Abstract
Breast cancer is the most common cancer in women, and incidence rates are rising, it is thought in part, due to increasing levels of obesity. Endocrine therapy (ET) remains the cornerstone of systemic therapy for early and advanced oestrogen receptor-positive (ER + ) breast cancer, but despite treatment advances, it is becoming more evident that obesity and insulin resistance are associated with worse outcomes. Here, we describe the current understanding of the relationship between both obesity and diabetes and the prevalence and outcomes for ER+ breast cancer. We also discuss the mechanisms associated with resistance to ET and the relationship to treatment toxicity.
Similar content being viewed by others
Introduction
The obesity epidemic is contributing to the rising incidence rates of breast cancer, which remains the most common cancer for women worldwide [1]. Furthermore, the relationship between breast cancer and metabolic disorders, specifically obesity and insulin resistance, increases the complexity of breast cancer treatment, posing unique challenges in managing toxicities and treatment resistance.
As described by the World Health Organisation (WHO), a body mass index (BMI) of 30 kg/m2 or greater is considered obese and is present in 13% of the world’s adult population, with greater prevalence in the Western world [2]. Obese patients with ER+ breast cancer are at a greater risk of cancer development, recurrence, and mortality [3,4,5,6], even after accounting for confounding variables such as concomitant diseases and chemotherapy underdosing [7]. Obesity also increases the risk of insulin resistance, characterised by cellular insensitivity to insulin, and is associated with a cluster of conditions, including hypertension, hyperglycaemia, central adiposity, and dyslipidaemia, known as metabolic syndrome [2]. These variables collectively raise the risk of developing Type 2 diabetes mellitus (T2DM), which in itself is associated with increased cancer risk [8].
In addition, there is growing evidence that the metabolic abnormalities associated with obesity and insulin resistance may have a detrimental impact on the efficacy of ET [4, 9, 10]. In this article, several hypotheses for this impaired efficacy have been explored, including the impaired regulation of aromatase in obesity as well as the role of PI3K, leptin, and FGFR1 signalling. Obesity and insulin resistance may also impact the altered toxicity profile of systemic cancer treatments.
In this paper, we consolidate the current understanding of the impact of obesity and diabetes on cancer risk, treatment outcomes, and toxicity in ER+ breast cancer.
Obesity, diabetes, breast cancer risk and outcome
In the last four decades, the rising incidence of breast cancer has been partly attributed to the introduction of national screening programmes worldwide, promoting the identification of small, early-stage tumours with favourable prognoses [11]. However, multiple epidemiological studies across diverse ethnic populations presented in Table 1, have associated the simultaneous rise in obesity levels with the increasing breast cancer incidence.
As body size and fat mass increase, endogenous oestrogen production is heightened, while sex hormone-binding globulin levels decrease. This hormonal imbalance is hypothesised to account for the link between obesity and an elevated risk of breast cancer in postmenopausal women [12, 13].
Although this link is well established in postmenopausal women, where increased androgen aromatisation in adipose tissue leads to higher oestrogen levels [14,15,16], the relationship between obesity and breast cancer risk in premenopausal women remains less clear. In this population, obesity is associated with a reduced incidence of breast cancer [14, 17]. For example, a large prospective multicentre analysis of over 700,000 premenopausal women showed that higher BMI during early adulthood is associated with a reduced risk of developing future breast cancer. This inverse association was stronger at younger ages and persisted across all BMI distributions, suggesting that increased adiposity early in life might have a protective effect against premenopausal breast cancer [18]. To explain this paradoxical risk reduction, it has been proposed that there is decreased oestrogen exposure in premenopausal obese women due to increased anovulatory menstrual cycles, a later decline in progesterone levels during menstruation, and longer menstruation [14,15,16, 19]. However, a longitudinal study did not find a clear relationship between BMI and ovulation-related variables like probable polycystic ovarian syndrome, oral contraceptives, and infertility secondary to an ovulatory disorder, which has cast doubt on this view [14, 18]. There is added complexity when considering findings from a pooled analysis of seven prospective studies, which investigated how circulating oestrogen and androgens affect premenopausal breast cancer risk. This study indicated that although total oestradiol levels were inversely related to BMI and positively associated with cancer risk, suggesting that the lower risk in obese women might stem from its reduced levels, free oestradiol, oestrone, and androgens such as DHEAS, testosterone, and free testosterone were found to be positively associated both with BMI and premenopausal breast cancer risk [20].
Aside from decreased oestrogen exposure, a net reduction in progesterone production is also hypothesised to account for decreased breast cancer risk in pre-menopause [21]. Progesterone is considered a major mitogen in the adult mammary epithelium in both mice and humans and has been linked to mammary carcinogenesis [22]. In the obese, premenopausal population, increased total oestrogen levels from adipose tissue and ovarian oestrogen production lead to enhanced negative feedback on hypothalamic pituitary-controlled gonadotropin release, therefore reducing ovarian steroid synthesis and progesterone production. Unlike postmenopausal women who produce no ovarian oestrogen [21], the oestrogen-progesterone imbalance in premenopausal obese women has been put forward as an explanation for the reduced breast cancer risk observed in this group.
In postmenopausal women, obesity has also been shown to increase breast cancer-related disease recurrence and mortality [7, 9, 23]. A meta-analysis of 82 follow-up studies demonstrated that breast cancer survivors with a higher BMI have worse overall and breast cancer-specific survival [7]. Overweight or obese breast cancer patients often present with larger tumours, higher-grade malignancy, and more positive lymph nodes at diagnosis. However, even after adjusting for these known prognostic factors, obesity independently raises the risk of distant metastases and breast cancer-related death [24].
Similarly, several meta-analyses in the last 20 years, as detailed in Table 2, have consistently shown that diabetes is associated with an increased incidence of breast cancer. It has been proposed that diabetes contributes to the onset of breast cancer via various mechanisms, such as mitogenic hyperinsulinaemia/insulin-like growth factor (IGF) pathway signalling, hyperglycaemia, inflammation caused by excess fat, and alterations in the levels of sex hormones [25]. These mechanisms are discussed further later in this article.
Furthermore, there is an approximate 40% increase in mortality following a breast cancer diagnosis among postmenopausal women with diabetes compared to women without diabetes. Nevertheless, this increase may, at least in part, be due to diabetes-related comorbidities [26]. Breast cancer-specific mortality also appears to be higher in diabetic women, although it is uncertain if mortality worsens with increasing severity of type 2 diabetes [27].
Genetic links and shared susceptibility in obesity, diabetes, and breast cancer
Various hypotheses have been proposed to explain the frequent co-occurrence of obesity, diabetes and breast cancer, with one of the most prominent being the shared genetic aetiology. Recent advancements in genetic research, particularly through large Genome-Wide Association Studies (GWAS), have revealed that specific genetic variants are associated with these complex diseases across different populations.
Several variants that are associated with T2DM have also been linked to breast cancer. Notable examples include polymorphisms mapping to loci at 10q25.2 and 9p21.3 at which transcription factor 7-like 2 (TCF7L2) and cyclin-dependent kinase inhibitor 2A/B (CDKN2A/B) have been proposed as the target genes and both of which are involved in signalling pathways that regulate cell-cycle progression and proliferation [28,29,30,31]. The first obesity susceptibility locus discovered by GWAS mapped to 16q12.2, proximal to the fat mass and obesity-associated (FTO) gene which has been recognised as a regulator in DNA repair mechanisms, DNA damage and inflammatory responses. Polymorphisms at this locus have also been associated with breast cancer risk [32,33,34,35,36]. Furthermore, the FTO-encoded protein, an RNA N6-methyladenosine (m6A) demethylase, has been implicated in breast tumourigenesis and progression [37, 38].
Recently, interest in the FTO gene has been renewed following a systematic analysis exploring the potential overlap of known GWAS risk variants for obesity, T2DM and breast cancer. This study identified 91 candidate variants in linkage disequilibrium using datasets from the 1000-Genomes Project to analyse candidate haplotypic blocks. Surprisingly, all variants were located within the vicinity of the FTO gene, thus highlighting the significant association of this locus with these diseases and strengthening the hypothesis of a shared genetic basis [39].
However, conflicting evidence from previous case–control studies in women of various ethnicities has questioned the potential pleiotropic effects of these risk variants on breast cancer, obesity and diabetes traits [40,41,42]. In addition, a case–control study involving U.S. Caucasian women found non-significant correlations between intronic and intergenic single nucleotide polymorphisms (SNPs) located in or near 29 diabetes-related genes and breast cancer incidence and mortality, casting further uncertainty on the functional significance of these variants in relation to breast cancer risk [43]. Finally, Mendelian randomisation analyses have been implemented to infer causality between genetic instruments associated with obesity and diabetes and breast cancer risk [44,45,46]. This method provides clearer insights into causal associations by reducing bias and confounding, as well as mitigating reverse causation. Nevertheless, further mechanistic studies will be required to elucidate the underlying biological pathways and interactions that drive these associations.
