Hostname: page-component-8448b6f56d-wq2xx Total loading time: 0 Render date: 2024-04-23T22:10:31.581Z Has data issue: false hasContentIssue false

Obesity-associated cancer: an immunological perspective

Published online by Cambridge University Press:  16 November 2015

Melissa J. Conroy
Affiliation:
Dept. Surgery, Trinity Centre for Health Sciences, St. James's Hospital and Trinity College Dublin, Dublin 8, Ireland
Margaret R. Dunne
Affiliation:
Dept. Surgery, Trinity Centre for Health Sciences, St. James's Hospital and Trinity College Dublin, Dublin 8, Ireland
Claire L. Donohoe
Affiliation:
Dept. Surgery, Trinity Centre for Health Sciences, St. James's Hospital and Trinity College Dublin, Dublin 8, Ireland
John V. Reynolds*
Affiliation:
Dept. Surgery, Trinity Centre for Health Sciences, St. James's Hospital and Trinity College Dublin, Dublin 8, Ireland
*
*Corresponding author: Professor John V. Reynolds, email reynoldsjv@STJAMES.IE
Rights & Permissions [Opens in a new window]

Abstract

Epidemiological studies have established an association between obesity, insulin resistance, type 2 diabetes and a number of cancer types. Research has focused predominantly on altered endocrine factors, growth factors and signalling pathways, with little known in man about the immune involvement in the relevant pathophysiological processes. Moreover, in an era of exciting new breakthroughs in cancer immunotherapy, there is also a need to study the safety and efficacy of immunotherapeutics in the complex setting of inflammatory-driven obesity-associated cancer. This review addresses key immune cell subsets underpinning obesity-associated inflammation and describes how such immune compartments might be targeted to prevent and treat obesity-associated cancer. We propose that the modulation, metabolism, migration and abundance of pro- and anti-inflammatory cells and tumour-specific T cells might be therapeutically altered to both restore immune balance, alleviating pathological inflammation, and to improve anti-tumour immune responses in obesity-associated cancer.

Type
Conference on ‘Nutrition at key life stages: new findings, new approaches’
Copyright
Copyright © The Authors 2015 

The burgeoning global health burden of obesity is of grave concern, affecting over half a billion adults worldwide, with approximately 3·5 million attributable deaths each year( 1 ). Despite national and international interventions to promote a healthy diet and lifestyle, recent reports predict that global obesity rates show no signs of abating, with predictions that half of all adults will be overweight or obese by 2030 (WHO). The worldwide prevalence of obesity almost doubled in the period 1980–2008. In 1980, 5 % of men and 8 % of women were obese and by 2008, these rates were 10 and 14 %, respectively. In many areas of the western world, the overweight phenotype is now the most prevalent body type. For example, in the USA in 2010 the prevalence of a BMI ≥ 25 was 69·2 %, with 35·9 % of people being obese (BMI ≥ 30)( Reference Flegal, Carroll, Ogden and Curtin 2 ). A cause of current concern is that more than one-third of children are overweight or obese in the USA, with adolescent obesity rates quadrupling over the past 30 years( Reference Ogden, Carroll, Kit and Flegal 3 ). WHO figures also show that one in three European children are overweight or obese and of these, 60 % are predicted to be obese in adulthood( 1 ).

Obesity may fuel pathological chronic inflammation and therefore a substantial proportion of adults and children worldwide are at risk of developing obesity-associated morbidities such as cardiovascular disease (CVD), type 2 diabetes mellitus (T2DM), non-alcoholic steatohepatitis and cancer( 1 , Reference Calle, Rodriguez, Walker-Thurmond and Thun 4 Reference Calle, Rodriguez, Walker-Thurmond and Thun 7 ). Obesity contributes to between 3 and 20 % of cancer deaths in western populations( Reference Renehan, Tyson, Egger, Heller and Zwahlen 8 , Reference Beddy, Howard, McMahon, Knox, De Blacam and Ravi 9 ). Since obesity is a pro-inflammatory state, an altered immune system may fuel this process, with visceral adipose tissue and liver being primary sources of cells and cytokines. Understanding the role of obesity and related metabolic syndrome, insulin resistance, and T2DM in carcinogenesis and tumour biology is consequently the focus of significant present research interest, with most lessons learned from experimental models but with an increasing focus on studies in human subjects. In an era of novel immunotherapeutics in cancer, when targeted therapies such as nivolumab and ipilimumab have shown promise for several tumour types, a greater understanding in man of the impact of such approaches on obesity-associated cancers is required( Reference Postow, Callahan and Wolchok 10 ). Such cancers represent a unique immunotherapeutic challenge because an enhanced T helper type 1 (Th1) immune response is required to augment anti-tumour immunity, but without exacerbating tumourigenic obesity-associated inflammation( Reference Postow, Callahan and Wolchok 10 ). Herein, we address the pathophysiological processes driving obesity-associated inflammation with a focus on both the innate and adaptive arms of the immune system, and discuss future prospects for treatment of obesity-associated malignancy.

Epidemiology

The epidemiological evidence underpinning the association between obesity and cancer incidence is now compelling. Large population-based prospective studies have demonstrated convincingly consistent increased cancer incidences per 5 kg/m2 increase in BMI( Reference Calle, Rodriguez, Walker-Thurmond and Thun 4 , Reference Renehan, Tyson, Egger, Heller and Zwahlen 8 , 11 Reference Reeves, Pirie, Beral, Green, Spencer and Bull 13 ). For example, a recent UK population based prospective cohort of 5·2 million patients with a median follow-up of 6 years recorded 166 966 incident cancers across twenty-two of the most common cancers( Reference Bhaskaran, Douglas, Forbes, Dos-Santos-Silva, Leon and Smeeth 12 ). BMI was positively associated with cancers at the following sites: colon, rectum, liver, gallbladder, pancreas, breast (post-menopausal only), cervix, ovary, uterus, kidney, thyroid and leukaemia( Reference Bhaskaran, Douglas, Forbes, Dos-Santos-Silva, Leon and Smeeth 12 ). The derived hazard ratios from this study are displayed in Table 1. The administrative dataset used in this study was unable to subtype cancers, for example both squamous and adenocarcinomas of the oesophagus were pooled as oesophageal cancer. This probably accounts for the lack of association between oesophageal adenocarcinoma and obesity, which has been consistent and marked in other studies (approximate hazard ratios 4·76, 95 % CI 2·96, 7·66; for patients with BMI ≥ 40 compared with normal BMI)( Reference Renehan, Tyson, Egger, Heller and Zwahlen 8 , Reference Calle and Kaaks 14 ). A meta-analysis prospective study of 282 000 cancer cases from population of 4·8 million patients demonstrated similarly consistent findings across disease sites and sexes( Reference Renehan, Tyson, Egger, Heller and Zwahlen 8 ). Applying the Bradford-Hill hypotheses of strength, consistency, specificity, temporality, biological gradient, plausibility, coherence, experimental evidence and analogy, obesity is a strong candidate to be a causal factor in the development of cancer as detailed in a review by Renehan et al. ( Reference Hill 15 , Reference Renehan, Egger and Zwahlen 16 ).

Table 1. Hazard ratio (HR) for cancers per 5 kg/m2 increase in BMI( Reference Bhaskaran, Douglas, Forbes, Dos-Santos-Silva, Leon and Smeeth 12 )

Further epidemiological studies may lead to refinement of the associated increases in relative risk of incident cancers, especially cancers at sites with a less frequent incidence. Current estimates are subject to wide CI at disease sites where few incident cancers are reported. Heterogeneity in already reported relative risks at different cancer sites indicates that there is probably not a universal mechanism by which obesity drives cancer development. For example, the attributable fraction of endometrial cancers is approximately 41 % v. 10 % for cancers of the colon( Reference Bhaskaran, Douglas, Forbes, Dos-Santos-Silva, Leon and Smeeth 12 ). The main caveat concerning the association between obesity and cancer is the fact that a number of other environmental factors including dietary composition, energy intake and physical activity represent important confounding factors in obese patients, and no specifically designed prospective studies have yet to demarcate the relative contribution of each.

Pathological inflammation in obesity is associated with insulin resistance and altered cellular energetics

Obesity-associated inflammation and cancer are inextricably linked and despite robust epidemiological evidence connecting obesity and cancer, the pathophysiological mechanisms underpinning the association remain poorly characterised( Reference Donohoe, Doyle and Reynolds 17 , Reference Donohoe, O'Farrell, Doyle and Reynolds 18 ). The expanded adipose tissue mass associated with an obese phenotype is deposited in both the intra-abdominal compartment, where it is known as central or visceral adiposity, and beneath the skin, where it is called subcutaneous adipose tissue. Visceral adipose tissue (VAT) is more highly correlated with an altered glucose and lipid metabolic profile as well as increased risk of CVD than subcutaneous adipose tissue( Reference Fujioka, Matsuzawa, Tokunaga and Tarui 19 Reference Nakamura, Tokunaga, Shimomura, Nishida, Yoshida and Kotani 21 ). Obese patients often have a relative abundance of fat in one or other compartment( Reference Vague 22 ). Those with excess visceral fat have an increased risk (independent of BMI) of breast cancer, oesophageal adenocarcinoma, colorectal adenocarcinoma and colorectal adenoma( Reference Beddy, Howard, McMahon, Knox, De Blacam and Ravi 9 , Reference Schapira, Clark, Wolff, Jarrett, Kumar and Aziz 23 Reference Nam, Kim, Han, Ryu, Park and Kim 26 ).

Adipose tissue in obese subjects has altered endocrine function and a secretory profile with predominantly pro-inflammatory cytokines, which are secreted by both adipocytes (and called adipokines) and immune cells. This results in a state of chronic low-grade inflammation which is thought to be pro-tumourigenic( Reference Esposito and Giugliano 27 Reference Das 29 ). Inflammation provides the selective pressure that may fuel the accumulation of mutations driving tumour growth and survival and could result in poor immune surveillance of tumours once they develop( Reference Coussens and Werb 30 ). The relative contribution of each individual adipokine to this systemic inflammation is not understood and it is likely that a complex interplay between the various pro- and anti-inflammatory immune cells and their secreted cytokines contributes to tumourigenesis.

Another consequence of systemic inflammation in the obese state is insulin resistance and T2DM( Reference Xu, Barnes, Yang, Tan, Yang and Chou 31 ). These conditions are associated with increased levels of inflammatory mediators including acute phase reactants such as fibrinogen, C-reactive protein, IL-6, plasminogen activator inhibitor-1( Reference Hotamisligil, Arner, Caro, Atkinson and Spiegelman 32 Reference Antuna-Puente, Feve, Fellahi and Bastard 34 ). The prevalence of insulin resistance increases as body weight increases and can be reversed with weight loss( Reference DeFronzo 35 , Reference Buchwald, Avidor, Braunwald, Jensen, Pories and Fahrbach 36 ). Epidemiological studies on the role of T2DM in carcinogenesis report consistent increases in cancer incidence at certain sites, which display considerable overlap with those sites associated with obesity-associated cancers as previously described, including liver in particular, as well as pancreas, endometrium, breast, colorectal, bladder, non-Hodgkin's lymphoma and kidney cancers( Reference Johnson, Carstensen, Witte, Bowker, Lipscombe and Renehan 37 ).

