy balance remains difficult and, as mentioned above, some normal BMI individuals harbor WATi, calling into question our ability to fully exploit caloric restriction as a general strategy to address inflammation. Several common medications have been investigated for their potential cancer chemopreventive properties that could be relevant in the context of obesity and inflammation.114-117 Notably, metformin, statins, nonsteroidal anti-inflammatory drugs (NSAIDs) and docosahexaenoic acid (DHA) have been extensively studied in several tumor types. Although retrospective observational studies have described reduced cancer incidence associated with several of these agents and preclinical studies highlight potential antineoplastic mechanisms, in general these agents have not consistently demonstrated anticancer effects in prospective studies. Numerous studies of metformin are ongoing (see Heckman-Stoddard et

18

al. in this volume). Docosahexaenoic acid (DHA), an omega-3 fatty acid, is thought to suppress inflammation by multiple mechanisms including inhibiting TLR4-activated signaling pathways that induce TNF-α and COX-2. Fatty fish and organ meats are the main sources of DHA, with smaller amounts coming from shellfish, eggs, and poultry. Diets high in fish oil have been associated with a reduced risk of breast cancer 118-121 and similar diets protect against experimental models of breast cancer.122,123 A multicenter, randomized, placebocontrolled phase II study in women with a history of premalignant breast lesions or invasive breast cancer, which is being conducted through the NCI/DCP MD Anderson Early Phase Chemoprevention Consortium, was designed to evaluate the anti-inflammatory effects of DHA on tissue level proinflammatory markers. This trial is currently open and enrolling patients (NCT01849250).124 The conflicting results reported in epidemiologic studies may be explained, in part by overtreatment resulting from the limitations associated with current risk stratification models. That is, until we can select the at-risk subset, we will be distributing an intervention across a broad population of potentially sensitive and insensitive patients, thereby diluting any potential favorable effect. This is why a key initiative must be the identification of high-risk populations to optimize the potential beneficial effects of these agents.93

19

IV.

Future Directions

It is clear that BMI alone is not an adequate surrogate of WAT health. As we have learned more about WATi, it has become clear that its associated metabolic aberrations are likely to go clinically unrecognized in large numbers of patients including a subset of individuals with normal BMI. A promising strategy to improve our current risk assessment abilities, would be the development of bloodbased biomarkers that can accurately identify dysfunctional and/or inflamed WAT. Such an approach could eliminate the need for serial invasive sampling of WAT to assess the impact of interventions and could identify individuals likely to benefit from mechanistically-based risk reduction and treatment strategies. Studies involving the collection of paired serum/plasma and WAT samples are ongoing, with the goal of developing a predictive model to accurately discriminate the presence or absence of WATi. The development of a predictive signature of WATi to aid in both patient selection and monitor response to treatment should be important in improving cancer prevention and post-diagnosis outcomes for the large and growing number of patients with adipose tissue dysfunction.

References 1.

World Health Organization: Obesity and overweight fact sheet.

http://www.who.int/mediacentre/factsheets/fs311/en/index.html, March 2011

20

2.

Levi J, Segal LM, St. Laurent R, et al: F as in Fat: How Obesity

Threatens America's Future 2012, Trust for America's Health, 2012 3.

Calle EE, Kaaks R: Overweight, obesity and cancer:

epidemiological evidence and proposed mechanisms. Nat Rev Cancer 4:579-91, 2004 4.

van Kruijsdijk RC, van der Wall E, Visseren FL: Obesity and

cancer: the role of dysfunctional adipose tissue. Cancer Epidemiol Biomarkers Prev 18:2569-78, 2009 5.

Polednak AP: Estimating the number of U.S. incident cancers

attributable to obesity and the impact on temporal trends in incidence rates for obesity-related cancers. Cancer Detection and Prevention 32:190-199, 2008 6.

Calle EE, Rodriguez C, Walker-Thurmond K, et al: Overweight,

obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med 348:1625-38, 2003 7.

Iyengar NM, Hudis CA, Dannenberg AJ: Obesity and

inflammation. Am Soc Clin Oncol Educ Book 2013:46-51, 2013 8.