Mechanisms of obesity-induced carcinogenesis and treatment resistance in breast cancer
Breast cancer development in the context of obesity has been linked to increased inflammation in adipose tissue, marked by macrophage infiltration and the formation of crown-like structures (CLS) around dead adipocytes. This increased inflammation within the adipose tissue microenvironment has been shown in obese mouse models and is associated with increased cell proliferation and higher levels of inflammatory cytokines, including TNF-alpha, IL-1β and Cox-2, as well as insulin resistance [47,48,49]. In a study of women undergoing mastectomy or breast cancer surgery, CLS was detected in 40% of cases, and was associated with higher levels of insulin, glucose, leptin, triglycerides, C-reactive protein and IL-6 [50]. The presence of CLS in breast tissue is linked to an increased risk of breast cancer and a poorer prognosis, with evidence showing associations with metastasis and decreased overall survival [49, 51, 52]. Several mechanisms may contribute to CLS formation and resistance to ET. Understanding and selectively targeting these mechanisms could affect both breast cancer development and associated insulin resistance. A summary of the mechanisms of breast cancer carcinogenesis in obesity is described in Fig. 1.
RANKL/TNF-alpha/NF-κB activation
The accumulation of macrophages in CLS is thought to be caused by a decrease in macrophage apoptosis within obese adipose tissue through the activation of the transcription factor NF-κB [53]. NF-κB has been found to be activated in human breast cancer cell lines and is considered critical in the genesis of ET resistance in ER+ breast cancer, as it has been shown to promote tamoxifen resistance, early recurrence, metastasis, and worse overall survival [53,54,55,56]. There also appears to be cross-talk between ER and NF-κB, potentially working in tandem to support breast cancer cell survival and transition to a more aggressive phenotype [57]. The upregulation of NF-κB is independently associated with hyperinsulinemia, and reduced β-cell function [58].
Novel therapies inhibiting NF-κB gene activation could therefore potentially prevent ER+ tumour recurrence and restore endocrine responsiveness. Preclinical studies indicate that suppressing NF-κB significantly enhances the sensitivity of resistant breast cancer tumour cells to tamoxifen [59, 60]. Riggins et al. demonstrated that pharmacologic inhibition of NF-κB by parthenolide, a small molecule inhibitor against NF-κB, could restore fulvestrant-mediated suppression of growth in breast cancer cell lines [61]. Despite this preclinical data, clinical trials exploring NF-κB inhibition have not been promising to date. Three Phase II studies investigating bortezomib, a proteosome inhibitor that blocks the NF-κB pathway, as a single agent or in combination with ET, did not elicit an objective tumour response in metastatic breast cancer patients [62,63,64].
Targeting upstream or downstream signals of NF-κB may provide more promising therapeutic prospects. RANK ligand (RANKL), a TNF-related molecule, has been shown to activate NF-κB in preclinical studies and thereby promote proliferative changes in the mammary epithelium as well as epithelial-mesenchymal transition, which induces tumour cell migration, invasion and metastasis [65,66,67]. Systemic and hepatic blockage of RANKL signalling can also improve hepatic insulin sensitivity and glucose tolerance [58]. Currently in clinical practice, the use of the RANKL inhibitor denosumab is not extended beyond the prevention or treatment of osteoporosis, primarily due to disappointment on its efficacy in improving disease-free survival (DFS) in patients [68, 69]. However, the potential of RANKL inhibitors, particularly denosumab, to counteract NF-κB-mediated resistance in ER+ breast cancer may still merit further clinical investigation.
Similarly, TNF-alpha, a cytokine acting upstream of NF-κB, has been shown to induce proliferation in murine mammary tumour cells [70]. By upregulating PTEN and suppressing the AKT/eNOS/NO signalling pathway, TNF-alpha also contributes to vascular insulin resistance [71]. Infliximab, which binds to an neutralises TNF-alpha, was found to be tolerable in patients with advanced cancer with some evidence of on-target activity [72]. In metastatic breast cancer, a Phase II clinical trial demonstrated the safety of another anti-TNF-alpha agent, etanercept, in heavily pre-treated patients, although more research is required to understand efficacy and any treatment role [73]. TNF-alpha blockade may also have a role in overcoming resistance to anti-PD-1 therapy, and combination therapy should be assessed for feasibility [74].
Hypoxia and induction of hypoxia-inducible factor 1-alpha (HIF-1α)
The activation of NF-κB in obesity-related breast cancer may be driven by adipocyte hypoxia [75]. It is hypothesised that adipocyte hypertrophy without hyperplasia leads to accelerated tissue growth with insufficient supportive angiogenesis [76]. Hypoxia in turn triggers the activation of hypoxia-inducible factors (HIFs), which are associated with increased proliferation and expression of ER and VEGF, suggesting a possible relationship with more aggressive tumours [77]. HIF-1α expression is associated with poorly differentiated breast cancer, a higher pathological stage, and poor treatment response and outcome [77, 78]. Obesity is also associated with elevated HIF-1α mRNA and protein in adipose tissue [79], while HIF-1α activation in macrophages is associated with the development of insulin resistance and glucose metabolism in addition to pro-tumour mechanisms [80]. This may be in part due to HIF-1α-induced upregulation of insulin receptor substrate 2 (IRS-2), which is an important mediator of insulin, glucose metabolism, and mitogenesis [81]. PI3K and downstream signalling effectors AKT and mTOR are activated through the recruitment of the IRS proteins.
Hypoxia is also a recognised driver of ET resistance, with elevated expression of HIF-2α observed in endocrine-resistant ERα-positive breast cancer cell lines [78, 82]. Introducing HIF-2 into previously sensitive cells leads to their development of resistance to antioestrogens and inhibiting HIF-2α signalling can restore sensitivity in cells that have become resistant to ET [82]. Additionally, established HIF inhibitors such as digoxin and acriflavine appear to have activity against breast cancer metastatic niche formation [83], and have been shown to decelerate diet-induced obesity by various mechanisms in mouse studies, including decreasing lipogenesis [84,85,86]. Therefore, focusing on HIF inhibitors may not only help overcome resistance to ET in obesity but also provide valuable insights into preventing diet-induced obesity.
PI3K–AKT–mTOR pathway activation
PI3K–AKT–mTOR is a key signal transduction pathway that mediates cell growth, metabolism, and cell survival. PI3K–AKT–mTOR integrates upstream signals, including those from insulin and insulin growth factors (IGF-1 and IGF2) as well as cellular nutrients, energy and oxygen levels (Fig. 2). There is cross-talk between the PI3K–AKT–mTOR pathway and the oestrogen receptor (ER) pathway at multiple levels [87, 88].
Genetic alterations affecting different nodes of the PI3K–AKT–mTOR pathway are common in ER+ breast cancer [89]. The international data sharing consortium, AACR Project GENIE, showed that genetic alterations in PIK3CA, PTEN and AKT1 occur in ~36%, 7% and 5% of breast cancer, respectively [90]. It is not known whether these activating mutations are more likely to arise in breast cancer for patients that have insulin resistance or may influence the response to fasting in the context of ET.
Activation of the insulin receptor (IR) promotes downstream PI3K–AKT–mTOR signalling (see Fig. 2). Increased insulin levels are associated with higher breast cancer incidence and mortality [91]. Studies with both insulin analogues, blocking and stimulating anti-IR antibodies, and small molecule inhibitors, have shown a role for insulin signalling in breast cancer development and progression [92]. IR expression in breast cancer is well described, and high IR expression has been implicated in poor prognosis [93]. IR is more commonly expressed in endocrine-resistant breast cancer, and low expression correlates with improved survival [94].
Several preclinical studies have shown that the PI3K–AKT–mTOR pathway plays a key role in mediating resistance to ET in breast cancer, and the concept of targeting the PI3K–AKT–mTOR pathway to augment ET has now been proven in clinical settings. The BOLERO-2 Phase 3 trial demonstrated an improvement in median progression-free survival when the mTOR inhibitor everolimus was combined with the aromatase inhibitor exemestane in patients already refractory to single-agent aromatase inhibitor therapy [95]. The SOLAR-1 Phase 3 study has now also shown that the combination of the PI3K inhibitor, alpelisib, with fulvestrant led to an improvement in progression-free survival versus fulvestrant alone in PIK3CA-mutant, ER+ metastatic breast cancer resistant to first-line ET [96]. The Phase 3 placebo-controlled CAPltello-291 trial reported an improvement in progression-free survival with the addition of the AKT inhibitor, capivasertib to fulvestrant in patients with ER+ advanced breast cancer, irrespective of PIK3CA mutation status [97]. Lastly, the combination of inavolisib (a novel PI3K inhibitor) + palbociclib + fulvestrant in ER+ve metastatic breast cancer showed a significant improvement in investigator-assessed progression-free survival [98].
