Next Article in Journal
A Topic Modeling Analysis of TCGA Breast and Lung Cancer Transcriptomic Data
Next Article in Special Issue
Deciphering the Role of Ca2+ Signalling in Cancer Metastasis: From the Bench to the Bedside
Previous Article in Journal
Microwave Ablation Versus Radiofrequency Ablation for Treatment of Hepatocellular Carcinoma: A Meta-Analysis of Randomized Controlled Trials
Previous Article in Special Issue
Prostate Cancer Metastases Are Strongly Inhibited by Agonistic Epha2 Ligands in an Orthotopic Mouse Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Obesity and Cancer Metastasis: Molecular and Translational Perspectives

School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Science, 123 St Stephen’s Green, D02 YN77 Dublin, Ireland
*
Author to whom correspondence should be addressed.
Submission received: 15 November 2020 / Revised: 11 December 2020 / Accepted: 14 December 2020 / Published: 16 December 2020
(This article belongs to the Special Issue Targeting Cancer Metastasis)

Abstract

:

Simple Summary

A major challenge in treating cancer is when the cancer spreads from its original site to other parts of the body, a process also known as metastasis. In order to survive, the cancer cells must communicate with their environment for survival. It is increasingly recognized that fat tissue supports the survival of metastatic cancer cells allowing tumors to form at distant sites. Obese patients therefore have a worse prognosis due to accelerated tumor spread. Preventing cancer cells from communicating with fat tissue could therefore lead to new treatments aimed at stopping this spread. In this review we discuss how dysfunctional fat tissue supports the metastatic process and evaluate new therapies and lifestyle interventions that aim to prevent the communication between cancer cells and fat tissue.

Abstract

Obesity is a modern health problem that has reached pandemic proportions. It is an established risk factor for carcinogenesis, however, evidence for the contribution of adipose tissue to the metastatic behavior of tumors is also mounting. Over 90% of cancer mortality is attributed to metastasis and metastatic tumor cells must communicate with their microenvironment for survival. Many of the characteristics observed in obese adipose tissue strongly mirror the tumor microenvironment. Thus in the case of prostate, pancreatic and breast cancer and esophageal adenocarcinoma, which are all located in close anatomical proximity to an adipose tissue depot, the adjacent fat provides an ideal microenvironment to enhance tumor growth, progression and metastasis. Adipocytes provide adipokines, fatty acids and other soluble factors to tumor cells whilst immune cells infiltrate the tumor microenvironment. In addition, there are emerging studies on the role of the extracellular vesicles secreted from adipose tissue, and the extracellular matrix itself, as drivers of obesity-induced metastasis. In the present review, we discuss the major mechanisms responsible for the obesity–metastatic link. Furthermore, understanding these complex mechanisms will provide novel therapies to halt the tumor–adipose tissue crosstalk with the ultimate aim of inhibiting tumor progression and metastatic growth.

1. Introduction

The metabolic and cardiovascular risks of obesity are well known. However, it is estimated that 40% of all cancer deaths are also attributable to obesity [1]. Indeed, globally, excess body weight is third behind smoking and infection as an attributable risk factor for cancer, and second to smoking in Western populations [2]. Obesity adversely effects cancer in two ways, (i) by promoting carcinogenesis resulting in a higher cancer incidence and (ii) cancer progression resulting in an increased risk of mortality [3]. In breast cancer, for example, obesity is only associated with an increased incidence of post-menopausal breast cancer, whilst obesity is a risk factor for progression in all breast cancer subtypes [4]. The global obesity rate in women is projected to reach 21% by 2025 and this is particularly alarming considering that 55% of all female cancers have an obesity associated mechanism [3]. Central obesity, resulting from the overgrowth of visceral white adipose tissue (WAT), has been specifically linked to cancer progression [5]. While diet is undoubtedly important in obesity, animal models have indicated that WAT overgrowth directly promotes cancer progression irrespective of diet [3,6]. Epidemiological studies have demonstrated a compelling association between cancer risk and obesity. Analysis of the association between body mass index (BMI) and early stage breast cancer outcomes in the Danish Breast Cancer Cooperative Group (n = 53,816 women) revealed that obese women have a 46% higher risk of developing distinct metastasis at 10 year follow up compared to normal weight women [7]. Furthermore, a meta-analysis of 82 studies found a 41% and 35% higher risk, respectively, of all-cause mortality and breast cancer specific mortality in obese women compared to normal weight women [2]. An umbrella review of 204 meta-analyses revealed a strong association between obesity and gastrointestinal cancers including esophageal adenocarcinoma (OAC) [8]. OAC was notable for a progressive increase in risk ratio (RR) for each 5 kg/m2 increase in BMI (RR 4.8 for a BMI ≥ 40 kg/m2) suggesting a dose–response effect [9]. The top two cancers demonstrating very strong associations with BMI are endometrial (RR 1.48) and OAC (RR 1.45) [10].
In obesity, white adipocytes become hypertrophic and hyperplasic, which results in physiologic changes, including elevated free fatty acids (FFAs) and triglycerides, increased blood glucose and insulin resistance. Obese adipose tissue increases production of proinflammatory cytokines, e.g., tumor necrosis factor (TNFα), interleukin-6 (IL-6), interleukin-1β (IL-1β) and adipokines (e.g., leptin) [3]. Metastasis is the primary cause of cancer morbidity and mortality and efforts to unravel the molecular mechanisms linking dysfunctional adipose tissue and the ability of tumor cells to acquire metastatic properties will lead to the discovery of novel targets for metastasis. Here we review the recent findings regarding the molecular processes underpinning the impact of adipose tissue on cancer metastasis and potential therapeutic interventions.

2. The Metastatic Cascade

The “hallmarks of cancer” define characteristics that are critical for cellular transformation [11]. Among the hallmarks there is only one defining factor, invasion, which distinguishes a malignant and benign tumor [11]. The metastatic cascade begins with local invasion before progressing to intravasation, arrest at distant organs, extravasation, micro metastasis formation and finally metastatic outgrowth (Figure 1).
In the early stages of tumor development, the primary tumor cell mass typically has an expansive phase in the absence of invasion, encapsulated in a dense fibrous network (i.e., desmoplasia) [12]. A subset of neoplastic cells acquire the ability to escape through the basement membrane and detach from the primary tumor [12]. The dissemination of cancer cells is a consequence of chromosomal instability that causes continuous errors in chromosome segregation during mitosis [13]. This in turns leads to the rupture of micronuclei and the secretion of genomic DNA into the cytosol, which activates DNA sensing pathways and NF-κB signaling [13]. In addition, epithelial–mesenchymal transition (EMT) is a transdifferentiating process, which permits epithelial cells to attain a mesenchymal phenotype with migratory potential [14]. Spontaneous EMT in primary tumor cells can be triggered by hypoxia, metabolic stressors and matrix stiffness [15,16]. Although some studies have cast doubts on the necessity of EMT during metastasis [17,18], there is evidence for cells expressing both epithelial and mesenchymal markers within the primary tumor, circulation and at a secondary metastatic site [19]. Therefore EMT is being increasingly understood as a spectrum of transitional stages between epithelial and mesenchymal phenotypes rather than a binary choice between an epithelial or mesenchymal phenotype [20,21]. There is evidence that cells in different EMT stages prefer certain microenvironments, for example, metastatic cells with a predominating mesenchymal phenotype proliferate near endothelial and inflammatory cells [21]. These tumor cells release factors to attract immune cells and stimulate angiogenesis, thus promoting an inflammatory and highly vascularized niche [21]. Although EMT is required for metastatic dissemination, the opposite process of mesenchymal to epithelial transition (MET) is required for metastatic colonization of distant sites [22]. There is also substantial evidence that disseminated tumor cells express stem cell markers, such as aldehyde dehydrogenase (ALDH), and functionally these cells, highly enriched in stem cells markers, have an enhanced ability to cause metastasis [19,23,24,25]. Furthermore, genome wide analysis of both cells undergoing EMT and circulating tumor cells has revealed a similar transcriptome to primary cancer stem cells (CSCs) thus indicating an overlapping subpopulation [19,26,27]. In pancreatic cancer, primary CD133+ CSCs demonstrated classic CSC characteristics such as tumor initiation and chemoresistance [28]. However, at the invasive front of the tumor CD133+ cells are enriched for CXC-chemokine receptor 4 (CXCR4) and the CD133+CXCR4+ population is more migratory than CD133+CXCR4-- [28]. Moreover, patients with increased CD133+CXCR4+ cells had more metastatic disease [28]. This indicates that microenvironmental cues within the tumor can trigger heterogeneity in CSCs and CD133+CXCR4+ and CD133+CXCR4- are not a distinct subpopulations but a gradient of stemness phenotypes [19,28]. CSCs are more resistant to chemotherapy due to higher expression of multidrug resistance (MDR) or detoxification proteins such as aldehyde dehydrogenase (ALDH) [29].
Prior to exiting the primary tumor mass, tumor cells communicate with other microenvironments, termed the premetastatic niche, and this niche is selectively primed by secreted factors and extracellular vesicles to induce vascular leakage, extra cellular matrix (ECM) remodeling and immunosuppression [30]. Cancer cells also adjust the niche themselves by remodeling the ECM leading to stromal tumorigenesis [20]. Cancer patients release large numbers of cancer cells into the circulation daily, however animal studies of melanoma suggest that <0.1% of these cells metastasize [31]. The most widely studied route of dissemination is through the bloodstream (hematogenous), however metastatic cells may also migrate along nerves, lymphatic vessels, across coelomic cavities or along the basal side of endothelial cells and never enter the lumen [12]. Intravasation, the dissemination of cancer cells to organs through the lumen of the vasculature can be active or passive depending on the tumor type, microenvironment and vasculature [32]. The migration of metastatic cells into the circulation relies on chemokines and complement components that direct tumor cells through the vasculature and metabolic factors that result in an antioxidant effect [20]. Furthermore, the dissemination of circulating tumor cells (CTCs) is supported by a close association with immune cells such as activated platelets, macrophages and neutrophils [20]. When CTCs pass into small capillaries they become trapped leading to microvascular rupture or the cell undergoes extravasation [20]. Establishing a vascular network is required for metastatic colonization and this can occur though angiogenesis, co-opting existing vessels or inducing vasculogenic mimicry [20,33]. Furthermore, cancer cells can also exploit neuronal signaling pathways for growth and adaptation [20]. Cancer dormancy is an arrest phase that can occur after invasion into secondary sites and in some cancer survivors, dormant cells result in relapse long after the removal of the primary tumor [20,34]. The subsequent reactivation is governed by self-renewal pathways (e.g., Wnt, Hedgehog and Notch) and cells exhibit increased levels of stem cell associated genes [35]. While dormant cancer cells downregulate the expression of immune cell recognizable antigens, persistent host organ inflammation and the establishment of neutrophils in extracellular traps may transform dormant cells into aggressive metastasis [35,36]. In many of the steps from dissemination to colonization, the microenvironment plays a major role and this can be significantly altered by dysfunctional adipose tissue in people living with obesity.

3. Mechanisms Linking Adipose Tissue to the Metastatic Cascade

3.1. Adipocytes and Adipokines

Adipocytes secrete more than 600 soluble factors, known as adipokines, and the most well characterized are leptin and adiponectin [37]. Intra-abdominal cancers such as ovarian, colon and gastric, preferentially metastasize to the omentum, a peritoneal organ largely composed of adipocytes, suggesting that adipocytes significantly contribute to the metastatic cascade [38]. Omental adipocytes promote migration and invasion of ovarian cancer cells by secreting cytokines [39]. Neutralization of these cytokines reduced in vivo homing of ovarian cancer cells to mouse omentum, suggesting that adipocytes promote the early stages of metastasis [39]. In addition, ovarian cancer cells metabolically adapt to the increased availability of lipids by utilizing energy from fatty acids for growth [39]. Fatty acid binding protein 4 (FABP4) was strongly expressed at the adipocyte–cancer cell interface and pharmacological inactivation of FABP4 decreased cancer cell lipid accumulation, invasiveness and omental metastasis [39]. Circulating levels of FABP4 are markedly increased in obese individuals due to release from an expanded adipose tissue depot and FABP4 can induce mammary tumor stem cells by enhancing ALDH1 activity via IL-6/STAT3 signaling [40,41]. In addition, FABP4 promoted aggressive acute myeloid leukemia (AML) in obesity by enhancing aberrant DNA methyltransferase 1 (DNMT1)-dependent DNA methylation of tumor cells [42]. Upon interaction with cancer cells, adipocytes dedifferentiate into preadipocytes or are reprogrammed into cancer-associated adipocytes (CAAs), which resemble fibroblasts and have dispersed lipid droplets [43]. In breast cancer, matrix metalloproteinase (MMP)-11 is induced in adipocytes by adjacent invading cancer cells. In the presence of MMP-11, the activated adipocyte dedifferentiates into a preadipocyte fibroblast-like cell, which can sustain cancer cell invasion [44,45]. Bone marrow adipocytes constitute approximately 15% of bone marrow volume in young adults, rising to 60% by the age of 65 years old [46]. They have a distinctive phenotype, which resembles both white and brown adipose tissue, and they secrete fatty acids, cytokines and adipokines, which influence the whole bone microenvironment [47]. The bone provides a supportive microenvironment for both solid tumor and hematological metastasis, including breast, prostate and multiple myeloma and bone metastatic cancers primarily occur in older adults whose bone marrow is heavily populated by adipocytes [48,49]. Similarly to the omentum, cancer cells are attracted to the adipocytes in the metabolically active red marrow and this creates a niche in the bone marrow for disseminated cancer cells to establish and progress [48]. In addition, leukemic stem cells expressing the fatty acid transporter CD36, induce lipolysis in gonadal adipose tissue to support their metabolism and evade chemotherapy [50]. Lipids can also be trafficked between bone marrow adipocytes and cancer cells by upregulation of FABP4 and fuel growth and invasiveness in metastatic tumor cells [51]. In addition, bone marrow adipocytes have been shown to promote the Warburg phenotype in metastatic prostate cancer cells through oxygen independent HIF-1α activation [52]. Bone marrow adipocytes are a major source of circulating adiponectin, much greater than WAT [53]. Adiponectin is reported to suppress many elements of the early metastatic cascade including adhesion, invasion, migration and stem cell properties via numerous signaling pathways including WNT, NF-κB and JAK/STAT [54]. In advanced cancer associated with cachexia, hyperadiponectinemia has been observed [55]. In addition, increased adiponectin signaling in dendritic cells can blunt anti-tumor immune responses in patients with metastatic disease [56]. However, the late increase in adiponectin has very little influence on the course of the disease, as its role is thought to be more prominent in early metastatic spread [54]. During the development of obesity, preadipocytes differentiate incorrectly leading to hypoxia and the induction of hypoxia induced factor-1 (HIF-1) [57]. This inhibits the expression of adiponectin and increases the expression of leptin, resulting in a reduced adiponectin to leptin ratio in obesity-associated adipose tissue [58,59]. A high leptin to adiponectin ratio has been reported to increase the risk of postmenopausal and triple negative breast cancer (TNBC) progression [60,61]. Leptin is another adipokine important in tumor progression and secretion of leptin is increased in CAAs compared to mature adipocytes [43]. Leptin levels are increased in the plasma of post-menopausal breast cancer patients, which correlated with a higher grade, advanced tumor stages and presence of distant metastasis [37]. Leptin exerts it effect through the transmembrane leptin receptor and only the full length isoform, ObRb, contains the intracellular domain required for JAK/STAT signaling [62]. ObRb was significantly overexpressed in metastatic lymph nodes compared to the primary tumor in ER-breast cancer patients [63]. Furthermore, TNBC patient derived xenografts (PDX) grown in the presence of primary adipose stem cells (ASCs) isolated from obese donors (obASCs) had increased HLA1+ human tumor cells and CD44+CD24 CSCs in the peripheral blood and metastasis compared to ASCs from lean women [64]. In addition, the knockdown of leptin expression in obASCs suppressed the prometastatic effect [64]. There is a plethora of evidence that leptin induces cell migration and invasion in breast cancer via JAK/STAT3 signaling [62]. In addition, the Notch signaling pathway is a key regulator of leptin induced cell migration in breast cancer obesity models [63]. Leptin has also been shown to promote the CSC phenotype though STAT3 activation and this in turn recruited a histone methyl transferase causing a repression of miR-200c by epigenetic silencing and the expansion of CSCs [65]. Autotaxin (ATX) is an adipocyte-derived lysophospholipase D that catalyzes the hydrolysis of circulating or cell associated lysophosphatidylcholine (LPC) to the bioactive lipid lysophosphatidic acid (LPA) [66]. Some tumor cells, such as in melanoma, glioblastoma and thyroid cancer, directly secrete ATX leading to a chronic inflammatory state and decreased acquired immune response [67]. In contrast, other tumor cells, such as breast, ovarian and pancreatic do not produce ATX and instead the tumor microenvironment is the primary source [67]. Interestingly, the ATX gene (ENPP2) was the second most upregulated gene in breast CSCs treated with paclitaxel whilst the LPP2 gene (PLPP2) was downregulated, indicating CSCs may favor an LPA-enriched microenvironment [68]. In a tissue microarray (TMA) of metastatic breast cancer, stromal ATX was highly expressed in bone metastasis [69]. In mice approximately 40% of ATX is produced by adipocytes and this increases when mice are fed a high fat diet [70]. Similarly, in humans ATX production increases in obesity, particularly in inflamed adipose tissue, and contributes to comorbidities including insulin resistance, hepatic steatosis and atherosclerosis [66,71,72,73]. In breast cancer cells both ATX and LPA are associated with mobility and invasive capacity via the JAK/STAT3 pathway or PI3K/MAPK pathways [74,75]. Therefore cross talk between adipose tissue derived ATX and tumors cells is a potential mechanism for tumor progression [76,77]. Indeed preclinical studies of an ATX inhibitor, ONO-843050 suppresses tumor growth and 60% of lung metastasis in a breast cancer mouse model [78]. Targeting other molecules in the ATX-LPA signaling pathways also results in decreased breast cancer metastasis formation in murine models [77,79,80]. The physiological upregulation of ATX occurs in response to inflammation and chronic activation of ATX-LPA signaling occurs in diseases such as pulmonary fibrosis, rheumatoid arthritis and inflammatory bowel diseases [81]. A first in class ATX inhibitor, GLPG1690, attenuated idiopathic pulmonary fibrosis in a Phase IIa clinical trial (NCT02738801) and two Phase III clinical trials are currently underway for this indication (NCT03711162; NCT03733444) [82,83]. In a preclinical breast cancer model, GLPG1690 acted synergistically with chemotherapy and radiotherapy to improve outcomes [84]. Considering the role of ATX in regulating CSCs and mobility, targeting ATX with inhibitors such as GLPG1690 may also prove to be beneficial in targeting metastasis, particularly in patients living with obesity, by preventing adipose tissue cross talk.

