Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Mar 29.
Published in final edited form as: Curr Opin Clin Nutr Metab Care. 2020 Nov;23(6):395–403. doi: 10.1097/MCO.0000000000000690

Current mechanisms in obesity and tumor progression

Andin Fosam a,b, Rachel J Perry a,b
PMCID: PMC10059279  NIHMSID: NIHMS1882574  PMID: 32868685

Abstract

Purpose of review

Hyperadiposity, as present in obesity, is a substantial threat to cancer risk and prognosis. Studies that have investigated the link between obesity and tumor progression have proposed several mechanistic frameworks, yet, these mechanisms are not fully defined. Further, a comprehensive understanding of how these various mechanisms may interact to create a dynamic disease state is lacking in the current literature.

Recent findings

Recent work has begun to explore not only discrete mechanisms by which obesity may promote tumor growth (for instance, metabolic and growth factor functions of insulin; inflammatory cytokines; adipokines; and others), but also how these putative tumor-promoting factors may interact.

Summary

This review will highlight the present understanding of obesity, as it relates to tumor development and progression. First, we will introduce the impact of obesity in cancer within the dynamic tumor microenvironment, which will serve as a theme to frame this review. The core of this review will discuss recently proposed mechanisms that implicate obesity in tumor progression, including chronic inflammation and the role of pro-inflammatory cytokines, adipokines, hormones, and genetic approaches. Furthermore, we intend to offer current insight in targeting adipose tissue during the development of cancer prevention and treatment strategies.

Keywords: cancer, cancer mechanisms, obesity, tumor microenvironment

INTRODUCTION

Obesity, characterized as a BMI of 30 or more and typically because of hyperadiposity [1], is a growing global disease and a risk factor for many complications, including metabolic syndrome, hepatic steatosis, cardiovascular disease, diabetes, poor bone health, and cancer [25]. Although epidemiologic links between obesity and cancer have been established, the pathophysiological mechanisms are not clear. Pro-inflammatory processes and obesity-related endocrine disorders have been widely studied in breast cancer, a tumor type, which is linked to obesity [6], but these mechanisms are not well defined in other cancer types. Further, although several physiologic links are offered, the molecular interactions underlying those mechanisms are still widely unknown.

Current studies suggest that most mechanisms underlying tumor growth are because of tumor microenvironment alterations [7]. This is often manifested as an upregulation of various factors including inflammatory cytokines and immune cells (IL-6, IL-1β, T cells, B cells, macrophages), disruption of normal extracellular matrix (ECM), and endothelial cell function [8].

In obesity, both the number and size of adipocytes increases, leading to chronic subclinical inflammation [9,10]. This low-grade inflammatory state in adipose tissue is comparable to the tumor microenvironment (TM), which may suggest its ability to support tumor growth. For example, recruitment of inflammatory factors and stimulation of the coagulation cascade is activated in obesity, as it is in the TM [2,11].

Here, we aim to compile current literature regarding obesity-related tumor progression. We discuss the present understanding of the mechanisms fundamental to these links within the framework of the TM. To conclude, we address current investigational therapeutics and other areas that warrant further investigation.

TUMOR MICROENVIRONMENT AND OBESITY

The TM is composed of diverse constituents, like cytotoxic immune cells, and antigen-presenting cells, like macrophages, which interact generating an inflamed state similar to an injured tissue [11]. During uncontrolled cell proliferation, cancer cells hijack the regulatory systems of noncancerous cells [9], initiating an infiltration of pro-inflammatory factors, like cytokines and growth factors, immune cells and injury-response processes like the coagulation cascade and platelet activation factors, often leading to endothelial dysfunction and fibrosis [10,12]. Breast cancer progression is sustained by hypoxia-related oxidative stress and infiltration of pro-inflammatory mediators, such as TNF-α, IL-6, and cyclooxygenase-2 (COX-2) [13].

Metabolic reprogramming within the TM also plays a role in tumor growth. The Warburg effect is a phenomenon whereby a tumor cell favors glycolysis over oxidative phosphorylation even in an aerobic environment [9]. Although glycolysis provides less ATP per glucose molecule, tumor cells rely on aerobic glycolysis to garner building blocks necessary for cell division and metastasis [14] as well as to promote lactate secretion. Changes to signaling cues within oncogenic pathways allow the tumor cell to outsource materials into the TM for survival, as well as to expand within surrounding tissues.

Obesity leads to chronic inflammation and an inappropriate secretion of adipokines and hormones, including leptin, adiponectin, and estrogen [15]. This altered, often unchecked, state within adipose tissue creates an inflamed environment susceptible to tumor infiltration. Chronic inflammation in adipose tissue allows cancer cells to co-opt pro-inflammatory substrates and tissue repair mechanisms to feed into their own energy demands [11], as discussed in the following section.

ADIPOSE TISSUE INFLAMMATION AND TUMOR GROWTH

Adipose tissue is a dynamic endocrine organ, broadly characterized as white (WAT) or brown (BAT) adipose tissue [2]. WAT is further distinguished as visceral (situated around ectopic organs) and subcutaneous (distributed under the skin)WAT. Activity of these fat depots is largely guided by their location. Initially considered solely an energy reservoir, adipose tissue is vital to maintaining energy homeostasis, immune function, and reproduction, and mediating systemic interactions involving lipid metabolism, insulin sensitivity, and angiogenesis [10,16].