Yoon HH, Lewis MA, Shi Q, et al: Prognostic impact of body

mass index stratified by smoking status in patients with esophageal adenocarcinoma. J Clin Oncol 29:4561-7, 2011 9.

Rajala MW, Scherer PE: Minireview: The adipocyte--at the

crossroads of energy homeostasis, inflammation, and atherosclerosis. Endocrinology 144:3765-73, 2003 10.

Guilherme A, Virbasius JV, Puri V, et al: Adipocyte dysfunctions

linking obesity to insulin resistance and type 2 diabetes. Nat Rev Mol Cell Biol 9:367-77, 2008 11.

Hursting SD, Digiovanni J, Dannenberg AJ, et al: Obesity, energy

balance, and cancer: new opportunities for prevention. Cancer Prev Res (Phila) 5:1260-72, 2012

21

12.

Hardy OT, Perugini RA, Nicoloro SM, et al: Body mass index-

independent inflammation in omental adipose tissue associated with insulin resistance in morbid obesity. Surg Obes Relat Dis 7:60-7, 2011 13.

Deepa M, Papita M, Nazir A, et al: Lean people with dysglycemia

have a worse metabolic profile than centrally obese people without dysglycemia. Diabetes Technol Ther 16:91-6, 2014 14.

Xu XJ, Gauthier MS, Hess DT, et al: Insulin sensitive and resistant

obesity in humans: AMPK activity, oxidative stress, and depot-specific changes in gene expression in adipose tissue. J Lipid Res 53:792-801, 2012 15.

Kloting N, Fasshauer M, Dietrich A, et al: Insulin-sensitive

obesity. Am J Physiol Endocrinol Metab 299:E506-15, 2010 16.

Denis GV, Obin MS: 'Metabolically healthy obesity': origins and

implications. Mol Aspects Med 34:59-70, 2013 17.

Arner P: Catecholamine-induced lipolysis in obesity. International

Journal of Obesity 23:10-13, 1999 18.

Vague J: The Degree of Masculine Differentiation of Obesities - a

Factor Determining Predisposition to Diabetes, Atherosclerosis, Gout, and Uric Calculous Disease. American Journal of Clinical Nutrition 4:20-34, 1956 19.

Khandekar MJ, Cohen P, Spiegelman BM: Molecular mechanisms

of cancer development in obesity. Nat Rev Cancer 11:886-95, 2011 20.

Pollak MN, Schernhammer ES, Hankinson SE: Insulin-like growth

factors and neoplasia. Nat Rev Cancer 4:505-18, 2004 21.

Goodwin PJ, Ennis M, Pritchard KI, et al: Fasting insulin and

outcome in early-stage breast cancer: results of a prospective cohort study. J Clin Oncol 20:42-51, 2002 22.

Ma J, Li H, Giovannucci E, et al: Prediagnostic body-mass index,

plasma C-peptide concentration, and prostate cancer-specific mortality in men with prostate cancer: a long-term survival analysis. Lancet Oncol 9:1039-47, 2008

22

23.

Barone BB, Yeh HC, Snyder CF, et al: Long-term all-cause

mortality in cancer patients with preexisting diabetes mellitus: a systematic review and meta-analysis. JAMA 300:2754-64, 2008 24.

Wong KK, Engelman JA, Cantley LC: Targeting the PI3K

signaling pathway in cancer. Curr Opin Genet Dev 20:87-90, 2010 25.

Zoncu R, Efeyan A, Sabatini DM: mTOR: from growth signal

integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol 12:21-35, 2011 26.

Rosen ED, Spiegelman BM: What we talk about when we talk

about fat. Cell 156:20-44, 2014 27.

Iyengar NM, Morris PG, Hudis CA, et al: Obesity, Inflammation,

and Breast Cancer, in Dannenberg AJ, Berger NA (eds): Obesity, Inflammation, and Cancer. Energy Balance and Cancer. New York, Springer, 2013, pp 181-217 28.