3.2. Immune Cells and Inflammatory Factors

CAAs secrete more inflammatory factors, such as monocyte chemoattractant protein (MCP-1), RANTES, IL-1β, IL-6 and TNFα than “normal” adipocytes and this can promote invasion and metastasis formation [43,85]. Furthermore, the recruitment and activation of immune cell subsets, particularly M1 macrophages, in obese WAT increases local and systemic levels of proinflammatory cytokines [86]. MCP-1 is elevated in obesity and secreted by many cells including tumor cells, fibroblasts, tumor infiltrating monocytes and endothelial cells [43,87]. In cancer cells, MCP-1 induces the expression of NOTCH1 and subsequently promotes the activity of CSCs and neovascularization [87,88]. RANTES expression in the peritumoral adipose tissue of women with TNBC correlated with lymph node and distant metastasis [89]. Furthermore, the RANTES inhibitors, maraviroc and vicriciroc, reduced invasion and pulmonary metastasis in a preclinical tumor model of breast cancer [90]. IL-6 is a pleotropic cytokine involved in immune regulation and tumorigenesis. One third of total circulating IL-6 originates from adipocytes and circulating levels are correlated with adiposity [91,92]. When adipocytes are cocultured with breast cancer cells, adipocytes increase secretion of IL-6, which in turn promotes invasion and migration of tumor cells [93]. Furthermore, IL-6 also plays a critical role in the biology of CSCs through activation of Notch and JAK/STAT signaling [94,95]. In addition to classical IL-6 signaling, IL-6 trans signaling is a major driver of obesity associated hepatocellular carcinoma (HCC), through inhibition of p53 induced apoptosis and enhanced angiogenesis [96]. IL-6 also promoted HCC progression via upregulation of osteopontin (OPN), a secretory ECM protein involved in the maintenance of the stemness phenotype [97].
The inflammasome is a highly regulated protein complex that triggers caspase-1 activation and subsequent secretion of IL-1β and IL-18 [98]. Obesity related factors, such as cholesterol crystals and fatty acids (palmitate and ceramide), can lead to unchecked activation of the inflammasome [98]. IL-1β expression in primary tumors is a potential biomarker for predicting breast cancer patients who are at increased risk of developing bone metastasis [99,100]. Furthermore, in vitro studies indicate that tumor cells expressing high levels of IL-1β specifically home to and colonize the bone [101]. A persistent increase of circulating levels of TNFα occurs in obesity, mainly due to the elevated number of M1 macrophages in obese WAT [86]. TNFα stimulates the secretion of MMPs in epithelial tumors and enhances EMT to promote invasion and migration of tumor cells [85,102,103]. Furthermore, TNFα increases liver metastasis through inducing expression of cell adhesion molecules, such as ICAM-1, E-selectin and VCAM-1, on liver specific endothelial cells, and thus enhancing tumor cell arrest and transendothelial migration [104,105].
Crown like structures (CLSs) are a histological feature of dead or dying adipocytes surrounded by macrophages and they are increased in obesity associated adipose tissue [37]. CLS are associated with free fatty acid (FFA) release, NF-κB activation and the generation of a proinflammatory microenvironment [106]. They are best characterized for their role in the initiation and progression of breast cancer [106] but they are also reported to play a role in endometrial cancer [107], prostate cancer [108] and non-alcoholic steatohepatitis (NASH)—a major risk factor for HCC [109]. In breast adipose tissue CLS are not only associated with inflammation but they also drive aromatase activity resulting in an increased ratio of estrogen:androgen in blood and local tissues [110]. The expression of programmed death-ligand 1 (PD-L1) in adipocytes prevents the antitumor function of cytotoxic CD8+ T cells [43]. It is therefore not surprising that treatment with anti PD-1/PD-L1 immunotherapy in patients with a BMI > 25 (i.e., overweight/obese) have improved clinical outcomes compared to normal weight patients [111].
CSCs will only proliferate in specific tumor environments indicating that environmental stimuli are critical to preserve their phenotypic plasticity, to protect them from the immune system and to facilitate metastatic potential [112]. High levels of proinflammatory cytokines from obese adipose tissue can stimulate CSC properties [113]. TNFα enhances the CSC phenotype in numerous cell types and is associated with upregulation of stem cell related genes, chemo resistance and tumorigenesis [114,115,116]. Furthermore, TNFα upregulates TAZ (a Hippo pathway effector) and Slug (an EMT mediator), which increase breast CSCs through both canonical and non-canonical NF-κB signaling [117,118]. Analysis of TNBC patient datasets reveals high tumor expression of the epigenetic reader methyl-CpG-binding domain protein 2 (MBD2), specifically the alternative splicing variant 2 (MBD2_v2) expression and high relapse rate and high BMI [119]. It is postulated that obesity drives high reactive oxygen species (ROS) levels, which subsequently promotes MBD2_v2 expression and an expansion of the CSC fraction [119].

3.3. Angiogenesis

The processes of angiogenesis and adipogenesis are closely linked. When preadipocytes differentiate into adipocytes and become adipose tissue, new blood vessels are also formed [120]. Conversely inhibition of adipocyte differentiation also reduces angiogenesis [121]. In breast cancer, leptin upregulates all components of the IL-1 system (IL-1α, IL-1β, IL-1Ra and IL-1R tI) and both leptin and IL-1 together promote angiogenesis through expression of VEGF/VEGFR [122]. Furthermore, leptin upregulation of VEGF/VEGFR2 was impaired by IL-1 signal blockage [122]. In addition, obesity leads to an increase in tumor infiltrating macrophages with activated NLRC4 inflammasome and increased IL-1β production in breast tumors [123]. This leads to a NLRC4/IL-1β dependent upregulation of adipocyte derived angiopoietin-like 4 and enhanced obesity associated tumor angiogenesis [123]. Anti VEGF therapy has fallen short of expectations, particularly in breast cancer where the FDA revoked approval for bevacizumab because of a lack of overall survival benefit [124]. Anti VEGF therapy resistance is partly driven through expression of proinflammatory and other alternative angiogenic factors, many of which are also increased in obesity [121,125]. Furthermore, breast cancer patients with obesity are less sensitive to anti VEGF treatment and they have increased systemic concentrations of IL-6 and fibroblast growth factor-2 (FGF-2) [126]. The elevated IL-6 was associated with increased IL-6 production from adipocytes and myeloid cells within tumors and IL-6 blockage abrogated obesity induced resistance to anti VEGF therapy at both primary and metastatic sites [126]. Vasculogenic mimicry (VM) is a tumor vascular system that is independent of angiogenesis of endothelial cells, and is associated with both poor survival in multiple tumor types and anti VEGF therapy resistance [127]. Notably, the adipose tissue secretome has been shown to induce melanoma cells to arrange in 3D vessel like structures, characteristic of vasculogenic mimicry [128], supporting a role of adipose tissue in this process.

3.4. Metabolic Repogramming

An essential function of adipocytes is energy mobilization and therefore a metabolic interaction between cancer cells and adipocytes is not surprising. The Warburg effect suggests that due to mitochondrial dysfunction, malignant cells prefer to produce adenosine triphosphate (ATP) via glycolysis instead of oxidative phosphorylation, even in the presence of oxygen [129]. In parallel, cancer cells are able to use alternative sources of energy such as amino acids and lactate from the microenvironment. Bone marrow adipocytes promoted the Warburg phenotype by increased expression of glycolytic enzymes, increased lactate production and decreased mitochondrial oxidative phosphorylation in metastatic prostate cells by paracrine signaling [52]. The “reverse Warburg effect” theory proposes that cancer cells induce oxidative stress in the neighboring stromal cells by secreting ROS and triggering aerobic glycolysis and production of high energy metabolites, especially lactate and pyruvate. These metabolites are then transported through the “lactate shuttle” to sustain the anabolic needs of adjacent cancer cells [130,131]. The effect has been mainly described in stromal fibroblast cells, however given that ASCs and CAA are fibroblast like cells, it is likely they are also important contributors. Indeed it has been reported that the “reverse Warburg effect” was induced during the co-culture of adrenocortical carcinoma cells with ASCs [132]. Ketone bodies are another catabolite produced and released by glycolytic adipocytes and they are an ideal substrate for ATP production by driving oxidative mitochondrial metabolism leading to enhanced tumor invasiveness [91,133].
A key characteristic of CAA is their loss of lipid content. The FFAs released by adipocytes after lipolysis are stored in tumor cells as triglycerides in lipid droplets [134]. Tumor cells then release FFAs from lipid droplets though an adipose triglyceride lipase dependent lipolysis (ATGL) pathway [134]. ATGL is upregulated in tumors on contact with adipocytes and it correlates with aggressiveness by stimulating tumor cell invasion [134]. The FFAs also act as structural units for newly synthesized membrane phospholipids and cancer cell membranes become enriched with saturated and/or mono unsaturated fats leading to changes in membrane dynamics [135]. This results in cancer cells that are more resistant to oxidative induced cell death and reduced the uptake of drugs [135]. Advanced metastatic melanomas frequently grow in subcutaneous tissues largely composed of adipocytes [136]. Adipocyte derived lipids are transferred to melanoma through the lipid transporter FATP1 and a small molecule inhibitor of FATPs reduced melanoma growth and invasion [136]. Furthermore in AML, leukemic blasts activate lipolysis in neighboring bone marrow adipocytes leading to the transfer of lipids to the blast through FABP4 [137].
Amino acids such as glutamine, glycerine, serine and proline also have important roles in the asymmetric metabolism of amino acids between cancer and stromal cells [43]. Glutamine is a pivotal source of the TCA cycle intermediates and ATP in cancer cells, and the substrate of the antioxidant glutathione [138,139]. Stromal cells within the tumor microenvironment harness carbon and nitrogen from non-canonical sources to synthesize glutamine and it is used by the tumor cells to promote growth and metastasis [140]. Glutamine is downregulated in obesity and is inversely associated with proinflammatory gene expression and macrophage infiltration [141]. Pancreatic ductal adenocarcinoma (PDAC) cells rely on glutamine utilization to fulfill their metabolic requirements and it is the most depleted amino acid within the PDAC microenvironment [142]. Glutamine deficiency leads to the induction of EMT through the upregulation of the master EMT regulator Slug [142]. In addition, as glutamine levels decline, tumor cells become more reliant on asparagine for proliferation and protein synthesis [143]. Asparagine affects the metastatic cascade at multiple stages. At the primary tumor level, asparagine promotes EMT and intravasation [144,145]. In the circulation asparagine helps circulating tumor cells survive shear and oxidative stress whilst at a distinct metastatic sites, asparagine facilitates cell growth and survival by inducing glutamine synthetase (GLUL) expression and glutamine biosynthesis [144,145].
Citrulline and nitric oxide are generated by tumor cells by catabolizing the semi essential amino acid arginine [146]. Nitric oxide facilitates glycolytic activity and suppresses oxidative phosphorylation to promote proliferation [146]. Citrulline is secreted into the ECM and is taken up by stromal adipocytes, before being converted back into arginine and released for cancer cells [146]. Depriving tumor cells of arginine has cytotoxic effects through apoptosis or autophagy depending upon the tumor type, and decreasing the ability of tumor cells to migrate and adhere to the ECM [147]. Arginine dependent migration requires arginine to be metabolized by two major enzymes, arginase (ARG1) and nitric oxide synthase (NOS) [147]. In HCC, higher expression of ARG1 is positively correlated to aggressive tumor growth and poor disease free survival [148]. In vitro studies revealed that overexpression of ARG1 enhanced arginase activity leading to multiple processes that contribute to progression including increased cell viability, migration, invasion and EMT [148]. Obesity coupled with PDAC results in accelerated tumor growth and enrichment in pathways regulating nitrogen metabolism. The mitochondrial form of arginase (ARG2) that hydrolyzes arginine into ornithine and urea is induced upon obesity and is accompanied by PDAC growth and increased nitrogen flux from 15N-glutamaine into the urea cycle, the principle pathway for ammonia detoxification [149]. Infusion of 15N-arginine in murine models demonstrates a shunting of arginine catabolism away from the urea cycle into creatine synthesis, resulting in ammonia accumulation specifically in obese tumors [149]. The biological consequences of ammonia accumulation in the tumor microenvironment is not fully understood but it has been shown to directly generate amino acids through glutamate dehydrogenase activity [150].
Hyperinsulinemia is a hallmark of chronic obesity and insulin stimulates the hepatic synthesis of the peptide IGF-1 [151]. Three quarters of breast cancer patients show activation of insulin/IGF-1 signaling and this rises to 87% in patients with invasive breast cancer [152]. In preclinical models, blocking IGF in combination with paclitaxel significantly reduced tumor cell proliferation and lung metastasis [152]. Due to the frequent dysregulation of the IGF system in cancer, various components of this system became attractive anticancer targets. However, clinical trials using IGF-1 receptor blocking antibodies failed to meet expectations, except in a small number of malignancies [153,154]. More recent developments reveal that dysregulation of the IGF system results in a substantial expansion of the cancer stem like subpopulation by supporting EMT and self-renewal signaling pathways [153]. IGF signaling regulates these pathways in multiple ways though i) stimulation of the transcription factors of the ZEB and the Snail family implicated in the EMT program, ii) interacting with pluripotency transcription factors (e.g., Oct-4, SOX2, Nanog, p53 and HMGA1 proteins) and iii) regulation of development signaling factors (e.g., Wnt/β-catenin, Notch and Shh pathways) classically involved in cell stemness [153]. Intriguingly, in HER2+ breast cancer patients, high IGF-1 in normal weight patients showed a superior recurrence free survival compared to low IGF-1 [155]. In contrast, high IGF-1 in overweight patients was associated with a reduced recurrence free survival [155]. Obese mice have a heightened inflammatory response in the liver and an increased incidence of metastatic colon carcinoma cells to the liver [156]. Moreover, liver inflammation induced by obesity was abrogated in liver specific IGF-I deficient mice leading to a significant reduction of in liver metastasis [156]. Furthermore, IGF-1 promotes neutrophil polarization to a tumor promoting phenotype and the induction of a prometastatic microenvironment in the liver [157].

3.5. Extracellular Matrix

In addition to adipocyte hypertrophy and dysregulated lipid metabolism, heightened inflammation, hypoxia and abnormal angiogenesis, obesity is also associated with ECM remodeling. Once a primary tumor is established, cells migrate and invade to form satellite tumors within centimeters of the original tumor mass and/or disseminate through the lymph nodes and vasculature to form secondary macrometastases at distance sites (Figure 1). This progression sequence is dependent on the ability of cancer cells to traverse the ECM. The adipose tissue ECM is a three-dimensional, non-cellular structural support of the numerous cell types that reside in the adipose tissue. It is composed primarily of collagens, fibronectin and to a lesser extent lamins that are supplied by a number of resident cell types including fibroblasts, adipocytes and preadipocytes [158]. In obesity, the highly dynamic adipose tissue ECM is constantly undergoing remodeling and reorganization to accommodate increased adipocyte numbers and adipocyte hypertrophy. A rapid increase in adipose tissue volume can result in regional hypoxia, which triggers excess deposition of fibrillar collagens by adipocytes and myofibroblasts, immune cell infiltration, adipose tissue fibrosis, a desmoplastic stroma and increased tissue stiffness, with overall behavior described as similar to “a wound that never heals”. This state of chronic low-grade inflammation within the adipose tissue drives obesity-associated pathologies including diabetes [159,160], cardiovascular disease [161] and cancer. Indeed, it is very similar to the microenvironment of a thriving tumor mass and thus trophic cancer cells that home to the adipose tissue are well supported by these suitable surroundings. Fibrosis is a hallmark of cancer, and desmoplasia within the tumor microenvironment, is a marker of poor prognosis in cancer [162,163] and can negatively impact drug delivery [164,165]. In the case of PDAC, obesity is associated with aggressive tumors with poor prognosis, and adipocyte accumulation in the malignant pancreas [166]. Obesity-induced accumulation of high adipocyte numbers in the pancreas has been shown to induce inflammation and excessive accumulation of ECM components, i.e., desmoplasia, which promotes tumor progression and resistance to chemotherapy [164].
Adipose tissue is the main component of the breast cancer microenvironment, crosstalk between the breast cancer cells and adipocytes or other adipose stromal cells stimulates the secretion of even larger quantities of ECM proteins, increasing matrix stiffness and scar formation, further enhancing EMT and local invasion of tumor cells. Within the adipose tissue, invading breast cancer cells manipulate adipocytes to form fibroblastic CAAs that secrete large volumes of ECM proteins including collagen I, III and IV, and the cleavage product of collagen IV, endotrophin, which is associated with breast cancer metastatic spread [85,167,168]. Furthermore adipocyte collagen IV has been shown to play a role in the early stages of tumor growth in breast cancer [169]. Recently, it has been shown that breast cancer secreted PAI-1 can stimulate CAA collagen biogenesis and reorganization via the induction of a lysyl hydroxylase protein, PLOD2, facilitating the migration of breast cancer cells along aligned collagen fibers, in vitro and in vivo, further promoting metastasis [170]. Additionally, tumors growing in the adipose tissue-rich microenvironment can induce morphological and functional changes in ADSCs so that they differentiate into a CAF-like myofibroblastic phenotype. Breast cancer cells can induce the differentiation of ADSCs to CAFs involving a mechanism dependent on TGF-β, and these ASC-derived CAF-like cells can promote breast cancer cell motility and invasion in vitro, and expressing high levels of stromal-derived factor 1 (SDF-1), a chemokine associated with a more aggressive and invasive cancer phenotype [171,172]. During obesity-induced fibrosis, as adipocytes become encased in the rigid ECM, necrosis ensues and dysfunctional adipocytes stimulate the recruitment of macrophages to the site, histologically forming CLS around the dying adipocytes [173]. The contribution of these activated macrophages to ECM production most likely occurs through their effects on other cell types. They are a major source of TGF-β, PDGF and other chemokines that attract and activate more ECM protein producing fibroblast type cells to the adipose tissue [174,175]. Finally, fibrosis dynamics are tightly regulated by the metalloproteinase (MMP) protein family, which cleave collagenous fibers, enabling matrix remodeling. There are many MMPs associated with obesity [176], in particular MMP-11 (also known as stromelysin-3/ST-3) has been shown to be overexpressed by adipocytes as a result of stimulation by invading breast cancer cells [177]. MMP-11 is important for collagen VI folding and it is also known to negatively regulate adipogenesis and can dedifferentiate adipocytes so that they can acquire a more fibroblast-like phenotype that benefits the invading tumor cells via involvement in adipose tissue fibrosis and ECM remodeling [45,177].