Figure 1 describes key alterations to adipose tissue that occur in obesity. adipose tissue dysfunction appears early in obesity and often includes WAT hypoxia [2,10]. A dysfunctional adipose tissue environment is much like a TM, leaving adjacent tissues and organs susceptible to infiltration and damage [17]. Interactions between adipose tissue and tumor cells allow tumor cells to acquire key energy substrates, such as lactate, fatty acids, and amino acids [2]. In breast cancer, hypoxic conditions within chronic adipose tissue inflammation leads to the activation of hypoxia-induced factor-1, which has been associated with poor outcomes in obese breast cancer patients [18]. Hypoxic conditions also drive leptin secretion from adipose tissue [2]. The role of leptin as a driver of tumor progression will be more comprehensively discussed later.

FIGURE 1.

FIGURE 1.

Obesity and adipose tissue inflammation. Obesity-induced inflammation in adipose tissue leads to dysfunction of several processes in adipose tissue. Obesity-induced inflammation increases in both the size and number of adipocytes, leading structural and functional damage, hypoxia, oxidative stress and increased cancer risk. Increase in hormone signaling leads to upregulation of metabolic pathways like JAK/STAT3 and AMPK. Inflamed adipose tissue incites abnormal adipokine secretion, including leptin and adiponectin. In a state of chronic injury, inflamed adipose tissue is infiltrated by pro-inflammatory factors, such as macrophages, IL-6 and TNFα. Systemic changes in obesity such as increased insulin resistance and cancer risk are mediated by this altered adipose tissue state. IL-6, interleukin-6; TNFα, tumor necrosis factor-α.

As adipose tissue dysfunction persists, disordered functioning is coupled with chronic tissue injury, activating proinflammatory processes that support tumor growth and metastasis (Fig. 2). WAT inflammation has been widely regarded as a key factor in tumor proliferation in multiple cancer types [10,17]. Adipose tissue inflammation arises and is perpetuated through increases in macrophages, lymphocytes, and mediators, such as IL-1β, COX-2, and TNFα. Abnormal expression and functioning of these molecules alter secretion of adipokines, which leads to dysregulation of key signaling pathways, like JAK/STAT3 and P13K. Further, pro-inflammatory factors can be used by adjacent tumor sites as substrates that feed into and support the tumor ecosystem [16,19].

FIGURE 2.

FIGURE 2.

Adipose tissue inflammation and the tumor microenvironment. In the presence of the tumor microenvironment, the obesity-induced altered adipose tissue state becomes a source of energy substrates and structural support for the growing tumor. Tumor tissue induce a transformation from adipocytes to CAAs where resources are shuttled to the tumor. Various mechanisms support tumor sustenance and metastasis, including increased adipocyte lipolysis, secretion of inflammatory cytokines, and metabolic reprogramming. The inflamed adipose tissue creates a pro-tumor environment where the tumor can adopt a more aggressive and invasive phenotype. AT, adipose tissue; CAA, cancer-associated adipocyte.

Cross-talk between inflamed WAT and cancer cells not only provides an avenue to siphon energy resources and metabolites to the tumor but also affects the TM itself. Under normal conditions, WAT plays a crucial role in secreting structural and mechanical support factors for the ECM surrounding adipose tissue. However, adipose tissue inflammation can lead to endocrine receptor stress and ECM stiffness, triggering fibrosis [10]. In the presence of a breast tumor, molecules secreted by the WAT, like collagen type VI, promote tumor progression [20]. Inflamed WAT secretion of these mediators in the presence of tumorcells also leads to destruction of the adipose tissue: matrix metalloproteinases (MMPs) actively suppress adipocyte differentiation and promote dedifferentiation, allowing tumor invasion.

AT inflammation in obesity contributes to tumor progression through the development of cancer-associated adipocytes (CAA) [18]. Due to pressure from malignant cells, CAAs repurpose adipocyte-derived factors like leptin and fatty acids in favor of tumor development [18]. Cancer cells take up CAA-secreted factors, which in turn promote metastasis [17]. CAAs’ function to sustain inflammation and fibrosis is often retained even when engulfed by the tumor. They lead to increases in IL-6, MMP-11, and HIF-1, all of which promote tumor progression [16,17]. In melanoma, CAAs are linked to malignant cells via exosomes. These membrane-bound vesicles are used to transfer functional material from the CAA to the tumor cell [18].

ADIPOKINES AND HORMONES IN CANCER

Excess adipose tissue contributes to the TM and tumor progression via secretion of endocrine-signaling factors. Among known adipokines, leptin and adiponectin are widely implicated as two prominent factors in tumorigenesis, especially as inflammatory mediators.

Leptin

Elevated leptin is derived from WAT, is almost universally present in obesity, and is responsible for regulating energy homeostasis by controlling food intake [2,21], and energy expenditure [22]. In addition, leptin contributes to tumor initiation and metastasis in various cancer types, including breast cancer, hepatocellular carcinoma, lung cancer, prostate cancer, colorectal cancer, and renal carcinoma [13,23] – most of which, interestingly, are obesity-associated tumor types. Recent studies have targeted overexpression of leptin and its receptor in its promotion of tumor migration, interactions with the ECM, and the epithelial–mesenchymal transition (EMT) [23]. Further, leptin has been shown to lead to the degradation of ECM components, via activation of proteolytic cleavage enzymes [23]. There are numerous studies correlating leptin to breast cancer, in which leptin effects pathways including JAK/STAT3, MAPK, and AMPK, promoting cancer proliferation [2]. Upregulation of these pathways leads to enhanced expression of tumorigenesis genes [20]. Additionally, leptin has been shown to participate in breast cancer cell growth by enhancing estrogen action via crosstalk with aromatase [24]. Current data demonstrate that leptin upregulates antiapoptotic proteins, inflammatory markers, and hypoxic factors, as well as potentiate estrogen signaling [8,13,25,26]. As breast cancer exists in an adipose tissue-rich milieu, leptin is a particularly attractive pro-cancer factor in this tumor type.