Drugs that lower circulating glucose and insulin levels, in particular metformin and SGLT2 inhibitors, have been proposed as treatments for breast cancer and could potentially synergise with ET by reducing PI3K–AKT–mTOR signalling (Fig. 2) [99, 100]. In particular, metformin has been extensively studied as a potential anticancer therapy, and a number of window-of-opportunity clinical trials have suggested that metformin may reduce cancer cell proliferation, and this effect may be greater in insulin-resistant women [100]. One meta-analysis of 11 observational studies has reported improved overall and cancer-specific survival in patients with breast cancer and diabetes who received metformin when compared with patients receiving other antidiabetic treatments [101]. However, another pharmacodynamic clinical study showed no clear link between metformin-induced reductions in circulating insulin levels and changes in tumour biology [102]. A large Phase 3 trial of 5 years of adjuvant metformin therapy in breast cancer showed no evidence of clinical benefit, although this study excluded patients with diabetes [103].
Aside from targeted therapies, dietary interventions have the potential to modulate the PI3K–AKT-mTOR pathway. Caffa et al. found that combining a periodic or fasting-mimicking diet (FMD) with hormone therapy, specifically fulvestrant and tamoxifen, enhanced anticancer effects in ER+ breast cancer mouse models by reducing leptin, insulin, and IGF-1 levels. Besides promoting sustained tumour regression, this approach could also revert acquired drug resistance [104].
Adipokine dysregulation and breast cancer
Investigating adipocyte biology, which goes beyond passive fat storage, is essential for comprehending the microenvironmental alterations linked to obesity. Adipocytes modulate the adipose tissue microenvironment through adipokine-mediated paracrine and autocrine signalling pathways. Two key adipokines involved in breast carcinogenesis are leptin and adiponectin.
Excess body fat increases leptin release from adipocytes, and BMI correlates with elevated leptin levels. By stimulating its receptor and a number of downstream pathways, including Jak2/Stat3, MAPK and PI3K–AKT, leptin likely promotes cell invasion and proliferation [104, 105]. A meta-analysis of 35 studies linked higher serum leptin levels with increased breast cancer risk, especially in postmenopausal women, suggesting its potential as a biomarker [106]. In addition, genetic variations in the leptin-coding genes LEP and ADIPOQ have been associated with elevated breast cancer risk [107]. Leptin has also been implicated in resistance mechanisms to tamoxifen and aromatase inhibitor treatment [108, 109].
Visceral adipose tissue (VAT) is known to produce leptin [50], but the effect of locally generated leptin from breast fat tissue compared to circulating leptin on breast tumour progression is not well understood.
On the other hand, low levels of adiponectin are associated with obesity and Type 2 diabetes, and studies suggest adiponectin may suppress cancer growth by modulating a number of intracellular metabolism and proliferation pathways associated with mitogenesis, including TNF-alpha, AMPK and SREBP-1 signalling [110, 111]. Unfavourable outcomes for breast cancer have been linked to both low adiponectin levels and increased leptin levels [112, 113], and it is speculated that the adiponectin:leptin ratio may be more important for breast cancer growth than the absolute levels [114].
Because high leptin levels are associated with an increased risk of breast cancer and may increase resistance to ET, as demonstrated in preclinical breast cancer models [115, 116], it has been speculated that lowering leptin levels through weight loss may improve outcomes for breast cancer survivors. Furthermore, a recent randomised study in this population demonstrated that both exercise and weight loss were associated with decreased leptin expression and improvements in the adipokine/leptin ratio [117], although whether this definitively translates to better clinical outcomes remains unanswered.
Obesity and FGF1, FGF2 and FGFR signalling
Another obesity-associated marker of elevated breast cancer risk, especially in the case of visceral fat, is fibroblast growth factor-2 (FGF2), which is released by adipose tissue. FGF2 binds to FGFR1 and FGFR2, and at least 10% of breast cancers harbour FGFR1 amplification, which is linked to early relapse and poor prognosis [118]. FGFR1 signalling directs healthy mammary duct development [119], and FGF2 levels are lowered in mice that have had a substantial fat pad removed, suggesting that FGF2 may have endocrine-mediated functions in addition to local ones [120]. Poor prognosis in breast cancer has been associated with elevated expression of the leptin receptor (LepR) and FGFR1 amplification, and co-expression of the FGFR1 gene and leptin protein copy number has been observed in primary breast tumours [121]. Antagonism of FGFR signalling in an obese mouse breast cancer model prevented outgrowth of pulmonary metastases [122] and FGFR inhibitors have already shown some promise in the clinic for the treatment of endocrine-resistant ER+ breast cancer [123].
In preclinical studies, elevated circulating levels of FGF2 have been linked to breast cancer development through the activation of oncogenic signalling pathways, including MAPK/ERK, cMYC and PI3K/AKT/mTOR. FGFR1 amplification is a key driver of ET resistance through MAPK signalling activation, and this therapeutic opportunity is currently being explored in clinical trials [124, 125]. Direct targeting of FGF2 is also being considered as a potential clinical approach [126].
FGF1 promotes adipocyte glucose uptake through AKT cross-talk as well as transcriptional promotion of glucose transporter protein type 1 (GLUT1), the primary glucose transporter [127,128,129]. GLUT1 is associated with higher grade, proliferation, as well as poorer prognosis in breast cancer [130, 131], although no link was observed between GLUT1 expression in breast cancer and background obesity or diabetes in one small study [132]. Notably, recent work has shown that FGF1 stimulates oestrogen receptor activation in obese mouse breast cancer models after oestrogen deprivation [128].
Aromatase overexpression in obesity
Aromatase inhibitors (AI) play a pivotal role in the treatment of ER+ breast cancer as a monotherapy in postmenopausal women. Postmenopausal status leads to a shift in the primary site of aromatase activity to the adipose tissue in the breast and gluteal areas. It is well described that AIs are less efficient at suppressing serum oestradiol levels in obese women [10]. A plausible explanation for this reduced efficiency is the observation that women with BMI >30, both with and without breast cancer, have elevated baseline oestrogen levels compared to those with BMI <22, and this may result in less effective suppression of oestrogen by an AI in postmenopausal women [133].
The formation of CLS in obesity is associated with heightened levels of gene transcription and increased activity of aromatase in mammary glands and visceral fat [51]. Aromatase expression is especially elevated in the adipose fibroblasts near breast tumours through the activation of proximal promoters, with immature fibroblasts primarily responsible for its production [134]. Furthermore, tumour cells in adipose tissue inhibit adipocyte differentiation by release of TNF-alpha and interleukin-11, thereby increasing the fibroblast:adipocyte ratio. This shift sustains elevated aromatase production, promoting local oestrogen synthesis and tumour progression [135].
A systematic review of three randomised controlled trials and five retrospective cohort studies suggested reduced efficacy of aromatase inhibitors in obesity, although the exact magnitude of this effect is not clearly established [136]. In a recent nationwide cohort study of 13,000 patients with hormone receptor-positive breast cancer, Harborg et al. showed that the risk of recurrence was higher among patients with obesity compared to those with a healthy weight (BMI 18.5–24.9) [4]. The ATAC study, which randomly assigned postmenopausal women with early-stage breast cancer to receive oral daily anastrozole alone, tamoxifen alone, or the combination, supported these findings. Specifically, women on anastrozole had a 27% lower recurrence rate compared to those taking tamoxifen, with women having a BMI <23 deriving an even greater benefit from treatment with an aromatase inhibitor [9].
In premenopausal women, AIs should be combined with ovarian suppression, typically with gonadotropin-releasing hormone (GnRh) analogues. Alternatively, selective oestrogen receptor modulators, such as tamoxifen, may be used alone or in combination with GnRh analogues. In premenopausal patients with HR-positive breast cancer who received adjuvant tamoxifen, a high BMI has been linked to a poorer prognosis [137]. However, to date, no similar association has been reported when ovarian suppression is used in conjunction with aromatase inhibitors.
AIs are usually provided at a standard dose that does not take specific inter-patient variation into consideration. Early studies investigating whether a larger dosage of AIs may improve outcomes for obese individuals with metastatic cancer suggested no additional benefits from an increased dose. However, these trials were conducted prior to the introduction of AIs as a standard-of-care option for postmenopausal ER+ breast cancer and therefore weight-dependent dosing may be revisited [138, 139].
Another issue demanding attention is the heightened insulin resistance and increased adiposity associated with postmenopausal women with breast cancer undergoing treatment with AIs. Studies in aromatase knockout mice and rare cases of congenital aromatase deficiency indicate a correlation with elevated adiposity, hepatic steatosis and insulin resistance [140,141,142]. Consequently, the advantages of ET must be weighed against the potential risks of obesity, metabolic syndrome and diabetes. This emphasizes the need for further investigation as to whether these changes in metabolism are associated with a worse prognosis and whether early drug or dietary intervention might be beneficial.