3.6. Extracellular Vesicles

In addition to the milieu of adipose tissue secreted factors discussed in the previous sections, there is now evidence highlighting the role that adipose tissue derived extracellular vesicles (EVs) play in guiding and enhancing the metastatic process. EVs are lipid membrane enclosed particles, measuring on the nanometer scale, that can be classified as either minute exosomes (<100 nM) or larger microvesicles (<1000 nM), which facilitate the horizontal transfer of cellular cargo, including nucleic acid, proteins, lipids and metabolites between communicating cells [178,179]. It is not clear whether obesity alters the content of EVs produced by adipocytes, however in the obese setting larger quantities of adipocyte-derived EVs are secreted compared to lean conditions [180]. While the role of cancer cell-derived EVs in manipulating cells in the tumor microenvironment including adipocytes is well established [181,182,183], very little attention has been given to the bidirectional role EVs play in adipocyte-cancer cell communication, and particularly the influence of adipocyte-derived EVs on tumor cell behavior. Nevertheless, a small number of recent studies demonstrate a link between adipocyte EVs and tumor progression in obesity-driven lung cancer [184], breast cancer [185,186] and melanoma [180,187].
One way in which adipocytes can promote tumor progression is through metabolic cooperation, by providing a local supply of fatty acids for the process of fatty acid oxidation (FAO) within tumor cells, an emerging favorable metabolic pathway that enhances tumor invasiveness, proliferation and stem cell properties [180,188,189]. In the later stages of tumor progression once the tumor cells have invaded the adipose tissue, secretion of tumor-derived soluble factors can stimulate adipocyte lipolysis and extracellular release of FFAs into the surrounding microenvironment [39,134,188]. Aside from direct release of FAs from adipocytes, EVs released by adipocytes can also be used as a method to transfer molecules including FA substrates and the protein machinery required for FAO to cancer cells either locally or over larger distances, e.g., through the circulation and other tissues.
Epidemiological studies have identified a link between melanoma aggressiveness and obesity [190,191]. Aptly, subcutaneous adipocytes are one of the main components of the tumor microenvironment of invading melanomas and indeed melanoma cells have been demonstrated to internalize naïve adipocyte (i.e., not previously exposed to cancer cells)-derived EVs, resulting in amplified FAO and an enhanced migratory and invasive tumor cell phenotype [180]. Lazar et al. demonstrated that melanoma cells cultured with adipocyte secreted EVs had an increased ability to form lung metastases in mice xenograft models, with a concomitant upregulation of tumor cell FAO. As previously mentioned obese-derived adipocytes secrete higher numbers of EVs compared to their lean counterparts, thus when comparing the effect of adipocyte EVs derived from obese versus lean conditions, equal concentrations of EV preparations were applied to melanoma cells. Noticeably, in obese conditions only adipocyte-derived EVs elicit a heightened effect on melanoma migration, in addition to enhancing clonogenicity and metastatic potential. Thus differences in the cargo content of these EVs as opposed to the sheer number of vesicles are most likely responsible for this heightened effect. Recent in-depth labeling experiments indicate that around 30% of the proteins within the adipocyte EVs are sufficiently transferred to melanoma tumor cells, and these include proteins involved in EV transport, the transport and storage of FAs, mitochondrial FAO and oxidative phosphorylation [187]. Efforts to understand how an obese state heightens the effect of adipocyte EVs on FAO-induced functions of melanoma cells have revealed that the level of FAO enzymes in melanoma tumor cells is unaltered following uptake of obese versus lean derived EVs. In contrast adipocyte EV supply of FAs and subsequent trafficking to melanoma cells was increased under obese conditions, resulting in enhanced substrate availability and FAO, and the altered mitochondrial dynamics that is critical to melanoma cell migration and invasion. Thus in the obese setting, it is the increased transfer of substrate (e.g., FA) and not machinery (FAO-related enzymes) that enhance FAO in recipient melanoma cancer cells [187].
In whole adipose tissue, aside from mature adipocytes, EVs are found in the supernatant ASCs. While adipocyte EVs are linked to enhanced tumor invasiveness and metastatic potential via lipid metabolism, current literature suggest ADSC secreted EVs play a role in angiogenesis [192,193,194], immune modulation [195,196,197] and tumor development [198]. When exosomes secreted from the preadipocyte cell line, 3T3-L1, are injected into the mammary fat together with breast cancer cells, primary tumor initiation and growth is enhanced. Of particular interest in this review, ASC derived exosomes have been shown to promote proliferation and migration, at least in part through the modulation of Wnt/β-catenin signaling, a key pathway in tumor stemness, EMT and metastasis [199,200]. ASCs are abundant in the microenvironment of highly metastatic osteosarcoma. Recently ASC exosomes has been shown to increase proliferation, invasion and migration of osteosarcoma cells in vitro, and growth and metastasis in vivo, via an induction of COLGALT2, a prometastatic gene that subsequently activates downstream EMT targets vimentin and MMP-2 and -9 [201]. Studies regarding adipose tissue secreted exosomes, be it adipocytes or ASCs, are limited to a small number of studies and a small number of tumor types. Further research is required to understand the role, if any, of adipose tissue-derived EVs in other cancer types, which share an intimate relationship with adipose tissue and are driven by obesity.

4. Targeting Metastasis through Adipose Tissue-Tumor Interactions

Obesity-associated cancer is a substantial public health problem worldwide and correcting the disharmony within dysfunctional adipose tissue represents an opportunity to halt disease progression, as summarized in Figure 2. Due to the importance of adiponectin in the progression of several cancer types, adiponectin receptors agonists have been developed [54]. An adiponectin-based short peptide named ADP 355 has high affinity with AdipoR1 and regulates canonical adiponectin signaling pathways (i.e., AMPK, Akt, STAT3, ERK1/2) to halt tumor growth [202,203]. Adiponectin has a larger role in early stage metastatic processes and there are concerns that its agonists may blunt antitumor immunity. Therefore cancer stage is an important consideration for adiponectin-based agonist therapy [54]. Interestingly, adiponectin may also be modulated with dietary and lifestyle factors. Daily fish intake, omega 3 and fiber supplementation have been shown to increase blood levels of adiponectin [204]. Furthermore, moderate intensity aerobic exercise was shown to elevate adiponectin levels to 260% within one week [205].
Inhibitors of leptin, another important adipokine, are also in development. Conditioned media (CM) from breast adipocytes significantly increased mammosphere formation, a marker of breast CSCs, and depletion of leptin from the CM completely abrogated this effect [206]. In addition, blocking leptin signaling using a full-length leptin receptor (ObR) antagonist also reduced mammosphere formation indicating a potential therapeutic target to block stromal-tumor interactions driving breast CSC-mediated disease progression [206]. The ObR antagonist, Allo-aca, a short leptin-based peptidomimetic inhibits leptin induced tumor cell proliferation and angiogenesis [207,208]. In addition, it has good biodistribution to the brain and could potentially target brain metastasis [209].
Adipose tissue significantly contributes to inflammatory cytokines in the tumor microenvironment that may be targeted therapeutically to treat metastasis. Agents targeting IL-6 or the IL-6 receptor have already received FDA approval for the treatment of inflammatory conditions or myeloproliferative neoplasms and are being evaluated in solid tumors [210]. However, despite promising preclinical activity, targeting IL-6 alone in unselected cancer patient populations has not shown benefit to patient outcomes [210]. Therefore, more selective formulations and selecting potentially sensitive patient populations, such as obese patients, are required. A novel-CD44 antibody mediated liposomal nanoparticle loaded with anti-IL6R antibody demonstrated specific antimetastatic efficacy in a preclinical mouse model by inhibiting IL-6/STAT3 signaling in CSCs and also reducing the SOX2+ and CD206+ cells in the tumor microenvironment of lung metastasis, thus demonstrating dual inhibitory activity on CSCs themselves and the metastatic niche microenvironment [211].
Inhibiting IL-1R signaling using the receptor antagonist Anakinra resulted in decreased bone metastasis in an in vivo TNBC model. Pretreatment of Anakinra did not prevent tumor cell numbers that arrived to the bone, but instead held the tumor cells in a dormant state, thereby preventing metastasis [212]. Furthermore, therapeutic treatment of bone metastasis in the model with Ankinira stalled tumor growth by reducing proliferation and angiogenesis [212]. Anakinra and other IL-1 signaling pathway inhibitors are currently in multiple clinical trials to target metastasis in solid tumors, multiple myeloma and plasma cell neoplasm, as summarized in [100]. In a clinical trial of patients with smoldering or indolent multiple myeloma at risk of progression to active myeloma, Anakinra treatment decreased myeloma proliferative rate and hs-CRP in responders leading to improved progression free survival [213]. A large cardiovascular randomized control trial of Canakinumab (Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS)), an interleukin (IL)-1β inhibitory antibody, noted a dramatic reduction in the incidence of lung cancer [214]. The trial of 10,500 patients was not designed to study lung cancer as an endpoint and the patients were extensively screened prior to recruitment [215]. These results indicate that IL-1β reduced the progression, invasiveness and metastatic spread of early stage lung cancers that were undiagnosed at the time of recruitment [215]. Therefore these studies suggest that anti IL-1β therapy may prevent the metastatic cascade if given early in the disease process, and further studies are required to examine if metastatic spread is also prevented in people living with obesity. However, there is emerging evidence that anti IL-1β therapy may promote metastatic spread if given later in disease development. Castano et al. eloquently reported that primary tumors elicit a systemic inflammatory response involving IL-1β expressing innate immune cells that infiltrate distant metastasis microenvironments [216]. At the metastatic site, IL-1β maintains metastasis initiating cells in a ZEB-1 positive differentiation state, but inhibition of IL-1 receptor relieves the differentiation block and resulted in metastatic colonization [216].
People living with obesity experience more chemoresistance due to altered drug pharmacokinetics and enhanced proinflammatory and adipokine secretions [217]. Therefore further research is required to investigate if inhibitors of downstream intracellular pathways associated with both obesity and chemo-resistance, such as AKT or NF-κB pathways, are clinically superior in combination with chemotherapy in people with obesity. On the other hand, immune checkpoint inhibitors appear to be more effective in the obese population compared to lean patients. The chronic inflammation and lipid accumulation induced by obesity leads to an “exhausted” T cell phenotype, which is associated with PD-1 upregulation [218]. Obese melanoma patients treated with immune checkpoints inhibitors have an improved overall survival and progression free survival compared to lean patients [219]. In addition, response to PD-1 inhibitors has been modest in TNBC however results from the JAVELIN trial suggests that obese TNBC patients with an exhausted immune response might benefit from checkpoint blockage [220,221,222]. However, aged mice with excess adiposity developed a lethal cytokine storm reaction following immunotherapy, potentially indicating that older people with obesity may be at a higher risk of adverse effects [223].
Adipose derived fatty acid binding proteins (FABPs) in the tumor microenvironment have important roles as intracellular lipid chaperones and pharmacological agents that modify FABP function are in development for obesity, diabetes and atherosclerosis [224]. FABP5 is not expressed in the normal prostate but is highly upregulated in metastatic prostate cancer and offers is a novel therapeutic target for pharmaceutical inhibition [225,226]. However, lipid metabolism is central to the function of regulatory T cells (Tregs). Inhibition of FABP5 in Tregs triggers mitochondrial DNA release, which induces cGAS-STING dependent type 1 interferon signaling and the production of IL-10. Overall this promotes Tregs immunosuppressant activity within the tumor microenvironment leading to enhanced tumor progression [227]. Treg activity is associated with a poor prognosis and therefore the effect of pharmacological inhibition of FABP5 would have to be carefully evaluated in all cells within the tumor microenvironment [228].
CD36 is a transmembrane glycoprotein that mediates lipid uptake and binds to a diverse range of ligands including apoptotic cells, thrombospondin-1 (TSP-1) and fatty acids [229]. High fat diet or palmitic acid enhances a subpopulation of CD36+ cells with metastatic potential in mouse models of human oral cancer [230]. The presence of CD36+ metastasis initiating cells also correlated with poor prognosis in a number of cancer types and anti-CD36 neutralizing antibodies induced regression of the lymph node and lung metastasis in murine models [230]. However, there is also evidence that loss of CD36 in stromal cells and surrounding endothelial cells can increase the metastatic potential of the niche and induce angiogenesis [231,232]. Furthermore, several agents targeting CD36 in clinical trials with thrombospondin mimetic peptides have been unsuccessful due to adverse effects and limited efficacy [229,232]. CD36 may be promising to specifically target metastasis however further knowledge of its role in stromal cells and downstream signaling pathways is required to develop more precise therapeutics.
As glutamine levels are decreased in obesity and cells adapt to low glutamine by becoming dependent on asparagine, an amino acid, which promotes multiple stages of the metastatic cascade, targeting asparagine may prevent obesity-induced metastasis [141,143,145]. Indeed, treatment with L-asparaginase, an enzyme that depletes asparagine, or alternatively dietary asparagine restriction, reduced metastasis in a breast cancer model without affecting the growth of the primary tumor [145]. L-asparagine is a mainstay treatment in childhood acute lymphoblastic leukemia (ALL) and children with obesity have a 50% greater risk of ALL relapse than lean counterparts [233]. Glutamine synthesis was markedly increased in bone marrow adipocytes following chemotherapy and obesity substantially impaired L-asparaginase efficacy due to adipocytes secreting glutamine [233]. In addition, obesity increases the risk of hepatic toxicity with L-asparaginase therapy by promoting a maladaptive integrated stress response [234]. Therefore, whilst inhibiting asparagine preferentially targeted metastasis, the experience of L-asparaginase in ALL suggests that it could be less effective and more toxic in people with obesity.
Another proposed method to target the metabolism of tumor cells is through short term dietary fasting. Fasting diets are currently in clinical trial to enhance chemotherapy efficacy and reduce toxicity however there is also evidence they may slow tumor progression [235]. In the fasting state IGF-1, a promoter of EMT, is inhibited, and clinical trials are underway to investigate if tumor progression can be inhibited [235]. Obesity induces lung neutrophilia leading to increased breast cancer lung metastasis through GM-CSF and IL-5 signaling. Interestingly, weight loss reversed this effect, leading to reduced serum GM-CSF and IL-5 in both mouse and humans. In addition, this suggests that the disruption of normal lung homeostasis is critical in obesity induced metastasis and pharmacological interventions that resolve lung inflammation may reduce metastatic risk [236]. Targeting IL-5 was shown to be sufficient to block lung neutrophil trafficking in obese mice and there are a number of anti-IL-5 therapies with FDA approval for severe hypereosinophilic asthma [236,237]. Therefore, obese cancer patients with increased neutrophilia, GM-CSF or IL-5, may benefit from anti-IL-5 therapies to prevent lung metastasis development.

5. Targeting Metastasis via the Treatment of Obesity

Complementary treatment of obesity may potentially improve outcomes in cancer patients [238]. Indeed, a systematic review and meta-analysis of animal models concluded that weight loss decreases cancer progression and metastasis [239]. The most common method of weight management is dietary calorie restriction (CR) and increasing energy expenditure though exercise. CR extends lifespan and reduces age related diseases, including cancer, in preclinical models and recently translational studies have tested the potential of CR as an adjuvant therapy in cancer. However, chronic CR is often contra indicated in cancer patients due to the risks of malnutrition and cachexia and other strategies, such as intermittent fasting, CR mimetic drugs or alterative diets (e.g., ketogenic diet) may be more suitable [240,241]. Clinical studies have shown that both adjuvant and neo-adjuvant chemotherapy is less effective in people with obesity [242,243,244]. CR, fasting or fasting mimicking diets (FMDs) may improve response to both chemotherapy and radiotherapy as well asreducing side effects associated with cytotoxic drugs [240]. In addition, obesity is associated with an increase desmoplasia surrounding the tumor tissue, contributing to chemoresistance, and CR can reduce the desmoplasia to facilitate better therapeutic drug delivery to the tumor cells [164,240]. CR, fasting and FMD also have been shown to reduce secreted factors within the tumor microenvironment including proinflammatory cytokines, leptin and IGF-1 [241]. Considering the key roles of these molecules to be dysregulated in obesity and promote metastasis, CR, fasting or FMD may reduce tumor progression through this mechanism. Interestingly, however, a preclinical study of colon cancer showed that an intermittent fasting diet expanded the CSCs population and enhanced EMT by promoting the Warburg/Crabtree effect following post-fasting food overconsumption [245]. Therefore, carefully controlled clinical studies of CR, fasting and FMD are required to determine if they have a role in preventing cancer progression without leading to nutritional deficiencies or cachexia. With this in mind, weight management strategies may be a more appropriate strategy to prevent reoccurrence during long-term cancer survivorship.
Physical inactivity is a well-known avoidable risk factor for developing cancer. However, there is also evidence that an increased level of physical activity between before to after diagnosis reduces cancer mortality [246]. In contrast, a decreased physical activity level between pre- to post- diagnosis is associated with higher cancer mortality [246]. Together this indicates that tumor progression could be slowed or prevented by physical activity and indeed physical activity regulates multiple steps of the metastatic cascade, as comprehensively reviewed in [247]. In summary, moderate intensity exercise appears to prevent tumor spread by normalizing angiogenesis, reducing circulating tumor cells and decreasing endothelial cell permeability [247,248,249]. On the contrary, high-intensity exercise may favor cancer dissemination and promote the formation of the premetastatic niche through excessive stress [247,250]. In overweight or obese breast cancer survivors, moderate to vigorous exercise three times a week for 16 weeks reduced systemic levels of insulin, IGF-1 and leptin, and increased adiponectin [251]. Therefore, chronic adaptations to moderate-intensity endurance exercise may be the most effective way to achieve a preventive effect of exercise on metastases [247]. Overall, exercise and nutritional strategies may be especially beneficial in overweight or obese cancer survivors to reduce both obesity-associated comorbidities and prevent cancer reoccurrence.
Bariatric surgery is the most effective treatment for obesity and its associated metabolic and cardiovascular complications. On the whole studies report that overall cancer incidence and mortality is reduced post bariatric surgery, particularly in women [252,253,254]. Obesity is responsible for increased estradiol production in adipose tissue thus driving hormone sensitive cancers and the augmented concentration of total serum estrogen following bariatric surgery is suggested to be the reason for reduced post-menopausal breast and endometrial cancer risk [254]. Indeed bariatric surgery may also reduce tumor progression by specifically treating hyperinsulinemia and reducing high circulating IGF-1 levels [254]. However, data is conflicting and there is a potential increase in cancer incidence following bariatric surgery [254,255]. There are reports suggesting bariatric surgery results in an increased risk of colorectal cancer (CRC) but the results are complicated as control groups were not standardized for weight changes, dietary changes and medications, which can have a profound impact on CRC incidence [255]. One hypothesis for the increased CRC incidence is based upon a study investigating colonic epithelial cell proliferation in subjects undergoing roux-en-Y-gastric bypass surgery [256]. Biopsies comparing the day of surgery and 6 months after surgery found an increase in the mitotic activity of colonic cells following surgery and an increased expression of proinflammatory and tumorigenic COX-1 and COX-2 enzymes [256]. Bariatric surgery can successfully achieve weight loss among cancer survivors, but long term effects have not been reported [257]. Overall, current evidence indicates bariatric surgery is more effective for cancer prevention in female hormone driven cancers compared to male cancers. Indeed, a study of women with BMI > 40 kg/m2 who underwent bariatric surgery resulted in a reduction of endothelial proliferation marker and resolved most cases of the precancerous condition atypical hyperplasia [258]. Randomized control trials are required to determine if bariatric surgery in obese women can increase breast or endometrial cancer survivorship and reduce tumor reoccurrence.