Adiponectin

Secreted by the adipose tissue, adiponectin improves insulin sensitivity and suppresses inflammation [15], but its levels decline in obesity. Adiponectin is a protective factor against multiple cancer types, including endometrial, prostate, thyroid, and ovarian cancer [15,2729], each of which has been associated with obesity (endometrial [3032], prostate [33,34], thyroid [35,36], ovarian [32]). Decreased local and intra-tumor adiponectin also promotes breast cancer via reduced inhibition of energy metabolism pathways, like AMPK/MTOR/SK6 [20,28,37]. Although adiponectin-based therapies have been posed as a potential tool in cancer treatment, the paradigm that adiponectin inhibits tumor growth must be further investigated.

Estrogen

The systemic effects of estrogen are primarily regulated by estrogen receptors α and β (ER-α and ER-β). The obesity-induced hyper-inflammatory state alters estrogen production and signaling, which leads to DNA damage, cell proliferation, angiogenesis, and mutagenesis in multiple cancers [8]. Increased estrogen bioavailability and estrogen signaling is variable and seemingly dependent on the tumor type. In breast cancer, ER-α activation leads to targeted expression of genes and metabolic pathways that regulate cell survival, growth, and plasticity, including upregulation of P13K and MAPK [38,39]. Similarly, ER-α promotes aggressive tumor phenotypes in endometrial carcinoma via stromal cell interactions [40]. Recent evidence has linked ER-α to promoting cell proliferation and migration in prostate cancer [41,42]. ER-α is suggested to be a prognostic marker of these cancer types. The role of ER-β in cancer disorders is less clear but is suggested to be antiproliferative and tumor-suppressive. There is evidence that ER-β-induced apoptosis and autophagy inhibits metastasis in osteosarcoma through regulation of various molecular factors, such as inhibitors of apoptotic proteins (IAP), caspase-3, LC3, and p62 [43]. In breast cancer, ER-β supports the EMT and perhaps reduces ER-α induced tumor proliferation and tumor growth [44]. ER-β also plays a protective role in colon cancer, where it is essential for maintaining cellular homeostasis, as wells as activating p53 signaling and increased apoptosis [44]. Further, within the TM, coordination between ER-α and ER-β lead to remodeling of the ECM by inducing changes in the expression profiles of ECM mediators, including MMPs and tyrosine kinase receptors [44].

Insulin

Hyperinsulinemia is common in obese individuals, usually caused by insulin resistance, and poses unique risks in cancer appearance and metastasis. Elevated insulin frequently occurs in concert with elevated insulin-like growth factor-1 (IGF-1). IGF-1 has antiapoptotic properties and also activates the P13K and MAPK pathways, all of which are cancer-promoting [2,7,8]. Obese patients with breast cancer who had poorer survival outcomes also presented with increased insulin and IGF-1 [8]. Insulin also interacts with leptin, adiponectin, and estrogen signaling to induce tumor promotion and survival via these metabolic pathways. Further, hyperinsulinemia is pro-inflammatory, leading to disruption to the TM, as described above. This state of metabolic instability and high oxidative stress is not remediated via processes that limit insulin utilization by other tissues (i.e. impaired insulin sensitivity), and consequently promotes tumor survival [45].

Targeting hyperinsulinemia within obesity-based cancers has provided some insight to potential insulin-dependent tumor growth mechanisms and insulin-based therapies. Huang et al. [46] suggest that hyperinsulinemia inhibits tumor autophagy. In hyperinsulinemic conditions, hepatocellular carcinoma cells exhibited increased growth, proliferation and invasion into HepG2 cells, in a concentration-dependent manner [46]. Hyperinsulinemia has been implicated in obesity-associated pancreatic cancer [47] via P13K/AKT and ERK1/2 activation in human pancreatic duct-derived cells, which led to tumor cell migration and invasion and reduced cell survival [48]. Rabin-Court et al. [49▪▪] suggest that obesity-related tumors can be distinguished by their physiologic response to insulin. When grown in insulin, the obesity-related (breast [6,32], colon [50,51], and prostate [33,34]) cancer cell lines increased glucose uptake and oxidation as well as cell division, in contrast to nonobesity-related (melanoma, lymphoma, and lung) cell lines, in which insulin did not affect glucose metabolism or cell division [49▪▪]. Nasiri et al. [52▪▪] describe an insulin-dependent promotion of breast and colon cancer progression in obese mice, which was reduced by SGLT2 inhibition in an insulin-dependent manner, suggesting a potential therapeutic role of glucose and insulin-lowering agents. Further, hyperinsulinemia and hyperglycemia promoted oncogenic signaling in bladder epithelial cells and were not abrogated by insulin-sensitizing and glucose-lowering agents, metformin and pioglitazone. Certain amino-acid signatures, like increased glutamine, glycine and serine, have also been proposed to mediate the links between insulin and obesity-related cancers [53].