Implications for toxicity
It has long been recognised that there is a challenge in correctly dosing obese patients with adjuvant chemotherapy, as the maximum tolerated dose for cytotoxic therapy will have typically been determined in a leaner population. Obese patients are thought to be often underdosed due to empirical dose reductions, contrary to guidelines recommending full weight-based dosing. Although there is limited clinical data, it is hypothesised that this may lead to worse outcomes [143, 144]. There is also considerable uncertainty when using drugs with a high risk of cumulative toxicity, such as doxorubicin, fluoropyrimidines and cyclophosphamide. Studies are ongoing to determine whether better measures of body composition can more accurately predict toxicity in early breast cancer treatment [145].
Furthermore, studies have shown that obese patients with breast cancer receiving ET experience increased adverse effects, such as increased joint symptoms and cardiovascular events, which could potentially lead to treatment discontinuation [146, 147]. Hence, lack of treatment compliance in this context may be a contributing factor to poorer outcomes. Notably, in the context of treatment with adjuvant CDK4/6 inhibitors, obese patients had lower rates of neutropaenia which translated into a reduced treatment discontinuation rate in the PALLAS trial. The investigators hypothesised that obese patients may have a lower distributional volume, although survival data are still immature [148]. This suggests a need for dosage adjustments based on body composition rather than standard weight-based protocols to maximise therapeutic effects in obese patients.
The use of mTOR and PI3K inhibitors in conjunction with ET for the treatment of breast cancer may be especially problematic in patients with obesity or insulin resistance. Using clinical trial data from two studies of PI3K inhibitors, Rodon et al. developed a risk prediction model for grade 3/4 hyperglycaemia, and identified five factors, including baseline fasting plasma glucose, HbA1c and BMI, as the strongest predictors for classifying patients as low or high risk [149]. Notably, preclinical research has shown that the insulin feedback causing hyperglycaemia can be prevented using dietary or pharmaceutical approaches, which greatly enhance the efficacy of treatment [150].
Conclusion
For some time, it has been understood that obesity and insulin resistance are associated with both an increased risk of developing ER+ breast cancer and poorer outcomes. Substantial preclinical evaluation has now provided greater insight into the mechanisms that drive these phenomena, and potential therapeutic strategies have been proposed. Clinical studies of interventions aimed at improving outcomes for breast cancer patients with metabolic disorders are warranted. More accurate measures of body composition beyond BMI and their association with patient outcome need to be assessed in the clinic and potential differences in treatment resistance between premenopausal and postmenopausal women in the context of obesity remain understudied. Lastly, the breast cancer community needs to evaluate strategies to effectively manage treatment toxicity in the context of obesity.
References
Arnold M, Morgan E, Rumgay H, Mafra A, Singh D, Laversanne M, et al. Current and future burden of breast cancer: Global statistics for 2020 and 2040. Breast. 2022;66:15–23.
Mili N, Paschou SA, Goulis DG, Dimopoulos MA, Lambrinoudaki I, Psaltopoulou T. Obesity, metabolic syndrome, and cancer: pathophysiological and therapeutic associations. Endocrine. 2021;74:478–97.
Munsell MF, Sprague BL, Berry DA, Chisholm G, Trentham-Dietz A. Body mass index and breast cancer risk according to postmenopausal estrogen-progestin use and hormone receptor status. Epidemiol Rev. 2014;36:114–36.
Harborg S, Cronin-Fenton D, Jensen MR, Ahern TP, Ewertz M, Borgquist S. Obesity and risk of recurrence in patients with breast cancer treated with aromatase inhibitors. JAMA Netw Open. 2023;6:e2337780.
Anderson GL, Neuhouser ML. Obesity and the risk for premenopausal and postmenopausal breast cancer. Cancer Prev Res. 2012;5:515–21.
Cecchini RS, Costantino JP, Cauley JA, Cronin WM, Wickerham DL, Land SR, et al. Body mass index and the risk for developing invasive breast cancer among high-risk women in NSABP P-1 and STAR breast cancer prevention trials. Cancer Prev Res. 2012;5:583–92.
Chan DSM, Vieira AR, Aune D, Bandera EV, Greenwood DC, McTiernan A, et al. Body mass index and survival in women with breast cancer-systematic literature review and meta-analysis of 82 follow-up studies. Ann Oncol. 2014;25:1901–14.
Kang C, LeRoith D, Gallagher EJ. Diabetes, obesity, and breast cancer. Endocrinology. 2018;159:3801–12.
Sestak I, Distler W, Forbes JF, Dowsett M, Howell A, Cuzick J. Effect of body mass index on recurrences in tamoxifen and anastrozole treated women: an exploratory analysis from the ATAC trial. J Clin Oncol. 2010;28:3411–5.
Pfeiler G, Königsberg R, Hadji P, Fitzal F, Maroske M, Dressel-Ban G, et al. Impact of body mass index on estradiol depletion by aromatase inhibitors in postmenopausal women with early breast cancer. Br J Cancer. 2013;109:1522–7.
Bleyer A, Welch HG. Effect of three decades of screening mammography on breast-cancer incidence. New Engl J Med. 2012;367:1998–2005.
Key T, Appleby P, Barnes I, Reeves G, Hormones E, Group BCC. Endogenous sex hormones and breast cancer in postmenopausal women: reanalysis of nine prospective studies. J Natl Cancer Inst. 2002;94:606–16.
Baglietto L, English DR, Hopper JL, MacInnis RJ, Morris HA, Tilley WD, et al. Circulating steroid hormone concentrations in postmenopausal women in relation to body size and composition. Breast Cancer Res Treat. 2009;115:171–9.
Michels KB, Terry KL, Willett WC. Longitudinal study on the role of body size in premenopausal breast cancer. Arch Intern Med. 2006;166:2395–402.
Baer HJ, Colditz GA, Rosner B. Body fatness during childhood and adolescence and incidence of breast cancer in premenopausal women: a prospective cohort study. Breast Cancer Res. 2005;7:R314.
Dehesh T, Fadaghi S, Seyedi M. The relation between obesity and breast cancer risk in women by considering menstruation status and geographical variations: a systematic review and meta-analysis. BMC Women’s Health. 2023;23:392.
Tran T, Chang Y, Choi H, Kwon R, Lim G, Kim E, et al. Adiposity, body composition measures, and breast cancer risk in Korean premenopausal women. JAMA Netw Open. 2024;7:e245423.
Group PBCC, Schoemaker MJ, Nichols HB, Wright LB, Brook MN, Jones ME, et al. Association of body mass index and age with subsequent breast cancer risk in premenopausal women. JAMA Oncol. 2018;4:e181771.
Key TJ, Pike MC. The role of oestrogens and progestagens in the epidemiology and prevention of breast cancer. Eur J Cancer Clin Oncol. 1988;24:29–43.
Hormones, Group BCC E, Key TJ, Appleby PN, Reeves GK, Travis RC, et al. Sex hormones and risk of breast cancer in premenopausal women: a collaborative reanalysis of individual participant data from seven prospective studies. Lancet Oncol. 2013;14:1009–19.
Dowsett M, Folkerd E. Reduced progesterone levels explain the reduced risk of breast cancer in obese premenopausal women: a new hypothesis. Breast Cancer Res Treat. 2015;149:1–4.
Brisken C. Progesterone signalling in breast cancer: a neglected hormone coming into the limelight. Nat Rev Cancer. 2013;13:385–96.
Petrelli F, Cortellini A, Indini A, Tomasello G, Ghidini M, Nigro O, et al. Association of obesity with survival outcomes in patients with cancer: a systematic review and meta-analysis. JAMA Netw Open. 2021;4:e213520.
Ewertz M, Jensen MB, Gunnarsdóttir KÁ, Højris I, Jakobsen EH, Nielsen D, et al. Effect of obesity on prognosis after early-stage breast cancer. J Clin Oncol. 2011;29:25–31.
Xiong F, Wang J, Nierenberg JL, Van Blarigan EL, Kenfield SA, Chan JM, et al. Diabetes mellitus and risk of breast cancer: a large-scale, prospective, population-based study. Br J Cancer. 2023;129:648–55.
Lipscombe LL, Goodwin PJ, Zinman B. The impact of diabetes on survival following breast cancer. Breast Cancer Res Treat. 2008;109:389–95.
Lee KN, Torres MA, Troeschel AN, He J, Gogineni K, McCullough LE. Type 2 diabetes, breast cancer specific and overall mortality: associations by metformin use and modification by race, body mass, and estrogen receptor status. PLoS ONE. 2020;15:e0232581.
Grarup N, Rose CS, Andersson EA, Andersen G, Nielsen AL, Albrechtsen A, et al. Studies of association of variants near the HHEX, CDKN2A/B, and IGF2BP2 genes with type 2 diabetes and impaired insulin release in 10,705 Danish subjects: validation and extension of genome-wide association studies. Diabetes. 2007;56:3105–11.