6. Conclusions

Obesity is a growing and significant global health crisis, with epidemiological studies demonstrating a compelling association between cancer risk and progression. A clearer understanding of the molecular processes associated with obesity-induced cancer is warranted and has the potential to uncover new targeting strategies. Indeed, potential new targets are already emerging. It is reasonable to posit that weight loss might be beneficial to reduce metastasis risk. However, it is also possible that weight loss alone is insufficient to reverse the effects of chronic obesity due to potential epigenetic reprogramming of tumor cells and/or stromal cells. In addition, patients with advanced disease often experience WAT loss and wasting (cachexia), which complicates treatment and adversely affects patient survival. The Breast Cancer Weight Loss (BWEL) trial (Identifier NCT02750826) is a phase III randomized trial currently underway assessing the impact of a weight loss intervention on disease recurrence in women with stage II to III HER2-negative breast cancer. The results of this trial will help provide answers to some of these questions. Given the limitations of BMI as a measurement of adiposity, novel methods of identifying people with metabolically unhealthy adipose tissue is required. Additional strategies may also benefit individuals who are not obese (BMI< 30) but who have signs of metabolic syndrome or adipose tissue dysfunction. Ultimately, there is a definitive need for more tailored interventions for obese patients to improve survival status in this patient population, especially considering the higher risk of drug resistance in the obese setting.

Funding

The authors are supported by grants from the National Children’s Research Centre and the Children’s Medical & Research Foundation, Crumlin, Ireland and the Science Foundation Ireland Strategic Partnership programme, Precision Oncology Ireland.

Conflicts of Interest

The authors declare no conflict of interests.