Lipid metabolism and free fatty acids

Obesity is associated with increased lipid metabolism, leading to higher circulating free fatty acids (FFA). Excess circulating FFAs are transported to the cancer cell and used as a metabolic substrate and/or as a structural element in the proliferating cancer membrane [10]. Fatty acid oxidation, induced by STAT3 activation, has been shown to be crucial in breast cancer tumor growth [54]. Further, cancer cells induce lipid metabolism from compromised adipocytes and CAAs and store them as long-chain fatty acids [2]. FFAs are increased in mice with prostate cancer, which was associated with increased tumor growth [55]. Similarly, Ha et al. [56] found a positive association between circulating FFA and cancer proliferation, migration and invasion. They postulate that this effect is mediated by an upregulation of peroxisome proliferator-activated receptor gamma, which promotes glucose and lipid metabolism in favor of the tumor cell [56].

MOLECULAR IMPLICATIONS

Genetic and molecular mechanisms, like long non-coding RNAs (lncRNA), are also implicated in tumor progression. LncRNAs, transcribed by RNA polymerase, were shown to be dysregulated in obese patients [14,57]. Oncogenic lncRNAs (e.g. h19, ANRIL, and HOTAIR) have been previously identified, and are well characterized as regulators of nutrient availability in obesity-related tumors [5760]. Recent in-vitro and in-vivo studies show upregulation of cancer-associated lncRNAs within obesity-related tumors leading to tumor metastasis [6169]. Liu et al. show that expression of lncRNAs for genes related to tumor proliferation were upregulated and genes responsible for mediating normal metabolic functioning and oxidative stress were down-regulated, suggesting potential effects at the genetic and epigenetic level [70]. In addition to the target genes of the predominant metabolic pathways in energy homeostasis (JAK/STAT, AMPK, MTOR), upregulation of fibroblast, inflammatory, ECM-specific genes are shown to be implicated in tumor pathogenesis [71,72▪▪,73].

FUTURE CONSIDERATIONS

The recent studies highlighted above have provided crucial insight into the pathophysiological mechanism linking obesity to tumor progression (Fig. 3). However, a majority of these studies explore only a few main obesity-related cancers (breast cancer, colon cancer, pancreatic cancer). Future research is needed to understand these mechanisms in the context of other obesity-related cancers. Additionally, work is needed to clarify the molecular interactions among these implicated mechanisms to better understand how they interact to form the dynamic disease state. As highlighted in Table 1, studies that outline potential therapeutics to combat tumor progression in obesity-related cancers help link known mechanism to clinical progress but extensiveness of these studies is lacking.

FIGURE 3.

FIGURE 3.

Mechanisms by which adipocyte dysfunction may promote tumor growth. In obesity, the hyperplastic, hypertrophic adipocytes – including CAAs – promote inflammation, ECM remodeling, and oxidative stress. These phenomena promote a variety of pathologic properties, including increases in adipokine and cytokine secretion, increased circulating insulin, leptin, and estrogen concentrations, and long noncoding RNAs. Together, each of these perturbations has been proposed to promote tumor pathogenesis. CAA, cancer-associated adipocyte; ECM, extracellular matrix; FFA, free fatty acids; lncRNAs, long noncoding RNAs.

Table 1.

Current findings: obesity and cancer therapeutics

Study Cancer type Subjects Study design Intervention Details

Cohen et al. [74] Ovarian/endometrial F (n = 45) Randomized case-controlled Ketogenic diet Ketogenic diet resulted in fat loss but retention in lean mass. Increased circulating ketones are suggested to be inhospitable to tumor microenvironment.
Cortesi et al. [75] Breast F (n = 430), obese Single-arm, experimental Weight loss, physical activity Improved survival outcomes in obese women following a lifestyle intervention including weight loss and physical activity
Wang et al. [76▪▪] LLC C57BL/6 mice, HFD-fed, injected with LLC In vivo Aspirin Aspirin reduced tumor growth in obese mice via targeting of glucose and glutamine metabolism
Pabona et al. [77] Endometrial Endometrial tumor cells derived from obese-nondiabetic women In vitro Metformin Metformin-treated EC tumor cells showed increased antitumor biomarkers via altered signaling
Guo et al. [78] Endometrial LKB1fl/fl p53fl/fl mouse model of endometrial cancer In vivo Metformin Metformin reversed upregulation of lipid and protein biosynthesis in obese mice
Dos Santos et al. [79] Melanoma Female HFD-fed mice In vivo Physical Activity Tumor growth rate reduced with implementation of an exercise regimen
Zell et al. [80] Colorectal M (n = 23);F (n = 9), obese nondiabetic Phase II clinical trial Metformin No differences in premetformin versus postmetformin rectal tissue mucosa following 12-week intervention
Jiang et al. [81] Pancreatic Human pancreatic BxPC-3 and CFPAC-1 cells In vitro Adiponectin Activation of adiponectin signaling attenuates cell proliferation and β-catenin signaling
Shafa et al. [82] Endometrial Obese woman, 54 years old Case study Surgery Combination of a hysterectomy and bariatric surgery resulted in sustained weight loss and improved tumor pathology
Demark-Wahnefried et al. [83] Breast F (n = 32), obese Single blind, two-arm, randomized case-controlled Weight Loss Inconsistent effects caloric restriction of circulating cancer-related biomarkers. Improvement of P13K signaling and apoptotic gene expression after physical activity
Hsieh et al. [84] Breast 4T1 breast cancer cells In vitro Aspirin Aspirin attenuated breast cancer cell growth and migration by inhibiting obesity-induced inflammation
Hao et al. [55] Prostate LAPC-4-implanted mice In vivo Arctigenin Arctigenin inhibits tumor growth in HF-fed mice. Induced reduction in serum FFA and subcutaneous fat depots
Yang et al. [85] Obesity-related Cancers Review Herbal Medications Review discusses novel findings regarding the antiproliferative effects of herbal medications including PD-L1 and resveratrol in obesity-related cancers
Kato et al. [86] Colorectal C57BL/KsJ–db/db mice In vivo Tofogliflozin Suppression of colorectal cancer tumorigenesis after SGLT2-inhibitor treatment in azoxymethane-injected obese mice

Recent publications exploring cancer therapeutics in obese populations. EC, endometrial cancer; F, female; FFA, free fatty acid; HF, high fat; HFD, high-fat diet; LLC, Lewis lung carcinoma; M, male; PD-L1, programmed death-ligand 1; SGLT2, sodium-glucose transport protein 2.