ShahidSales S, Mehramiz M, Ghasemi F, Aledavood A, Shamsi M, Hassanian SM, et al. A genetic variant in CDKN2A/B gene is associated with the increased risk of breast cancer. J Clin Lab Anal. 2018;32:e22190.
Burwinkel B, Shanmugam KS, Hemminki K, Meindl A, Schmutzler RK, Sutter C, et al. Transcription factor 7-like 2 (TCF7L2) variant is associated with familial breast cancer risk: a case-control study. BMC Cancer. 2006;6:268.
Zhang M, Tang M, Fang Y, Cui H, Chen S, Li J, et al. Cumulative evidence for relationships between multiple variants in the VTI1A and TCF7L2 genes and cancer incidence. Int J Cancer. 2018;142:498–513.
Frayling TM, Timpson NJ, Weedon MN, Zeggini E, Freathy RM, Lindgren CM, et al. A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science. 2007;316:889–94.
Liu W, Yasui M, Sassa A, You X, Wan J, Cao Y, et al. FTO regulates the DNA damage response via effects on cell-cycle progression. Mutat Res Genet Toxicol Environ Mutagen. 2023;887:503608.
Xu Y, Ye S, Zhang N, Zheng S, Liu H, Zhou K, et al. The FTO/miR-181b-3p/ARL5B signaling pathway regulates cell migration and invasion in breast cancer. Cancer Commun. 2020;40:484–500.
Kaklamani V, Yi N, Sadim M, Siziopikou K, Zhang K, Xu Y, et al. The role of the fat mass and obesity associated gene (FTO) in breast cancer risk. BMC Med Genet. 2011;12:52.
Gholamalizadeh M, Jarrahi AM, Akbari ME, Bourbour F, Mokhtari Z, Salahshoornezhad S, et al. Association between FTO gene polymorphisms and breast cancer: the role of estrogen. Expert Rev Endocrinol Metab. 2020;15:115–21.
Jeschke J, Collignon E, Al Wardi C, Krayem M, Bizet M, Jia Y, et al. Downregulation of the FTO m6A RNA demethylase promotes EMT-mediated progression of epithelial tumors and sensitivity to Wnt inhibitors. Nat Cancer. 2021;2:611–28.
Niu Y, Lin Z, Wan A, Chen H, Liang H, Sun L, et al. RNA N6-methyladenosine demethylase FTO promotes breast tumor progression through inhibiting BNIP3. Mol Cancer. 2019;18:46.
Gholami M. FTO is a major genetic link between breast cancer, obesity, and diabetes. Breast Cancer Res Treat. 2024;204:159–69.
Chen F, Wilkens LR, Monroe KR, Stram DO, Kolonel LN, Henderson BE, et al. No association of risk variants for diabetes and obesity with breast cancer: the Multiethnic Cohort and PAGE studies. Cancer Epidemiol Biomark Prev. 2011;20:1039–42.
Hou N, Zheng Y, Gamazon ER, Ogundiran TO, Adebamowo C, Nathanson KL, et al. Genetic susceptibility to type 2 diabetes and breast cancer risk in women of European and African ancestry. Cancer Epidemiol Biomark Prev. 2012;21:552–6.
Zhao Z, Wen W, Michailidou K, Bolla MK, Wang Q, Zhang B, et al. Association of genetic susceptibility variants for type 2 diabetes with breast cancer risk in women of European ancestry. Cancer Causes Control. 2016;27:679–93.
Parada HJ, Cleveland RJ, North KE, Stevens J, Teitelbaum SL, Neugut AI, et al. Genetic polymorphisms of diabetes-related genes, their interaction with diabetes status, and breast cancer incidence and mortality: the Long Island Breast Cancer Study Project. Mol Carcinog. 2019;58:436–46.
Shu X, Wu L, Khankari NK, Shu XO, Wang TJ, Michailidou K, et al. Associations of obesity and circulating insulin and glucose with breast cancer risk: a Mendelian randomization analysis. Int J Epidemiol. 2019;48:795–806.
Guo Y, Warren Andersen S, Shu XO, Michailidou K, Bolla MK, Wang Q, et al. Genetically predicted body mass index and breast cancer risk: Mendelian randomization analyses of data from 145,000 women of European descent. PLoS Med. 2016;13:e1002105.
Au Yeung SL, Schooling CM. Impact of glycemic traits, type 2 diabetes and metformin use on breast and prostate cancer risk: a Mendelian randomization study. BMJ Open Diabetes Res Care. 2019;7:e000872.
Harris BHL, Macaulay VM, Harris DA, Klenerman P, Karpe F, Lord SR, et al. Obesity: a perfect storm for carcinogenesis. Cancer Metastasis Rev. 2022;41:491–515.
Savva C, Copson E, Johnson PWM, Cutress RI, Beers SA. Obesity is associated with immunometabolic changes in adipose tissue that may drive treatment resistance in breast cancer: immune-metabolic reprogramming and novel therapeutic strategies. Cancers. 2023;15:2440.
Chang MC, Eslami Z, Ennis M, Goodwin PJ. Crown-like structures in breast adipose tissue of breast cancer patients: associations with CD68 expression, obesity, metabolic factors and prognosis. NPJ Breast Cancer. 2021;7:97.
Iyengar NM, Zhou XK, Gucalp A, Morris PG, Howe LR, Giri DD, et al. Systemic correlates of white adipose tissue inflammation in early-stage breast cancer. Clin Cancer Res. 2016;22:2283–9.
Morris PG, Hudis CA, Giri D, Morrow M, Falcone DJ, Zhou XK, et al. Inflammation and increased aromatase expression occur in the breast tissue of obese women with breast cancer. Cancer Prev Res. 2011;4:1021–9.
Birts CN, Savva C, Laversin SA, Lefas A, Krishnan J, Schapira A, et al. Prognostic significance of crown-like structures to trastuzumab response in patients with primary invasive HER2 + breast carcinoma. Sci Rep. 2022;12:7802.
Hill AA, Anderson-Baucum EK, Kennedy AJ, Webb CD, Yull FE, Hasty AH. Activation of NF-κB drives the enhanced survival of adipose tissue macrophages in an obesogenic environment. Mol Metab. 2015;4:665–77.
Kastrati I, Joosten SEP, Semina SE, Alejo LH, Brovkovych SD, Stender JD, et al. The NF-κB pathway promotes tamoxifen tolerance and disease recurrence in estrogen receptor-positive breast cancers. Mol Cancer Res: MCR. 2020;18:1018–27.
Zhou Y, Yau C, Gray JW, Chew K, Dairkee SH, Moore DH, et al. Enhanced NF kappa B and AP-1 transcriptional activity associated with antiestrogen resistant breast cancer. BMC Cancer. 2007;7:59.
Wu JT, Kral JG. The NF-kappaB/IkappaB signaling system: a molecular target in breast cancer therapy. J Surg Res. 2005;123:158–69.
Frasor J, Weaver A, Pradhan M, Dai Y, Miller LD, Lin CY, et al. Positive cross-talk between estrogen receptor and NF-kappaB in breast cancer. Cancer Res. 2009;69:8918–25.
Lê KA, Mahurkar S, Alderete TL, Hasson RE, Adam TC, Kim JS, et al. Subcutaneous adipose tissue macrophage infiltration is associated with hepatic and visceral fat deposition, hyperinsulinemia, and stimulation of NF-κB stress pathway. Diabetes. 2011;60:2802–9.
deGraffenried LA, Chandrasekar B, Friedrichs WE, Donzis E, Silva J, Hidalgo M, et al. NF-kappa B inhibition markedly enhances sensitivity of resistant breast cancer tumor cells to tamoxifen. Ann Oncol. 2004;15:885–90.
de Groot AF, Appelman-Dijkstra NM, van der Burg SH, Kroep JR. The anti-tumor effect of RANKL inhibition in malignant solid tumors—a systematic review. Cancer Treat Rev. 2018;62:18–28.
Riggins RB, Zwart A, Nehra R, Clarke R. The nuclear factor kappa B inhibitor parthenolide restores ICI 182,780 (Faslodex; fulvestrant)-induced apoptosis in antiestrogen-resistant breast cancer cells. Mol Cancer Ther. 2005;4:33–41.
Trinh XB, Sas L, Van Laere SJ, Prové A, Deleu I, Rasschaert M, et al. A phase II study of the combination of endocrine treatment and bortezomib in patients with endocrine-resistant metastatic breast cancer. Oncol Rep. 2012;27:657–63.
Engel RH, Brown JA, Von Roenn JH, O’Regan RM, Bergan R, Badve S, et al. A phase II study of single agent bortezomib in patients with metastatic breast cancer: a single institution experience. Cancer Investig. 2007;25:733–7.
Yang CH, Gonzalez-Angulo AM, Reuben JM, Booser DJ, Pusztai L, Krishnamurthy S, et al. Bortezomib (VELCADE) in metastatic breast cancer: pharmacodynamics, biological effects, and prediction of clinical benefits. Ann Oncol. 2006;17:813–7.