References

  1. Steele, C.B.; Thomas, C.C.; Henley, S.J.; Massetti, G.M.; Galuska, D.A.; Agurs-Collins, T.; Puckett, M.; Richardson, L.C. Vital Signs: Trends in Incidence of Cancers Associated with Overweight and Obesity—United States, 2005–2014. MMWR Morb. Mortal. Wkly. Rep. 2017, 66, 1052–1058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Chan, D.S.M.; Vieira, A.R.; Aune, D.; Bandera, E.V.; Greenwood, D.C.; McTiernan, A.; Rosenblatt, D.N.; Thune, I.; Vieira, R.; Norat, T. Body mass index and survival in women with breast cancer—Systematic literature review and meta-analysis of 82 follow-up studies. Ann. Oncol. 2014, 25, 1901–1914. [Google Scholar] [CrossRef] [PubMed]
  3. Lengyel, E.; Makowski, L.; DiGiovanni, J.; Kolonin, M.G. Cancer as a Matter of Fat: The Crosstalk between Adipose Tissue and Tumors. Trends Cancer 2018, 4, 374–384. [Google Scholar] [CrossRef] [PubMed]
  4. Renehan, A.G.; Zwahlen, M.; Egger, M. Adiposity and cancer risk: New mechanistic insights from epidemiology. Nat. Rev. Cancer 2015, 15, 484–498. [Google Scholar] [CrossRef]
  5. Allott, E.H.; Masko, E.M.; Freedland, S.J. Obesity and prostate cancer: Weighing the evidence. Eur. Urol. 2013, 63, 800–809. [Google Scholar] [CrossRef] [Green Version]
  6. Huffman, D.M.; Johnson, M.S.; Watts, A.; Elgavish, A.; Eltoum, I.A.; Nagy, T.R. Cancer progression in the transgenic adenocarcinoma of mouse prostate mouse is related to energy balance, body mass, and body composition, but not food intake. Cancer Res. 2007, 67, 417–424. [Google Scholar] [CrossRef] [Green Version]
  7. Ewertz, M.; Jensen, M.B.; Gunnarsdóttir, K.Á.; Højris, I.; Jakobsen, E.H.; Nielsen, D.; Stenbygaard, L.E.; Tange, U.B.; Cold, S. Effect of obesity on prognosis after early-stage breast cancer. J. Clin. Oncol. 2011, 29, 25–31. [Google Scholar] [CrossRef]
  8. Kyrgiou, M.; Kalliala, I.; Markozannes, G.; Gunter, M.J.; Paraskevaidis, E.; Gabra, H.; Martin-Hirsch, P.; Tsilidis, K.K. Adiposity and cancer at major anatomical sites: Umbrella review of the literature. BMJ 2017, 356, 477. [Google Scholar] [CrossRef] [Green Version]
  9. O’Sullivan, J.; Lysaght, J.; Donohoe, C.L.; Reynolds, J.V. Obesity and gastrointestinal cancer: The interrelationship of adipose and tumour microenvironments. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 699–714. [Google Scholar] [CrossRef]
  10. Fang, X.; Wei, J.; He, X.; Lian, J.; Han, D.; An, P.; Zhou, T.; Liu, S.; Wang, F.; Min, J. Quantitative association between body mass index and the risk of cancer: A global Meta-analysis of prospective cohort studies. Int. J. Cancer 2018, 143, 1595–1603. [Google Scholar] [CrossRef] [Green Version]
  11. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Welch, D.R.; Hurst, D.R. Defining the Hallmarks of Metastasis. Cancer Res. 2019, 79, 3011–3027. [Google Scholar] [CrossRef] [PubMed]
  13. Bakhoum, S.F.; Ngo, B.; Laughney, A.M.; Cavallo, J.A.; Murphy, C.J.; Ly, P.; Shah, P.; Sriram, R.K.; Watkins, T.B.K.; Taunk, N.K.; et al. Chromosomal instability drives metastasis through a cytosolic DNA response. Nature 2018, 553, 467–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Ye, X.; Weinberg, R.A. Epithelial-Mesenchymal Plasticity: A Central Regulator of Cancer Progression. Trends Cell Biol. 2015, 25, 675–686. [Google Scholar] [CrossRef] [Green Version]
  15. Rankin, E.B.; Giaccia, A.J. Hypoxic control of metastasis. Science 2016, 352, 175–180. [Google Scholar] [CrossRef] [Green Version]
  16. Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanisms of epithelial-mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2014, 15, 178–196. [Google Scholar] [CrossRef] [Green Version]
  17. Zheng, X.; Carstens, J.L.; Kim, J.; Scheible, M.; Kaye, J.; Sugimoto, H.; Wu, C.-C.; LeBleu, V.S.; Kalluri, R. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 2015, 527, 525–530. [Google Scholar] [CrossRef] [Green Version]
  18. Fischer, K.R.; Durrans, A.; Lee, S.; Sheng, J.; Li, F.; Wong, S.T.C.; Choi, H.; El Rayes, T.; Ryu, S.; Troeger, J.; et al. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature 2015, 527, 472–476. [Google Scholar] [CrossRef]
  19. Lytle, N.K.; Barber, A.G.; Reya, T. Stem cell fate in cancer growth, progression and therapy resistance. Nat. Rev. Cancer 2018, 18, 669–680. [Google Scholar] [CrossRef]
  20. Fares, J.; Fares, M.Y.; Khachfe, H.H.; Salhab, H.A.; Fares, Y. Molecular principles of metastasis: A hallmark of cancer revisited. Signal Transduct. Target. Ther. 2020, 5, 1–17. [Google Scholar] [CrossRef]
  21. Pastushenko, I.; Brisebarre, A.; Sifrim, A.; Fioramonti, M.; Revenco, T.; Boumahdi, S.; Van Keymeulen, A.; Brown, D.; Moers, V.; Lemaire, S.; et al. Identification of the tumour transition states occurring during EMT. Nature 2018, 556, 463–468. [Google Scholar] [CrossRef] [PubMed]
  22. Jolly, M.K.; Ware, K.E.; Gilja, S.; Somarelli, J.A.; Levine, H. EMT and MET: Necessary or permissive for metastasis? Mol. Oncol. 2017, 11, 755–769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Fox, R.G.; Lytle, N.K.; Jaquish, D.V.; Park, F.D.; Ito, T.; Bajaj, J.; Koechlein, C.S.; Zimdahl, B.; Yano, M.; Kopp, J.L.; et al. Image-based detection and targeting of therapy resistance in pancreatic adenocarcinoma. Nature 2016, 534, 407–411. [Google Scholar] [CrossRef] [Green Version]
  24. Balic, M.; Lin, H.; Young, L.; Hawes, D.; Giuliano, A.; McNamara, G.; Datar, R.H.; Cote, R.J. Most early disseminated cancer cells detected in bone marrow of breast cancer patients have a putative breast cancer stem cell phenotype. Clin. Cancer Res. 2006, 12, 5615–5621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Grillet, F.; Bayet, E.; Villeronce, O.; Zappia, L.; Lagerqvist, E.L.; Lunke, S.; Charafe-Jauffret, E.; Pham, K.; Molck, C.; Rolland, N.; et al. Circulating tumour cells from patients with colorectal cancer have cancer stem cell hallmarks in ex vivo culture. Gut 2017, 66, 1802–1810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Aktas, B.; Tewes, M.; Fehm, T.; Hauch, S.; Kimmig, R.; Kasimir-Bauer, S. Stem cell and epithelial-mesenchymal transition markers are frequently overexpressed in circulating tumor cells of metastatic breast cancer patients. Breast Cancer Res. 2009, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Baccelli, I.; Schneeweiss, A.; Riethdorf, S.; Stenzinger, A.; Schillert, A.; Vogel, V.; Klein, C.; Saini, M.; Bäuerle, T.; Wallwiener, M.; et al. Identification of a population of blood circulating tumor cells from breast cancer patients that initiates metastasis in a xenograft assay. Nat. Biotechnol. 2013, 31, 539–544. [Google Scholar] [CrossRef]
  28. Hermann, P.C.; Huber, S.L.; Herrler, T.; Aicher, A.; Ellwart, J.W.; Guba, M.; Bruns, C.J.; Heeschen, C. Distinct Populations of Cancer Stem Cells Determine Tumor Growth and Metastatic Activity in Human Pancreatic Cancer. Cell Stem Cell 2007, 1, 313–323. [Google Scholar] [CrossRef] [Green Version]
  29. Phi, L.T.H.; Sari, I.N.; Yang, Y.G.; Lee, S.H.; Jun, N.; Kim, K.S.; Lee, Y.K.; Kwon, H.Y. Cancer stem cells (CSCs) in drug resistance and their therapeutic implications in cancer treatment. Stem Cells Int. 2018, 2018. [Google Scholar] [CrossRef] [Green Version]
  30. Peinado, H.; Zhang, H.; Matei, I.R.; Costa-Silva, B.; Hoshino, A.; Rodrigues, G.; Psaila, B.; Kaplan, R.N.; Bromberg, J.F.; Kang, Y.; et al. Pre-metastatic niches: Organ-specific homes for metastases. Nat. Rev. Cancer 2017, 17, 302–317. [Google Scholar] [CrossRef]
  31. Luzzi, K.J.; MacDonald, I.C.; Schmidt, E.E.; Kerkvliet, N.; Morris, V.L.; Chambers, A.F.; Groom, A.C. Multistep nature of metastatic inefficiency: Dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am. J. Pathol. 1998, 153, 865–873. [Google Scholar] [CrossRef]
  32. Bockhorn, M.; Jain, R.K.; Munn, L.L. Active versus passive mechanisms in metastasis: Do cancer cells crawl into vessels, or are they pushed? Lancet Oncol. 2007, 8, 444–448. [Google Scholar] [CrossRef] [Green Version]
  33. Hillen, F.; Griffioen, A.W. Tumour vascularization: Sprouting angiogenesis and beyond. Cancer Metastasis Rev. 2007, 26, 489–502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Park, S.Y.; Nam, J.S. The force awakens: Metastatic dormant cancer cells. Exp. Mol. Med. 2020, 52, 569–581. [Google Scholar] [CrossRef] [Green Version]
  35. Giancotti, F.G. XMechanisms governing metastatic dormancy and reactivation. Cell 2013, 155, 750. [Google Scholar] [CrossRef] [Green Version]
  36. Albrengues, J.; Shields, M.A.; Ng, D.; Park, C.G.; Ambrico, A.; Poindexter, M.E.; Upadhyay, P.; Uyeminami, D.L.; Pommier, A.; Küttner, V.; et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science 2018, 361. [Google Scholar] [CrossRef] [Green Version]
  37. Choi, J.; Cha, Y.J.; Koo, J.S. Adipocyte biology in breast cancer: From silent bystander to active facilitator. Prog. Lipid Res. 2018, 69, 11–20. [Google Scholar] [CrossRef]
  38. Gerber, S.A.; Rybalko, V.Y.; Bigelow, C.E.; Lugade, A.A.; Foster, T.H.; Frelinger, J.G.; Lord, E.M. Preferential attachment of peritoneal tumor metastases to omental immune aggregates and possible role of a unique vascular microenvironment in metastatic survival and growth. Am. J. Pathol. 2006, 169, 1739–1752. [Google Scholar] [CrossRef] [Green Version]
  39. Nieman, K.M.; Kenny, H.A.; Penicka, C.V.; Ladanyi, A.; Buell-Gutbrod, R.; Zillhardt, M.R.; Romero, I.L.; Carey, M.S.; Mills, G.B.; Hotamisligil, G.S.; et al. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat. Med. 2011, 17, 1498–1503. [Google Scholar] [CrossRef] [Green Version]
  40. Cao, H.; Sekiya, M.; Ertunc, M.E.; Burak, M.F.; Mayers, J.R.; White, A.; Inouye, K.; Rickey, L.M.; Ercal, B.C.; Furuhashi, M.; et al. Adipocyte lipid chaperone aP2 Is a secreted adipokine regulating hepatic glucose production. Cell Metab. 2013, 17, 768–778. [Google Scholar] [CrossRef] [Green Version]
  41. Hao, J.; Zhang, Y.; Yan, X.; Yan, F.; Sun, Y.; Zeng, J.; Waigel, S.; Yin, Y.; Fraig, M.M.; Egilmez, N.K.; et al. Circulating Adipose Fatty Acid Binding Protein Is a New Link Underlying Obesity-Associated Breast/Mammary Tumor Development. Cell Metab. 2018, 28, 689–705.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Yan, F.; Shen, N.; Pang, J.X.; Zhang, Y.W.; Rao, E.Y.; Bode, A.M.; Al-Kali, A.; Zhang, D.E.; Litzow, M.R.; Li, B.; et al. Fatty acid-binding protein FABP4 mechanistically links obesity with aggressive AML by enhancing aberrant DNA methylation in AML cells. Leukemia 2017, 31, 1434–1442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Wu, Q.; Li, B.; Li, Z.; Li, J.; Sun, S.; Sun, S. Cancer-associated adipocytes: Key players in breast cancer progression. J. Hematol. Oncol. 2019, 12, 1–15. [Google Scholar] [CrossRef] [PubMed]
  44. Guerrero, J.; Tobar, N.; Cáceres, M.; Espinoza, L.; Escobar, P.; Dotor, J.; Smith, P.C.; Martínez, J. Soluble factors derived from tumor mammary cell lines induce a stromal mammary adipose reversion in human and mice adipose cells. Possible role of TGF-β1 and TNF-α. Breast Cancer Res. Treat. 2010, 119, 497–508. [Google Scholar] [CrossRef]
  45. Andarawewa, K.L.; Motrescu, E.R.; Chenard, M.P.; Gansmuller, A.; Stoll, I.; Tomasetto, C.; Rio, M.C. Stromelysin-3 is a potent negative regulator of adipogenesis participating to cancer cell-adipocyte interaction/crosstalk at the tumor invasive front. Cancer Res. 2005, 65, 10862–10871. [Google Scholar] [CrossRef] [Green Version]
  46. Meunier, P.; Aaron, J.; Edouard, C.; Vignon, G. Osteoporosis and the replacement of cell populations of the marrow by adipose tissue. A quantitative study of 84 iliac bone biopsies. Clin. Orthop. Relat. Res. 1971, 80, 147–154. [Google Scholar] [CrossRef]
  47. Scheller, E.L.; Rosen, C.J. What’s the matter with MAT? Marrow adipose tissue, metabolism, and skeletal health. Ann. N. Y. Acad. Sci. 2014, 1311, 14–30. [Google Scholar] [CrossRef] [Green Version]
  48. Morris, E.V.; Edwards, C.M. The role of bone marrow adipocytes in bone metastasis. J. Bone Oncol. 2016, 5, 121–123. [Google Scholar] [CrossRef] [Green Version]
  49. Olechnowicz, S.W.Z.; Edwards, C.M. Contributions of the host microenvironment to cancer-induced bone disease. Cancer Res. 2014, 74, 1625–1631. [Google Scholar] [CrossRef] [Green Version]
  50. Woolthuis, C.M.; Stranahan, A.W.; Park, C.Y.; Minhajuddin, M.; Gasparetto, M.; Stevens, B.; Pei, S.; Jordan, C.T. Leukemic Stem Cells Evade Chemotherapy by Metabolic Adaptation to an Adipose Tissue Niche. Cell Stem Cell 2016, 19, 23–37. [Google Scholar] [CrossRef] [Green Version]
  51. Herroon, M.K.; Rajagurubandara, E.; Hardaway, A.L.; Powell, K.; Turchick, A.; Feldmann, D.; Podgorski, I. Bone marrow adipocytes promote tumor growth in bone via FABP4-dependent mechanisms. Oncotarget 2013, 4, 2108–2123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Diedrich, J.D.; Rajagurubandara, E.; Herroon, M.K.; Mahapatra, G.; Hüttemann, M.; Podgorski, I. Bone marrow adipocytes promote the warburg phenotype in metastatic prostate tumors via HIF-1α activation. Oncotarget 2016, 7, 64854–64877. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Cawthorn, W.P.; Scheller, E.L.; Learman, B.S.; Parlee, S.D.; Simon, B.R.; Mori, H.; Ning, X.; Bree, A.J.; Schell, B.; Broome, D.T.; et al. Bone marrow adipose tissue is an endocrine organ that contributes to increased circulating adiponectin during caloric restriction. Cell Metab. 2014, 20, 368–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Katira, A.; Tan, P.H. Evolving role of adiponectin in cancer-controversies and update. Cancer Biol. Med. 2016, 13, 101–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Batista, M.L.; Olivan, M.; Alcantara, P.S.M.; Sandoval, R.; Peres, S.B.; Neves, R.X.; Silverio, R.; Maximiano, L.F.; Otoch, J.P.; Seelaender, M. Adipose tissue-derived factors as potential biomarkers in cachectic cancer patients. Cytokine 2013, 61, 532–539. [Google Scholar] [CrossRef] [Green Version]
  56. Tan, P.H.; Tyrrell, H.E.J.; Gao, L.; Xu, D.; Quan, J.; Gill, D.; Rai, L.; Ding, Y.; Plant, G.; Chen, Y.; et al. Adiponectin receptor signaling on dendritic cells blunts antitumor immunity. Cancer Res. 2014, 74, 5711–5722. [Google Scholar] [CrossRef] [Green Version]
  57. Trayhurn, P. Hypoxia and adipose tissue function and dysfunction in obesity. Physiol. Rev. 2013, 93, 1–21. [Google Scholar] [CrossRef] [Green Version]
  58. Chen, B.; Lam, K.S.L.; Wang, Y.; Wu, D.; Lam, M.C.; Shen, J.; Wong, L.; Hoo, R.L.C.; Zhang, J.; Xu, A. Hypoxia dysregulates the production of adiponectin and plasminogen activator inhibitor-1 independent of reactive oxygen species in adipocytes. Biochem. Biophys. Res. Commun. 2006, 341, 549–556. [Google Scholar] [CrossRef]
  59. Wang, B.; Wood, I.S.; Trayhurn, P. Hypoxia induces leptin gene expression and secretion in human preadipocytes: Differential effects of hypoxia on adipokine expression by preadipocytes. J. Endocrinol. 2008, 198, 127–134. [Google Scholar] [CrossRef] [Green Version]
  60. Chen, D.C.; Chung, Y.F.; Yeh, Y.T.; Chaung, H.C.; Kuo, F.C.; Fu, O.Y.; Chen, H.Y.; Hou, M.F.; Yuan, S.S.F. Serum adiponectin and leptin levels in Taiwanese breast cancer patients. Cancer Lett. 2006, 237, 109–114. [Google Scholar] [CrossRef]
  61. Sultana, R.; Kataki, A.C.; Borthakur, B.B.; Basumatary, T.K.; Bose, S. Imbalance in leptin-adiponectin levels and leptin receptor expression as chief contributors to triple negative breast cancer progression in Northeast India. Gene 2017, 621, 51–58. [Google Scholar] [CrossRef]
  62. Barone, I.; Giordano, C.; Bonofiglio, D.; Andò, S.; Catalano, S. The weight of obesity in breast cancer progression and metastasis: Clinical and molecular perspectives. Semin. Cancer Biol. 2020, 60, 274–284. [Google Scholar] [CrossRef] [PubMed]
  63. Alshaker, H.; Krell, J.; Frampton, A.E.; Waxman, J.; Blyuss, O.; Zaikin, A.; Winkler, M.; Stebbing, J.; Yagüe, E.; Pchejetski, D. Leptin induces upregulation of sphingosine kinase 1 in oestrogen receptor-negative breast cancer via Src family kinase-mediated, janus kinase 2-independent pathway. Breast Cancer Res. 2014, 16, 426. [Google Scholar] [CrossRef] [PubMed]
  64. Sabol, R.A.; Bowles, A.C.; Côté, A.; Wise, R.; O’Donnell, B.; Matossian, M.D.; Hossain, F.M.; Burks, H.E.; Del Valle, L.; Miele, L.; et al. Leptin produced by obesity-altered adipose stem cells promotes metastasis but not tumorigenesis of triple-negative breast cancer in orthotopic xenograft and patient-derived xenograft models. Breast Cancer Res. 2019, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Chang, C.C.; Wu, M.J.; Yang, J.Y.; Camarillo, I.G.; Chang, C.J. Leptin-STAT3-G9a signaling promotes obesity-mediated breast cancer progression. Cancer Res. 2015, 75, 2375–2386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Brandon, J.A.; Kraemer, M.; Vandra, J.; Halder, S.; Ubele, M.; Morris, A.J.; Smyth, S.S. Adipose-derived autotaxin regulates inflammation and steatosis associated with diet-induced obesity. PLoS ONE 2019, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Benesch, M.G.K.; Tang, X.; Brindley, D.N. Autotaxin and breast cancer: Towards overcoming treatment barriers and sequelae. Cancers 2020, 12, 374. [Google Scholar] [CrossRef] [Green Version]
  68. Gupta, P.B.; Onder, T.T.; Jiang, G.; Tao, K.; Kuperwasser, C.; Weinberg, R.A.; Lander, E.S. Identification of Selective Inhibitors of Cancer Stem Cells by High-Throughput Screening. Cell 2009, 138, 645–659. [Google Scholar] [CrossRef] [Green Version]
  69. Shim, S.J.; Shin, E.; Lee, C.-S.; Koo, J.S. The expressions of autotaxin-lysophosphatidate signaling-related proteins in metastatic breast cancer. Int. J. Clin. Exp. Pathol. 2019, 12, 2920–2930. [Google Scholar]
  70. Dusaulcy, R.; Rancoule, C.; Grès, S.; Wanecq, E.; Colom, A.; Guigné, C.; Van Meeteren, L.A.; Moolenaar, W.H.; Valet, P.; Saulnier-Blache, J.S. Adipose-specific disruption of autotaxin enhances nutritional fattening and reduces plasma lysophosphatidic acid. J. Lipid Res. 2011, 52, 1247–1255. [Google Scholar] [CrossRef] [Green Version]
  71. Rancoule, C.; Dusaulcy, R.; Tréguer, K.; Grès, S.; Guigné, C.; Quilliot, D.; Valet, P.; Saulnier-Blache, J.S. Depot-specific regulation of autotaxin with obesity in human adipose tissue. J. Physiol. Biochem. 2012, 68, 635–644. [Google Scholar] [CrossRef] [PubMed]