CONCLUSION

In summary, there are several intertwined mechanisms that link obesity to tumor development and progression. Many of the involved factors, like chronic inflammation and upregulation of key circulating molecules, induce instability in the TM, promoting tumor survival. Upregulated adipokines and circulatory factors, like leptin, adiponectin, insulin and estrogen, also enhance tumor growth through activation of target genes and metabolic pathways, like JAK/STAT3, P13K/AKT, and MAPK. These recent findings lay groundwork to understanding the role of obesity in tumor growth and to developing targeted therapies.

KEY POINTS.

  • Obesity impacts tumor progression via several mechanisms, including chronic inflammation, upregulation of secreted adipokines and circulatory factors, and genetic associations.

  • Many of these mechanisms coexist and interact to alter the tumor microenvironment in support of tumor growth and metastasis.

  • Therapeutic frameworks targeting these mechanisms in obesity-related cancers provide insight to tumor suppression but warrant further investigation.

Financial support and sponsorship

This work was supported by grants from the U.S. Public Health Service (K99/R00 CA215315, Medical Scientist Training Grant T32GM007205), a Lion Heart Pilot Award, and a Yale SPORE in Melanoma Award.

Footnotes

Conflicts of interest

There are no conflicts of interest.

REFERENCES AND RECOMMENDED READING

Papers of particular interest, published within the annual period of review, have been highlighted as:

▪ of special interest

▪▪ of outstanding interest

  • 1.Niemiro GM, Rewane A, Algotar AM. Obesity, exercise and fitness. Treasure Island, Florida: StatPearls; 2020. [PubMed] [Google Scholar]
  • 2.Jiramongkol Y, Lam EW. Multifaceted oncogenic role of adipocytes in the tumour microenvironment. Adv Exp Med Biol 2020; 1219:125–142. [DOI] [PubMed] [Google Scholar]
  • 3.Jung SY, Kim YA, Jo M, et al. Prediagnosis obesity and secondary primary cancer risk in female cancer survivors: a national cohort study. Cancer Med 2019; 8:824–838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kwon H, Chang Y, Cho A, et al. Metabolic obesity phenotypes and thyroid cancer risk: a cohort study. Thyroid 2019; 29:349–358. [DOI] [PubMed] [Google Scholar]
  • 5.Sharma DK, Anderson PH, Morris HA, Clifton PM. Visceral fat is a negative determinant of bone health in obese postmenopausal women. Int J Environ Res Public Health 2020; 17:3996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kolb R, Zhang W. Obesity and breast cancer: a case of inflamed adipose tissue. Cancers (Basel) 2020; 12:1686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Hopkins BD, Goncalves MD, Cantley LC. Insulin-PI3K signalling: an evolutionarily insulated metabolic driver of cancer. Nat Rev Endocrinol 2020; 16:276–283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Atoum MF, Alzoughool F, Al-Hourani H. Linkage between obesity leptin and breast cancer. Breast Cancer (Auckl) 2020; 14:1178223419898458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Park J, Morley TS, Kim M, et al. Obesity and cancer–mechanisms underlying tumour progression and recurrence. Nat Rev Endocrinol 2014; 10:455–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Pallegar NK, Christian SL. Adipocytes in the tumour microenvironment. Adv Exp Med Biol 2020; 1234:1–13. [DOI] [PubMed] [Google Scholar]
  • 11.Iyengar NM, Gucalp A, Dannenberg AJ, Hudis CA. Obesity and cancer mechanisms: tumor microenvironment and inflammation. J Clin Oncol 2016; 34:4270–4276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kompella P, Vasquez KM. Obesity and cancer: a mechanistic overview of metabolic changes in obesity that impact genetic instability. Mol Carcinog 2019; 58:1531–1550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sanchez-Jimenez F, Perez-Perez A, de la Cruz-Merino L, Sanchez-Margalet V. Obesity and breast cancer: role of leptin. Front Oncol 2019; 9:596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Li Z, Sun X. Non-coding RNAs operate in the crosstalk between cancer metabolic reprogramming and metastasis. Front Oncol 2020; 10:810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Tumminia A, Vinciguerra F, Parisi M, et al. Adipose tissue; obesity and adiponectin: role in endocrine cancer risk. Int J Mol Sci 2019; 20:2863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zewdu A, Casadei L, Pollock RE, Braggio D. Adipose tumor microenvironment. Adv Exp Med Biol 2020; 1226:73–86. [DOI] [PubMed] [Google Scholar]
  • 17. Wang YX, Zhu N, Zhang CJ, et al. Friend or foe: multiple roles of adipose tissue in cancer formation and progression. J Cell Physiol 2019; 234:21436–21449. ▪ Insightful discussion of risks and potential benefits of hyperadiposity to tumor growth.
  • 18. Munteanu R, Onaciu A, Moldovan C, et al. Adipocyte-based cell therapy in oncology: the role of cancer-associated adipocytes and their reinterpretation as delivery platforms. Pharmaceutics 2020; 12:402. ▪ Discussion of cancer-associated adipocytes and potential therapeutic roles.
  • 19.Rasha F, Ramalingam L, Menikdiwela K, et al. Renin angiotensin system inhibition attenuates adipocyte-breast cancer cell interactions. Exp Cell Res 2020; 394:112114. [DOI] [PubMed] [Google Scholar]
  • 20.Christodoulatos GS, Spyrou N, Kadillari J, et al. The role of adipokines in breast cancer: current evidence and perspectives. Curr Obes Rep 2019; 8:413–433. [DOI] [PubMed] [Google Scholar]
  • 21.Perry RJ, Resch JM, Douglass AM, et al. Leptin’s hunger-suppressing effects are mediated by the hypothalamic-pituitary-adrenocortical axis in rodents. Proc Natl Acad Sci U S A 2019; 116:13670–13679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Perry RJ, Lyu K, Rabin-Court A, et al. Leptin mediates postprandial increases in body temperature through hypothalamus-adrenal medulla-adipose tissue crosstalk. J Clin Invest 2020; 130:2001–2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ghasemi A, Saeidi J, Azimi-Nejad M, Hashemy SI. Leptin-induced signaling pathways in cancer cell migration and invasion. Cell Oncol (Dordr) 2019; 42:243–260. [DOI] [PubMed] [Google Scholar]
  • 24.Engin AB, Engin A, Gonul II. The effect of adipocyte-macrophage crosstalk in obesity-related breast cancer. J Mol Endocrinol 2019; 62:R201–R222. [DOI] [PubMed] [Google Scholar]
  • 25.Singh S, Mayengbam SS, Chouhan S, et al. Role of TNFalpha and leptin signaling in colon cancer incidence and tumor growth under obese phenotype. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165660. [DOI] [PubMed] [Google Scholar]
  • 26.Angelucci A, Clementi L, Alesse E. Leptin in tumor microenvironment. Adv Exp Med Biol 2020; 1259:89–112. [DOI] [PubMed] [Google Scholar]
  • 27.Ellis PE, Barron GA, Bermano G. Adipocytokines and their relationship to endometrial cancer risk: a systematic review and meta-analysis. Gynecol Oncol 2020; 158:507–516. [DOI] [PubMed] [Google Scholar]
  • 28.Ando S, Naimo GD, Gelsomino L, et al. Novel insights into adiponectin action in breast cancer: evidence of its mechanistic effects mediated by ERalpha expression. Obes Rev 2020; 21:e13004. [DOI] [PubMed] [Google Scholar]
  • 29.Hu X, Hu C, Zhang C, et al. Role of adiponectin in prostate cancer. Int Braz J Urol 2019; 45:220–228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.McDonald ME, Bender DP. Endometrial cancer: obesity, genetics, and targeted agents. Obstet Gynecol Clin North Am 2019; 46:89–105. [DOI] [PubMed] [Google Scholar]
  • 31.Yang X, Wang J. The role of metabolic syndrome in endometrial cancer: a review. Front Oncol 2019; 9:744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ishihara BP, Farah D, Fonseca MCM, Nazario A. The risk of developing breast, ovarian, and endometrial cancer in obese women submitted to bariatric surgery: a meta-analysis. Surg Obes Relat Dis 2020; 80:In Press. [DOI] [PubMed] [Google Scholar]