Gonzalez-Suarez E, Jacob AP, Jones J, Miller R, Roudier-Meyer MP, Erwert R, et al. RANK ligand mediates progestin-induced mammary epithelial proliferation and carcinogenesis. Nature. 2010;468:103–7.
Schramek D, Leibbrandt A, Sigl V, Kenner L, Pospisilik JA, Lee HJ, et al. Osteoclast differentiation factor RANKL controls development of progestin-driven mammary cancer. Nature. 2010;468:98–102.
Tsubaki M, Komai M, Fujimoto S, Itoh T, Imano M, Sakamoto K, et al. Activation of NF-κB by the RANKL/RANK system up-regulates snail and twist expressions and induces epithelial-to-mesenchymal transition in mammary tumor cell lines. J Exp Clin Cancer Res: CR. 2013;32:62.
Gnant M, Pfeiler G, Steger GG, Egle D, Greil R, Fitzal F, et al. Adjuvant denosumab in postmenopausal patients with hormone receptor-positive breast cancer (ABCSG-18): disease-free survival results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2019;20:339–51.
Coleman R, Finkelstein DM, Barrios C, Martin M, Iwata H, Hegg R, et al. Adjuvant denosumab in early breast cancer (D-CARE): an international, multicentre, randomised, controlled, phase 3 trial. Lancet Oncol. 2020;21:60–72.
Rivas MA, Carnevale RP, Proietti CJ, Rosemblit C, Beguelin W, Salatino M, et al. TNF alpha acting on TNFR1 promotes breast cancer growth via p42/P44 MAPK, JNK, Akt and NF-kappa B-dependent pathways. Exp Cell Res. 2008;314:509–29.
da Costa RM, Neves KB, Mestriner FL, Louzada-Junior P, Bruder-Nascimento T, Tostes RC. TNF-α induces vascular insulin resistance via positive modulation of PTEN and decreased Akt/eNOS/NO signaling in high fat diet-fed mice. Cardiovasc Diabetol. 2016;15:119.
Brown ER, Charles KA, Hoare SA, Rye RL, Jodrell DI, Aird RE, et al. A clinical study assessing the tolerability and biological effects of infliximab, a TNF-alpha inhibitor, in patients with advanced cancer. Ann Oncol. 2008;19:1340–6.
Madhusudan S, Foster M, Muthuramalingam SR, Braybrooke JP, Wilner S, Kaur K, et al. A phase II study of etanercept (Enbrel), a tumor necrosis factor alpha inhibitor in patients with metastatic breast cancer. Clin Cancer Res. 2004;10:6528–34.
Bertrand F, Montfort A, Marcheteau E, Imbert C, Gilhodes J, Filleron T, et al. TNFα blockade overcomes resistance to anti-PD-1 in experimental melanoma. Nat Commun. 2017;8:2256.
D’Ignazio L, Rocha S. Hypoxia Induced NF-κB. Cells. 2016;5:10.
Anvari G, Bellas E. Hypoxia induces stress fiber formation in adipocytes in the early stage of obesity. Sci Rep. 2021;11:21473.
Bos R, Zhong H, Hanrahan CF, Mommers EC, Semenza GL, Pinedo HM, et al. Levels of hypoxia-inducible factor-1 alpha during breast carcinogenesis. J Natl Cancer Inst. 2001;93:309–14.
Generali D, Berruti A, Brizzi MP, Campo L, Bonardi S, Wigfield S, et al. Hypoxia-inducible factor-1alpha expression predicts a poor response to primary chemoendocrine therapy and disease-free survival in primary human breast cancer. Clin Cancer Res. 2006;12:4562–8.
He Q, Gao Z, Yin J, Zhang J, Yun Z, Ye J. Regulation of HIF-1{alpha} activity in adipose tissue by obesity-associated factors: adipogenesis, insulin, and hypoxia. Am J Physiol Endocrinol Metab. 2011;300:E877–85.
Takikawa A, Mahmood A, Nawaz A, Kado T, Okabe K, Yamamoto S, et al. HIF-1α in myeloid cells promotes adipose tissue remodeling toward insulin resistance. Diabetes. 2016;65:3649–59.
Lero MW, Shaw LM. Diversity of insulin and IGF signaling in breast cancer: implications for therapy. Mol Cell Endocrinol. 2021;527:111213.
Alam MW, Persson CU, Reinbothe S, Kazi JU, Rönnstrand L, Wigerup C, et al. HIF2α contributes to antiestrogen resistance via positive bilateral crosstalk with EGFR in breast cancer cells. Oncotarget. 2016;7:11238–50.
Wong CC, Zhang H, Gilkes DM, Chen J, Wei H, Chaturvedi P, et al. Inhibitors of hypoxia-inducible factor 1 block breast cancer metastatic niche formation and lung metastasis. J Mol Med. 2012;90:803–15.
Sun K, Halberg N, Khan M, Magalang UJ, Scherer PE. Selective inhibition of hypoxia-inducible factor 1α ameliorates adipose tissue dysfunction. Mol Cell Biol. 2013;33:904–17.
Tang YA, Chen YF, Bao Y, Mahara S, Yatim SMJM, Oguz G, et al. Hypoxic tumor microenvironment activates GLI2 via HIF-1α and TGF-β2 to promote chemoresistance in colorectal cancer. Proc Natl Acad Sci USA. 2018;115:E5990–E9.
Shin MK, Drager LF, Yao Q, Bevans-Fonti S, Yoo DY, Jun JC, et al. Metabolic consequences of high-fat diet are attenuated by suppression of HIF-1α. PLoS ONE. 2012;7:e46562.
Bostner J, Karlsson E, Pandiyan MJ, Westman H, Skoog L, Fornander T, et al. Activation of Akt, mTOR, and the estrogen receptor as a signature to predict tamoxifen treatment benefit. Breast Cancer Res Treat. 2013;137:397–406.
Hasson SP, Rubinek T, Ryvo L, Wolf I. Endocrine resistance in breast cancer: focus on the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin signaling pathway. Breast Care. 2013;8:248–55.
Campbell RA, Bhat-Nakshatri P, Patel NM, Constantinidou D, Ali S, Nakshatri H. Phosphatidylinositol 3-kinase/AKT-mediated activation of estrogen receptor alpha: a new model for anti-estrogen resistance. J Biol Chem. 2001;276:9817–24.
Consortium APG. AACR Project GENIE: powering precision medicine through an international consortium. Cancer Discov. 2017;7:818–31.
Goodwin PJ, Ennis M, Pritchard KI, Trudeau ME, Koo J, Madarnas Y, et al. Fasting insulin and outcome in early-stage breast cancer: results of a prospective cohort study. J Clin Oncol. 2002;20:42–51.
Hopkins BD, Goncalves MD, Cantley LC. Insulin-PI3K signalling: an evolutionarily insulated metabolic driver of cancer. Nat Rev Endocrinol. 2020;16:276–83.
Mathieu MC, Clark GM, Allred DC, Goldfine ID, Vigneri R. Insulin receptor expression and clinical outcome in node-negative breast cancer. Proc Assoc Am Physicians. 1997;109:565–71.
Gradishar WJ, Yardley DA, Layman R, Sparano JA, Chuang E, Northfelt DW, et al. Clinical and translational results of a phase II, randomized trial of an anti-IGF-1R (cixutumumab) in women with breast cancer that progressed on endocrine therapy. Clin Cancer Res. 2016;22:301–9.
Baselga J, Campone M, Piccart M, Burris HA 3rd, Rugo HS, et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. New Engl J Med. 2012;366:520–9.
Andre F, Ciruelos E, Rubovszky G, Campone M, Loibl S, Rugo HS, et al. Alpelisib for PIK3CA-mutated, hormone receptor-positive advanced breast cancer. New Engl J Med. 2019;380:1929–40.
Turner NC, Oliveira M, Howell SJ, Dalenc F, Cortes J, Gomez Moreno HL, et al. Capivasertib in hormone receptor-positive advanced breast cancer. New Engl J Med. 2023;388:2058–70.
Juric D, Kalinsky K, Turner NC, Jhaveri KL, Schmid P, Loi S, et al. First-line inavolisib/placebo + palbociclib + fulvestrant (Inavo/Pbo+Palbo+Fulv) in patients with PIK3CA-mutated, hormone receptor-positive, HER2-negative locally advanced/metastatic breast cancer who relapsed during/within 12 months of adjuvant endocrine therapy completion: INAVO120 phase III randomized trial additional analyses. J Clin Oncol. 2024;42:1003.
Dutka M, Bobiński R, Francuz T, Garczorz W, Zimmer K, Ilczak T, et al. SGLT-2 inhibitors in cancer treatment-mechanisms of action and emerging new perspectives. Cancers. 2022;14:5811.
Lord SR, Harris AL. Is it still worth pursuing the repurposing of metformin as a cancer therapeutic? Br J Cancer. 2023;128:958–66.