Hofbauer KG: Molecular pathways to obesity. Int J Obes Relat

Metab Disord 26 Suppl 2:S18-27, 2002 29.

Alberti KG, Eckel RH, Grundy SM, et al: Harmonizing the

metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation 120:1640-5, 2009 30.

Pothiwala P, Jain SK, Yaturu S: Metabolic syndrome and cancer.

Metab Syndr Relat Disord 7:279-88, 2009 31.

Capasso I, Esposito E, Pentimalli F, et al: Metabolic syndrome

affects breast cancer risk in postmenopausal women: National Cancer Institute of Naples experience. Cancer Biol Ther 10:1240-3, 2010 32.

Porto LA, Lora KJ, Soares JC, et al: Metabolic syndrome is an

independent risk factor for breast cancer. Arch Gynecol Obstet 284:1271-6, 2011

23

33.

Rosato V, Bosetti C, Talamini R, et al: Metabolic syndrome and

the risk of breast cancer in postmenopausal women. Ann Oncol 22:2687-92, 2011 34.

Tilg H, Moschen AR: Adipocytokines: mediators linking adipose

tissue, inflammation and immunity. Nat Rev Immunol 6:772-83, 2006 35.

Housa D, Housova J, Vernerova Z, et al: Adipocytokines and

cancer. Physiol Res 55:233-44, 2006 36.

Bub JD, Miyazaki T, Iwamoto Y: Adiponectin as a growth

inhibitor in prostate cancer cells. Biochem Biophys Res Commun 340:1158-66, 2006 37.

Kim AY, Lee YS, Kim KH, et al: Adiponectin represses colon

cancer cell proliferation via AdipoR1- and -R2-mediated AMPK activation. Mol Endocrinol 24:1441-52, 2010 38.

Cong L, Gasser J, Zhao J, et al: Human adiponectin inhibits cell

growth and induces apoptosis in human endometrial carcinoma cells, HEC-1-A and RL95 2. Endocr Relat Cancer 14:713-20, 2007 39.

Grossmann ME, Nkhata KJ, Mizuno NK, et al: Effects of

adiponectin on breast cancer cell growth and signaling. Br J Cancer 98:370-9, 2008 40.

Kamada Y, Matsumoto H, Tamura S, et al: Hypoadiponectinemia

accelerates hepatic tumor formation in a nonalcoholic steatohepatitis mouse model. Journal of Hepatology 47:556-564, 2007 41.

Fujisawa T, Endo H, Tomimoto A, et al: Adiponectin suppresses

colorectal carcinogenesis under the high-fat diet condition. Gut 57:1531-8, 2008 42.

Miyoshi Y, Funahashi T, Kihara S, et al: Association of serum

adiponectin levels with breast cancer risk. Clin Cancer Res 9:5699-704, 2003 43.

Soliman PT, Wu D, Tortolero-Luna G, et al: Association between

adiponectin, insulin resistance, and endometrial cancer. Cancer 106:2376-81, 2006

24

44.

Xu XT, Xu Q, Tong JL, et al: Meta-analysis: circulating

adiponectin levels and risk of colorectal cancer and adenoma. J Dig Dis 12:23444, 2011 45.

Michalakis K, Williams CJ, Mitsiades N, et al: Serum adiponectin

concentrations and tissue expression of adiponectin receptors are reduced in patients with prostate cancer: a case control study. Cancer Epidemiol Biomarkers Prev 16:308-13, 2007 46.

Li H, Stampfer MJ, Mucci L, et al: A 25-year prospective study of

plasma adiponectin and leptin concentrations and prostate cancer risk and survival. Clin Chem 56:34-43, 2010 47.

Kaklamani VG, Wisinski KB, Sadim M, et al: Variants of the

adiponectin (ADIPOQ) and adiponectin receptor 1 (ADIPOR1) genes and colorectal cancer risk. JAMA 300:1523-31, 2008 48.

Kaklamani VG, Sadim M, Hsi A, et al: Variants of the adiponectin

and adiponectin receptor 1 genes and breast cancer risk. Cancer Res 68:3178-84, 2008 49.