Diet-induced insulin resistance develops as a metabolic adaptation to increased circulating levels of non-esterified fatty acids (NEFA), which are constantly released from adipose tissue, especially from visceral fat stores( Reference Calle and Kaaks 14 ). Increased NEFA levels force liver, muscle and other tissues to shift towards increased storage and oxidation of fats for their energy production( Reference Bergman and Ader 38 ). The compensatory effect is a reduced capacity of these tissues to absorb, store and metabolise glucose. Patients with insulin resistance also exhibit a reduction in cellular insulin-receptor levels and reduced responsiveness of some intracellular transduction pathways mediating the effects of insulin binding to its receptor( Reference Moller and Flier 39 ). Interestingly, insulin resistance leads to increased insulin production and the mitogenic effects of insulin have been associated with several cancers( Reference Schoen, Tangen, Kuller, Burke, Cushman and Tracy 24 , Reference Trevisan, Liu, Muti, Misciagna, Menotti and Fucci 40 , Reference Colangelo, Gapstur, Gann, Dyer and Liu 41 ). The insulin-like growth factor (IGF) axis comprising two growth factors (IGF-1 and IGF-2) as well as a number of binding proteins and two receptors (IGF-1R and IGF-2R), sharing homology with the insulin receptor, has been cited as a mediator of hyperinsulinaemia on cell growth and energy metabolism( Reference Pollak 42 ). Increased expression of IGF-1R is reported in a number of cancer subtypes but a large systematic review and meta-analysis of studies of serum concentrations of IGF-1 and insulin as biomarkers reveals there to be significant bias found in the studies which have been published in the literature( Reference Hellawell, Turner, Davies, Poulsom, Brewster and Macaulay 43 Reference Tsilidis, Travis, Appleby, Allen, Lindstrom and Schumacher 46 ). New prospective specifically-designed epidemiological studies are warranted to help clarify the relative contribution of insulin resistance, inflammation and obesity to cancer development.

Changes in cellular energetics and dysregulated metabolism are hallmarks of cancer and it is not surprising that associations between metabolic changes and obesity are being uncovered( Reference Hanahan and Weinberg 47 ). Recent findings of our group show that secreted factors from VAT of obesity-associated cancer patients induce metabolic changes in oesophageal adenocarcinoma cell lines; increasing mitochondrial mass, mitochondrial membrane potential and inducing a shift to glycolysis( Reference Lynam-Lennon, Maher, Maguire, Phelan, Muldoon and Reynolds 48 ). Moreover, these studies identified a positive correlation between obesity and the expression of the glycolytic marker pyruvate kinase M2 in the tumours of oesophageal adenocarcinoma patients supporting a shift to glycolysis in obesity. Further analysis revealed that the VAT of obese oesophageal adenocarcinoma patients exhibited a glycolytic profile with increased expression of lactate, an established carcinogen( Reference Lynam-Lennon, Maher, Maguire, Phelan, Muldoon and Reynolds 48 , Reference Groff, Stugard, Mays, Koopmans and DiGirolamo 49 ). Therefore, these data present a possible link between obesity and the dysregulated bioenergetics that are characteristic of cancer, thus providing a mechanism through which increased VAT mass might promote tumourigenesis.

The sex differential in cancer incidence in obese patients has also been repeatedly reproduced in epidemiological studies( Reference Renehan, Tyson, Egger, Heller and Zwahlen 8 , Reference Bhaskaran, Douglas, Forbes, Dos-Santos-Silva, Leon and Smeeth 12 ). This, as well as the marked increased incidence in endometrial cancers in obese females in particular, has led to the hypothesis that sex hormones play a role in the carcinogenic process in obesity, at least for some sites. There is a well-recognised association between the development of endometrial cancer and oestrogen excess( Reference Lukanova, Lundin, Zeleniuch-Jacquotte, Muti, Mure and Rinaldi 50 , Reference Zeleniuch-Jacquotte, Akhmedkhanov, Kato, Koenig, Shore and Kim 51 ). A factor analysis of inflammatory, insulin-regulated physiological axes and sex steroids measured from the pre-diagnostic serum levels of patients enrolled in the European Prospective Investigation into Cancer and Nutrition cohort compared patients who developed endometrial cancer v. matched controls( Reference Dossus, Lukanova, Rinaldi, Allen, Cust and Becker 52 ). This allowed an analysis of these highly correlated biomarkers for their relative contribution to endometrial cancer development and demonstrated a contribution to cancer risk in all three of these axes.

In light of the metabolic changes that occur with increasing visceral adipose tissue mass, it stands to reason that obesity may be a confounding factor when developing immune-based anti-cancer therapies for obesity-associated malignancies. However, an improved understanding of the interplay between the immunity–obesity–cancer axis may allow novel insights into cancer development, and can allow improvement of current strategies for personalised therapy.

Adipokines and immune function

In addition to their direct roles in cellular energetics and metabolism( Reference Cao 53 ), emerging evidence demonstrates roles for adipokines in immune system regulation. For example, leptin, produced by adipose tissue, is a long-term regulator of weight and acts via its receptor to decrease appetite and food intake and increase energy consumption( Reference Maffei, Halaas, Ravussin, Pratley, Lee and Zhang 54 ). Leptin is also a pro-inflammatory cytokine inducing activation of T cells and Th1 cell differentiation( Reference Lord, Matarese, Howard, Baker, Bloom and Lechler 55 ). It also modulates the function of all other immune cell lineages (but only in the presence of other non-specific immunostimulants)( Reference Matarese, Moschos and Mantzoros 56 ). Leptin is induced by inflammatory cytokines such as IL-1, IL-6 and lipopolysaccharide( Reference Faggioni, Feingold and Grunfeld 57 ). It could be hypothesised that the relative lack of some subpopulations of T cells (CD3+, CD4+CD45RO+, CD8+)( Reference Tanaka, Isoda, Ishihara, Kimura and Yamakawa 58 ) could be related to the leptin resistant state prevalent in obese subjects( Reference Considine, Sinha, Heiman, Kriauciunas, Stephens and Nyce 59 ). Adiponectin is an abundant adipokine secreted by adipocytes in VAT. Circulating levels are inversely associated with obesity( Reference Kadowaki and Yamauchi 60 ) and decreased expression of adiponectin is closely correlated with insulin resistance( Reference Weyer, Funahashi, Tanaka, Hotta, Matsuzawa and Pratley 61 , Reference Yamauchi, Kamon, Waki, Terauchi, Kubota and Hara 62 ). Adiponectin inhibits macrophage phagocytosis and reduces macrophage production of pro-inflammatory IL-6 and TNF-α and increases production of anti-inflammatory IL-10 and IL-1R( Reference Lago, Dieguez, Gómez-Reino and Gualillo 63 ).

Innate and adaptive immunity in obesity and cancer

Obesity induces a state of pathological chronic inflammation, marked by elevated levels of NEFA, abnormal cytokine production and activation of inflammatory signalling pathways( Reference Fain 64 Reference Wellen and Hotamisligil 68 ). The expanded vasculature and increased blood supply that accompanies the expansion of VAT in obesity, results in increased inflammatory immune cell infiltration, enhanced pro-inflammatory cytokine release and ultimately local and systemic tumourigenic inflammation( Reference Karin 69 , Reference Ramos-Nino 70 ). The alterations in immune cell repertoires in the peripheral blood, VAT and proximal tissues, in particular liver, continue to be investigated in an effort to elucidate the extent of immune dysregulation in obesity. Immunotherapeutic strategies to reset the balance between pro- and anti-inflammatory cells could also be crucial to restoring immune control, attenuating obesity-associated inflammation and treating/preventing resultant disease. However the immune dysregulation in obesity with underlying malignancy could significantly negatively impact on anti-tumour immunity. In fact, ideal immunotherapeutic approaches in obesity-associated cancer should attenuate chronic systemic inflammation while simultaneously augmenting specific immunity at the tumour site. Research is required to fully characterise the immune cell repertoires in the periphery, VAT and tumour of obesity-associated cancer patients to distinguish between the subsets that drive tumourigenic inflammation, those that control it and those that are crucial for tumour eradication. For the purpose of this review, we will focus on the present knowledge of conventional T lymphocytes, innate lymphocytes and macrophages in obesity and cancer (Table 2).

Table 2. Summary of immunological impact of obesity in human and potential therapeutic targets

iNKT, invariant natural killer T cells; MAIT, mucosal associated invariant T cells; NK, natural killer; VAT, visceral adipose tissue.

Conventional T cells

CD4+ and CD8+ T cells comprise the antigen-specific effector arm of the adaptive immune response and elicit their effector functions through cytokine production and cytotoxic activity. In the CD4+ T cell compartment, Th1 cells produce interferon (IFN)-γ, TNF-α and IL-2, while T helper type 2 (Th2) cells express IL-4, IL-5 and IL-13( Reference Ikeda, Chamoto, Tsuji, Suzuki, Wakita and Takeshima 91 , Reference Foster, Martinez-Moczygemba, Huston and Corry 92 ). Inflammatory T helper type 17 (Th17) cells express IL-17 and IL-22 and regulatory T (Treg) cells express IL-10 and transforming growth factor β ( Reference Ciric, El-behi, Cabrera, Zhang and Rostami 93 , Reference Cottrez and Groux 94 ). CD8+ T cells are the key cytotoxic T lymphocytes but are also potent cytokine producers. Therefore in the context of malignancy, tumour infiltration of the appropriate CD4+ and CD8+ T cell milieu is crucial for tumour eradication( Reference Kalams and Walker 95 ). In fact, the ratio of cytotoxic T lymphocytes to Treg cells can be predictive of outcome( Reference Kono, Kawaida, Takahashi, Sugai, Mimura and Miyagawa 96 , Reference Shen, Zhou, Wang, Li, Zhong and Liang 97 ). In obesity, our group and others have reported enrichments of activated and effector pro-inflammatory CD4+ and CD8+ T cells in VAT and have identified these cells as key players in inflammatory macrophage recruitment and inflammatory cytokine production, thus contributing to the initiation and maintenance of obesity-associated inflammation( Reference Lysaght, Allott, Donohoe, Howard, Pidgeon and Reynolds 65 , Reference Winer, Chan, Paltser, Truong, Tsui and Bahrami 73 , Reference Nishimura, Manabe, Nagasaki, Eto, Yamashita and Ohsugi 80 ). Furthermore, we have previously reported significantly higher frequencies of such IFN-γ-producing T cells in the VAT of obesity-associated cancer patients, compared with non-cancer control subjects( Reference Lysaght, Allott, Donohoe, Howard, Pidgeon and Reynolds 65 ). In recent unpublished studies, we have also observed enrichments of TNF-α and IL-17-producing T cells in the VAT, demonstrating that more than one inflammatory T cell subset is contributing to adipose tissue inflammation in obesity and obesity-associated cancer. In contrast, while Treg cells are enriched in lean VAT, their frequency is significantly diminished in obese VAT( Reference Feuerer, Herrero, Cipolletta, Naaz, Wong and Nayer 98 ). Therefore, VAT in obesity is rich in pro-inflammatory cytokine-producing effector T cells and a relative deficiency of Treg cells( Reference Lysaght, van der Stok, Allott, Casey, Donohoe and Howard 66 , Reference Nishimura, Manabe, Nagasaki, Eto, Yamashita and Ohsugi 80 , Reference Feuerer, Herrero, Cipolletta, Naaz, Wong and Nayer 98 ). Such approaches to reset the balance between regulatory and inflammatory T cell subsets may potentially attenuate pathological inflammation in obesity.