Xu H, Chen K, Jia X, Tian Y, Dai Y, Li D, et al. Metformin use is associated with better survival of breast cancer patients with diabetes: a meta-analysis. Oncologist. 2015;20:1236–44.
Lord SR, Cheng WC, Liu D, Gaude E, Haider S, Metcalf T, et al. Integrated pharmacodynamic analysis identifies two metabolic adaption pathways to metformin in breast cancer. Cell Metab. 2018;28:679–88.e4.
Goodwin PJ, Chen BE, Gelmon KA, Whelan TJ, Ennis M, Lemieux J, et al. Effect of metformin vs placebo on invasive disease-free survival in patients with breast cancer: the MA.32 randomized clinical trial. J Am Med Assoc. 2022;327:1963–73.
Caffa I, Spagnolo V, Vernieri C, Valdemarin F, Becherini P, Wei M, et al. Author Correction: Fasting-mimicking diet and hormone therapy induce breast cancer regression. Nature. 2020;588:E33.
Vona-Davis L, Rose DP. Adipokines as endocrine, paracrine, and autocrine factors in breast cancer risk and progression. Endocr Relat Cancer. 2007;14:189–206.
Pan H, Deng LL, Cui JQ, Shi L, Yang YC, Luo JH, et al. Association between serum leptin levels and breast cancer risk: an updated systematic review and meta-analysis. Medicine. 2018;97:e11345.
Peng WZ, Liu X, Li CF, Zhao J. Genetic alterations in LEP and ADIPOQ genes and risk for breast cancer: a meta-analysis. Front Oncol. 2023;13:1125189.
Chen X, Zha X, Chen W, Zhu T, Qiu J, Røe OD, et al. Leptin attenuates the anti-estrogen effect of tamoxifen in breast cancer. Biomed Pharmacother = Biomed Pharmacother. 2013;67:22–30.
Gelsomino L, Giordano C, Camera G, Sisci D, Marsico S, Campana A, et al. Leptin signaling contributes to aromatase inhibitor resistant breast cancer cell growth and activation of macrophages. Biomolecules. 2020;10:543.
Grossmann ME, Nkhata KJ, Mizuno NK, Ray A, Cleary MP. Effects of adiponectin on breast cancer cell growth and signaling. Br J Cancer. 2008;98:370–9.
Ziemke F, Mantzoros CS. Adiponectin in insulin resistance: lessons from translational research. Am J Clin Nutr. 2010;91:258S–61S.
Duggan C, Irwin ML, Xiao L, Henderson KD, Smith AW, Baumgartner RN, et al. Associations of insulin resistance and adiponectin with mortality in women with breast cancer. J Clin Oncol. 2011;29:32–9.
Goodwin PJ, Ennis M, Pritchard KI, Trudeau ME, Koo J, Taylor SK, et al. Insulin- and obesity-related variables in early-stage breast cancer: correlations and time course of prognostic associations. J Clin Oncol. 2012;30:164–71.
Cleary MP, Ray A, Rogozina OP, Dogan S, Grossmann ME. Targeting the adiponectin:leptin ratio for postmenopausal breast cancer prevention. Front Biosci. 2009;1:329–57.
Caffa I, Spagnolo V, Vernieri C, Valdemarin F, Becherini P, Wei M, et al. Fasting-mimicking diet and hormone therapy induce breast cancer regression. Nature. 2020;583:620–4.
Garofalo C, Sisci D, Surmacz E. Leptin interferes with the effects of the antiestrogen ICI 182,780 in MCF-7 breast cancer cells. Clin Cancer Res. 2004;10:6466–75.
Lin D, Sturgeon KM, Gordon BR, Brown JC, Sears DD, Sarwer DB, et al. WISER survivor trial: combined effect of exercise and weight loss interventions on adiponectin and leptin levels in breast cancer survivors with overweight or obesity. Nutrients. 2023;15:3453.
Benham V, Chakraborty D, Bullard B, Bernard JJ. A role for FGF2 in visceral adiposity-associated mammary epithelial transformation. Adipocyte. 2018;7:113–20.
Zhang X, Martinez D, Koledova Z, Qiao G, Streuli CH, Lu P. FGF ligands of the postnatal mammary stroma regulate distinct aspects of epithelial morphogenesis. Development. 2014;141:3352–62.
Chakraborty D, Benham V, Bullard B, Kearney T, Hsia HC, Gibbon D, et al. Fibroblast growth factor receptor is a mechanistic link between visceral adiposity and cancer. Oncogene. 2017;36:6668–79.
Boothby-Shoemaker W, Benham V, Paithankar S, Shankar R, Chen B, Bernard JJ. The relationship between leptin, the leptin receptor and FGFR1 in primary human breast tumors. Cells. 2020;9:2224.
Kulkoyluoglu Cotul E, Safdar MH, Paez SJ, Kulkarni A, Ayers MG, Lin H, et al. FGFR1 signaling facilitates obesity-driven pulmonary outgrowth in metastatic breast cancer. Mol Cancer Res. 2024;22:254–67.
Coombes RC, Badman PD, Lozano-Kuehne JP, Liu X, Macpherson IR, Zubairi I, et al. Results of the phase IIa RADICAL trial of the FGFR inhibitor AZD4547 in endocrine resistant breast cancer. Nat Commun. 2022;13:3246.
Turner N, Pearson A, Sharpe R, Lambros M, Geyer F, Lopez-Garcia MA, et al. FGFR1 amplification drives endocrine therapy resistance and is a therapeutic target in breast cancer. Cancer Res. 2010;70:2085–94.
Saridogan T, Akcakanat A, Zhao M. Efficacy of futibatinib, an irreversible fibroblast growth factor receptor inhibitor, in FGFR-altered breast cancer. Sci Rep. 2023;13:20223.
Gordon-Weeks AN, Lim SY, Yuzhalin AE, Jones K, Markelc B, Kim KJ, et al. Neutrophils promote hepatic metastasis growth through fibroblast growth factor 2-dependent angiogenesis in mice. Hepatology. 2017;65:1920–35.
Sankofi BM, Valencia-Rincón E, Sekhri M, Ponton-Almodovar AL, Bernard JJ, Wellberg EA. The impact of poor metabolic health on aggressive breast cancer: adipose tissue and tumor metabolism. Front Endocrinol. 2023;14:1217875.
Castillo-Castrejon M, Sankofi BM, Murguia SJ, Udeme AA, Cen HH, Xia YH, et al. FGF1 supports glycolytic metabolism through the estrogen receptor in endocrine-resistant and obesity-associated breast cancer. Breast Cancer Res. 2023;25:99.
Nies VJM, Struik D, Liu S, Liu W, Kruit JK, Downes M, et al. Autocrine FGF1 signaling promotes glucose uptake in adipocytes. Proc Natl Acad Sci USA. 2022;119:e2122382119.
Oh S, Kim H, Nam K, Shin I. Glut1 promotes cell proliferation, migration and invasion by regulating epidermal growth factor receptor and integrin signaling in triple-negative breast cancer cells. BMB Rep. 2017;50:132–7.
Barbosa AM, Martel F. Targeting glucose transporters for breast cancer therapy: the effect of natural and synthetic compounds. Cancers. 2020;12:154.
Suteau V, Bukasa-Kakamba J, Virjogh-Cenciu B, Adenis A, Sabbah N, Drak Alsibai K. Pathological significance of GLUT-1 expression in breast cancer cells in diabetic and obese patients: the French Guiana Study. Cancers. 2022;14:437.
McTiernan A, Rajan K, Tworoger S, Irwin M, Bernstein L, Baumgartner R, et al. Adiposity and sex hormones in postmenopausal breast cancer survivors. J Clin Oncol. 2003;21:1961–6.
Zhou J, Gurates B, Yang S, Sebastian S, Bulun S. Malignant breast epithelial cells stimulate aromatase expression via promoter II in human adipose fibroblasts: an epithelial-stromal interaction in breast tumors mediated by CCAAT/enhancer binding protein beta. Cancer Res. 2001;61:2328–34.
Meng L, Zhou J, Sasano H, Suzuki T, Zeitoun K, Bulun S. Tumor necrosis factor alpha and interleukin 11 secreted by malignant breast epithelial cells inhibit adipocyte differentiation by selectively down-regulating CCAAT/enhancer binding protein alpha and peroxisome proliferator-activated receptor gamma: mechanism of desmoplastic reaction. Cancer Res. 2001;61:2250–5.
Ioannides SJ, Barlow PL, Elwood JM, Porter D. Effect of obesity on aromatase inhibitor efficacy in postmenopausal, hormone receptor-positive breast cancer: a systematic review. Breast Cancer Res Treat. 2014;147:237–48.
Ozaki Y, Masuda J, Kataoka A, Kogawa T, Abe T, Morizono H, et al. The impact of obesity and endocrine therapy on the prognosis of premenopausal women with hormone receptor-positive breast cancer: a single-institute retrospective study. Cancer Rep. 2023;6:e1695.