  72. Benesch, M.G.K.; Macintyre, I.T.K.; McMullen, T.P.W.; Brindley, D.N. Coming of age for autotaxin and lysophosphatidate signaling: Clinical applications for preventing, detecting and targeting tumor-promoting inflammation. Cancers 2018, 10, 73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Reeves, V.L.; Trybula, J.S.; Wills, R.C.; Goodpaster, B.H.; Dubé, J.J.; Kienesberger, P.C.; Kershaw, E.E. Serum Autotaxin/ENPP2 correlates with insulin resistance in older humans with obesity. Obesity 2015, 23, 2371–2376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Azare, J.; Doane, A.; Leslie, K.; Chang, Q.; Berishaj, M.; Nnoli, J.; Mark, K.; Al-Ahmadie, H.; Gerald, W.; Hassimi, M.; et al. Stat3 mediates expression of autotaxin in breast cancer. PLoS ONE 2011, 6. [Google Scholar] [CrossRef] [Green Version]
  75. Du, J.; Sun, C.; Hu, Z.; Yang, Y.; Zhu, Y.; Zheng, D.; Gu, L.; Lu, X. Lysophosphatidic Acid Induces MDA-MB-231 Breast Cancer Cells Migration through Activation of PI3K/PAK1/ERK Signaling. PLoS ONE 2010, 5, e15940. [Google Scholar] [CrossRef]
  76. Wu, T.; Van der Kooi, C.; Shah, P.; Charnigo, R.; Huang, C.; Smyth, S.S.; Morris, A.J. Integrin-mediated cell surface recruitment of autotaxin promotes persistent directional cell migration. FASEB J. 2014, 28, 861–870. [Google Scholar] [CrossRef] [Green Version]
  77. Cha, Y.J.; Koo, J.S. Adipokines as therapeutic targets in breast cancer treatment. Expert Opin. Ther. Targets 2018, 22, 941–953. [Google Scholar] [CrossRef]
  78. Benesch, M.G.K.; Tang, X.; Maeda, T.; Ohhata, A.; Zhao, Y.Y.; Kok, B.P.C.; Dewald, J.; Hitt, M.; Curtis, J.M.; McMullen, T.P.W.; et al. Inhibition of autotaxin delays breast tumor growth and lung metastasis in mice. FASEB J. 2014, 28, 2655–2666. [Google Scholar] [CrossRef]
  79. Boucharaba, A.; Serre, C.-M.; Grès, S.; Saulnier-Blache, J.S.; Bordet, J.-C.; Guglielmi, J.; Clézardin, P.; Peyruchaud, O. Platelet-derived lysophosphatidic acid supports the progression of osteolytic bone metastases in breast cancer. J. Clin. Invest. 2004, 114, 1714–1725. [Google Scholar] [CrossRef] [Green Version]
  80. Zhang, H.; Xu, X.; Gajewiak, J.; Tsukahara, R.; Fujiwara, Y.; Liu, J.; Fells, J.I.; Perygin, D.; Parrill, A.L.; Tigyi, G.; et al. Dual activity lysophosphatidic acid receptor pan-antagonist/autotaxin inhibitor reduces breast cancer cell migration in vitro and causes tumor regression in vivo. Cancer Res. 2009, 69, 5441–5449. [Google Scholar] [CrossRef] [Green Version]
  81. Magkrioti, C.; Galaris, A.; Kanellopoulou, P.; Stylianaki, E.A.; Kaffe, E.; Aidinis, V. Autotaxin and chronic inflammatory diseases. J. Autoimmun. 2019, 104, 102327. [Google Scholar] [CrossRef] [PubMed]
  82. Maher, T.M.; Kreuter, M.; Lederer, D.J.; Brown, K.K.; Wuyts, W.; Verbruggen, N.; Stutvoet, S.; Fieuw, A.; Ford, P.; Abi-Saab, W.; et al. Rationale, design and objectives of two phase III, randomised, placebo-controlled studies of GLPG1690, a novel autotaxin inhibitor, in idiopathic pulmonary fibrosis (ISABELA 1 and 2). BMJ Open Respir. Res. 2019, 6. [Google Scholar] [CrossRef]
  83. Maher, T.M.; van der Aar, E.M.; Van de Steen, O.; Allamassey, L.; Desrivot, J.; Dupont, S.; Fagard, L.; Ford, P.; Fieuw, A.; Wuyts, W. Safety, tolerability, pharmacokinetics, and pharmacodynamics of GLPG1690, a novel autotaxin inhibitor, to treat idiopathic pulmonary fibrosis (FLORA): A phase 2a randomised placebo-controlled trial. Lancet Respir. Med. 2018, 6, 627–635. [Google Scholar] [CrossRef]
  84. Tang, X.; Wuest, M.; Benesch, M.G.K.; Dufour, J.; Zhao, Y.Y.; Curtis, J.M.; Monjardet, A.; Heckmann, B.; Murray, D.; Wuest, F.; et al. Inhibition of autotaxin with GLPG1690 increases the efficacy of radiotherapy and chemotherapy in a mouse model of breast cancer. Mol. Cancer Ther. 2020, 19, 63–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Dirat, B.; Bochet, L.; Dabek, M.; Daviaud, D.; Dauvillier, S.; Majed, B.; Wang, Y.Y.; Meulle, A.; Salles, B.; Le Gonidec, S.; et al. Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Res. 2011, 71, 2455–2465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Kern, L.; Mittenbühler, M.J.; Vesting, A.J.; Ostermann, A.L.; Wunderlich, C.M.; Wunderlich, F.T. Obesity-induced TNFα and IL-6 signaling: The missing link between obesity and inflammation-driven liver and colorectal cancers. Cancers 2019, 11, 24. [Google Scholar] [CrossRef] [Green Version]
  87. Arendt, L.M.; McCready, J.; Keller, P.J.; Baker, D.D.; Naber, S.P.; Seewaldt, V.; Kuperwasser, C. Obesity promotes breast cancer by CCL2-mediated macrophage recruitment and angiogenesis. Cancer Res. 2013, 73, 6080–6093. [Google Scholar] [CrossRef] [Green Version]
  88. Tsuyada, A.; Chow, A.; Wu, J.; Somlo, G.; Chu, P.; Loera, S.; Luu, T.; Li, A.X.; Wu, X.; Ye, W.; et al. CCL2 mediates cross-talk between cancer cells and stromal fibroblasts that regulates breast cancer stem cells. Cancer Res. 2012, 72, 2768–2779. [Google Scholar] [CrossRef] [Green Version]
  89. D’Esposito, V.D.; Liguoro, D.; Ambrosio, M.R.; Collina, F.; Cantile, M.; Spinelli, R.; Raciti, G.A.; Miele, C.; Valentino, R.; Campiglia, P.; et al. Adipose microenvironment promotes triple negative breast cancer cell invasiveness and dissemination by producing CCL5. Oncotarget 2016, 7, 24495–24509. [Google Scholar] [CrossRef] [Green Version]
  90. Velasco-Velázquez, M.; Jiao, X.; De La Fuente, M.; Pestell, T.G.; Ertel, A.; Lisanti, M.P.; Pestell, R.G. CCR5 antagonist blocks metastasis of basal breast cancer cells. Cancer Res. 2012, 72, 3839–3850. [Google Scholar] [CrossRef] [Green Version]
  91. Rybinska, I.; Agresti, R.; Trapani, A.; Tagliabue, E.; Triulzi, T. Adipocytes in Breast Cancer, the Thick and the Thin. Cells 2020, 9, 560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Bastard, J.-P.; Jardel, C.; Bruckert, E.; Blondy, P.; Capeau, J.; Laville, M.; Vidal, H.; Hainque, B. Elevated Levels of Interleukin 6 Are Reduced in Serum and Subcutaneous Adipose Tissue of Obese Women after Weight Loss*. J. Clin. Endocrinol. Metab. 2000, 85, 3338–3342. [Google Scholar] [CrossRef] [PubMed]
  93. Kim, H.S.; Jung, M.; Choi, S.K.; Woo, J.; Piao, Y.J.; Hwang, E.H.; Kim, H.; Kim, S.J.; Moon, W.K. IL-6-mediated cross-talk between human preadipocytes and ductal carcinoma in situ in breast cancer progression. J. Exp. Clin. Cancer Res. 2018, 37, 200. [Google Scholar] [CrossRef] [PubMed]
  94. Sansone, P.; Storci, G.; Tavolari, S.; Guarnieri, T.; Giovannini, C.; Taffurelli, M.; Ceccarelli, C.; Santini, D.; Paterini, P.; Marcu, K.B.; et al. IL-6 triggers malignant features in mammospheres from human ductal breast carcinoma and normal mammary gland. J. Clin. Invest. 2007, 117, 3988–4002. [Google Scholar] [CrossRef]
  95. Marotta, L.L.C.; Almendro, V.; Marusyk, A.; Shipitsin, M.; Schemme, J.; Walker, S.R.; Bloushtain-Qimron, N.; Kim, J.J.; Choudhury, S.A.; Maruyama, R.; et al. The JAK2/STAT3 signaling pathway is required for growth of CD44 +CD24- stem cell-like breast cancer cells in human tumors. J. Clin. Invest. 2011, 121, 2723–2735. [Google Scholar] [CrossRef]
  96. Bergmann, J.; Müller, M.; Baumann, N.; Reichert, M.; Heneweer, C.; Bolik, J.; Lücke, K.; Gruber, S.; Carambia, A.; Boretius, S.; et al. IL-6 trans-signaling is essential for the development of hepatocellular carcinoma in mice. Hepatology 2017, 65, 89–103. [Google Scholar] [CrossRef]
  97. Wang, C.-Q.; Sun, H.-T.; Gao, X.-M.; Ren, N.; Sheng, Y.-Y.; Wang, Z.; Zheng, Y.; Wei, J.-W.; Zhang, K.-L.; Yu, X.-X.; et al. Interleukin-6 enhances cancer stemness and promotes metastasis of hepatocellular carcinoma via up-regulating osteopontin expression. Am. J. Cancer Res. 2016, 6, 1873–1889. [Google Scholar]
  98. Lukens, J.R.; Dixit, V.D.; Kanneganti, T.D. Inflammasome activation in obesity-related inflammatory diseases and autoimmunity. Discov. Med. 2011, 12, 65–74. [Google Scholar]
  99. Coleman, R.E.; Marshall, H.; Cameron, D.; Dodwell, D.; Burkinshaw, R.; Keane, M.; Gil, M.; Houston, S.J.; Grieve, R.J.; Barrett-Lee, P.J.; et al. Breast-cancer adjuvant therapy with zoledronic acid. N. Engl. J. Med. 2011, 365, 1396–1405. [Google Scholar] [CrossRef] [Green Version]
  100. Tulotta, C.; Ottewell, P. The role of IL-1B in breast cancer bone metastasis. Endocr. Relat. Cancer 2018, 25, R421–R434. [Google Scholar] [CrossRef] [Green Version]
  101. Nutter, F.; Holen, I.; Brown, H.K.; Cross, S.S.; Evans, C.A.; Walker, M.; Coleman, R.E.; Westbrook, J.A.; Selby, P.J.; Brown, J.E.; et al. Different molecular profiles are associated with breast cancer cell homing compared with colonisation of bone: Evidence using a novel bone-seeking cell line. Endocr. Relat. Cancer 2014, 21, 327–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Hagemann, T.; Wilson, J.; Kulbe, H.; Li, N.F.; Leinster, D.A.; Charles, K.; Klemm, F.; Pukrop, T.; Binder, C.; Balkwill, F.R. Macrophages Induce Invasiveness of Epithelial Cancer Cells Via NF-κB and JNK. J. Immunol. 2005, 175, 1197–1205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Chua, H.L.; Bhat-Nakshatri, P.; Clare, S.E.; Morimiya, A.; Badve, S.; Nakshatri, H. NF-κB represses E-cadherin expression and enhances epithelial to mesenchymal transition of mammary epithelial cells: Potential involvement of ZEB-1 and ZEB-2. Oncogene 2007, 26, 711–724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Sturm, J.W.; Magdeburg, R.; Berger, K.; Petruch, B.; Samel, S.; Bönninghoff, R.; Keese, M.; Hafner, M.; Post, S. Influence of TNFA on the formation of liver metastases in a syngenic mouse model. Int. J. Cancer 2003, 107, 11–21. [Google Scholar] [CrossRef]
  105. Ham, B.; Fernandez, M.C.; D’Costa, Z.; Brodt, P. The diverse roles of the TNF axis in cancer progression and metastasis. Trends Cancer Res. 2016, 11, 1–27. [Google Scholar]
  106. Faria, S.S.; Corrêa, L.H.; Heyn, G.S.; de Sant’Ana, L.P.; Almeida, R.D.N.; Magalhães, K.G. Obesity and breast cancer: The role of crown-like structures in breast adipose tissue in tumor progression, prognosis, and therapy. J. Breast Cancer 2020, 23, 233–245. [Google Scholar] [CrossRef]
  107. Berstein, L.M.; Iyevleva, A.G.; Mukhina, M.S.; Vasilyev, D.A.; Poroshina, T.E. Features of omental adipose tissue in endometrial cancer patients with ‘standard’ or ‘metabolically healthy’ obesity: Associations with tumor process characteristics. Springerplus 2016, 5, 1900. [Google Scholar] [CrossRef] [Green Version]
  108. Gucalp, A.; Iyengar, N.M.; Zhou, X.K.; Giri, D.D.; Falcone, D.J.; Wang, H.; Williams, S.; Krasne, M.D.; Yaghnam, I.; Kunzel, B.; et al. Periprostatic adipose inflammation is associated with high-grade prostate cancer. Prostate Cancer Prostatic Dis. 2017, 20, 418–423. [Google Scholar] [CrossRef]
  109. Itoh, M.; Kato, H.; Suganami, T.; Konuma, K.; Marumoto, Y.; Terai, S.; Sakugawa, H.; Kanai, S.; Hamaguchi, M.; Fukaishi, T.; et al. Hepatic crown-like structure: A unique histological feature in non-alcoholic steatohepatitis in mice and humans. PLoS ONE 2013, 8. [Google Scholar] [CrossRef] [Green Version]
  110. Mullooly, M.; Yang, H.P.; Falk, R.T.; Nyante, S.J.; Cora, R.; Pfeiffer, R.M.; Radisky, D.C.; Visscher, D.W.; Hartmann, L.C.; Carter, J.M.; et al. Relationship between crown-like structures and sex-steroid hormones in breast adipose tissue and serum among postmenopausal breast cancer patients. Breast Cancer Res. 2017, 19. [Google Scholar] [CrossRef] [Green Version]
  111. Cortellini, A.; Bersanelli, M.; Buti, S.; Cannita, K.; Santini, D.; Perrone, F.; Giusti, R.; Tiseo, M.; Michiara, M.; Di Marino, P.; et al. A multicenter study of body mass index in cancer patients treated with anti-PD-1/PD-L1 immune checkpoint inhibitors: When overweight becomes favorable. J. Immunother. Cancer 2019, 7, 57. [Google Scholar] [CrossRef] [PubMed]
  112. Plaks, V.; Kong, N.; Werb, Z. The Cancer Stem Cell Niche: How Essential Is the Niche in Regulating Stemness of Tumor Cells? Cell Stem Cell 2015, 16, 225–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Zhang, S.; Yang, X.; Wang, L.; Zhang, C. Interplay between inflammatory tumor microenvironment and cancer stem cells (Review). Oncol. Lett. 2018, 16, 679–686. [Google Scholar] [CrossRef] [PubMed]
  114. Lee, S.H.; Hong, H.S.; Liu, Z.X.; Kim, R.H.; Kang, M.K.; Park, N.-H.; Shin, K.-H. TNFα enhances cancer stem cell-like phenotype via Notch-Hes1 activation in oral squamous cell carcinoma cells. Biochem. Biophys. Res. Commun. 2012, 424, 58–64. [Google Scholar] [CrossRef] [Green Version]
  115. Ostyn, P.; El Machhour, R.; Begard, S.; Kotecki, N.; Vandomme, J.; Flamenco, P.; Segard, P.; Masselot, B.; Formstecher, P.; Touil, Y.; et al. Transient TNF regulates the self-renewing capacity of stem-like label-retaining cells in sphere and skin equivalent models of melanoma. Cell Commun. Signal. 2014, 12. [Google Scholar] [CrossRef] [PubMed]
  116. Zhao, X.; Ma, L.; Dai, L.; Zuo, D.; Li, X.; Zhu, H.; Xu, F. TNF-α promotes the malignant transformation of intestinal stem cells through the NF-κB and Wnt/β-catenin signaling pathways. Oncol. Rep. 2020, 44, 577–588. [Google Scholar] [CrossRef]
  117. Storci, G.; Sansone, P.; Mari, S.; D’Uva, G.; Tavolari, S.; Guarnieri, T.; Taffurelli, M.; Ceccarelli, C.; Santini, D.; Chieco, P.; et al. TNFalpha up-regulates SLUG via the NF-kappaB/HIF1alpha axis, which imparts breast cancer cells with a stem cell-like phenotype. J. Cell. Physiol. 2010, 225, 682–691. [Google Scholar] [CrossRef] [Green Version]
  118. Liu, W.; Lu, X.; Shi, P.; Yang, G.; Zhou, Z.; Li, W.; Mao, X.; Jiang, D.; Chen, C. TNF-α increases breast cancer stem-like cells through up-regulating TAZ expression via the non-canonical NF-κB pathway. Sci. Rep. 2020, 10, 1–11. [Google Scholar] [CrossRef] [Green Version]
  119. Teslow, E.A.; Mitrea, C.; Bao, B.; Mohammad, R.M.; Polin, L.A.; Dyson, G.; Purrington, K.S.; Bollig-Fischer, A. Obesity-induced MBD2_v2 expression promotes tumor-initiating triple-negative breast cancer stem cells. Mol. Oncol. 2019, 13, 894–908. [Google Scholar] [CrossRef]
  120. Fukumura, D.; Ushiyama, A.; Duda, D.G.; Xu, L.; Tam, J.; Krishna, V.; Chatterjee, K.; Garkavtsev, I.; Jain, R.K. Paracrine regulation of angiogenesis and adipocyte differentiation during in vivo adipogenesis. Circ. Res. 2003, 93. [Google Scholar] [CrossRef] [Green Version]
  121. Fukumura, D.; Incio, J.; Shankaraiah, R.C.; Jain, R.K. Obesity and Cancer: An Angiogenic and Inflammatory Link. Microcirculation 2016, 23, 191–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Zhou, W.; Guo, S.; Gonzalez-Perez, R.R. Leptin pro-angiogenic signature in breast cancer is linked to IL-1 signalling. Br. J. Cancer 2011, 104, 128–137. [Google Scholar] [CrossRef] [PubMed]
  123. Kolb, R.; Kluz, P.; Tan, Z.W.; Borcherding, N.; Bormann, N.; Vishwakarma, A.; Balcziak, L.; Zhu, P.; Davies, B.S.; Gourronc, F.; et al. Obesity-associated inflammation promotes angiogenesis and breast cancer via angiopoietin-like 4. Oncogene 2019, 38, 2351–2363. [Google Scholar] [CrossRef] [PubMed]
  124. Lohmann, A.E.; Chia, S. Patients with metastatic breast cancer using bevacizumab as a treatment: Is there still a role for it? Curr. Treat. Options Oncol. 2012, 13, 249–262. [Google Scholar] [CrossRef]
  125. Jain, R.K.; Duda, D.G.; Willett, C.G.; Sahani, D.V.; Zhu, A.X.; Loeffler, J.S.; Batchelor, T.T.; Sorensen, A.G. Biomarkers of response and resistance to antiangiogenic therapy. Nat. Rev. Clin. Oncol. 2009, 6, 327–338. [Google Scholar] [CrossRef]
  126. Incio, J.; Ligibel, J.A.; McManus, D.T.; Suboj, P.; Jung, K.; Kawaguchi, K.; Pinter, M.; Babykutty, S.; Chin, S.M.; Vardam, T.D.; et al. Obesity promotes resistance to anti-VEGF therapy in breast cancer by up-regulating IL-6 and potentially FGF-2. Sci. Transl. Med. 2018, 10, 945. [Google Scholar] [CrossRef] [Green Version]
  127. Fernández-Cortés, M.; Delgado-Bellido, D.; Oliver, F.J. Vasculogenic Mimicry: Become an Endothelial Cell “But Not So Much”. Front. Oncol. 2019, 9, 803. [Google Scholar] [CrossRef] [Green Version]
  128. Coelho, P.; Almeida, J.; Prudêncio, C.; Fernandes, R.; Soares, R. Effect of Adipocyte Secretome in Melanoma Progression and Vasculogenic Mimicry. J. Cell. Biochem. 2016, 117, 1697–1706. [Google Scholar] [CrossRef] [Green Version]
  129. Marín, D.; Sabater, B. The cancer Warburg effect may be a testable example of the minimum entropy production rate principle. Phys. Biol. 2017, 14, 024001. [Google Scholar] [CrossRef]
  130. Fu, Y.; Liu, S.; Yin, S.; Niu, W.; Xiong, W.; Tan, M.; Li, G.; Zhou, M. The reverse Warburg effect is likely to be an Achilles’ heel of cancer that can be exploited for cancer therapy. Oncotarget 2017, 8, 57813–57825. [Google Scholar] [CrossRef] [Green Version]
  131. Pavlides, S.; Whitaker-Menezes, D.; Castello-Cros, R.; Flomenberg, N.; Witkiewicz, A.K.; Frank, P.G.; Casimiro, M.C.; Wang, C.; Fortina, P.; Addya, S.; et al. The reverse Warburg effect: Aerobic glycolysis in cancer associated fibroblasts and the tumor stroma. Cell Cycle 2009, 8, 3984–4001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Armignacco, R.; Cantini, G.; Poli, G.; Guasti, D.; Nesi, G.; Romagnoli, P.; Mannell, M.; Luconi, M. The adipose stem cell as a novel metabolic actor in adrenocortical carcinoma progression: Evidence from an in vitro tumor microenvironment crosstalk model. Cancers 2019, 11, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Martinez-Outschoorn, U.E.; Lin, Z.; Whitaker-Menezes, D.; Howell, A.; Sotgia, F.; Lisanti, M.P. Ketone body utilization drives tumor growth and metastasis. Cell Cycle 2012, 11, 3964–3971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Wang, Y.Y.; Attané, C.; Milhas, D.; Dirat, B.; Dauvillier, S.; Guerard, A.; Gilhodes, J.; Lazar, I.; Alet, N.; Laurent, V.; et al. Mammary adipocytes stimulate breast cancer invasion through metabolic remodeling of tumor cells. JCI Insight 2017, 2. [Google Scholar] [CrossRef] [Green Version]
  135. Zaidi, N.; Lupien, L.; Kuemmerle, N.B.; Kinlaw, W.B.; Swinnen, J.V.; Smans, K. Lipogenesis and lipolysis: The pathways exploited by the cancer cells to acquire fatty acids. Prog. Lipid Res. 2013, 52, 585–589. [Google Scholar] [CrossRef] [Green Version]