Natural killer cells

Natural killer (NK) cells are MHC-unrestricted lymphocytes and comprise a major component of innate lymphocytes in human blood and liver, comprising 10–15 and 30 % of total lymphocytes, respectively( Reference Robertson and Ritz 99 ). NK cells display powerful cytotoxic activities once activated, and are potent producers of pro-inflammatory cytokines including IFN-γ and TNF-α which can contribute to the modulation of adaptive immune responses and direct anti-tumour effects( Reference Robertson and Ritz 99 Reference Ing and Stevenson 101 ). In the context of inflammatory disease, NK cells have been implicated in the pathogenesis of inflammatory bowel disease and psoriasis( Reference Yadav, Chen and Liu 102 , Reference Gardiner and Dunphy 103 ). However as of yet, their role has not been fully elucidated in the chronic inflammation that is characteristic of obesity. Lynch et al. have reported significant reductions of circulating NK cells in metabolically unhealthy obese subjects, suggesting that a lack of NK cells in the periphery is detrimental for metabolic health( Reference Lynch, O'Connell, Kwasnik, Cawood, O'Farrelly and O'Shea 83 ). Lynch et al. also reported significant expansions of circulating inhibitory-receptor bearing and CD69+ NK cells in metabolically unhealthy obese individuals, compared with healthy controls, suggesting that peripheral NK cells in metabolically unhealthy obese patients were activated but unable to elicit effector functions, making such individuals more susceptible to disease( Reference Lynch, O'Connell, Kwasnik, Cawood, O'Farrelly and O'Shea 83 ). Furthermore, circulating NK cells from obese subjects were found to be less effective at killing tumour cells than their counterparts in lean subjects, suggesting that obese individuals have impaired defences against malignancy( Reference O'Shea, Cawood, O'Farrelly and Lynch 104 ). More recently, a study by O’ Rourke et al. revealed expansions of the CD56BRIGHT (main cytokine producing subset) of NK cells in obese VAT, indicating that cytokine-producing fractions of NK cells are more prevalent in obesity( Reference O'Rourke, Gaston, Meyer, White and Marks 105 ). Moreover, NK cells were identified as key players in macrophage recruitment to adipose tissue but did not affect overall secreted inflammatory cytokine levels( Reference O'Rourke 106 ). In fact, the macrophage population recruited to the VAT by NK cells in this study was not the macrophage subset implicated in VAT inflammation and therefore, the role of NK cells in adipose tissue inflammation was unclear. However, new data gathered by Wensveen et al. demonstrates that IFN-γ-producing NK cells are pivotal in M1 macrophage polarisation in the adipose tissue of obese mice and subsequent insulin resistance, but similar results have yet to be reported in human subjects( Reference Wensveen, Jelenčić, Valentić, Šestan, Wensveen and Theurich 107 ). Interestingly, this study was the first to report a direct interaction between NK cells and adipocytes through the mouse equivalent of NK receptor P46 (NKP46) natural cytotoxicity receptor 1 (NCR1), and shows that NK cells and adipocytes together drive inflammatory macrophage recruitment and contributes to adipose tissue inflammation in mice. With respect to therapeutic avenues, NKP46 might be a potential target to attenuate NK cell and adipocyte-mediated inflammation. Since mouse and human NK cells are characterised differently, it is first important to fully investigate the NK repertoires in human blood and VAT, elucidate their interactions with adipocytes, their cytokine profiles and subsequent contribution to inflammatory cell recruitment and adipose tissue inflammation. Furthermore, the investigation of the NK cell milieu in blood, VAT and tumour of patients with obesity-associated malignancy is warranted to completely understand the changes that NK cells undergo in human obesity and how this impacts anti-tumour immunity and patient outcomes.

Invariant natural killer T cells

Invariant NKT (iNKT) cells are a subset of MHC-unrestricted innate lymphocytes that respond rapidly, elicit potent IFN-γ production and cytotoxic activity and modulate adaptive immune responses, thus making them key players in anti-tumour immunity. They express a restricted T cell receptor repertoire consisting of a Vα14Jα18 α-chain in mice and a Vα24Jα18 α-chain in human subjects, paired with a limited number of β-chains( Reference Bendelac, Rivera, Park and Roark 108 ). Such cells also express a number of cell-surface markers typically expressed on NK cells, hence the name NKT cell, and recognise lipid antigen presented by the MHC-like glycoprotein, CD1d( Reference Bendelac, Rivera, Park and Roark 108 Reference Gumperz 111 ). The most potent activator of iNKT cells isolated to date is the marine sponge-derived glycolipid α-galactosylceramide (α-GalCer), which has been shown to induce anti-tumour cytotoxic activity and cytokine production in mice leading to the inhibition of tumour growth( Reference Kawano, Cui, Koezuka, Toura, Kaneko and Motoki 112 ). However, α-GalCer has proven much less effective in human subjects due to the lower abundance of iNKT cells in human subjects compared with mice( Reference Kenna, Golden-Mason, Norris, Hegarty, O'Farrelly and Doherty 113 ). VAT is the only human tissue reported to have a high proportion of iNKT cells, averaging 10 % in healthy subjects( Reference Lynch, O'Connell, Kwasnik, Cawood, O'Farrelly and O'Shea 83 ). However, in the setting of obesity the frequencies of iNKT cells are diminished and this depletion is reversed following weight loss( Reference Lynch, Nowak, Varghese, Clark, Hogan and Toxavidis 114 ). VAT iNKT cells preferentially secrete higher levels of IL-10, compared with the Th1 cytokine profile they exhibit in other tissues, suggesting that iNKT cells in adipose tissue predominantly serve an immune regulatory role( Reference Lynch, Nowak, Varghese, Clark, Hogan and Toxavidis 114 ). Furthermore, the adoptive transfer of iNKT cells into obese mice can induce weight loss and improve insulin sensitivity, implying that iNKT cells are pivotal for metabolic health and protective against insulin resistance. This may suggest that metabolically unhealthy individuals might benefit from iNKT cell-based therapies and that IL-10-producing VAT iNKT cells may prove useful in the attenuation of obesity-associated inflammation. However, iNKT cell therapeutic potential in obesity-associated cancer has yet to be uncovered.

Gamma/delta T cells

Human γδ T cells are another type of innate or unconventional T cell subset, typically comprising 1–5 % of circulating adult T cells, and are also found within the gut( Reference Dunne, Elliott, Hussey, Mahmud, Kelly and Doherty 115 ), skin( Reference Laggner, Di Meglio, Perera, Hundhausen, Lacy and Ali 116 ), lung( Reference Lahn 117 ) and uterus ( Reference Exley and Boyson 118 ). In human subjects there are three main γδ T cell types, defined by their T cell receptor delta chain (called Vδ1, Vδ2 or Vδ3, respectively), which can be paired with various gamma chains( Reference Hayday 119 ). γδ T cells respond rapidly to infection or stress signals and display anti-viral, anti-bacterial, anti-parasitic and anti-tumour functions, and a strong Th1 predominance( Reference Dunne, Mangan, Madrigal-Estebas and Doherty 120 ). The main subset of circulating human γδ T cells (Vγ9Vδ2 T cells) recognises pyrophosphate antigens derived from bacterial or endogenous sources. The precise antigens recognised and modes of antigen recognition for other γδ T cell subsets are presently unclear but it is known that MHC-mediated antigen presentation is not required( Reference Crowley, Reich, Mavaddat, Altman and Chien 121 , Reference Singhal, Mori and De Libero 122 ). In mouse models, γδ T cells produce copious amounts of IL-17( Reference Lockhart, Green and Flynn 123 ), and comprise the predominant T cell subset found in tissues( Reference Bergstresser, Sullivan, Streilein and Tigelaar 124 ), whereas this is not the case in human subjects. Furthermore, the predominant γδ T cell subset in human blood, the Vγ9Vδ2 T cell subset, is only found in human subjects and other higher primates. All three γδ T cell subsets in the human demonstrate potent and specific anti-tumour cytotoxicity against a range of different tumour types( Reference Hayday 119 , Reference Siegers, Dhamko, Wang, Mathieson, Kosaka and Felizardo 125 , Reference Mangan, Dunne, O'Reilly, Dunne, Exley and O'Shea 126 ) and as a result, their use as immunotherapeutic agents is presently under investigation( Reference Gomes, Martins and Silva-Santos 88 ). Meta-analysis of clinical trial data shows limited success with present immunotherapeutic approaches used to date however, most of which involve adoptive transfer of Vγ9Vδ2 T cells. It is hoped that an improved understanding of γδ T cell biology and subset differences will improve future immunotherapeutic strategies.

In obese individuals, there is an inverse correlation between the frequency of circulating γδ T cells and increasing BMI( Reference Costanzo, Taylor, Dutt, Han, Fujioka and Jameson 87 ), with obese individuals having, on average, more than four times fewer circulating Vγ9Vδ2 T cells than non-obese donors. Remaining γδ T cells have a skewed maturation phenotype similar to that observed in aged populations, a blunted IFN-γ response and reduced IL-2 receptor α chain expression. Constanzo and colleagues reported that this dysregulation could be overcome by stimulation of cells with pyrophosphate antigens and addition of IL-2( Reference Costanzo, Taylor, Dutt, Han, Fujioka and Jameson 87 ). IL-2 acts as a growth factor for γδ T cells, suggesting that cellular dysfunction in obesity may be via growth factor deprivation rather than cell incapacitation. In mouse models of obesity, γδ T cells are shown to promote insulin resistance and inflammation via recruitment of macrophages to adipose tissue( Reference Mehta, Nuotio-Antar and Smith 127 ), though whether this also applies in human subjects remains to be investigated. The role of γδ T cells in the underlying inflammation and anti-tumour responses in the setting of obesity-associated cancer has yet to be elucidated. Our group are currently investigating the distribution and functional phenotype of γδ T cell subsets in the periphery, VAT and tumour of obesity-associated cancer patients in order to identify their usefulness in the prophylaxis and treatment of such malignancies.

Mucosal associated invariant T cells

Mucosal associated invariant T (MAIT) cells are a population of invariant T cells comprising 1–5 % of T cells in human blood, and are defined by expression of an invariant Vα7·2-Jα33 chain and high levels of the NK cell marker CD161( Reference Treiner, Duban, Bahram, Radosavljevic, Wanner and Tilloy 128 ). MAIT cells are enriched in human adipose tissue and in mucosal and inflamed tissues, where they recognise vitamin B2 metabolites produced by bacteria and yeasts( Reference Dusseaux, Martin, Serriari, Péguillet, Premel and Louis 129 ). MAIT cells thus recognise and kill bacteria-infected cells( Reference Le Bourhis, Dusseaux, Bohineust, Bessoles, Martin and Premel 130 ). While MAIT cells have been described in breast cancer and are reportedly resistant to chemotherapy treatment( Reference Dusseaux, Martin, Serriari, Péguillet, Premel and Louis 129 ), their role in cancer is as yet unknown. MAIT cell frequency is dramatically decreased in the blood and adipose tissues of patients with T2DM and/or severe obesity compared with control donors, and this depletion is associated with diabetic status( Reference Carolan, Tobin, Mangan, Corrigan, Gaoatswe and Byrne 89 , Reference Magalhaes, Pingris, Poitou, Bessoles, Venteclef and Kiaf 90 ). Moreover, in both patient groups, circulating MAIT cells display an activated phenotype that is associated with elevated Th1 and Th17 cytokine production. In obese patients, MAIT cells are more abundant in adipose tissue than in blood and exhibit a striking IL-17 profile, a cytokine implicated in insulin resistance. Similar results were also observed in an obese paediatric cohort( Reference Carolan, Tobin, Mangan, Corrigan, Gaoatswe and Byrne 89 ). MAIT cell frequencies increase while their cytokine production decreases in obese patients after bariatric surgery, in line with improvements in metabolic parameters. These studies reveal profound abnormalities in MAIT cells in patients harbouring metabolic disorders, suggesting their potential role in these pathologies, yet future work remains to determine the role for MAIT cells in obesity-associated cancers.