Kaklamani V, Yi N, Zhang K, et al: Polymorphisms of ADIPOQ

and ADIPOR1 and prostate cancer risk. Metabolism 60:1234-43, 2011 50.

Jarde T, Perrier S, Vasson MP, et al: Molecular mechanisms of

leptin and adiponectin in breast cancer. Eur J Cancer 47:33-43, 2011 51.

Fogarty S, Hardie DG: Development of protein kinase activators:

AMPK as a target in metabolic disorders and cancer. Biochim Biophys Acta 1804:581-91, 2010 52.

Jarde T, Caldefie-Chezet F, Damez M, et al: Leptin and leptin

receptor involvement in cancer development: a study on human primary breast carcinoma. Oncol Rep 19:905-11, 2008 53.

Friedman JM, Halaas JL: Leptin and the regulation of body weight

in mammals. Nature 395:763-70, 1998

25

54.

Sharma D, Saxena NK, Vertino PM, et al: Leptin promotes the

proliferative response and invasiveness in human endometrial cancer cells by activating multiple signal-transduction pathways. Endocr Relat Cancer 13:629-40, 2006 55.

Choi JH, Park SH, Leung PC, et al: Expression of leptin receptors

and potential effects of leptin on the cell growth and activation of mitogenactivated protein kinases in ovarian cancer cells. J Clin Endocrinol Metab 90:20710, 2005 56.

Hoda MR, Keely SJ, Bertelsen LS, et al: Leptin acts as a mitogenic

and antiapoptotic factor for colonic cancer cells. Br J Surg 94:346-54, 2007 57.

Onuma M, Bub JD, Rummel TL, et al: Prostate cancer cell-

adipocyte interaction: leptin mediates androgen-independent prostate cancer cell proliferation through c-Jun NH2-terminal kinase. J Biol Chem 278:42660-7, 2003 58.

Chia VM, Newcomb PA, Lampe JW, et al: Leptin concentrations,

leptin receptor polymorphisms, and colorectal adenoma risk. Cancer Epidemiol Biomarkers Prev 16:2697-703, 2007 59.

Uddin S, Bu R, Ahmed M, et al: Overexpression of leptin receptor

predicts an unfavorable outcome in Middle Eastern ovarian cancer. Molecular Cancer 8, 2009 60.

Ishikawa M, Kitayama J, Nagawa H: Enhanced expression of

leptin and leptin receptor (OB-R) in human breast cancer. Clinical Cancer Research 10:4325-4331, 2004 61.

Grossmann ME, Mizuno NK, Bonorden MJ, et al: Role of the

adiponectin leptin ratio in prostate cancer. Oncol Res 18:269-77, 2009 62.

Sharma D, Wang J, Fu PP, et al: Adiponectin antagonizes the

oncogenic actions of leptin in hepatocellular carcinogenesis. Hepatology 52:171322, 2010

26

63.

Ashizawa N, Yahata T, Quan JH, et al: Serum leptin-adiponectin

ratio and endometrial cancer risk in postmenopausal female subjects. Gynecologic Oncology 119:65-69, 2010 64.

Grossmann ME, Ray A, Nkhata KJ, et al: Obesity and breast

cancer: status of leptin and adiponectin in pathological processes. Cancer and Metastasis Reviews 29:641-653, 2010 65.

Mistry T, Digby JE, Desai KM, et al: Leptin and adiponectin

interact in the regulation of prostate cancer cell growth via modulation of p53 and bcl-2 expression. Bju International 101:1317-1322, 2008 66.

Chen J, Tan B, Karteris E, et al: Secretion of adiponectin by

human placenta: differential modulation of adiponectin and its receptors by cytokines. Diabetologia 49:1292-1302, 2006 67.

Cleary MP, Grossmann ME: Minireview: Obesity and breast

cancer: the estrogen connection. Endocrinology 150:2537-42, 2009 68.

Reeves GK, Pirie K, Beral V, et al: Cancer incidence and mortality

in relation to body mass index in the Million Women Study: cohort study. BMJ 335:1134, 2007 69.