  • 33.Narita S, Nara T, Sato H, et al. Research evidence on high-fat diet-induced prostate cancer development and progression. J Clin Med 2019; 8:597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Fujita K, Hayashi T, Matsushita M, et al. Obesity, inflammation, and prostate cancer. J Clin Med 2019; 8:201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Matrone A, Ferrari F, Santini F, Elisei R. Obesity as a risk factor for thyroid cancer. Curr Opin Endocrinol Diabetes Obes 2020. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 36.Harikrishna A, Ishak A, Ellinides A, et al. The impact of obesity and insulin resistance on thyroid cancer: a systematic review. Maturitas 2019; 125:45–49. [DOI] [PubMed] [Google Scholar]
  • 37.Yu Z, Tang S, Ma H, et al. Association of serum adiponectin with breast cancer: a meta-analysis of 27 case-control studies. Medicine (Baltimore) 2019; 98:e14359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Vonka P, Hrstka R. Overview of current findings about the role of oestrogen receptor alpha in cancer cell signalling pathways. Klin Onkol 2019; 32(Suppl 3):34–38. [DOI] [PubMed] [Google Scholar]
  • 39.Bhardwaj P, Au CC, Benito-Martin A, et al. Estrogens and breast cancer: mechanisms involved in obesity-related development, growth and progression. J Steroid Biochem Mol Biol 2019; 189:161–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Yoriki K, Mori T, Kokabu T, et al. Estrogen-related receptor alpha induces epithelial-mesenchymal transition through cancer-stromal interactions in endometrial cancer. Sci Rep 2019; 9:6697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Lombardi APG, Vicente CM, Porto CS. Estrogen receptors promote migration, invasion and colony formation of the androgen-independent prostate cancer cells PC-3 through beta-catenin pathway. Front Endocrinol (Lausanne) 2020; 11:184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Valcarcel-Jimenez L, Macchia A, Crosas-Molist E, et al. PGC1alpha suppresses prostate cancer cell invasion through ERRalpha transcriptional control. Cancer Res 2019; 79:6153–6165. [DOI] [PubMed] [Google Scholar]
  • 43. Yang ZM, Yang MF, Yu W, Tao HM. Molecular mechanisms of estrogen receptor beta-induced apoptosis and autophagy in tumors: implication for treating osteosarcoma. J Int Med Res 2019; 47:4644–4655. ▪ Introduces increased autophagy as a down-stream effect of altered estrogen signaling in tumors.
  • 44.Piperigkou Z, Karamanos NK. Estrogen receptor-mediated targeting of the extracellular matrix network in cancer. Semin Cancer Biol 2020; 62:116–124. [DOI] [PubMed] [Google Scholar]
  • 45.Schwartz SS, Grant SFA, Herman ME. Intersections and clinical translations of diabetes mellitus with cancer promotion, progression and prognosis. Postgrad Med 2019; 131:597–606. [DOI] [PubMed] [Google Scholar]
  • 46.Huang J, Liu Y, Liu T, et al. Dual-targeting nanotherapeutics antagonize hyperinsulinemia-promoted tumor growth via activating cell autophagy. J Mater Chem B 2019; 7:6751–6758. [DOI] [PubMed] [Google Scholar]
  • 47.Brocco D, Florio R, De Lellis L, et al. The role of dysfunctional adipose tissue in pancreatic cancer: a molecular perspective. Cancers (Basel) 2020; 12:E1849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wang G, Yin L, Peng Y, et al. Insulin promotes invasion and migration of KRAS(G12D) mutant HPNE cells by upregulating MMP-2 gelatinolytic activity via ERK- and PI3K-dependent signalling. Cell Prolif 2019; 52:e12575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Rabin-Court A, Rodrigues MR, Zhang XM, Perry RJ. Obesity-associated, but not obesity-independent, tumors respond to insulin by increasing mitochondrial glucose oxidation. PLoS One 2019; 14:e0218126. ▪▪ Discusses associative role of the metabolic effect of obesity in hyperinsulinemic states.
  • 50.Kern L, Mittenbuhler MJ, Vesting AJ, et al. Obesity-induced TNFalpha and IL-6 signaling: the missing link between obesity and inflammation-driven liver and colorectal cancers. Cancers (Basel) 2018; 11:24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Tarasiuk A, Mosinska P, Fichna J. The mechanisms linking obesity to colon cancer: an overview. Obes Res Clin Pract 2018; 12:251–259. [DOI] [PubMed] [Google Scholar]
  • 52. Nasiri AR, Rodrigues MR, Li Z, et al. SGLT2 inhibition slows tumor growth in mice by reversing hyperinsulinemia. Cancer Metab 2019; 7:10. ▪▪ Suggests SGLT2 inhibitors as a potential obesity-related cancer therapeutic.
  • 53.Dioguardi FS, Flati V, Corsetti G, et al. Is the response of tumours dependent on the dietary input of some amino acids or ratios among essential and non-essential amino Acids? All that glitters is not gold. Int J Mol Sci 2018; 19:3631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Zhang C, Yue C, Herrmann A, et al. STAT3 activation-induced fatty acid oxidation in CD8(þ) T effector cells is critical for obesity-promoted breast tumor growth. Cell Metab 2020; 31:148.e5–61.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Hao Q, Diaz T, Verduzco ADR, et al. Arctigenin inhibits prostate tumor growth in high-fat diet fed mice through dual actions on adipose tissue and tumor. Sci Rep 2020; 10:1403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Ha X, Wang J, Chen K, et al. Free fatty acids promote the development of prostate cancer by upregulating peroxisome proliferator-activated receptor gamma. Cancer Manag Res 2020; 12:1355–1369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Yau MY, Xu L, Huang CL, Wong CM. Long non-coding RNAs in obesity-induced cancer. Noncoding RNA 2018; 4:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Chen TJ, Gao F, Yang T, et al. LncRNA HOTAIRM1 inhibits the proliferation and invasion of lung adenocarcinoma cells via the miR-498/WWOX axis. Cancer Manag Res 2020; 12:4379–4390. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 59.Kong Y, Hsieh CH, Alonso LC. ANRIL: a lncRNA at the CDKN2A/B locus with roles in cancer and metabolic disease. Front Endocrinol (Lausanne) 2018; 9:405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Mai H, Zhou B, Liu L, et al. Molecular pattern of lncRNAs in hepatocellular carcinoma. J Exp Clin Cancer Res 2019; 38:198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Wang J, Huang F, Shi Y, et al. RP11–323N12.5 promotes the malignancy and immunosuppression of human gastric cancer by increasing YAP1 transcription. Gastric Cancer 2020. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 62.Han Y, Sun G. Overexpression of lncRNA TINCR is associated with high-grade, invasive, and recurring tumors, and facilitates proliferation in vitro and in vivo of urothelial carcinoma of the bladder. Urol Oncol 2020; 38:738.e1–738.e8. [DOI] [PubMed] [Google Scholar]