Macrophages

While macrophages play a crucial role in innate immunity, their polarisation to an M1 phenotype positions them as key drivers of pathological inflammation, while a dominant M2 phenotype can facilitate their immune suppressive role in the tumour microenvironment( Reference Mantovani, Sica, Sozzani, Allavena, Vecchi and Locati 131 , Reference Verreck, de Boer, Langenberg, Hoeve, Kramer and Vaisberg 132 ). M1, or classically-activated macrophages, are regarded as the more inflammatory subset, while M2 macrophages are considered anti-inflammatory and immunoregulatory. Macrophages recruited to the tumour microenvironment can maintain an M1 phenotype and elicit potent IFN-γ, TNF-α, IL-12 and chemokine production, promote T cell activation and mediate anti-tumour activity( Reference Lamagna, Aurrand-Lions and Imhof 133 ). However, upon recruitment to the tumour microenvironment, macrophages are often polarised toward an M2 phenotype and become tumour-associated macrophages which produce immunomodulatory and pro-angiogenic factors including IL-10, TGF-β, vascular endothelial growth factor and matrix metallopeptidase 9( Reference Lamagna, Aurrand-Lions and Imhof 133 ). In the obese setting, several studies have implicated M1 macrophages as drivers of adipose tissue inflammation, with T cells and NK cells contributing to their recruitment and polarisation( Reference Xu, Barnes, Yang, Tan, Yang and Chou 31 , Reference Nishimura, Manabe, Nagasaki, Eto, Yamashita and Ohsugi 80 , Reference Wensveen, Jelenčić, Valentić, Šestan, Wensveen and Theurich 107 , Reference Weisberg, McCann, Desai, Rosenbaum, Leibel and Ferrante 134 ). In fact, M1 macrophages are believed to be the main producers of IL-1β and TNF in obesity and key players in insulin resistance( Reference Patsouris, Li, Thapar, Chapman, Olefsky and Neels 135 ). Furthermore, while the chemokine system plays a central role in macrophage recruitment to adipose tissue and the development of insulin resistance, the inhibition of one chemokine alone is not enough to block macrophage accumulation in murine models of obesity( Reference Surmi, Webb, Ristau and Hasty 136 , Reference Ota 137 ). Therefore, future studies might investigate the inhibition of more than one chemokine pathway to block M1 macrophage accumulation in obese VAT and alleviate chronic inflammation. Blocking chemokine-guided recruitment of macrophages to tumour might attenuate tumour-associated macrophage-mediated immune suppression and angiogenesis in cancer and prove useful in cancer immunotherapy. However, it is now understood that tumour-associated macrophage phenotype and function is critically influenced by the tumour microenvironment and therefore exploiting such environmental cues might be a preferable therapeutic approach( Reference Ostuni, Kratochvill, Murray and Natoli 138 ).

Immunometabolism in obesity and cancer

In recent years, research has revealed how immune cell metabolism supports and controls immune function, and has led to a focus on exploiting immunometabolism to treat inflammation and cancer. In brief, more activated and rapidly dividing immune cells rely on glycolysis to maintain their biomass and energy levels, while more quiescent and regulatory immune cells mainly utilise oxidative phosphorylation (OxPhos)( Reference Mockler, Conroy and Lysaght 139 ). Depending on the stage of their development, T cell subsets possess strikingly different metabolic profiles which reflect their energetic and biosynthetic requirements, which in turn support their function( Reference Fox, Hammerman and Thompson 140 , Reference Pearce and Pearce 141 ). Naive T cells are usually metabolically quiescent and use OxPhos as the main means of generating ATP, while rapidly proliferating effector CD4+ Th1, Th2 and Th17 cells and CD8+ cytotoxic T cells utilise significantly elevated levels of glycolysis to meet their energy requirements( Reference Fox, Hammerman and Thompson 140 Reference Bental and Deutsch 146 ). Following resolution of the immune response, the remaining pool of memory T cells mainly utilise OxPhos and lipid oxidation to meet their lower turnover rates and to support their longevity, as do Treg cells( Reference Pearce, Walsh, Cejas, Harms, Shen and Wang 147 , Reference Michalek, Gerriets, Jacobs, Macintyre, MacIver and Mason 148 ). Manipulation of such cellular metabolic pathways might represent a novel means of therapeutically skewing immune responses. Since our group and others have shown that pro-inflammatory Th1 cells are key players in obesity-associated inflammation while Treg cells are depleted, a targeted therapy to alter T cell metabolic profile and attenuate T cell-mediated inflammation in the VAT may present favourable outcomes in obesity and obesity-associated cancer( Reference Lysaght, van der Stok, Allott, Casey, Donohoe and Howard 66 , Reference Nishimura, Manabe, Nagasaki, Eto, Yamashita and Ohsugi 80 , Reference Gregor and Hotamisligil 149 , Reference Gilbert and Slingerland 150 ). Similarly, inflammatory M1 macrophages, other key players in adipose tissue inflammation, and antibody-secreting B cells also rely on glycolytic processes to fuel their inflammatory functions while M2 macrophages use fatty acids to support OxPhos( Reference Weisberg, McCann, Desai, Rosenbaum, Leibel and Ferrante 134 , Reference O'Neill 151 , Reference Murray, Allen, Biswas, Fisher, Gilroy and Goerdt 152 ).

Exploitation of immune cell metabolism might be utilised as a prophylactic to prevent obesity-associated disease or to treat obesity-associated cancer. For instance, inhibition of both the mammalian target of rapamycin and mitogen-activated protein kinase (extracellular signal-regulated kinase) pathways using rapamycin and the mitogen-activated protein kinase inhibitor PD325901 robustly blocked effector CD4+ T cell proliferation and decreased disease severity in a mouse model of arthritis( Reference Lin, Stein, Wong, Kalpathy, Su and Utz 153 ). Since our group have found pro-inflammatory effector T cells to be enriched in the inflamed omentum in obesity, extracellular signal-regulated kinase and mammalian target of rapamycin inhibitors might elicit a similar anti-inflammatory effect in obesity( Reference Lysaght, van der Stok, Allott, Casey, Donohoe and Howard 66 ). Others have found that beauvericin-mediated inhibition of T cell activation and pro-inflammatory cytokine production in an animal model of colitis was achieved by targeting P13 K and AKT and resulted in reduced weight loss, diarrhoea and mortality( Reference Wu, Wu, Huang, Liu, Ye and Huang 154 ). More recently, inhibition of AKT in expanded tumour-infiltrating lymphocytes resulted in increased rates of OxPhos and fatty acid oxidation( Reference Crompton, Sukumar, Roychoudhuri, Clever, Gros and Eil 155 ). This led to enhanced in vivo persistence of such memory T cells and augmented anti-tumour immunity following adoptive transfer( Reference Crompton, Sukumar and Restifo 156 ). Therefore, AKT inhibition might serve to dampen pro-inflammatory effector T cells in the VAT and simultaneously promote tumour-specific memory T cell responses at the tumour site, thus possibly offering a novel immuno-metabolomic therapeutic approach for chronically inflamed obesity-associated cancer patients. However, there is first an urgent need to perform focussed studies to elucidate the metabolic profiles of pro- and anti-inflammatory immune cells in the VAT and tumour of such patients.

Future prospects for obesity-associated cancer: prevention v. treatment

It is estimated that one in every four cancer cases is preventable by implementing lifestyle changes. Furthermore, studies such as the Women's Intervention Nutrition Study have shown that reduction of fat in the diet of women with breast cancer and associated weight loss reduced the relative risk of cancer recurrence by 24 %( Reference Chlebowski, Blackburn, Thomson, Nixon, Shapiro and Hoy 157 ). Prevention and management of obesity is therefore a major therapeutic goal. Intervention before the pathological inflammatory cascade ensues and immune system becomes excessively dysregulated is the ideal prophylactic approach. However, there have already been multiple government campaigns, legislation amendments and educational reforms to promote healthier lifestyle but with escalating obesity rates, a more aggressive approach is certainly needed. Griggs & Sabel argue for the benefits of aggressive promotion of lifestyle changes by physicians as part of the cancer patient treatment plan( Reference Griggs and Sabel 158 ). However, obesity is a complex variable, and BMI is not always a reliable measure of metabolic health. Schmitz et al. suggest that further clarification of the biological-social-environmental feedback loop is required in order to elucidate the combined and independent contributions of race, ethnicity, co-morbidities and obesity on cancer survival and adverse treatment effects( Reference Schmitz, Neuhouser, Agurs-Collins, Zanetti, Cadmus-Bertram and Dean 159 ).

This review highlights the critical importance of understanding the obesity–immunity–cancer axis and the fact that obesity driven immunological dysregulation should no longer be ignored when developing immunotherapeutic strategies. Immunotherapies such as pembrolizumab (Merck), nivolumab and ipilimumab (Bristol Meyer Squibb) have recently gained Food and Drug Administration approval and are so far, proving effective for a number of different cancers (www.clinicaltrials.gov). However, their suitability has not been thoroughly scrutinised under the pre-existing inflammatory condition of obesity. The challenge of using checkpoint inhibitors in obesity-associated cancer is to alleviate immune suppression and augment anti-tumour responses without inducing excessive pro-tumourigenic inflammatory cytokine production by T cells, which have already been shown to drive adipose tissue inflammation in obesity( Reference Lysaght, Allott, Donohoe, Howard, Pidgeon and Reynolds 65 , Reference Nishimura, Manabe, Nagasaki, Eto, Yamashita and Ohsugi 80 ). In the context of chronic obesity-associated inflammation, there is cause for concern with such immunotherapies, since blocking programmed death-ligand 1 has previously been shown to enhance monocyte function by decreasing IL-10 and enhancing inflammatory cytokine production( Reference Curiel, Wei, Dong, Alvarez, Cheng and Mottram 160 ). Furthermore, aged obese mice were observed to have the most extreme pathological responses to anti-CD40 and IL-2 therapy, causing cytokine storms, organ pathology and eventual death( Reference Mirsoian, Bouchlaka, Sckisel, Chen, Pai and Maverakis 161 ). Therefore, the use of such elegant immunotherapeutic approaches might be theoretically desirable for obesity-associated malignancy but first, their potential to exacerbate inflammation in obesity must be fully investigated. In the context of obesity-associated cancer, targeting the correct tissue rather than a systemic immunotherapeutic approach is also of paramount importance, when aiming to enhance anti-tumour immunity without contributing to adipose tissue and systemic inflammation. For instance, augmenting anti-tumour immunity without exacerbating inflammation might be achieved through the adoptive transfer of T or NK cells, already primed with a chemotactic signal toward the tumour site and not the adipose tissue, as demonstrated by Wennerberg et al. ( Reference Wennerberg, Kremer, Childs and Lundqvist 162 ). Similarly, approaches might combine adoptive transfer with chemokine treatments to enhance anti-inflammatory cell migration to the VAT and attenuate pathological inflammation. For instance, M2 macrophages, Treg cell and iNKT cell numbers might be replenished in the obese VAT to restore immune balance and prevent progression to inflammation-driven disease. Already, adoptive transfer of iNKT cells has been shown to reduce weight loss and improve insulin sensitivity( Reference Lynch, Nowak, Varghese, Clark, Hogan and Toxavidis 114 ). Furthermore, the Th2 proportion of CD4+ T cells adoptively transferred into mouse models of obesity has also been shown to reverse weight gain and insulin resistance( Reference Winer and Winer 163 ). However the specific chemokine pathways guiding the key T, NK and iNKT cells to the tumour and to inflamed tissue sites must first be elucidated before such combination immunotherapies can be considered.