Jonat W, Howell A, Blomqvist C, Eiermann W, Winblad G, Tyrrell C, et al. A randomised trial comparing two doses of the new selective aromatase inhibitor anastrozole (Arimidex) with megestrol acetate in postmenopausal patients with advanced breast cancer. Eur J Cancer. 1996;32A:404–12.
Buzdar A, Jonat W, Howell A, Plourde P. ARIMIDEX: a potent and selective aromatase inhibitor for the treatment of advanced breast cancer. J Steroid Biochem Mol Biol. 1997;61:145–9.
Gibb FW, Dixon JM, Clarke C, Homer NZ, Faqehi AMM, Andrew R, et al. Higher insulin resistance and adiposity in postmenopausal women with breast cancer treated with aromatase inhibitors. J Clin Endocrinol Metab. 2019;104:3670–8.
Takeda K, Toda K, Saibara T, Nakagawa M, Saika K, Onishi T, et al. Progressive development of insulin resistance phenotype in male mice with complete aromatase (CYP19) deficiency. J Endocrinol. 2003;176:237–46.
Hewitt KN, Pratis K, Jones ME, Simpson ER. Estrogen replacement reverses the hepatic steatosis phenotype in the male aromatase knockout mouse. Endocrinology. 2004;145:1842–8.
Carroll J, Protani M, Walpole E, Martin J. Effect of obesity on toxicity in women treated with adjuvant chemotherapy for early-stage breast cancer: a systematic review. Breast Cancer Res Treat. 2012;136:323–30.
Griggs J, Bohlke K, Balaban E, Dignam J, Hall E, Harvey R. Appropriate systemic therapy dosing for obese adult patients with cancer: ASCO guideline update. J Clin Oncol. 2021;39:2037–48.
Durkin K, Heetun A, Ewings S, Munday R, Wootton SA, Turner L, et al. Body composition and chemotherapy toxicity in women with early breast cancer (CANDO-3): protocol for an observational cohort study. BMJ Open. 2022;12:e054412.
Sestak I, Cuzick J, Sapunar F, Eastell R, Forbes J, Bianco A, et al. Risk factors for joint symptoms in patients enrolled in the ATAC trial: a retrospective, exploratory analysis. Lancet Oncol. 2008;9:866–72.
Boszkiewicz K, Piwowar A, Petryszyn P. Aromatase inhibitors and risk of metabolic and cardiovascular adverse effects in breast cancer patients—a systematic review and meta-analysis. J Clin Med. 2022;11:3133.
Pfeiler G, Hlauschek D, Mayer E, Deutschmann C, Kacerovsky-Strobl S, Martin M. Impact of BMI in patients with early hormone receptor-positive breast cancer receiving endocrine therapy with or without palbociclib in the PALLAS trial. J Clin Oncol. 2023;41:5118–30.
Rodón J, Demanse D, Rugo H, Burris H, Simó R, Farooki A. A risk analysis of alpelisib-induced hyperglycemia in patients with advanced solid tumors and breast cancer. Breast Cancer Res. 2024;26:36.
Hopkins BD, Pauli C, Du X, Wang DG, Li X, Wu D, et al. Suppression of insulin feedback enhances the efficacy of PI3K inhibitors. Nature. 2018;560:499–503.
Park J, Han K, Shin D, Yeo Y, Chang J, Yoo J, et al. Obesity and breast cancer risk for pre- and postmenopausal women among over 6 million Korean women. Breast Cancer Res Treat. 2021;185:495–506.
Chen M, Wu W, Yen A, Fann J, Chen S, Chiu S, et al. Body mass index and breast cancer: analysis of a nation-wide population-based prospective cohort study on 1,393,985 Taiwanese women. Int J Obes. 2016;40:524–30.
Reeves GK, Pirie K, Beral V, Green J, Spencer E, Bull D, et al. Cancer incidence and mortality in relation to body mass index in the Million Women Study: cohort study. BMJ. 2007;335:1134.
Yang TO, Cairns BJ, Pirie K, Green J, Beral V, Floud S, et al. Body size in early life and the risk of postmenopausal breast cancer. BMC Cancer. 2022;22:232.
Lahmann P, Hoffmann K, Allen N, van Gils C, Khaw K, Tehard B, et al. Body size and breast cancer risk: findings from the European Prospective Investigation into Cancer And Nutrition (EPIC). Int J Cancer. 2004;111:762–71.
da Silva MWE, Licaj I, Lissner L, Rylander C. Excess body weight, weight gain and obesity-related cancer risk in women in Norway: the Norwegian Women and Cancer study. Br J Cancer. 2018;119:646–56.
Ahn J, Schatzkin A, Lacey JV Jr, Albanes D, Ballard-Barbash R. Adiposity, adult weight change, and postmenopausal breast cancer risk. Arch Intern Med. 2007;167:2091–102.
Morimoto L, White E, Chen Z, Chlebowski R, Hays J, Kuller L, et al. Obesity, body size, and risk of postmenopausal breast cancer: the Women’s Health Initiative (United States). Cancer Causes Control. 2002;13:741–51.
Suzuki R, Rylander-Rudqvist T, Ye W, Saji S, Wolk A. Body weight and postmenopausal breast cancer risk defined by estrogen and progesterone receptor status among Swedish women: a prospective cohort study. Int J Cancer. 2006;119:1683–9.
White A, Nichols H, Bradshaw P, Sandler D. Overall and central adiposity and breast cancer risk in the Sister Study. Cancer. 2015;121:3700–8.
Suzuki S, Kojima M, Tokudome S, Mori M, Sakauchi F, Wakai K, et al. Obesity/weight gain and breast cancer risk: findings from the Japan collaborative cohort study for the evaluation of cancer risk. J Epidemiol. 2013;23:139–45.
Gathirua-Mwangi WG, Palmer JR, Champion V, Castro-Webb N, Stokes AC, Adams-Campbell L, et al. Maximum and time-dependent body mass index and breast cancer incidence among postmenopausal women in the Black Women’s Health Study. Am J Epidemiol. 2022;191:646–54.
Xiong F, Dai Q, Zhang S, Bent S, Tahir P, Van Blarigan EL, et al. Diabetes and incidence of breast cancer and its molecular subtypes: a systematic review and meta-analysis. Diabetes Metab Res Rev. 2024;40:e3709.
Lu Y, Hajjar A, Cryns V, Trentham-Dietz A, Gangnon R, Heckman-Stoddard B, et al. Breast cancer risk for women with diabetes and the impact of metformin: a meta-analysis. Cancer Med. 2023;12:11703–18.
Hardefeldt P, Edirimanne S, Eslick G. Diabetes increases the risk of breast cancer: a meta-analysis. Endocr Relat Cancer. 2012;19:793–803.
Boyle P, Boniol M, Koechlin A, Robertson C, Valentini F, Coppens K, et al. Diabetes and breast cancer risk: a meta-analysis. Br J Cancer. 2012;107:1608–17.
Liao S, Li J, Wei W, Wang L, Zhang Y, Li J, et al. Association between diabetes mellitus and breast cancer risk: a meta-analysis of the literature. Asian Pac J Cancer Prev. 2011;12:1061–5.
Larsson SC, Mantzoros CS, Wolk A. Diabetes mellitus and risk of breast cancer: a meta-analysis. Int J Cancer. 2007;121:856–62.
Funding
SL holds project grants from the NIHR, Cancer Research UK, the World Cancer Research Fund and a fellowship from Against Breast Cancer supporting his work.
Author information
Authors and Affiliations
Contributions
SRJ, AS, MZZ and SL wrote and edited the manuscript. AS designed the figures and legends (Figures created with BioRender.com).
Corresponding author
Ethics declarations
Competing interests
SL—Honoraria: Eisai, Prosigna, Roche, Pfizer, Novartis, Sanofi. Advisory: Shionogi, Sanofi, GLG Consulting: Rejuversen, Oxford Biodynamics. Research grant funding: CRUK, NIHR, Against Breast Cancer, Pathios Therapeutics. Travel/Accommodation/ Expenses: Pfizer, Roche, Synthon, Piqur Therapeutics. Stock holding: Mitox Therapeutics. Previous employment: Pfizer. My institution has received funding for clinical trials for which I am chief investigator or principal investigator from: Cancer Research UK, Boehringer Ingelheim, Piqur Therapeutics, AstraZeneca, Carrick Therapeutics, Sanofi, Merck KGaA, Synthon, Roche, Exscientia, BioInvent, RS Oncology. The remaining authors declare no competing interests.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Javed, S.R., Skolariki, A., Zameer, M.Z. et al. Implications of obesity and insulin resistance for the treatment of oestrogen receptor-positive breast cancer. Br J Cancer (2024). https://doi.org/10.1038/s41416-024-02833-1
Received:
Revised:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41416-024-02833-1