  • 63.Lin C, Xiang Y, Sheng J, et al. Long noncoding RNA CRNDE promotes malignant progression of hepatocellular carcinoma through the miR-33a-5p/CDK6 axis. J Physiol Biochem 2020; 76:469–481. [DOI] [PubMed] [Google Scholar]
  • 64.Jin W, Zhang H, Li M, Lin S. Long noncoding RNA regulator of reprogramming regulates cell growth, metastasis, and cisplatin resistance in gastric cancer via miR-519d-3p/HMGA2 axis. Cancer Biother Radiopharm 2020. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 65.Li D, Wen S. Silencing of lncRNA LINC00346 inhibits the proliferation and promotes the apoptosis of colorectal cancer cells through inhibiting JAK1/STAT3 signaling. Cancer Manag Res 2020; 12:4605–4614. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 66.Sun W, Nie W, Wang Z, et al. Lnc HAGLR promotes colon cancer progression through sponging miR-185–5p and activating CDK4 and CDK6 in vitro and in vivo. Onco Targets Ther 2020; 13:5913–5925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Liu X, Zhu Q, Guo Y, et al. LncRNA LINC00689 promotes the growth, metastasis and glycolysis of glioma cells by targeting miR-338–3p/PKM2 axis. Biomed Pharmacother 2019; 117:109069. [DOI] [PubMed] [Google Scholar]
  • 68.Wang Y, Yang L, Chen T, et al. A novel lncRNA MCM3AP-AS1 promotes the growth of hepatocellular carcinoma by targeting miR-194–5p/FOXA1 axis. Mol Cancer 2019; 18:28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wang F, Rong L, Zhang Z, et al. LncRNA H19-derived miR-675–3p promotes epithelial-mesenchymal transition and stemness in human pancreatic cancer cells by targeting the STAT3 pathway. J Cancer 2020; 11:4771–4782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Liu L, He C, Liu H, et al. Transcriptomic profiling of long noncoding RNAs in nonvirus associated hepatocellular carcinoma. Cell Biochem Biophys 2020. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 71.Asakawa M, Itoh M, Suganami T, et al. Upregulation of cancer-associated gene expression in activated fibroblasts in a mouse model of nonalcoholic steatohepatitis. Sci Rep 2019; 9:19601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Boughanem H, Cabrera-Mulero A, Hernandez-Alonso P, et al. The expression/methylation profile of adipogenic and inflammatory transcription factors in adipose tissue are linked to obesity-related colorectal cancer. Cancers (Basel) 2019; 11:1629. ▪▪ Offers insight to the genetic implications of hyperadiposity in colorectal cancer.
  • 73.Hermano E, Goldberg R, Rubinstein AM, et al. Heparanase accelerates obesity-associated breast cancer progression. Cancer Res 2019; 79: 5342–5354. [DOI] [PubMed] [Google Scholar]
  • 74.Cohen CW, Fontaine KR, Arend RC, et al. A ketogenic diet reduces central obesity and serum insulin in women with ovarian or endometrial cancer. J Nutr 2018; 148:1253–1260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Cortesi L, Sebastiani F, Iannone A, et al. Lifestyle intervention on body weight and physical activity in patients with breast cancer can reduce the risk of death in obese women: the EMILI Study. Cancers (Basel) 2020; 12:1709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Wang JD, Chen WY, Li JR, et al. Aspirin mitigated tumor growth in obese mice involving metabolic inhibition. Cells 2020; 9:569. ▪▪ Aspirin reduced tumor growth in obese mouse models of LLC, generally known as a nonobesity-related cancer.
  • 77.Pabona JMP, Burnett AF, Brown DM, et al. Metformin promotes antitumor biomarkers in human endometrial cancer cells. Reprod Sci 2020; 27: 267–277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Guo H, Kong W, Zhang L, et al. Reversal of obesity-driven aggressiveness of endometrial cancer by metformin. Am J Cancer Res 2019; 9:2170–2193. [PMC free article] [PubMed] [Google Scholar]
  • 79.Dos Santos CMM, Diniz VLS, Bachi ALL, et al. Moderate physical exercise improves lymphocyte function in melanoma-bearing mice on a high-fat diet. Nutr Metab (Lond) 2019; 16:63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Zell JA, McLaren CE, Morgan TR, et al. A phase IIa trial of metformin for colorectal cancer risk reduction among individuals with history of colorectal adenomas and elevated body mass index. Cancer Prev Res (Phila) 2020; 13:203–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Jiang J, Fan Y, Zhang W, et al. Adiponectin suppresses human pancreatic cancer growth through attenuating the beta-catenin signaling pathway. Int J Biol Sci 2019; 15:253–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Shafa A, Kumar A, Torres D, McKenzie TJ. Minimally invasive hysterectomy and bariatric surgery to improve endometrial cancer survivorship. Obstet Gynecol 2019; 134:570–572. [DOI] [PubMed] [Google Scholar]
  • 83.Demark-Wahnefried W, Rogers LQ, Gibson JT, et al. Randomized trial of weight loss in primary breast cancer: impact on body composition, circulating biomarkers and tumor characteristics. Int J Cancer 2020; 146:2784–2796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Hsieh CC, Chiu HH, Wang CH, Kuo CH. Aspirin modifies inflammatory mediators and metabolomic profiles and contributes to the suppression of obesity-associated breast cancer cell growth. Int J Mol Sci 2020; 21:4652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Yang YSH, Li ZL, Shih YJ, et al. Herbal medicines attenuate PD-L1 expression to induce anti-proliferation in obesity-related cancers. Nutrients 2019; 11:2979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kato J, Shirakami Y, Ohnishi M, et al. Suppressive effects of the sodiumglucose cotransporter 2 inhibitor tofogliflozin on colorectal tumorigenesis in diabetic and obese mice. Oncol Rep 2019; 42:2797–2805. [DOI] [PubMed] [Google Scholar]

RESOURCES