It is now evident that several critical factors must be taken into consideration when developing successful immunotherapeutic approaches. Firstly, evaluation of the immunological environment must be considered. Overwhelming data shows a dearth of innate lymphocytes or their inactivation and general dysregulation in obesity (e.g. γδ T cells and MAIT cells are reduced with increasing BMI)( Reference Costanzo, Taylor, Dutt, Han, Fujioka and Jameson 87 , Reference Carolan, Tobin, Mangan, Corrigan, Gaoatswe and Byrne 89 , Reference Magalhaes, Pingris, Poitou, Bessoles, Venteclef and Kiaf 90 ). It may be useful to evaluate the level of immune involvement in the tumour microenvironment, and evaluate the degree of immune cell dysregulation prior to devising a patient treatment plan. Indeed, evaluation of immunological parameters is presently in development as a prognostic aid, and shows superior predictive ability compared with traditional histopathological staging methods in colorectal cancer( Reference Pagès, Berger, Camus, Sanchez-Cabo, Costes and Molidor 164 Reference Park, McMillan, Powell, Richards, Horgan and Edwards 166 ). Galon et al. show that evaluation of the type, density and location of immune cell infiltration into tumours can allow prediction of patient survival( Reference Galon, Costes, Sanchez-Cabo, Kirilovsky, Mlecnik and Lagorce-Pagès 165 ). A greater understanding of the underlying biology behind immunotherapy is also urgently required. Checkpoint inhibitors such as ipilimumab and tremelimumab have been hailed as breakthrough drugs, but show no benefit in the majority of patients and there is a paucity of published data in the obesity-associated cancer space( Reference Snyder, Makarov, Merghoub, Yuan, Zaretsky and Desrichard 167 ). Recent work shows there may be a genetic basis for clinical responses in a cohort of melanoma patients( Reference Snyder, Makarov, Merghoub, Yuan, Zaretsky and Desrichard 167 ). Improved understanding of the immunological landscape in obesity and cancer will undoubtedly allow development of further breakthroughs, for example combination immunotherapy approaches may be advantageous, combining selective cytokine or chemokine replenishment or depletion strategies with checkpoint inhibitors, adoptive transfer and traditional chemoradiotherapy strategies or hormone therapies for maximum effect. Some success has already been reported when anti-PD-1 (programmed cell death protein 1) and anti-CTLA4 (cytotoxic T lymphocytes antigen-4) therapies are administered together( Reference Curran, Montalvo, Yagita and Allison 168 ). Replenishment of growth factors that are lacking in obesity may be a useful addition to existing immunotherapeutic strategies, such as reconstituting IL-2 levels in order to boost γδ T cell responses. Simultaneous neutralisation of certain pro-inflammatory cytokines (e.g. IL-17, IL-1, IL-6) may be also be warranted in obese patients to fully overcome immunological dysregulation. Optimal dosing and timing strategies will also require further consideration, particularly in regard to treating patients with elevated BMI( Reference Drake 169 ).

In conclusion, global health is now plagued by pathologies that have arisen from an obesity epidemic that shows little sign of abating. This review has focused on the potential role of a dysregulated immune system in a growing number of obesity-associated cancers. With a new generation of immunotherapies which includes the checkpoint inhibitors nivolumab and ipilimumab anticipated to change the face of cancer treatment and improve survival for a number of cancer types, it is important that the chronic inflammation underpinning obesity-associated cancer is better understood. Over the past 10 years, there has been an increase in studies in human subjects investigating the phenotypes and functions of immune cells in adipose tissue, together with their role in obesity-associated disease, including cancer, however at this time there is scant understanding of how this knowledge can be used to therapeutic advantage with targeted approaches. Accordingly, further research investigating the immune compartments and chemokine and cytokine networks underpinning inflammation-driven obesity-related malignancy in human subjects is of immediate relevance and importance. Also, in the wake of multiple campaigns which have failed to halt growing obesity rates, more hard-line approaches are urgently required to promote lifestyle change and nutritional education. Childhood obesity must be tackled to prevent a further upsurge in obesity-associated cancer incidence among future generations. In addition to laboratory-based research, a joint effort between clinicians, research scientists, dietitians and governing bodies is needed to focus on the preventability of obesity through diet and lifestyle. Exploring these avenues will allow a multi-pronged approach to combating this burgeoning health crisis.

Conclusion

While there is a plethora of research implicating a dysregulated immune system in obesity-associated inflammation and related pathologies, further work is urgently required to identify novel approaches to prevent and treat obesity-associated diseases such as cancer. Evidence suggests that a combination of both immunotherapy- and lifestyle-based approaches may reduce incidence and improve outcomes for obesity-associated malignancy.

Fig. 1. Future approaches for the prevention and treatment of obesity-associated cancer (10,11,139). iNKT, invariant natural killer T cells; Treg, regulatory T cells; mTOR, mammalian target of rapamycin; MAPK (ERK), mitogen-activated protein kinase (extracellular signal-regulated kinase); VAT, visceral adipose tissue.

Acknowledgements

The authors would like to acknowledge the research team in the Department of Surgery at St. James's Hospital, Dublin.

Financial support

The work reported here was funded by Health Research Award HRA_POR/2011/91, Irish Research Council Government of Ireland Award 12999, and CROSS charity (Reg. Charity 389874).

Conflict of interest

None.

Authorship

J. V. R. presented the work and was responsible for the concept, direction and supervision of the manuscript. C. L. D. and M. R. D. researched and prepared sections for the manuscript. M. J. C. researched and prepared sections for the manuscript and was responsible for manuscript assembly.