  136. Zhang, M.; Di Martino, J.S.; Bowman, R.L.; Campbell, N.R.; Baksh, S.C.; Simon-Vermot, T.; Kim, I.S.; Haldeman, P.; Mondal, C.; Yong-Gonzales, V.; et al. Adipocyte-derived lipids mediate melanoma progression via FATP proteins. Cancer Discov. 2018, 8, 1006–1025. [Google Scholar] [CrossRef] [Green Version]
  137. Shafat, M.S.; Oellerich, T.; Mohr, S.; Robinson, S.D.; Edwards, D.R.; Marlein, C.R.; Piddock, R.E.; Fenech, M.; Zaitseva, L.; Abdul-Aziz, A.; et al. Leukemic blasts program bone marrow adipocytes to generate a protumoral microenvironment. Blood 2017, 129, 1320–1332. [Google Scholar] [CrossRef]
  138. Martinez-Outschoorn, U.E.; Peiris-Pagés, M.; Pestell, R.G.; Sotgia, F.; Lisanti, M.P. Cancer metabolism: A therapeutic perspective. Nat. Rev. Clin. Oncol. 2017, 14, 11–31. [Google Scholar] [CrossRef]
  139. Cluntun, A.A.; Lukey, M.J.; Cerione, R.A.; Locasale, J.W. Glutamine Metabolism in Cancer: Understanding the Heterogeneity. Trends Cancer 2017, 3, 169–180. [Google Scholar] [CrossRef] [Green Version]
  140. Yang, L.; Achreja, A.; Yeung, T.L.; Mangala, L.S.; Jiang, D.; Han, C.; Baddour, J.; Marini, J.C.; Ni, J.; Nakahara, R.; et al. Targeting Stromal Glutamine Synthetase in Tumors Disrupts Tumor Microenvironment-Regulated Cancer Cell Growth. Cell Metab. 2016, 24, 685–700. [Google Scholar] [CrossRef] [Green Version]
  141. Petrus, P.; Lecoutre, S.; Dollet, L.; Wiel, C.; Sulen, A.; Gao, H.; Tavira, B.; Laurencikiene, J.; Rooyackers, O.; Checa, A.; et al. Glutamine Links Obesity to Inflammation in Human White Adipose Tissue. Cell Metab. 2020, 31, 375–390. [Google Scholar] [CrossRef] [PubMed]
  142. Recouvreux, M.V.; Moldenhauer, M.R.; Galenkamp, K.M.O.; Jung, M.; James, B.; Zhang, Y.; Lowy, A.; Bagchi, A.; Commisso, C. Glutamine depletion regulates Slug to promote EMT and metastasis in pancreatic cancer. J. Exp. Med. 2020, 217. [Google Scholar] [CrossRef] [PubMed]
  143. Pavlova, N.N.; Hui, S.; Ghergurovich, J.M.; Fan, J.; Intlekofer, A.M.; White, R.M.; Rabinowitz, J.D.; Thompson, C.B.; Zhang, J. As Extracellular Glutamine Levels Decline, Asparagine Becomes an Essential Amino Acid. Cell Metab. 2018, 27, 428–438.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Luo, M.; Brooks, M.; Wicha, M.S. Asparagine and Glutamine: Co-conspirators Fueling Metastasis. Cell Metab. 2018, 27, 947–949. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Knott, S.R.V.; Wagenblast, E.; Khan, S.; Kim, S.Y.; Soto, M.; Wagner, M.; Turgeon, M.O.; Fish, L.; Erard, N.; Gable, A.L.; et al. Asparagine bioavailability governs metastasis in a model of breast cancer. Nature 2018, 554, 378–381. [Google Scholar] [CrossRef] [PubMed]
  146. Rizi, B.S.; Caneba, C.; Nowicka, A.; Nabiyar, A.W.; Liu, X.; Chen, K.; Klopp, A.; Nagrath, D. Nitric oxide mediates metabolic coupling of omentum-derived adipose stroma to ovarian and endometrial cancer cells. Cancer Res. 2015, 75, 456–471. [Google Scholar] [CrossRef] [Green Version]
  147. Al-Koussa, H.; El Mais, N.; Maalouf, H.; Abi-Habib, R.; El-Sibai, M. Arginine deprivation: A potential therapeutic for cancer cell metastasis? A review. Cancer Cell Int. 2020, 20, 150. [Google Scholar] [CrossRef]
  148. You, J.; Chen, W.; Chen, J.; Zheng, Q.; Dong, J.; Zhu, Y. The oncogenic role of ARG1 in progression and metastasis of hepatocellular carcinoma. BioMed Res. Int. 2018, 2018. [Google Scholar] [CrossRef]
  149. Zaytouni, T.; Tsai, P.Y.; Hitchcock, D.S.; Dubois, C.D.; Freinkman, E.; Lin, L.; Morales-Oyarvide, V.; Lenehan, P.J.; Wolpin, B.M.; Mino-Kenudson, M.; et al. Critical role for arginase 2 in obesity-Associated pancreatic cancer. Nat. Commun. 2017, 8, 1–12. [Google Scholar] [CrossRef]
  150. Spinelli, J.B.; Yoon, H.; Ringel, A.E.; Jeanfavre, S.; Clish, C.B.; Haigis, M.C. Metabolic recycling of ammonia via glutamate dehydrogenase supports breast cancer biomass. Science 2017, 358, 941–946. [Google Scholar] [CrossRef] [Green Version]
  151. Thomas, D.D.; Corkey, B.E.; Istfan, N.W.; Apovian, C.M. Hyperinsulinemia: An early indicator of metabolic dysfunction. J. Endocr. Soc. 2019, 3, 1727–1747. [Google Scholar] [CrossRef] [PubMed]
  152. Ireland, L.; Santos, A.; Campbell, F.; Figueiredo, C.; Hammond, D.; Ellies, L.G.; Weyer-Czernilofsky, U.; Bogenrieder, T.; Schmid, M.; Mielgo, A. Blockade of insulin-like growth factors increases efficacy of paclitaxel in metastatic breast cancer. Oncogene 2018, 37, 2022–2036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Malaguarnera, R.; Belfiore, A. The emerging role of insulin and insulin-like growth factor signaling in cancer stem cells. Front. Endocrinol. 2014, 5, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Baserga, R. The decline and fall of the IGF-I receptor. J. Cell. Physiol. 2013, 228, 675–679. [Google Scholar] [CrossRef]
  155. Tong, Y.; Wu, J.; Huang, O.; He, J.; Zhu, L.; Chen, W.; Li, Y.; Chen, X.; Shen, K. IGF-1 Interacted With Obesity in Prognosis Prediction in HER2-Positive Breast Cancer Patients. Front. Oncol. 2020, 10. [Google Scholar] [CrossRef]
  156. Wu, Y.; Brodt, P.; Sun, H.; Mejia, W.; Novosyadlyy, R.; Nunez, N.; Chen, X.; Mendoza, A.; Hong, S.H.; Khanna, C.; et al. Insulin-like growth factor-I regulates the liver microenvironment in obese mice and promotes liver metastasis. Cancer Res. 2010, 70, 57–67. [Google Scholar] [CrossRef] [Green Version]
  157. Rayes, R.F.; Milette, S.; Fernandez, M.C.; Ham, B.; Wang, N.; Bourdeau, F.; Perrino, S.; Yakar, S.; Brodt, P. Loss of neutrophil polarization in colon carcinoma liver metastases of mice with an inducible, liver-specific IGF-I deficiency. Oncotarget 2018, 9, 15691–15704. [Google Scholar] [CrossRef] [Green Version]
  158. Ruiz-Ojeda, F.J.; Méndez-Gutiérrez, A.; Aguilera, C.M.; Plaza-Díaz, J. Extracellular matrix remodeling of adipose tissue in obesity and metabolic diseases. Int. J. Mol. Sci. 2019, 20, 4888. [Google Scholar] [CrossRef] [Green Version]
  159. Buechler, C.; Krautbauer, S.; Eisinger, K. Adipose tissue fibrosis. World J. Diabetes 2015, 6, 548. [Google Scholar] [CrossRef]
  160. Hasegawa, Y.; Ikeda, K.; Chen, Y.; Alba, D.L.; Stifler, D.; Shinoda, K.; Hosono, T.; Maretich, P.; Yang, Y.; Ishigaki, Y.; et al. Repression of Adipose Tissue Fibrosis through a PRDM16-GTF2IRD1 Complex Improves Systemic Glucose Homeostasis. Cell Metab. 2018, 27, 180–194.e6. [Google Scholar] [CrossRef] [Green Version]
  161. Oikonomou, E.K.; Antoniades, C. The role of adipose tissue in cardiovascular health and disease. Nat. Rev. Cardiol. 2019, 16, 83–99. [Google Scholar] [CrossRef] [PubMed]
  162. Moorman, A.M.; Vink, R.; Heijmans, H.J.; Van Der Palen, J.; Kouwenhoven, E.A. The prognostic value of tumour-stroma ratio in triple-negative breast cancer. Eur. J. Surg. Oncol. 2012, 38, 307–313. [Google Scholar] [CrossRef] [PubMed]
  163. Zippi, M.; De Toma, G.; Minervini, G.; Cassieri, C.; Pica, R.; Colarusso, D.; Stock, S.; Crispino, P. Desmoplasia influenced recurrence of disease and mortality in stage III colorectal cancer within five years after surgery and adjuvant therapy. Saudi J. Gastroenterol. 2017, 23, 39. [Google Scholar] [CrossRef] [PubMed]
  164. Incio, J.; Liu, H.; Suboj, P.; Chin, S.M.; Chen, I.X.; Pinter, M.; Ng, M.R.; Nia, H.T.; Grahovac, J.; Kao, S.; et al. Obesity-induced inflammation and desmoplasia promote pancreatic cancer progression and resistance to chemotherapy. Cancer Discov. 2016, 6, 852–869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Verset, L.; Tommelein, J.; Lopez, X.M.; Decaestecker, C.; Boterberg, T.; De Vlieghere, E.; Salmon, I.; Mareel, M.; Bracke, M.; De Wever, O.; et al. Impact of neoadjuvant therapy on cancer-associated fibroblasts in rectal cancer. Radiother. Oncol. 2015, 116, 449–454. [Google Scholar] [CrossRef] [PubMed]
  166. Arkan, M.C. Cancer: Fat and the fate of pancreatic tumours. Nature 2016, 536, 157–158. [Google Scholar] [CrossRef] [PubMed]
  167. Bochet, L.; Lehuédé, C.; Dauvillier, S.; Wang, Y.Y.; Dirat, B.; Laurent, V.; Dray, C.; Guiet, R.; Maridonneau-Parini, I.; Le Gonidec, S.; et al. Adipocyte-derived fibroblasts promote tumor progression and contribute to the desmoplastic reaction in breast cancer. Cancer Res. 2013, 73, 5657–5668. [Google Scholar] [CrossRef] [Green Version]
  168. Park, J.; Scherer, P.E. Adipocyte-derived endotrophin promotes malignant tumor progression. J. Clin. Invest. 2012, 122, 4243–4256. [Google Scholar] [CrossRef] [Green Version]
  169. Iyengar, P.; Espina, V.; Williams, T.W.; Lin, Y.; Berry, D.; Jelicks, L.A.; Lee, H.; Temple, K.; Graves, R.; Pollard, J.; et al. Adipocyte-derived collagen VI affects early mammary tumor progression in vivo, demonstrating a critical interaction in the tumor/stroma microenvironment. J. Clin. Invest. 2005, 115, 1163–1176. [Google Scholar] [CrossRef] [Green Version]
  170. Wei, X.; Li, S.; He, J.; Du, H.; Liu, Y.; Yu, W.; Hu, H.; Han, L.; Wang, C.; Li, H.; et al. Tumor-secreted PAI-1 promotes breast cancer metastasis via the induction of adipocyte-derived collagen remodeling. Cell Commun. Signal. 2019, 17, 58. [Google Scholar] [CrossRef] [Green Version]
  171. Jotzu, C.; Alt, E.; Welte, G.; Li, J.; Hennessy, B.T.; Devarajan, E.; Krishnappa, S.; Pinilla, S.; Droll, L.; Song, Y.H. Adipose tissue derived stem cells differentiate into carcinoma-associated fibroblast-like cells under the influence of tumor derived factors. Cell. Oncol. 2011, 34, 55–67. [Google Scholar] [CrossRef] [PubMed]
  172. Kang, H.; Watkins, G.; Parr, C.; Douglas-Jones, A.; Mansel, R.E.; Jiang, W.G. Stromal cell derived factor-1: Its influence on invasiveness and migration of breast cancer cells in vitro, and its association with prognosis and survival in human breast cancer. Breast Cancer Res. 2005, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Cozzo, A.J.; Fuller, A.M.; Makowski, L. Contribution of adipose tissue to development of cancer. Compr. Physiol. 2018, 8, 237–282. [Google Scholar] [CrossRef]
  174. Wynn, T.A.; Barron, L. Macrophages: Master regulators of inflammation and fibrosis. Semin. Liver Dis. 2010, 30, 245–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Corrêa, L.H.; Corrêa, R.; Farinasso, C.M.; de Sant’Ana, D.L.P.; Magalhães, K.G. Adipocytes and macrophages interplay in the orchestration of tumor microenvironment: New implications in cancer progression. Front. Immunol. 2017, 8, 1. [Google Scholar] [CrossRef] [PubMed]
  176. Jaoude, J.; Koh, Y. Matrix metalloproteinases in exercise and obesity. Vasc. Health Risk Manag. 2016, 12, 287–295. [Google Scholar] [PubMed] [Green Version]
  177. Motrescu, E.R.; Blaise, S.; Etique, N.; Messaddeq, N.; Chenard, M.P.; Stoll, I.; Tomasetto, C.; Rio, M.C. Matrix metalloproteinase-11/stromelysin-3 exhibits collagenolytic function against collagen VI under normal and malignant conditions. Oncogene 2008, 27, 6347–6355. [Google Scholar] [CrossRef] [Green Version]
  178. De Lope, L.R.; Alcíbar, O.L.; López, A.A.; Hergueta-Redondo, M.; Peinado, H. Tumour–adipose tissue crosstalk: Fuelling tumour metastasis by extracellular vesicles. Philos. Trans. R. Soc. B Biol. Sci. 2018, 373, 20160485. [Google Scholar] [CrossRef]
  179. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
  180. Lazar, I.; Clement, E.; Dauvillier, S.; Milhas, D.; Ducoux-Petit, M.; LeGonidec, S.; Moro, C.; Soldan, V.; Dalle, S.; Balor, S.; et al. Adipocyte Exosomes Promote Melanoma Aggressiveness through Fatty Acid Oxidation: A Novel Mechanism Linking Obesity and Cancer. Cancer Res. 2016, 76, 4051–4057. [Google Scholar] [CrossRef] [Green Version]
  181. Milane, L.; Singh, A.; Mattheolabakis, G.; Suresh, M.; Amiji, M.M. Exosome mediated communication within the tumor microenvironment. J. Control. Release 2015, 219, 278–294. [Google Scholar] [CrossRef] [PubMed]
  182. Sun, S.; Wu, Q.; Li, J.; Li, Z.; Sun, S.; Zhu, S.; Wang, L.; Wu, J.; Yuan, J.; Zhang, Y.; et al. Exosomes from the tumour-adipocyte interplay stimulate beige/brown differentiation and reprogram metabolism in stromal adipocytes to promote tumour progression. J. Exp. Clin. Cancer Res. 2019, 38, 223. [Google Scholar] [CrossRef] [Green Version]
  183. Sagar, G.; Sah, R.P.; Javeed, N.; Dutta, S.K.; Smyrk, T.C.; Lau, J.S.; Giorgadze, N.; Tchkonia, T.; Kirkland, J.L.; Chari, S.T.; et al. Pathogenesis of pancreatic cancer exosome-induced lipolysis in adipose tissue. Gut 2016, 65, 1165–1174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Wang, J.; Wu, Y.; Guo, J.; Fei, X.; Yu, L.; Ma, S. Adipocyte-derived exosomes promote lung cancer metastasis by increasing MMP9 activity via transferring MMP3 to lung cancer cells. Oncotarget 2017, 8, 81880–81891. [Google Scholar] [CrossRef]
  185. Wang, S.; Su, X.; Xu, M.; Xiao, X.; Li, X.; Li, H.; Keating, A.; Zhao, R.C. Exosomes secreted by mesenchymal stromal/stem cell-derived adipocytes promote breast cancer cell growth via activation of Hippo signaling pathway. Stem Cell Res. Ther. 2019, 10, 117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Benito-Martin, A.; Paik, P.; Mushannen, M.; Bhardwaj, P.; Oshchepkova, S.; Spector, J.; Brown, K.A. SAT-126 Breast Adipose Tissue Extracellular Vesicles from Obese Women Increase Breast Cancer Aggressiveness—A Novel Mechanism for the Obesity-Breast Cancer Link. J. Endocr. Soc. 2020, 4. [Google Scholar] [CrossRef]
  187. Clement, E.; Lazar, I.; Attané, C.; Carrié, L.; Dauvillier, S.; Ducoux-Petit, M.; Menneteau, T.; Moutahir, M.; Le Gonidec, S.; Dalle, S.; et al. Adipocyte vesicles: ‘all-in-one’ packages that stimulate tumor mitochondrial metabolism and dynamics. bioRxiv 2019, 649327. [Google Scholar] [CrossRef]
  188. Balaban, S.; Shearer, R.F.; Lee, L.S.; van Geldermalsen, M.; Schreuder, M.; Shtein, H.C.; Cairns, R.; Thomas, K.C.; Fazakerley, D.J.; Grewal, T.; et al. Adipocyte lipolysis links obesity to breast cancer growth: Adipocyte-derived fatty acids drive breast cancer cell proliferation and migration. Cancer Metab. 2017, 5, 1. [Google Scholar] [CrossRef] [Green Version]
  189. Kuo, C.Y.; Ann, D.K. When fats commit crimes: Fatty acid metabolism, cancer stemness and therapeutic resistance. Cancer Commun. 2018, 38, 47. [Google Scholar] [CrossRef] [Green Version]
  190. Skowron, F.; Bérard, F.; Balme, B.; Maucort-Boulch, D. Role of obesity on the thickness of primary cutaneous melanoma. J. Eur. Acad. Dermatol. Venereol. 2015, 29, 262–269. [Google Scholar] [CrossRef]
  191. Clement, E.; Lazar, I.; Muller, C.; Nieto, L. Obesity and melanoma: Could fat be fueling malignancy? Pigment Cell Melanoma Res. 2017, 30, 294–306. [Google Scholar] [CrossRef] [Green Version]
  192. Lopatina, T.; Bruno, S.; Tetta, C.; Kalinina, N.; Porta, M.; Camussi, G. Platelet-derived growth factor regulates the secretion of extracellular vesicles by adipose mesenchymal stem cells and enhances their angiogenic potential. Cell Commun. Signal. 2014, 12, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Ren, S.; Chen, J.; Duscher, D.; Liu, Y.; Guo, G.; Kang, Y.; Xiong, H.; Zhan, P.; Wang, Y.; Wang, C.; et al. Microvesicles from human adipose stem cells promote wound healing by optimizing cellular functions via AKT and ERK signaling pathways. Stem Cell Res. Ther. 2019, 10, 47. [Google Scholar] [CrossRef] [PubMed]
  194. An, Y.; Zhao, J.; Nie, F.; Qin, Z.; Xue, H.; Wang, G.; Li, D. Exosomes from Adipose-Derived Stem Cells (ADSCs) Overexpressing miR-21 Promote Vascularization of Endothelial Cells. Sci. Rep. 2019, 9, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Liu, Z.; Xu, Y.; Wan, Y.; Gao, J.; Chu, Y.; Li, J. Exosomes from adipose-derived mesenchymal stem cells prevent cardiomyocyte apoptosis induced by oxidative stress. Cell Death Discov. 2019, 5, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Blazquez, R.; Sanchez-Margallo, F.M.; de la Rosa, O.; Dalemans, W.; Álvarez, V.; Tarazona, R.; Casado, J.G. Immunomodulatory potential of human adipose mesenchymal stem cells derived exosomes on in vitro stimulated T cells. Front. Immunol. 2014, 5, 556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Hong, P.; Yang, H.; Wu, Y.; Li, K.; Tang, Z. The functions and clinical application potential of exosomes derived from adipose mesenchymal stem cells: A comprehensive review. Stem Cell Res. Ther. 2019, 10, 1–12. [Google Scholar] [CrossRef]
  198. Gernapudi, R.; Yao, Y.; Zhang, Y.; Wolfson, B.; Roy, S.; Duru, N.; Eades, G.; Yang, P.; Zhou, Q. Targeting exosomes from preadipocytes inhibits preadipocyte to cancer stem cell signaling in early-stage breast cancer. Breast Cancer Res. Treat. 2015, 150, 685–695. [Google Scholar] [CrossRef] [Green Version]
  199. Lin, R.; Wang, S.; Zhao, R.C. Exosomes from human adipose-derived mesenchymal stem cells promote migration through Wnt signaling pathway in a breast cancer cell model. Mol. Cell. Biochem. 2013, 383, 13–20. [Google Scholar] [CrossRef]
  200. Zhan, T.; Rindtorff, N.; Boutros, M. Wnt signaling in cancer. Oncogene 2017, 36, 1461–1473. [Google Scholar] [CrossRef]
  201. Wang, Y.; Chu, Y.; Li, K.; Zhang, G.; Guo, Z.; Wu, X.; Qiu, C.; Li, Y.; Wan, X.; Sui, J.; et al. Exosomes Secreted by Adipose-Derived Mesenchymal Stem Cells Foster Metastasis and Osteosarcoma Proliferation by Increasing COLGALT2 Expression. Front. Cell Dev. Biol. 2020, 8, 353. [Google Scholar] [CrossRef] [PubMed]
  202. Otvos, L.; Haspinger, E.; La Russa, F.; Maspero, F.; Graziano, P.; Kovalszky, I.; Lovas, S.; Nama, K.; Hoffmann, R.; Knappe, D.; et al. Design and development of a peptide-based adiponectin receptor agonist for cancer treatment. BMC Biotechnol. 2011, 11, 90. [Google Scholar] [CrossRef] [Green Version]
  203. Gelsomino, L.; Naimo, G.D.; Catalano, S.; Mauro, L.; Andò, S. The emerging role of adiponectin in female malignancies. Int. J. Mol. Sci. 2019, 20, 2127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Silva, F.M.; De Almeida, J.C.; Feoli, A.M. Effect of diet on adiponectin levels in blood. Nutr. Rev. 2011, 69, 599–612. [Google Scholar] [CrossRef] [PubMed]
  205. Kriketos, A.D.; Gan, S.K.; Poynten, A.M.; Furler, S.M.; Chisholm, D.J.; Campbell, L.V. Exercise Increases Adiponectin Levels and Insulin Sensitivity in Humans. Diabetes Care 2004, 27, 629–630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Giordano, C.; Chemi, F.; Panza, S.; Barone, I.; Bonofiglio, D.; Lanzino, M.; Cordella, A.; Campana, A.; Hashim, A.; Rizza, P.; et al. Leptin as a mediator of tumor-stromal interactions promotes breast cancer stem cell activity. Oncotarget 2016, 7, 1262–1275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Otvos, L.; Kovalszky, I.; Riolfi, M.; Ferla, R.; Olah, J.; Sztodola, A.; Nama, K.; Molino, A.; Piubello, Q.; Wade, J.D.; et al. Efficacy of a leptin receptor antagonist peptide in a mouse model of triple-negative breast cancer. Eur. J. Cancer 2011, 47, 1578–1584. [Google Scholar] [CrossRef] [PubMed]