Li CI, Daling JR, Malone KE: Incidence of invasive breast cancer

by hormone receptor status from 1992 to 1998. J Clin Oncol 21:28-34, 2003 70.

Key TJ, Appleby PN, Reeves GK, et al: Body mass index, serum

sex hormones, and breast cancer risk in postmenopausal women. J Natl Cancer Inst 95:1218-26, 2003 71.

Lorincz AM, Sukumar S: Molecular links between obesity and

breast cancer. Endocr Relat Cancer 13:279-92, 2006 72.

Giovannucci E, Rimm EB, Liu Y, et al: Body mass index and risk

of prostate cancer in U.S. health professionals. J Natl Cancer Inst 95:1240-4, 2003 73.

Jayachandran J, Banez LL, Aronson WJ, et al: Obesity as a

predictor of adverse outcome across black and white race: results from the Shared

27

Equal Access Regional Cancer Hospital (SEARCH) Database. Cancer 115:526371, 2009 74.

Parker AS, Thiel DD, Bergstralh E, et al: Obese men have more

advanced and more aggressive prostate cancer at time of surgery than non-obese men after adjusting for screening PSA level and age: results from two independent nested case-control studies. Prostate Cancer Prostatic Dis 16:352-6, 2013 75.

Lonergan PE, Tindall DJ: Androgen receptor signaling in prostate

cancer development and progression. J Carcinog 10:20, 2011 76.

Dhindsa S, Miller MG, McWhirter CL, et al: Testosterone

concentrations in diabetic and nondiabetic obese men. Diabetes Care 33:1186-92, 2010 77.

Olefsky JM, Glass CK: Macrophages, inflammation, and insulin

resistance. Annu Rev Physiol 72:219-46, 2010 78.

Pierce BL, Ballard-Barbash R, Bernstein L, et al: Elevated

biomarkers of inflammation are associated with reduced survival among breast cancer patients. J Clin Oncol 27:3437-44, 2009 79.

Mantovani A, Allavena P, Sica A, et al: Cancer-related

inflammation. Nature 454:436-44, 2008 80.

Coussens LM, Werb Z: Inflammation and cancer. Nature 420:860-

81.

Osborn O, Olefsky JM: The cellular and signaling networks

7, 2002

linking the immune system and metabolism in disease. Nat Med 18:363-74, 2012 82.

Monteiro R, Azevedo I: Chronic inflammation in obesity and the

metabolic syndrome. Mediators Inflamm 2010, 2010 83.

Subbaramaiah K, Howe LR, Bhardwaj P, et al: Obesity is

associated with inflammation and elevated aromatase expression in the mouse mammary gland. Cancer Prev Res (Phila) 4:329-46, 2011

28

84.

Morris PG, Hudis CA, Giri D, et al: Inflammation and increased

aromatase expression occur in the breast tissue of obese women with breast cancer. Cancer Prev Res (Phila) 4:1021-9, 2011 85.

Bachelot T, Ray-Coquard I, Menetrier-Caux C, et al: Prognostic

value of serum levels of interleukin 6 and of serum and plasma levels of vascular endothelial growth factor in hormone-refractory metastatic breast cancer patients. Br J Cancer 88:1721-6, 2003 86.

Dandona P, Weinstock R, Thusu K, et al: Tumor necrosis factor-

alpha in sera of obese patients: fall with weight loss. J Clin Endocrinol Metab 83:2907-10, 1998 87.

Vozarova B, Weyer C, Hanson K, et al: Circulating interleukin-6

in relation to adiposity, insulin action, and insulin secretion. Obes Res 9:414-7, 2001 88.

Sun X, Casbas-Hernandez P, Bigelow C, et al: Normal breast

tissue of obese women is enriched for macrophage markers and macrophageassociated gene expression. Breast Cancer Res Treat 131:1003-12, 2012 89.

Subbaramaiah K, Morris PG, Zhou XK, et al: Increased levels of

COX-2 and prostaglandin E2 contribute to elevated aromatase expression in inflamed breast tissue of obese women. Cancer Discov 2:356-65, 2012 90.

Zhao Y, Agarwal VR, Mendelson CR, et al: Estrogen biosynthesis

proximal to a breast tumor is stimulated by PGE2 via cyclic AMP, leading to activation of promoter II of the CYP19 (aromatase) gene. Endocrinology 137:5739-42, 1996 91.

Irahara N, Miyoshi Y, Taguchi T, et al: Quantitative analysis of

aromatase mRNA expression derived from various promoters (I.4, I.3, PII and I.7) and its association with expression of TNF-alpha, IL-6 and COX-2 mRNAs in human breast cancer. Int J Cancer 118:1915-21, 2006

29

92.

Iyengar NM, Morris PG, Zhou XK, et al: Menopause is a

Determinant of Breast Adipose Inflammation. Cancer Prev Res (Phila), 2015 93.

Hudis CA, Subbaramaiah K, Morris PG, et al: Breast cancer risk

reduction: no pain, no gain? J Clin Oncol 30:3436-8, 2012 94.

Howe LR, Subbaramaiah K, Hudis CA, et al: Molecular Pathways:

Adipose Inflammation as a Mediator of Obesity-Associated Cancer. Clin Cancer Res 19:6074-83, 2013 95.

Cinti S, Mitchell G, Barbatelli G, et al: Adipocyte death defines

macrophage localization and function in adipose tissue of obese mice and humans. J Lipid Res 46:2347-55, 2005 96.

Clement K, Viguerie N, Poitou C, et al: Weight loss regulates

inflammation-related genes in white adipose tissue of obese subjects. FASEB J 18:1657-69, 2004 97.

Ligibel JA, Goodwin PJ: NEW and RENEW: Building the Case

for Weight Loss in Breast Cancer. J Clin Oncol 30:2294-6, 2012 98.

Demark-Wahnefried W, Morey MC, Sloane R, et al: Reach out to

enhance wellness home-based diet-exercise intervention promotes reproducible and sustainable long-term improvements in health behaviors, body weight, and physical functioning in older, overweight/obese cancer survivors. J Clin Oncol 30:2354-61, 2012 99.

Campbell KL, Foster-Schubert KE, Alfano CM, et al: Reduced-

calorie dietary weight loss, exercise, and sex hormones in postmenopausal women: randomized controlled trial. J Clin Oncol 30:2314-26, 2012 100.

Chlebowski RT, Aiello E, McTiernan A: Weight loss in breast

cancer patient management. J Clin Oncol 20:1128-43, 2002 101.

Holmes MD, Chen WY, Feskanich D, et al: Physical activity and

survival after breast cancer diagnosis. JAMA 293:2479-86, 2005

30

102.

Holick CN, Newcomb PA, Trentham-Dietz A, et al: Physical

activity and survival after diagnosis of invasive breast cancer. Cancer Epidemiol Biomarkers Prev 17:379-86, 2008 103.

Irwin ML, Smith AW, McTiernan A, et al: Influence of pre- and

postdiagnosis physical activity on mortality in breast cancer survivors: the health, eating, activity, and lifestyle study. J Clin Oncol 26:3958-64, 2008 104.

Chen X, Lu W, Zheng W, et al: Exercise after diagnosis of breast

cancer in association with survival. Cancer Prev Res (Phila) 4:1409-18, 2011 105.

Chlebowski RT, Blackburn GL, Thomson CA, et al: Dietary fat

reduction and breast cancer outcome: interim efficacy results from the Women's Intervention Nutrition Study. J Natl Cancer Inst 98:1767-76, 2006 106.

Bhardwaj P, Du B, Zhou XK, et al: Caloric restriction reverses

obesity-induced mammary gland inflammation in mice. Cancer Prev Res (Phila) 6:282-9, 2013 107.

Bastard JP, Jardel C, Bruckert E, et al: Elevated levels of

interleukin 6 are reduced in serum and subcutaneous adipose tissue of obese women after weight loss. J Clin Endocrinol Metab 85:3338-42, 2000 108.

Esposito K, Pontillo A, Di Palo C, et al: Effect of weight loss and

lifestyle changes on vascular inflammatory markers in obese women: a randomized trial. JAMA 289:1799-804, 2003 109.

Imayama I, Ulrich CM, Alfano CM, et al: Effects of a caloric

restriction weight loss diet and exercise on inflammatory biomarkers in overweight/obese postmenopausal women: a randomized controlled trial. Cancer Res 72:2314-26, 2012 110.

van Rossum EF, Nicklas BJ, Dennis KE, et al: Leptin responses to

weight loss in postmenopausal women: relationship to sex-hormone binding globulin and visceral obesity. Obes Res 8:29-35, 2000

31

111.

Cancello R, Henegar C, Viguerie N, et al: Reduction of

macrophage infiltration and chemoattractant gene expression changes in white adipose tissue of morbidly obese subjects after surgery induced weight loss. Diabetes 54:2277-2286, 2005 112.

Adams TD, Stroup AM, Gress RE, et al: Cancer Incidence and

Mortality After Gastric Bypass Surgery. Obesity 17:796-802, 2009 113.

Sjostrom L: Review of the key results from the Swedish Obese

Subjects (SOS) trial - a prospective controlled intervention study of bariatric surgery. Journal of Internal Medicine 273:219-234, 2013 114.

Goodwin PJ, Pritchard KI, Ennis M, et al: Insulin-lowering effects

of metformin in women with early breast cancer. Clin Breast Cancer 8:501-5, 2008 115.

Dannenberg AJ, Altorki NK, Boyle JO, et al: Cyclo-oxygenase 2: a

pharmacological target for the prevention of cancer. Lancet Oncol 2:544-51, 2001 116.

Higgins MJ, Prowell TM, Blackford AL, et al: A short-term

biomarker modulation study of simvastatin in women at increased risk of a new breast cancer. Breast Cancer Res Treat 131:915-24, 2012 117.

Gucalp A, Morris PG, Zhou XK, et al: A multicenter phase II

study of docosahexaenoic acid (DHA) in patients (pts) with a history of breast cancer (BC), premalignant lesions, or benign breast disease. ASCO Meeting Abstracts 32:TPS1615, 2014 118.

Kaizer L, Boyd NF, Kriukov V, et al: Fish consumption and breast

cancer risk: an ecological study. Nutr Cancer 12:61-8, 1989 119.

Gago-Dominguez M, Yuan JM, Sun CL, et al: Opposing effects of

dietary n-3 and n-6 fatty acids on mammary carcinogenesis: The Singapore Chinese Health Study. Br J Cancer 89:1686-92, 2003

32

120.

Hirose K, Takezaki T, Hamajima N, et al: Dietary factors

protective against breast cancer in Japanese premenopausal and postmenopausal women. Int J Cancer 107:276-82, 2003 121.

Brasky TM, Lampe JW, Potter JD, et al: Specialty supplements

and breast cancer risk in the VITamins And Lifestyle (VITAL) Cohort. Cancer Epidemiol Biomarkers Prev 19:1696-708, 2010 122.

Rose DP, Connolly JM: Omega-3 fatty acids as cancer

chemopreventive agents. Pharmacol Ther 83:217-44, 1999 123.

Yee LD, Young DC, Rosol TJ, et al: Dietary (n-3) polyunsaturated

fatty acids inhibit HER-2/neu-induced breast cancer in mice independently of the PPARgamma ligand rosiglitazone. J Nutr 135:983-8, 2005 124.

Docosahexaenoic Acid in Preventing Recurrence in Breast Cancer

Survivors. https://clinicaltrials.gov/ct2/show/NCT01849250?term=DHA+breast+cancer&ran k=6, March 2015 125.

Iyengar NM, Hudis CA, Dannenberg AJ: Obesity and Cancer:

Local and Systemic Mechanisms. Annual Review of Medicine 66:297-309, 2015

33

Figure 1

Local and systemic mechanisms underlying obesity and cancer promotion. Reprinted with permission.125

34