References

1. WHO. 2015, Obesity and overweight. Fact sheet. Available at: http://www.who.int/mediacentre/factsheets/fs311/en/.Google Scholar
2. Flegal, KM, Carroll, MD, Ogden, CL & Curtin, LR (2010) Prevalence and trends in obesity among US adults, 1999–2008. JAMA 303, 235241.CrossRefGoogle ScholarPubMed
3. Ogden, CL, Carroll, MD, Kit, BK & Flegal, KM (2014) Prevalence of childhood and adult obesity in the United States, 2011–2012. JAMA 311, 806814; available from: http://www.ncbi.nlm.nih.gov/pubmed/24570244 Google Scholar
4. Calle, EE, Rodriguez, C, Walker-Thurmond, K & Thun, MJ (2003) Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med 348, 16251638.Google Scholar
5. Roberts, DL, Dive, C & Renehan, AG (2010) Biological mechanisms linking obesity and cancer risk: new perspectives. Annu Rev Med 61, 301316.Google Scholar
6. Scheen, AJ & Luyckx, FH (2002) Obesity and liver disease. Best Pr Res Clin 16, 703716.Google Scholar
7. Goran, MI, Ball, GDC & Cruz, ML (2003) Obesity and risk of type 2 diabetes and cardiovascular disease in children and adolescents. J Clin Endocrinol Metab 88, 14171427.Google Scholar
8. Renehan, AG, Tyson, M, Egger, M, Heller, RF & Zwahlen, M (2008) Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet 371, 569578.Google Scholar
9. Beddy, P, Howard, J, McMahon, C, Knox, M, De Blacam, C, Ravi, N et al. (2010) Association of visceral adiposity with oesophageal and junctional adenocarcinomas. Br J Surg 97, 10281034.Google Scholar
10. Postow, MA, Callahan, MK & Wolchok, JD (2015) Immune checkpoint blockade in cancer therapy. J Clin Oncol 33, 19741982; available from: http://jco.ascopubs.org/cgi/doi/10.1200/JCO.2014.59.4358 Google Scholar
11. World Cancer Research Fund (2007) Food, nutrition, physical activity, and the prevention of cancer: a global perspective. American Institute of Cancer Research.Google Scholar
12. Bhaskaran, K, Douglas, I, Forbes, H, Dos-Santos-Silva, I, Leon, DA & Smeeth, L (2014) Body-mass index and risk of 22 specific cancers: a population-based cohort study of 5·24 million UK adults. The Lancet 384, 755765.Google Scholar
13. Reeves, GK, Pirie, K, Beral, V, Green, J, Spencer, E & Bull, D (2007) Cancer incidence and mortality in relation to body mass index in the million women study: cohort study. BMJ 335, 1134.CrossRefGoogle ScholarPubMed
14. Calle, EE & Kaaks, R (2004) Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat Rev Cancer 4, 579591.CrossRefGoogle ScholarPubMed
15. Hill, AB (2015) The environment and disease: association or causation? 1965. J R Soc Med 108, 3237; available from: http://www.ncbi.nlm.nih.gov/pubmed/25572993 CrossRefGoogle ScholarPubMed
16. Renehan, AG, Egger, M & Zwahlen, M (2010) Body mass index and cancer risk: the evidence for causal association~!2009-04-22~!2009-06-16~!2010-07-13~!. Open Obes J 2, 1222; available from: http://www.bentham-open.org/pages/content.php?TOOBESJ/2010/00000002/00000002/12TOOBESJ.SGM Google Scholar
17. Donohoe, CL, Doyle, SL & Reynolds, JV (2011) Visceral adiposity, insulin resistance and cancer risk. Diabetol Metab Syndr 3, 12.Google Scholar
18. Donohoe, CL, O'Farrell, NJ, Doyle, SL & Reynolds, JV (2014) The role of obesity in gastrointestinal cancer: evidence and opinion. Therap Adv Gastroenterol 7, 3850; available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3871278&tool=pmcentrez&rendertype=abstract CrossRefGoogle ScholarPubMed
19. Fujioka, S, Matsuzawa, Y, Tokunaga, K & Tarui, S (1987) Contribution of intra-abdominal fat accumulation to the impairment of glucose and lipid metabolism in human obesity. Metabolism 36, 5459.Google Scholar
20. Seidell, JC, Björntorp, P, Sjöström, L, Kvist, H & Sannerstedt, R (1990) Visceral fat accumulation in men is positively associated with insulin, glucose, and C-peptide levels, but negatively with testosterone levels. Metabolism 39, 897901.Google Scholar
21. Nakamura, T, Tokunaga, K, Shimomura, I, Nishida, M, Yoshida, S, Kotani, K et al. (1994) Contribution of visceral fat accumulation to the development of coronary artery disease in non-obese men. Atherosclerosis 107, 239246.Google Scholar
22. Vague, J (1956) The degree of masculine differentiation of obesities: a factor determining predisposition to diabetes, atherosclerosis, gout, and uric calculous disease. Am J Clin Nutr 4, 2034.Google Scholar
23. Schapira, DV, Clark, RA, Wolff, PA, Jarrett, AR, Kumar, NB & Aziz, NM (1994) Visceral obesity and breast cancer risk. Cancer 74, 632639.Google Scholar
24. Schoen, RE, Tangen, CM, Kuller, LH, Burke, GL, Cushman, M & Tracy, RP et al. (1999) Increased blood glucose and insulin, body size, and incident colorectal cancer. J Natl Cancer Inst 91, 11471154.Google Scholar
25. Yamaji, T, Iwasaki, M, Sasazuki, S, Kurahashi, N, Mutoh, M, Yamamoto, S et al. (2009) Visceral fat volume and the prevalence of colorectal adenoma. Am J Epidemiol 170, 15021511.CrossRefGoogle ScholarPubMed
26. Nam, SY, Kim, BC, Han, KS, Ryu, KH, Park, BJ, Kim, HB et al. (2010) Abdominal visceral adipose tissue predicts risk of colorectal adenoma in both sexes. Clin Gastroenterol Hepatol 8, 44350.e1–2.Google Scholar
27. Esposito, K & Giugliano, D (2004) The metabolic syndrome and inflammation: association or causation? Nutrition, metabolism, and cardiovascular diseases. NMCD 228232.Google Scholar
28. Wajchenberg, BL (2000) Subcutaneous and visceral adipose tissue: their relation to the metabolic syndrome. Endocr Rev 697738.Google Scholar
29. Das, UN (2001) Is obesity an inflammatory condition? Nutrition 953966.Google Scholar
30. Coussens, LM, Werb, Z (2002) Inflammation and cancer. Nature 420, 860867.Google Scholar
31. Xu, H, Barnes, GT, Yang, Q, Tan, G, Yang, D, Chou, CJ et al. (2003) Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest 112, 18211830.Google Scholar
32. Hotamisligil, GS, Arner, P, Caro, JF, Atkinson, RL & Spiegelman, BM (1995) Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest 95, 24092415.Google Scholar
33. Shoelson, SE, Lee, J & Goldfine, AB (2006) Inflammation and insulin resistance. J Clin Invest 17931801.Google Scholar
34. Antuna-Puente, B, Feve, B, Fellahi, S & Bastard, J-P (2008) Adipokines: the missing link between insulin resistance and obesity. Diabetes Metab 34, 211.Google Scholar
35. DeFronzo, RA (2010) Insulin resistance, lipotoxicity, type 2 diabetes and atherosclerosis: the missing links. The Claude Bernard lecture 2009. Diabetologia 12701287.Google Scholar
36. Buchwald, H, Avidor, Y, Braunwald, E, Jensen, MD, Pories, W, Fahrbach, K et al. (2004) Bariatric surgery: a systematic review and meta-analysis. JAMA 292, 17241737.Google Scholar
37. Johnson, JA, Carstensen, B, Witte, D, Bowker, SL, Lipscombe, L & Renehan, AG (2012) Diabetes and cancer (1): evaluating the temporal relationship between type 2 diabetes and cancer incidence. Diabetologia 16071618.Google Scholar
38. Bergman, RN & Ader, M (2000) Free fatty acids and pathogenesis of type 2 diabetes mellitus. Trends Endocrinol Metab 11, 351356.Google Scholar
39. Moller, DE & Flier, JS (1991) Insulin resistance – mechanisms, syndromes, and implications. N Engl J Med 325, 938948.Google Scholar
40. Trevisan, M, Liu, J, Muti, P, Misciagna, G, Menotti, A & Fucci, F (2001) Markers of insulin resistance and colorectal cancer mortality. Cancer Epidemiol Biomarkers Prev 10, 937941.Google Scholar
41. Colangelo, LA, Gapstur, SM, Gann, PH, Dyer, AR & Liu, K (2002) Colorectal cancer mortality and factors related to the insulin resistance syndrome. Cancer Epidemiol Biomarkers Prev 11, 385391.Google Scholar
42. Pollak, M (2012) The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nat Rev Cancer.Google Scholar
43. Hellawell, GO, Turner, GDH, Davies, DR, Poulsom, R, Brewster, SF & Macaulay, VM (2002) Expression of the type 1 insulin-like growth factor receptor is up-regulated in primary prostate cancer and commonly persists in metastatic disease. Cancer Res 62, 29422950.Google Scholar
44. Law, JH, Habibi, G, Hu, K, Masoudi, H, Wang, MYC, Stratford, AL et al. (2008) Phosphorylated insulin-like growth factor-I/insulin receptor is present in all breast cancer subtypes and is related to poor survival. Cancer Res 68, 1023810246.CrossRefGoogle ScholarPubMed
45. Donohoe, CL, Doyle, SL, McGarrigle, S, Cathcart, MC, Daly, E, O'Grady, A et al. (2012) Role of the insulin-like growth factor 1 axis and visceral adiposity in oesophageal adenocarcinoma. Br J Surg 99, 387396; available from: http://www.ncbi.nlm.nih.gov/pubmed/22241325 Google Scholar
46. Tsilidis, KK, Travis, RC, Appleby, PN, Allen, NE, Lindstrom, S, Schumacher, FR et al. (2012) Interactions between genome-wide significant genetic variants and circulating concentrations of insulin-like growth factor 1, sex hormones, and binding proteins in relation to prostate cancer risk in the national cancer institute breast and prostate cancer. Am J Epidemiol 175, 926935.Google Scholar
47. Hanahan, D & Weinberg, RA (2011) Hallmarks of cancer: the next generation. Cell 144, 646674; available from: http://www.ncbi.nlm.nih.gov/pubmed/21376230 Google Scholar
48. Lynam-Lennon, N, Maher, SG, Maguire, A, Phelan, J, Muldoon, C, Reynolds, JV et al. (2014) Altered mitochondrial function and energy metabolism is associated with a radio resistant phenotype in oesophageal adenocarcinoma. PLoS ONE 9 (6) e100738.CrossRefGoogle Scholar
49. Groff, JL, Stugard, CE, Mays, CJ, Koopmans, HS & DiGirolamo, M (1992) Glucose metabolism in isolated rat adipocytes: estimate of total recovery by the product summation method. J Lab Clin Med 119, 216220.Google Scholar
50. Lukanova, A, Lundin, E, Zeleniuch-Jacquotte, A, Muti, P, Mure, A, Rinaldi, S et al. (2004) Body mass index, circulating levels of sex-steroid hormones, IGF-I and IGF-binding protein-3: a cross-sectional study in healthy women. Eur J Endocrinol 150, 161171.Google Scholar
51. Zeleniuch-Jacquotte, A, Akhmedkhanov, A, Kato, I, Koenig, KL, Shore, RE, Kim, MY et al. (2001) Postmenopausal endogenous oestrogens and risk of endometrial cancer: results of a prospective study. Br J Cancer 84, 975981.Google Scholar
52. Dossus, L, Lukanova, A, Rinaldi, S, Allen, N, Cust, AE, Becker, S et al. (2013) Hormonal, metabolic, and inflammatory profiles and endometrial cancer risk within the EPIC cohort – A factor analysis. Am J Epidemiol 177, 787799.Google Scholar
53. Cao, H (2014) Adipokines in obesity and metabolic disease. J Endocrinol 220, T47T59.Google Scholar
54. Maffei, M, Halaas, J, Ravussin, E, Pratley, RE, Lee, GH, Zhang, Y et al. (1995) Leptin levels in human and rodent: measurement of plasma leptin and ob RNA in obese and weight-reduced subjects. Nat Med 1, 11551161.Google Scholar
55. Lord, GM, Matarese, G, Howard, JK, Baker, RJ, Bloom, SR & Lechler, RI (1998) Leptin modulates the T-cell immune response and reverses starvation-induced immunosuppression. Nature 394, 897901.Google Scholar
56. Matarese, G, Moschos, S & Mantzoros, CS (2005) Leptin in immunology. J Immunol 174, 31373142; available from: http://www.jimmunol.org/cgi/doi/10.4049/jimmunol.174.6.3137 Google Scholar
57. Faggioni, R, Feingold, KR & Grunfeld, C (2001) Leptin regulation of the immune response and the immunodeficiency of malnutrition. FASEB J 15, 25652571.Google Scholar
58. Tanaka, S, Isoda, F, Ishihara, Y, Kimura, M & Yamakawa, T (2001) T lymphopaenia in relation to body mass index and TNF-alpha in human obesity: adequate weight reduction can be corrective. Clin Endocrinol (Oxf) 54, 347354.Google ScholarPubMed
59. Considine, RV, Sinha, MK, Heiman, ML, Kriauciunas, A, Stephens, TW, Nyce, MR et al. (1996) Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N Engl J Med 334, 292295.CrossRefGoogle ScholarPubMed
60. Kadowaki, T & Yamauchi, T (2005) Adiponectin and adiponectin receptors. Endocr Rev 439451.Google Scholar
61. Weyer, C, Funahashi, T, Tanaka, S, Hotta, K, Matsuzawa, Y, Pratley, RE et al. (2001) Hypoadiponectinemia in obesity and type 2 diabetes: close association with insulin resistance and hyperinsulinemia. J Clin Endocrinol Metab 86, 19301935.Google Scholar
62. Yamauchi, T, Kamon, J, Waki, H, Terauchi, Y, Kubota, N, Hara, K et al. (2001) The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat Med 7, 941946.Google Scholar
63. Lago, F, Dieguez, C, Gómez-Reino, J & Gualillo, O (2007) Adipokines as emerging mediators of immune response and inflammation. Nat Clin Pract Rheumatol 3, 716724; available from: http://www.nature.com/doifinder/10.1038/ncprheum0674 CrossRefGoogle ScholarPubMed
64. Fain, JN (2006) Release of interleukins and other inflammatory cytokines by human adipose tissue is enhanced in obesity and primarily due to the nonfat cells. Vitam Horm 74, 443477.Google Scholar
65. Lysaght, J, Allott, EH, Donohoe, CL, Howard, JM, Pidgeon, GP, Reynolds, JV (2011) T lymphocyte activation in visceral adipose tissue of patients with oesophageal adenocarcinoma. Br J Surg 98, 964974.Google Scholar
66. Lysaght, J, van der Stok, EP, Allott, EH, Casey, R, Donohoe, CL, Howard, JM et al. (2011) Pro-inflammatory and tumour proliferative properties of excess visceral adipose tissue. Cancer Lett 312, 6272.Google Scholar
67. Lutz, CT, Quinn, LS (2012) Sarcopenia, obesity, and natural killer cell immune senescence in aging: altered cytokine levels as a common mechanism. Aging 4, 535546.CrossRefGoogle ScholarPubMed
68. Wellen, KE, Hotamisligil, GS (2003) Obesity-induced inflammatory changes in adipose tissue. J Clin Invest 17851788.Google Scholar
69. Karin, M (2009) NF-kappaB as a critical link between inflammation and cancer. Cold Spring Harb Perspect Biol. 1, a000141.CrossRefGoogle ScholarPubMed
70. Ramos-Nino, ME (2013) The role of chronic inflammation in obesity-associated cancers. ISRN Oncol 125; available from: http://www.hindawi.com/isrn/oncology/2013/697521/ Google Scholar
71. Zhang, X, Zhou, M, Guo, Y, Song, Z & Liu, B (2015) 1,25-Dihydroxyvitamin D3 promotes high glucose-induced M1 macrophage switching to M2 via the VDR-PPARγ signaling pathway. Biomed Res Int 157834; available from: http://www.ncbi.nlm.nih.gov/pubmed/25961000 Google Scholar
72. Fridlender, ZG, Jassar, A, Mishalian, I, Wang, L-C, Kapoor, V, Cheng, G et al. (2013) Using macrophage activation to augment immunotherapy of established tumours. Br J Cancer 108, 12881297.Google Scholar
73. Winer, S, Chan, Y, Paltser, G, Truong, D, Tsui, H, Bahrami, J et al. (2009) Normalization of obesity-associated insulin resistance through immunotherapy. Nat Med 15, 921929.CrossRefGoogle ScholarPubMed
74. Knutson, KL & Disis, ML (2005) Tumor antigen-specific T helper cells in cancer immunity and immunotherapy. Cancer Immunol Immunother 721728.Google Scholar
75. Zanetti, M (2015) Tapping CD4 T cells for cancer immunotherapy: the choice of personalized genomics. J Immunol 194, 20492056; available from: http://www.ncbi.nlm.nih.gov/pubmed/25710958 Google Scholar
76. Deiuliis, J, Shah, Z, Shah, N, Needleman, B, Mikami, D, Narula, V et al. (2011) Visceral adipose inflammation in obesity is associated with critical alterations in tregulatory cell numbers. Brusic V, editor. PLoS ONE 6, e16376; available from: http://dx.plos.org/10.1371/journal.pone.0016376 Google Scholar
77. Bertola, A, Ciucci, T, Rousseau, D, Bourlier, V, Duffaut, C, Bonnafous, S et al. (2012) Identification of adipose tissue dendritic cells correlated with obesity-associated insulin-resistance and inducing Th17 responses in mice and patients. Diabetes 61, 22382247.Google Scholar
78. Zou, W & Restifo, NP (2010) T(H)17 cells in tumour immunity and immunotherapy. Nat Rev Immunol 10, 248256.Google Scholar
79. Nishikawa, H, Sakaguchi, S (2014) Regulatory T cells in cancer immunotherapy. Curr Opin Immunol 17.Google Scholar
80. Nishimura, S, Manabe, I, Nagasaki, M, Eto, K, Yamashita, H, Ohsugi, M et al. (2009) CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med 15, 914920.Google Scholar
81. Jackson, SR, Yuan, J & Teague, RM (2014) Targeting CD8+ T-cell tolerance for cancer immunotherapy. Immunotherapy 6, 833852; available from: http://www.ncbi.nlm.nih.gov/pubmed/25290416 Google Scholar
82. Klebanoff, CA, Gattinoni, L & Restifo, NP (2006) CD8+ T-cell memory in tumor immunology and immunotherapy. Immunol Rev 211, 214224; available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=1501075&tool=pmcentrez&rendertype=abstract Google Scholar
83. Lynch, LA, O'Connell, JM, Kwasnik, AK, Cawood, TJ, O'Farrelly, C & O'Shea, DB (2009) Are natural killer cells protecting the metabolically healthy obese patient? Obesity (Silver Spring) 17, 601605.Google Scholar
84. Ljunggren, H-G & Malmberg, K-J (2007) Prospects for the use of NK cells in immunotherapy of human cancer. Nat Rev Immunol 7, 329339.Google Scholar
85. Kawano, T, Nakayama, T, Kamada, N, Kaneko, Y, Harada, M, Ogura, N et al. (1999) Antitumor cytotoxicity mediated by ligand-activated human Vα24 NKT cells. Cancer Res 59, 51025105.Google Scholar
86. Schilbach, K, Frommer, K, Meier, S, Handgretinger, R, Eyrich, M (0000) Immune response of human propagated gammadelta-T-cells to neuroblastoma recommend the Vdelta1+ subset for gammadelta-T-cell-based immunotherapy. J Immunother 31, 896905.Google Scholar
87. Costanzo, AE, Taylor, KR, Dutt, S, Han, PP, Fujioka, K & Jameson, JM (2015) Obesity impairs γδ T cell homeostasis and antiviral function in humans. PLoS ONE 10, e0120918; available from: http://www.ncbi.nlm.nih.gov/pubmed/25785862 Google Scholar
88. Gomes, AQ, Martins, DS & Silva-Santos, B (2010) Targeting γδ T lymphocytes for cancer immunotherapy: from novel mechanistic insight to clinical application. Cancer Res 70, 1002410027; available from: http://www.ncbi.nlm.nih.gov/pubmed/21159627 Google Scholar
89. Carolan, E, Tobin, LM, Mangan, BA, Corrigan, M, Gaoatswe, G, Byrne, G et al. (2015) Altered distribution and increased IL-17 production by mucosal-associated invariant T cells in adult and childhood obesity. J Immunol 194, 57755780; available from: http://www.ncbi.nlm.nih.gov/pubmed/25980010 Google Scholar
90. Magalhaes, I, Pingris, K, Poitou, C, Bessoles, S, Venteclef, N, Kiaf, B et al. (2015) Mucosal-associated invariant T cell alterations in obese and type 2 diabetic patients. J Clin Invest 125, 17521762; available from: http://www.ncbi.nlm.nih.gov/pubmed/25751065 Google Scholar
91. Ikeda, H, Chamoto, K, Tsuji, T, Suzuki, Y, Wakita, D, Takeshima, T et al. (2004) The critical role of type-1 innate and acquired immunity in tumor immunotherapy. Cancer Sci 697703.Google Scholar
92. Foster, PS, Martinez-Moczygemba, M, Huston, DP & Corry, DB (2002) Interleukins-4, −5, and −13: emerging therapeutic targets in allergic disease. Pharmacol Ther 253264.Google Scholar
93. Ciric, B, El-behi, M, Cabrera, R, Zhang, G-X & Rostami, A (2009) IL-23 drives pathogenic IL-17-producing CD8+ T cells. J Immunol 182, 52965305; available from: http://www.jimmunol.org/cgi/doi/10.4049/jimmunol.0900036 Google Scholar
94. Cottrez, F & Groux, H (2001) Regulation of TGF-beta response during T cell activation is modulated by IL-10. J Immunol 167, 773778.Google Scholar
95. Kalams, SA & Walker, BD (1998) The critical need for CD4 help in maintaining effective cytotoxic T lymphocyte responses. J Exp Med 188, 21992204.Google Scholar
96. Kono, K, Kawaida, H, Takahashi, A, Sugai, H, Mimura, K, Miyagawa, N et al. (2006) CD4(+)CD25high regulatory T cells increase with tumor stage in patients with gastric and esophageal cancers. Cancer Immunol Immunother 55, 10641071.Google Scholar
97. Shen, Z, Zhou, S, Wang, Y, Li, RL, Zhong, C, Liang, C et al. (2010) Higher intratumoral infiltrated Foxp3+ Treg numbers and Foxp3+/CD8+ ratio are associated with adverse prognosis in resectable gastric cancer. J Cancer Res Clin Oncol 136, 15851595.Google Scholar
98. Feuerer, M, Herrero, L, Cipolletta, D, Naaz, A, Wong, J, Nayer, A et al. (2009) Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med 15, 930939.Google Scholar
99. Robertson, MJ & Ritz, J (1990) Biology and clinical relevance of human natural killer cells. Blood 76, 24212438.Google Scholar
100. Harizi, H (2013) Reciprocal crosstalk between dendritic cells and natural killer cells under the effects of PGE2 in immunity and immunopathology. Cell Mol Immunol 10, 213221; available from: http://www.ncbi.nlm.nih.gov/pubmed/23524652 Google Scholar
101. Ing, R & Stevenson, MM (2009) Dendritic cell and NK cell reciprocal cross talk promotes gamma interferon-dependent immunity to blood-stage Plasmodium chabaudi AS infection in mice. Infect Immun 77, 770782.Google Scholar
102. Yadav, PK, Chen, C, Liu, Z (2011) Potential Role of NK Cells in the pathogenesis of inflammatory bowel disease. J Biomed Biotechnol 16; available from: http://www.hindawi.com/journals/bmri/2011/348530/ Google Scholar
103. Gardiner, CM & Dunphy, S (2011) NK cells and psoriasis. J Biomed Biotechnol. Available from: http://dx.doi.org/10.1155/2011/248317 Google Scholar
104. O'Shea, D, Cawood, TJ, O'Farrelly, C & Lynch, L (2010) Natural killer cells in obesity: impaired function and increased susceptibility to the effects of cigarette smoke. PLoS ONE 5.Google Scholar
105. O'Rourke, RW, Gaston, GD, Meyer, KA, White, AE & Marks, DL (2013) Adipose tissue NK cells manifest an activated phenotype in human obesity. Metabolism 62, 15571561.Google Scholar
106. O'Rourke, RW (2014) Obesity and cancer: at the crossroads of cellular metabolism and proliferation. Surg Obes Relat Dis 10, 12081219; available from: http://www.ncbi.nlm.nih.gov/pubmed/25264328 Google Scholar
107. Wensveen, FM, Jelenčić, V, Valentić, S, Šestan, M, Wensveen, TT, Theurich, S et al. (2015) NK cells link obesity-induced adipose stress to inflammation and insulin resistance. Nat Immunol 16; available from: http://www.ncbi.nlm.nih.gov/pubmed/25729921 Google Scholar
108. Bendelac, A, Rivera, MN, Park, SH, Roark, JH (1997) Mouse CD1-specific NK1 T cells: development, specificity, and function. Annu Rev Immunol 15, 535562.Google Scholar
109. Exley, M, Garcia, J, Balk, SP & Porcelli, S (1997) Requirements for CD1d recognition by human invariant Valpha24+ CD4-CD8- T cells. J Exp Med 186, 109120.Google Scholar
110. Brigl, M & Brenner, MB (2004) CD1: antigen presentation and T cell function. Annu Rev Immunol 22, 817890.Google Scholar
111. Gumperz, JE (2006) The ins and outs of CD1 molecules: bringing lipids under immunological surveillance. Traffic 213.Google Scholar
112. Kawano, T, Cui, J, Koezuka, Y, Toura, I, Kaneko, Y, Motoki, K et al. (1997) CD1d-restricted and TCR-mediated activation of valpha14 NKT cells by glycosylceramides. Science 278, 16261629.Google Scholar
113. Kenna, T, Golden-Mason, L, Norris, S, Hegarty, JE, O'Farrelly, C & Doherty, DG (2004) Distinct subpopulations of gamma delta T cells are present in normal and tumor-bearing human liver. Clin Immunol 113, 5663; available from: http://www.ncbi.nlm.nih.gov/pubmed/15380530 Google Scholar
114. Lynch, L, Nowak, M, Varghese, B, Clark, J, Hogan, AE, Toxavidis, V et al. (2012) Adipose tissue invariant NKT cells protect against diet-induced obesity and metabolic disorder through regulatory cytokine production. Immunity 37, 574587; available from: http://linkinghub.elsevier.com/retrieve/pii/S1074761312003792 Google Scholar
115. Dunne, MR, Elliott, L, Hussey, S, Mahmud, N, Kelly, J, Doherty, DG et al. (2013) Persistent changes in circulating and intestinal γδ T cell subsets, invariant natural killer T cells and mucosal-associated invariant T cells in children and adults with coeliac disease. PLoS ONE 8, e76008.Google Scholar
116. Laggner, U, Di Meglio, P, Perera, GK, Hundhausen, C, Lacy, KE, Ali, N et al. (2011) Identification of a novel proinflammatory human skin-homing Vγ9 Vδ2 T cell subset with a potential role in psoriasis. J Immunol 187, 27832793.Google Scholar
117. Lahn, M (2000) The role of gammadelta T cells in the airways. J Mol Med (Berl) 78, 409425.Google Scholar
118. Exley, MA & Boyson, JE (2011) Protective role of regulatory decidual γδ T cells in pregnancy. Clin Immunol 141, 236239.Google Scholar
119. Hayday, AC (2000) [gamma][delta] cells: a right time and a right place for a conserved third way of protection. Annu Rev Immunol 18, 9751026.Google Scholar
120. Dunne, MR, Mangan, BA, Madrigal-Estebas, L & Doherty, DG (2010) Preferential Th1 cytokine profile of phosphoantigen-stimulated human Vγ9Vδ2 T cells. Mediators Inflamm 704941.Google Scholar
121. Crowley, MP, Reich, Z, Mavaddat, N, Altman, JD & Chien, Y (1997) The recognition of the nonclassical major histocompatibility complex (MHC) class I molecule, T10, by the gammadelta T cell, G8. J Exp Med 185, 12231230.Google Scholar
122. Singhal, A, Mori, L, De Libero, GT (2013) cell recognition of non-peptidic antigens in infectious diseases. Indian J Med Res 620631.Google Scholar
123. Lockhart, E, Green, AM & Flynn, JL (2006) IL-17 production is dominated by gammadelta T cells rather than CD4 T cells during Mycobacterium tuberculosis infection. J Immunol 177, 46624669.Google Scholar
124. Bergstresser, PR, Sullivan, S, Streilein, JW & Tigelaar, RE (1985) Origin and function of Thy-1+ dendritic epidermal cells in mice. J Invest Dermatol 85(Suppl. 1), 85s90s.Google Scholar
125. Siegers, GM, Dhamko, H, Wang, X-H, Mathieson, AM, Kosaka, Y, Felizardo, TC et al. (2011) Human Vδ1 γδ T cells expanded from peripheral blood exhibit specific cytotoxicity against B-cell chronic lymphocytic leukemia-derived cells. Cytotherapy 13, 753764.Google Scholar
126. Mangan, BA, Dunne, MR, O'Reilly, VP, Dunne, PJ, Exley, MA, O'Shea, D et al. (2013) Cutting edge: CD1d restriction and Th1/Th2/Th17 cytokine secretion by human Vδ3 T cells. J Immunol 191, 3034; available from: http://www.ncbi.nlm.nih.gov/pubmed/23740951 Google Scholar
127. Mehta, P, Nuotio-Antar, AM & Smith, CW (2015) γδ T cells promote inflammation and insulin resistance during high fat diet-induced obesity in mice. J Leukoc Biol 97, 121134; available from: http://www.ncbi.nlm.nih.gov/pubmed/25395302 Google Scholar
128. Treiner, E, Duban, L, Bahram, S, Radosavljevic, M, Wanner, V, Tilloy, F et al. (2003) Selection of evolutionarily conserved mucosal-associated invariant T cells by MR1. Nature 422, 164169; available from: http://www.ncbi.nlm.nih.gov/pubmed/12634786 Google Scholar
129. Dusseaux, M, Martin, E, Serriari, N, Péguillet, I, Premel, V, Louis, D et al. (2011) Human MAIT cells are xenobiotic-resistant, tissue-targeted, CD161hi IL-17-secreting T cells. Blood 117, 12501259; available from: http://www.ncbi.nlm.nih.gov/pubmed/21084709 Google Scholar
130. Le Bourhis, L, Dusseaux, M, Bohineust, A, Bessoles, S, Martin, E, Premel, V et al. (2013) MAIT cells detect and efficiently lyse bacterially-infected epithelial cells. PLoS Pathog 9.Google Scholar
131. Mantovani, A, Sica, A, Sozzani, S, Allavena, P, Vecchi, A & Locati, M (2004) The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol 677686.Google Scholar
132. Verreck, FAW, de Boer, T, Langenberg, DML, Hoeve, MA, Kramer, M, Vaisberg, E et al. (2004) Human IL-23-producing type 1 macrophages promote but IL-10-producing type 2 macrophages subvert immunity to (myco)bacteria. Proc Natl Acad Sci USA 101, 45604565.Google Scholar
133. Lamagna, C, Aurrand-Lions, M & Imhof, BA (2006) Dual role of macrophages in tumor growth and angiogenesis. J Leukoc Biol 80, 705713.Google Scholar
134. Weisberg, SP, McCann, D, Desai, M, Rosenbaum, M, Leibel, RL & Ferrante, AW (2003) Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest 112, 17961808.Google Scholar
135. Patsouris, D, Li, PP, Thapar, D, Chapman, J, Olefsky, JM & Neels, JG (2008) Ablation of CD11c-positive cells normalizes insulin sensitivity in obese insulin resistant animals. Cell Metab 8, 301309.Google Scholar
136. Surmi, BK, Webb, CD, Ristau, AC & Hasty, AH (2010) Absence of macrophage inflammatory protein-1{alpha} does not impact macrophage accumulation in adipose tissue of diet-induced obese mice. Am J Physiol Endocrinol Metab 299, E437E445.Google Scholar
137. Ota, T (2013) Chemokine systems link obesity to insulin resistance. Diabetes Metab J 37, 165172.Google Scholar
138. Ostuni, R, Kratochvill, F, Murray, PJ, Natoli, G (2015) Macrophages and cancer: from mechanisms to therapeutic implications. Trends Immunol 36, 229239; available from: http://www.ncbi.nlm.nih.gov/pubmed/25770924 Google Scholar
139. Mockler, MB, Conroy, MJ & Lysaght, J (2014) Targeting T cell immunometabolism for cancer immunotherapy; understanding the impact of the tumor microenvironment. Front Oncol 4; available from: http://journal.frontiersin.org/article/10.3389/fonc.2014.00107/abstract Google Scholar
140. Fox, CJ, Hammerman, PS & Thompson, CB (2005) Fuel feeds function: energy metabolism and the T-cell response. Nat Rev Immunol 5, 844852.Google Scholar
141. Pearce, E & Pearce, E (2013) Metabolic pathways in immune cell activation and quiescence. Immunity 633643.CrossRefGoogle ScholarPubMed
142. Guppy, M, Greiner, E & Brand, K (1993) The role of the Crabtree effect and an endogenous fuel in the energy metabolism of resting and proliferating thymocytes. Eur J Biochem 212, 9599.Google Scholar
143. Greiner, EF, Guppy, M & Brand, K (1994) Glucose is essential for proliferation and the glycolytic enzyme induction that provokes a transition to glycolytic energy production. J Biol Chem 269, 3148431490.Google Scholar
144. Carr, EL, Kelman, A, Wu, GS, Gopaul, R, Senkevitch, E, Aghvanyan, A et al. (2010) Glutamine uptake and metabolism are coordinately regulated by ERK/MAPK during T lymphocyte activation. J Immunol 185, 10371044; available from: http://www.jimmunol.org/cgi/doi/10.4049/jimmunol.0903586 Google Scholar
145. Van Stipdonk, MJB, Hardenberg, G, Bijker, MS, Lemmens, EE, Droin, NM, Green, DR et al. (2003) Dynamic programming of CD8+ T lymphocyte responses. Nat Immunol 4, 361365.Google Scholar
146. Bental, M & Deutsch, C (1993) Metabolic changes in activated T cells: an NMR study of human peripheral blood lymphocytes. Magn Reson Med 29, 317326.Google Scholar
147. Pearce, EL, Walsh, MC, Cejas, PJ, Harms, GM, Shen, H, Wang, L-S et al. (2009) Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature 460, 103107.Google Scholar
148. Michalek, RD, Gerriets, VA, Jacobs, SR, Macintyre, AN, MacIver, NJ, Mason, EF et al. (2011) Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol 186, 32993303.Google Scholar
149. Gregor, MF, Hotamisligil, GS (2011) Inflammatory mechanisms in obesity. Annu Rev Immunol 29, 415445; available from: http://www.annualreviews.org/doi/abs/10.1146/annurev-immunol-031210-101322 Google Scholar
150. Gilbert, Ca, Slingerland, JM (2013) Cytokines, obesity, and cancer: new insights on mechanisms linking obesity to cancer risk and progression. Annu Rev Med 64, 4557; available from: http://www.ncbi.nlm.nih.gov/pubmed/23121183 Google Scholar
151. O'Neill, LAJ (2015) A broken krebs cycle in macrophages. Immunity 42, 393394; available from: http://linkinghub.elsevier.com/retrieve/pii/S1074761315000928 CrossRefGoogle ScholarPubMed
152. Murray, PJ, Allen, JE, Biswas, SK, Fisher, EA, Gilroy, DW, Goerdt, S et al. (2014) Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41, 1420.Google Scholar
153. Lin, JT, Stein, EA, Wong, MT, Kalpathy, KJ, Su, LL, Utz, PJ et al. (2012) Differential mTOR and ERK pathway utilization by effector CD4 T cells suggests combinatorial drug therapy of arthritis. Clin Immunol 142, 127138.Google Scholar
154. Wu, D, Wu, P, Huang, Q, Liu, Y, Ye, J & Huang, J (2013) Interleukin-17: a promoter in colorectal cancer progression. Clin Dev Immunol 436307; available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3870650&tool=pmcentrez&rendertype=abstract Google Scholar
155. Crompton, JG, Sukumar, M, Roychoudhuri, R, Clever, D, Gros, A, Eil, RL et al. (2015) Akt inhibition enhances expansion of potent tumor-specific lymphocytes with memory cell characteristics. Cancer Res 75, 296305; available from: http://www.ncbi.nlm.nih.gov/pubmed/25432172 Google Scholar
156. Crompton, JG, Sukumar, M & Restifo, NP (2014) Uncoupling T-cell expansion from effector differentiation in cell-based immunotherapy. Immunol Rev 257, 264276.Google Scholar
157. Chlebowski, RT, Blackburn, GL, Thomson, CA, Nixon, DW, Shapiro, A, Hoy, MK et al. (2006) Dietary fat reduction and breast cancer outcome: interim efficacy results from the women's intervention nutrition study. J Natl Cancer Inst 98, 17671776.Google Scholar
158. Griggs, JJ & Sabel, MS (2008) Obesity and cancer treatment: weighing the evidence. J Clin Oncol 26, 40604062; available from: http://jco.ascopubs.org/cgi/doi/10.1200/JCO.2008.17.4250 Google Scholar
159. Schmitz, KH, Neuhouser, ML, Agurs-Collins, T, Zanetti, KA, Cadmus-Bertram, L, Dean, LT et al. (2013) Impact of obesity on cancer survivorship and the potential relevance of race and ethnicity. JNCI J Natl Cancer Inst 105, 13441354; available from: http://jnci.oxfordjournals.org/cgi/doi/10.1093/jnci/djt223 Google Scholar
160. Curiel, TJ, Wei, S, Dong, H, Alvarez, X, Cheng, P, Mottram, P et al. (2003) Blockade of B7-H1 improves myeloid dendritic cell–mediated antitumor immunity. Nat Med 9, 562567; available from: http://www.nature.com/doifinder/10.1038/nm863 Google Scholar
161. Mirsoian, A, Bouchlaka, MN, Sckisel, GD, Chen, M, Pai, C-CS, Maverakis, E et al. (2014) Adiposity induces lethal cytokine storm after systemic administration of stimulatory immunotherapy regimens in aged mice. J Exp Med 211, 23732383; available from: http://www.ncbi.nlm.nih.gov/pubmed/25366964 Google Scholar
162. Wennerberg, E, Kremer, V, Childs, R & Lundqvist, A (2015) CXCL10-induced migration of adoptively transferred human natural killer cells toward solid tumors causes regression of tumor growth in vivo . Cancer Immunol Immunother 64, 225235.Google Scholar
163. Winer, S & Winer, DA (2012) The adaptive immune system as a fundamental regulator of adipose tissue inflammation and insulin resistance. Immunol Cell Biol 90, 755762.Google Scholar
164. Pagès, F, Berger, A, Camus, M, Sanchez-Cabo, F, Costes, A, Molidor, R et al. (2005) Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med 353, 26542666.Google Scholar
165. Galon, J, Costes, A, Sanchez-Cabo, F, Kirilovsky, A, Mlecnik, B, Lagorce-Pagès, C et al. (2006) Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 313, 19601964; available from: http://www.ncbi.nlm.nih.gov/pubmed/17008531 Google Scholar
166. Park, JH, McMillan, DC, Powell, AG, Richards, CH, Horgan, PG, Edwards, J et al. (2015) Evaluation of a tumor microenvironment-based prognostic score in primary operable colorectal cancer. Clin Cancer Res 21, 882888; available from: http://www.ncbi.nlm.nih.gov/pubmed/25473000 Google Scholar
167. Snyder, A, Makarov, V, Merghoub, T, Yuan, J, Zaretsky, JM, Desrichard, A et al. (2014) Genetic basis for clinical response to CTLA-4 blockade in Melanoma. N Engl J Med available from: http://www.ncbi.nlm.nih.gov/pubmed/25409260 Google Scholar
168. Curran, MA, Montalvo, W, Yagita, H & Allison, JP (2010) PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci USA 107, 42754280.Google Scholar
169. Drake, CG (2012) Combination immunotherapy approaches. Ann Oncol. 23, Suppl. 8, vii 4146.Google Scholar
Figure 0

Table 1. Hazard ratio (HR) for cancers per 5 kg/m2 increase in BMI(12)

Figure 1

Table 2. Summary of immunological impact of obesity in human and potential therapeutic targets

Figure 2

Fig. 1. Future approaches for the prevention and treatment of obesity-associated cancer (10,11,139). iNKT, invariant natural killer T cells; Treg, regulatory T cells; mTOR, mammalian target of rapamycin; MAPK (ERK), mitogen-activated protein kinase (extracellular signal-regulated kinase); VAT, visceral adipose tissue.