  208. Ferla, R.; Bonomi, M.; Otvos, L.; Surmacz, E. Glioblastoma-derived Leptin Induces Tube Formation and Growth of Endothelial Cells: Comparison with VEGF Effects. BMC Cancer 2011, 11, 303. [Google Scholar] [CrossRef] [Green Version]
  209. Otvos, L.; Surmacz, E. Targeting the leptin receptor: A potential new mode of treatment for breast cancer. Expert Rev. Anticancer Ther. 2011, 11, 1147–1150. [Google Scholar] [CrossRef]
  210. Johnson, D.E.; O’Keefe, R.A.; Grandis, J.R. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat. Rev. Clin. Oncol. 2018, 15, 234–248. [Google Scholar] [CrossRef]
  211. Guo, C.; Chen, Y.; Gao, W.; Chang, A.; Ye, Y.; Shen, W.; Luo, Y.; Yang, S.; Sun, P.; Xiang, R.; et al. Liposomal nanoparticles carrying anti-IL6R antibody to the tumour microenvironment inhibit metastasis in two molecular subtypes of breast cancer mouse models. Theranostics 2017, 7, 775–788. [Google Scholar] [CrossRef] [PubMed]
  212. Holen, I.; Lefley, D.V.; Francis, S.E.; Rennicks, S.; Bradbury, S.; Coleman, R.E.; Ottewell, P. IL-1 drives breast cancer growth and bone metastasis in vivo. Oncotarget 2016, 7, 75571–75584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Lust, J.A.; Lacy, M.Q.; Zeldenrust, S.R.; Dispenzieri, A.; Gertz, M.A.; Witzig, T.E.; Kumar, S.; Hayman, S.R.; Russell, S.J.; Buadi, F.K.; et al. Induction of a chronic disease state in patients with smoldering or indolent multiple myeloma by targeting interleukin 1β-induced interleukin 6 production and the myeloma proliferative component. Mayo Clin. Proc. 2009, 84, 114–122. [Google Scholar] [CrossRef] [PubMed]
  214. Ridker, P.M.; MacFadyen, J.G.; Thuren, T.; Everett, B.; Libby, P.; Glynn, R.J.; Ridker, P.; Lorenzatti, A.; Krum, H.; Varigos, J.; et al. Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: Exploratory results from a randomised, double-blind, placebo-controlled trial. Lancet 2017, 390, 1833–1842. [Google Scholar] [CrossRef]
  215. Baker, K.J.; Houston, A.; Brint, E. IL-1 family members in cancer; two sides to every story. Front. Immunol. 2019, 10, 1197. [Google Scholar] [CrossRef] [Green Version]
  216. Castaño, Z.; Juan, B.P.S.; Spiegel, A.; Pant, A.; DeCristo, M.J.; Laszewski, T.; Ubellacker, J.M.; Janssen, S.R.; Dongre, A.; Reinhardt, F.; et al. IL-1β inflammatory response driven by primary breast cancer prevents metastasis-initiating cell colonization. Nat. Cell Biol. 2018, 20, 1084–1097. [Google Scholar] [CrossRef]
  217. Lashinger, L.M.; Rossi, E.L.; Hursting, S.D. Obesity and resistance to cancer chemotherapy: Interacting roles of inflammation and metabolic dysregulation. Clin. Pharmacol. Ther. 2014, 96, 458–463. [Google Scholar] [CrossRef]
  218. Wang, Z.; Aguilar, E.G.; Luna, J.I.; Dunai, C.; Khuat, L.T.; Le, C.T.; Mirsoian, A.; Minnar, C.M.; Stoffel, K.M.; Sturgill, I.R.; et al. Paradoxical effects of obesity on T cell function during tumor progression and PD-1 checkpoint blockade. Nat. Med. 2019, 25, 141–151. [Google Scholar] [CrossRef]
  219. McQuade, J.L.; Daniel, C.R.; Hess, K.R.; Mak, C.; Wang, D.Y.; Rai, R.R.; Park, J.J.; Haydu, L.E.; Spencer, C.; Wongchenko, M.; et al. Association of body-mass index and outcomes in patients with metastatic melanoma treated with targeted therapy, immunotherapy, or chemotherapy: A retrospective, multicohort analysis. Lancet Oncol. 2018, 19, 310–322. [Google Scholar] [CrossRef] [Green Version]
  220. Vonderheide, R.H.; Domchek, S.M.; Clark, A.S. Immunotherapy for breast cancer: What are we missing? Clin. Cancer Res. 2017, 23, 2640–2646. [Google Scholar] [CrossRef] [Green Version]
  221. Dirix, L.Y.; Takacs, I.; Jerusalem, G.; Nikolinakos, P.; Arkenau, H.T.; Forero-Torres, A.; Boccia, R.; Lippman, M.E.; Somer, R.; Smakal, M.; et al. Avelumab, an anti-PD-L1 antibody, in patients with locally advanced or metastatic breast cancer: A phase 1b JAVELIN solid tumor study. Breast Cancer Res. Treat. 2018, 167, 671–686. [Google Scholar] [CrossRef] [Green Version]
  222. Naik, A.; Monjazeb, A.M.; Decock, J. The obesity paradox in cancer, tumor immunology, and immunotherapy: Potential therapeutic implications in triple negative breast cancer. Front. Immunol. 2019, 10, 1940. [Google Scholar] [CrossRef] [PubMed]
  223. Mirsoian, A.; Bouchlaka, M.N.; Sckisel, G.D.; Chen, M.; Pai, C.C.S.; Maverakis, E.; Spencer, R.G.; Fishbein, K.W.; Siddiqui, S.; Monjazeb, A.M.; et al. Adiposity induces lethal cytokine storm after systemic administration of stimulatory immunotherapy regimens in aged mice. J. Exp. Med. 2014, 211, 2373–2383. [Google Scholar] [CrossRef] [PubMed]
  224. Furuhashi, M.; Hotamisligil, G.S. Fatty acid-binding proteins: Role in metabolic diseases and potential as drug targets. Nat. Rev. Drug Discov. 2008, 7, 489–503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Carbonetti, G.; Wilpshaar, T.; Kroonen, J.; Studholme, K.; Converso, C.; d’Oelsnitz, S.; Kaczocha, M. FABP5 coordinates lipid signaling that promotes prostate cancer metastasis. Sci. Rep. 2019, 9. [Google Scholar] [CrossRef] [Green Version]
  226. O’Sullivan, S.E.; Kaczocha, M. FABP5 as a novel molecular target in prostate cancer. Drug Discov. Today 2020. [Google Scholar] [CrossRef]
  227. Field, C.S.; Baixauli, F.; Kyle, R.L.; Pearce, E.J.; Blazar, B.R.; Pearce, E.L. Mitochondrial Integrity Regulated by Lipid Metabolism Is a Cell-Intrinsic Checkpoint for Treg Suppressive Function. Cell Metab. 2020, 31, 422–437. [Google Scholar] [CrossRef] [Green Version]
  228. Verma, A.; Mathur, R.; Farooque, A.; Kaul, V.; Gupta, S.; Dwarakanath, B.S. T-regulatory cells in tumor progression and therapy. Cancer Manag. Res. 2019, 11, 10731–10747. [Google Scholar] [CrossRef] [Green Version]
  229. Wang, J.; Li, Y. CD36 tango in cancer: Signaling pathways and functions. Theranostics 2019, 9, 4893–4908. [Google Scholar] [CrossRef]
  230. Pascual, G.; Avgustinova, A.; Mejetta, S.; Martín, M.; Castellanos, A.; Attolini, C.S.O.; Berenguer, A.; Prats, N.; Toll, A.; Hueto, J.A.; et al. Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature 2017, 541, 41–45. [Google Scholar] [CrossRef]
  231. DeFilippis, R.A.; Chang, H.; Dumont, N.; Rabban, J.T.; Chen, Y.Y.; Fontenay, G.V.; Berman, H.K.; Gauthier, M.L.; Zhao, J.; Hu, D.; et al. CD36 repression activates a multicellular stromal program shared by high mammographic density and tumor tissues. Cancer Discov. 2012, 2, 826–839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Enciu, A.M.; Radu, E.; Popescu, I.D.; Hinescu, M.E.; Ceafalan, L.C. Targeting CD36 as Biomarker for Metastasis Prognostic: How Far from Translation into Clinical Practice? BioMed Res. Int. 2018, 2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Ehsanipour, E.A.; Sheng, X.; Behan, J.W.; Wang, X.; Butturini, A.; Avramis, V.I.; Mittelman, S.D. Adipocytes cause leukemia cell resistance to l-asparaginase via release of glutamine. Cancer Res. 2013, 73, 2998–3006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Nikonorova, I.A.; Al-Baghdadi, R.J.T.; Mirek, E.T.; Wang, Y.; Goudie, M.P.; Wetstein, B.B.; Dixon, J.L.; Hine, C.; Mitchell, J.R.; Adams, C.M.; et al. Obesity challenges the hepatoprotective function of the integrated stress response to asparaginase exposure in mice. J. Biol. Chem. 2017, 292, 6786–6798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Zhang, J.; Deng, Y.; Khoo, B.L. Fasting to enhance Cancer treatment in models: The next steps. J. Biomed. Sci. 2020, 27, 1–14. [Google Scholar] [CrossRef] [PubMed]
  236. Quail, D.F.; Olson, O.C.; Bhardwaj, P.; Walsh, L.A.; Akkari, L.; Quick, M.L.; Chen, I.C.; Wendel, N.; Ben-Chetrit, N.; Walker, J.; et al. Obesity alters the lung myeloid cell landscape to enhance breast cancer metastasis through IL5 and GM-CSF. Nat. Cell Biol. 2017, 19, 974–987. [Google Scholar] [CrossRef]
  237. Nagase, H.; Ueki, S.; Fujieda, S. The roles of IL-5 and anti-IL-5 treatment in eosinophilic diseases: Asthma, eosinophilic granulomatosis with polyangiitis, and eosinophilic chronic rhinosinusitis. Allergol. Int. 2020, 69, 178–186. [Google Scholar] [CrossRef]
  238. Sirin, O.; Kolonin, M.G. Treatment of obesity as a potential complementary approach to cancer therapy. Drug Discov. Today 2013, 18, 567–573. [Google Scholar] [CrossRef]
  239. Lv, M.; Zhu, X.; Wang, H.; Wang, F.; Guan, W. Roles of Caloric Restriction, Ketogenic Diet and Intermittent Fasting during Initiation, Progression and Metastasis of Cancer in Animal Models: A Systematic Review and Meta-Analysis. PLoS ONE 2014, 9, e115147. [Google Scholar] [CrossRef]
  240. O’Flanagan, C.H.; Smith, L.A.; McDonell, S.B.; Hursting, S.D. When less may be more: Calorie restriction and response to cancer therapy. BMC Med. 2017, 15. [Google Scholar] [CrossRef]
  241. De Groot, S.; Pijl, H.; Van Der Hoeven, J.J.M.; Kroep, J.R. Effects of short-term fasting on cancer treatment. J. Exp. Clin. Cancer Res. 2019, 38, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Bonsang-Kitzis, H.; Chaltier, L.; Belin, L.; Savignoni, A.; Rouzier, R.; Sablin, M.-P.; Lerebours, F.; Bidard, F.-C.; Cottu, P.; Sastre-Garau, X.; et al. Beyond Axillary Lymph Node Metastasis, BMI and Menopausal Status Are Prognostic Determinants for Triple-Negative Breast Cancer Treated by Neoadjuvant Chemotherapy. PLoS ONE 2015, 10, e0144359. [Google Scholar] [CrossRef] [PubMed]
  243. Litton, J.K.; Gonzalez-Angulo, A.M.; Warneke, C.L.; Buzdar, A.U.; Kau, S.W.; Bondy, M.; Mahabir, S.; Hortobagyi, G.N.; Brewster, A.M. Relationship between obesity and pathologic response to neoadjuvant chemotherapy among women with operable breast cancer. J. Clin. Oncol. 2008, 26, 4072–4077. [Google Scholar] [CrossRef]
  244. De Azambuja, E.; McCaskill-Stevens, W.; Francis, P.; Quinaux, E.; Crown, J.P.A.; Vicente, M.; Giuliani, R.; Nordenskjöld, B.; Gutiérez, J.; Andersson, M.; et al. The effect of body mass index on overall and disease-free survival in node-positive breast cancer patients treated with docetaxel and doxorubicin-containing adjuvant chemotherapy: The experience of the BIG 02-98 trial. Breast Cancer Res. Treat. 2010, 119, 145–153. [Google Scholar] [CrossRef] [Green Version]
  245. Kusuoka, O.; Fujiwara-Tani, R.; Nakashima, C.; Fujii, K.; Ohmori, H.; Mori, T.; Kishi, S.; Miyagawa, Y.; Goto, K.; Kawahara, I.; et al. Intermittent calorie restriction enhances epithelial-mesenchymal transition through the alteration of energy metabolism in a mouse tumor model. Int. J. Oncol. 2018, 52, 413–423. [Google Scholar] [CrossRef] [PubMed]
  246. Schmid, D.; Leitzmann, M.F. Association between physical activity and mortality among breast cancer and colorectal cancer survivors: A systematic review and meta-analysis. Ann. Oncol. 2014, 25, 1293–1311. [Google Scholar] [CrossRef]
  247. De ten Ryen, S.D.; Deldicque, L. The regulation of the metastatic cascade by physical activity: A narrative review. Cancers 2020, 12, 153. [Google Scholar] [CrossRef] [Green Version]
  248. Buss, L.A.; Dachs, G.U. Effects of Exercise on the Tumour Microenvironment. In Advances in Experimental Medicine and Biology; Springer: Berlin/Heidelberg, Germany, 2020; Volume 1225, pp. 31–51. [Google Scholar]
  249. Faustino-Rocha, A.I.; Silva, A.; Gabriel, J.; da Costa, R.M.G.; Moutinho, M.; Oliveira, P.A.; Gama, A.; Ferreira, R.; Ginja, M. Long-term exercise training as a modulator of mammary cancer vascularization. Biomed. Pharmacother. 2016, 81, 273–280. [Google Scholar] [CrossRef] [Green Version]
  250. Jones, L.W.; Fels, D.R.; West, M.; Allen, J.D.; Broadwater, G.; Barry, W.T.; Wilke, L.G.; Masko, E.; Douglas, P.S.; Dash, R.C.; et al. Modulation of circulating angiogenic factors and tumor biology by aerobic training in breast cancer patients receiving neoadjuvant chemotherapy. Cancer Prev. Res. 2013, 6, 925–937. [Google Scholar] [CrossRef] [Green Version]
  251. Dieli-Conwright, C.M.; Courneya, K.S.; Demark-Wahnefried, W.; Sami, N.; Lee, K.; Buchanan, T.A.; Spicer, D.V.; Tripathy, D.; Bernstein, L.; Mortimer, J.E. Effects of aerobic and resistance exercise on metabolic syndrome, sarcopenic obesity, and circulating biomarkers in overweight or obese survivors of breast cancer: A randomized controlled trial. J. Clin. Oncol. 2018, 36, 875–883. [Google Scholar] [CrossRef]
  252. Sjöström, L.; Gummesson, A.; Sjöström, C.D.; Narbro, K.; Peltonen, M.; Wedel, H.; Bengtsson, C.; Bouchard, C.; Carlsson, B.; Dahlgren, S.; et al. Effects of bariatric surgery on cancer incidence in obese patients in Sweden (Swedish Obese Subjects Study): A prospective, controlled intervention trial. Lancet Oncol. 2009, 10, 653–662. [Google Scholar] [CrossRef]
  253. Schauer, D.P.; Feigelson, H.S.; Koebnick, C.; Caan, B.; Weinmann, S.; Leonard, A.C.; Powers, J.D.; Yenumula, P.R.; Arterburn, D.E. Bariatric Surgery and the Risk of Cancer in a Large Multisite Cohort. Ann. Surg. 2019, 269, 95–101. [Google Scholar] [CrossRef] [PubMed]
  254. Castagneto-Gissey, L.; Casella-Mariolo, J.; Casella, G.; Mingrone, G. Obesity Surgery and Cancer: What Are the Unanswered Questions? Front. Endocrinol. 2020, 11, 213. [Google Scholar] [CrossRef] [PubMed]
  255. Bruno, D.S.; Berger, N.A. Impact of bariatric surgery on cancer risk reduction. Ann. Transl. Med. 2020, 8, S13. [Google Scholar] [CrossRef]
  256. Sainsbury, A.; Goodlad, R.A.; Perry, S.L.; Pollard, S.G.; Robins, G.G.; Hull, M.A. Increased colorectal epithelial cell proliferation and crypt fission associated with obesity and Roux-en-Y gastric bypass. Cancer Epidemiol. Biomark. Prev. 2008, 17, 1401–1410. [Google Scholar] [CrossRef] [Green Version]
  257. Philip, E.J.; Torghabeh, M.H.; Strain, G.W. Bariatric surgery in cancer survivorship: Does a history of cancer affect weight loss outcomes? Surg. Obes. Relat. Dis. 2015, 11, 1105–1108. [Google Scholar] [CrossRef] [Green Version]
  258. MacKintosh, M.L.; Derbyshire, A.E.; McVey, R.J.; Bolton, J.; Nickkho-Amiry, M.; Higgins, C.L.; Kamieniorz, M.; Pemberton, P.W.; Kirmani, B.H.; Ahmed, B.; et al. The impact of obesity and bariatric surgery on circulating and tissue biomarkers of endometrial cancer risk. Int. J. Cancer 2019, 144, 641–650. [Google Scholar] [CrossRef]
Figure 1. Schematic diagram depicting the keys steps of the metastatic cascade from initial presentation as an in situ tumor mass at the primary site to macroscopically detected metastatic lesions at secondary sites.
Figure 1. Schematic diagram depicting the keys steps of the metastatic cascade from initial presentation as an in situ tumor mass at the primary site to macroscopically detected metastatic lesions at secondary sites.
Cancers 12 03798 g001
Figure 2. Summary of cross talk between adipose tissue and cancer cells that promote metastasis. Hyperinsulinemia in obesity induces insulin growth factor (IGF)-1 signaling, which promotes epithelial to mesenchymal transition (EMT) and cancer stem cells (CSCs) though ZEB and SNAIL transcription factors leading to an increase in pluripotency transcription factors (OCT4, SOX2 and NANOG) and the developmental signaling pathways (Wnt and NOTCH). Interleukin 1β (IL-1β) promotes promote tumor cell homing to the bone marrow and angiogenesis through vascular endothelial growth factor (VEGF) signaling. Interleukin 6 (IL-6) and leptin induces epithelial to mesenchymal transition (EMT) and CSCsthough JAK/STAT3 signaling. Tumor necrosis factor α (TNFα) induces trans endothelial migration and CSCs though nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling. Fatty acid binding proteins (FABP) modulates fatty acid metabolism from neighboring adipocytes to provide fuel for tumor cells to metastasis. Matrix metalloproteinase 11 (MMP-11) dedifferentiates mature adipocytes into cancer associated adipocytes (CAAs) leading to extra cellular matrix (ECM) remodeling and fibrosis. Asparagine promotes EMT in primary tumors and increases tumor cell proliferation at distinct metastatic sites.
Figure 2. Summary of cross talk between adipose tissue and cancer cells that promote metastasis. Hyperinsulinemia in obesity induces insulin growth factor (IGF)-1 signaling, which promotes epithelial to mesenchymal transition (EMT) and cancer stem cells (CSCs) though ZEB and SNAIL transcription factors leading to an increase in pluripotency transcription factors (OCT4, SOX2 and NANOG) and the developmental signaling pathways (Wnt and NOTCH). Interleukin 1β (IL-1β) promotes promote tumor cell homing to the bone marrow and angiogenesis through vascular endothelial growth factor (VEGF) signaling. Interleukin 6 (IL-6) and leptin induces epithelial to mesenchymal transition (EMT) and CSCsthough JAK/STAT3 signaling. Tumor necrosis factor α (TNFα) induces trans endothelial migration and CSCs though nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling. Fatty acid binding proteins (FABP) modulates fatty acid metabolism from neighboring adipocytes to provide fuel for tumor cells to metastasis. Matrix metalloproteinase 11 (MMP-11) dedifferentiates mature adipocytes into cancer associated adipocytes (CAAs) leading to extra cellular matrix (ECM) remodeling and fibrosis. Asparagine promotes EMT in primary tumors and increases tumor cell proliferation at distinct metastatic sites.
Cancers 12 03798 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Annett, S.; Moore, G.; Robson, T. Obesity and Cancer Metastasis: Molecular and Translational Perspectives. Cancers 2020, 12, 3798. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12123798

AMA Style

Annett S, Moore G, Robson T. Obesity and Cancer Metastasis: Molecular and Translational Perspectives. Cancers. 2020; 12(12):3798. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12123798

Chicago/Turabian Style

Annett, Stephanie, Gillian Moore, and Tracy Robson. 2020. "Obesity and Cancer Metastasis: Molecular and Translational Perspectives" Cancers 12, no. 12: 3798. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12123798

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop