Obesity represents one of the most significant public health challenges worldwide. Current clinical practice relies on body mass index (BMI) to define the obesity status of an individual, even though the index has long been recognized for its limitations as a measure of body fat. In normal BMI individuals, increased central adiposity has been associated with worse health outcomes, including increased risks of cardiovascular disease and metabolic disorders. The condition leading to these outcomes has been described as metabolic obesity in the normal weight (MONW). More recent evidence suggests that MONW is associated with increased risk of several obesity-related malignancies, including postmenopausal breast, endometrial, colorectal, and liver cancers. In MONW patients, the false reassurance of a normal range BMI can lead to lost opportunities for implementing preventive interventions that may benefit a substantial number of people. A growing body of literature has documented the increased risk profile of MONW individuals and demonstrated practical uses for body composition and biochemical analyses to identify this at-risk population. In this review, we survey the current literature on MONW and cancer, summarize pathophysiology and oncogenic mechanisms, highlight potential strategies for diagnosis and treatment, and suggest directions for future research.

Obesity is a well-documented risk factor for a wide variety of medical conditions. Although body mass index (BMI) (calculated as weight (kg)/height; m2) is used to diagnose obesity, it is widely recognized to be a flawed indicator of an individual's health status (1). A growing body of literature has identified subgroups of normal BMI individuals (18.5–24.9 kg/m2 for non-Asians and 18.5–22.9 kg/m2 for Asians) who suffer from negative health consequences similar to those seen in the phenotypically overtly obese (≥30 kg/m2 for non-Asians and ≥27.5 kg/m2 for Asians; ref. 2). The condition, known as metabolic obesity in the normal weight (MONW), has been associated with excess body fat, dyslipidemia, insulin resistance, elevated blood pressure, and low-grade inflammation (3–5). In addition to higher risks of cardiovascular disease (CVD) and metabolic disorders, growing evidence has shown that MONW individuals also face increased risks of developing several cancers typically associated with obesity (6–9).

Despite these serious health implications, MONW remains under-recognized clinically. Research studies have employed different combinations of anthropometric and laboratory parameters to describe MONW. The absence of a common definition makes measuring the true prevalence of MONW difficult, with findings ranging from under 1% to over 30% of individuals with a normal BMI (10, 11). Variations in prevalence can also be attributed to differences in body composition patterns across ethnicities. Although much MONW research has been conducted in Asian populations, that are known to have more body fat at a lower BMI than other ethnicities (12, 13), studies have revealed similar patterns in body composition and clinical profiles among MONW individuals from European, Latin American, and North American populations (11, 14–17). As such, MONW should be considered a public health problem on a global scale. The failure to recognize it and develop effective interventions represents a missed opportunity for reducing the risk of potentially life-threatening diseases.

In this review, we examine the literature on MONW and its impact on cancer risk, pathophysiology, diagnostic approaches, and intervention strategies (Fig. 1). Given the range of definitions used in MONW research, our review includes studies with criteria based on body composition only, cardiometabolic abnormalities only, and combinations of both. We focus on the emerging evidence around MONW's association with cancer, an area of active investigation not yet addressed by existing reviews. By synthesizing current knowledge, we aim to identify challenges in the diagnosis and treatment of MONW and propose directions for future research.

Figure 1.

Metabolic obesity in the normal weight and its clinical significance. Abbreviations: BMI, body mass index; MHNW, metabolically healthy normal weight; MONW, metabolically obese normal weight; OW/OB, overweight/obese; TG, triglyceride; LDL-C, low-density lipoprotein cholesterol; HDL-C, high-density lipoprotein cholesterol; CRP, C-reactive protein; SHBG, sex hormone binding globulin; DXA, dual-energy X-ray absorptiometry; BIA, bioelectrical impedance analysis.

Figure 1.

Metabolic obesity in the normal weight and its clinical significance. Abbreviations: BMI, body mass index; MHNW, metabolically healthy normal weight; MONW, metabolically obese normal weight; OW/OB, overweight/obese; TG, triglyceride; LDL-C, low-density lipoprotein cholesterol; HDL-C, high-density lipoprotein cholesterol; CRP, C-reactive protein; SHBG, sex hormone binding globulin; DXA, dual-energy X-ray absorptiometry; BIA, bioelectrical impedance analysis.

Close modal

MONW and cancer risk

Obesity is a risk factor for the development of 13 types of cancer and is associated with higher risk of mortality from many of these cancers (18, 19). Evidence from Mendelian randomization (MR) studies on obesity and cancer, which employ known genetic predictors of risk factors to seek evidence consistent with causal effect, has demonstrated significant associations between higher BMI and a number of obesity-related cancers (20). However, because most studies in this field have relied on BMI for defining obesity, the existing evidence has largely highlighted increased cancer risk in those with BMIs ≥ 30 kg/m2. In individuals with normal BMI, increased adiposity has been associated with risk factors such as insulin resistance, dyslipidemia, and elevated inflammatory markers (11, 14, 15). In MONW populations, elevated cancer risk and increased all-cancer and all-cause mortality rates have been observed (Table 1; refs. 21–23).

Table 1.

MONW and associated cancer risks.

Cohort
Cancer TypeNameCharacteristicsMONW ParametersAdiposity MeasuresCancer risk, MONW vs. MHNWReference
Breast cancer, postmenopausal Women's Health Initiative (USA) 3,460 NW F BMI 18.5–24.9 kg/m2 Upper quartiles total fat mass (>22 kg), trunk fat mass (>9.3 kg) DXA Q4 vs. Q1 Iyengar et al. (2019) 
     Total fat mass [HR = 1.89 (1.21–2.95)]  
     Trunk fat mass [HR = 1.88 (1.18–2.98)]  
 The Sister Study (USA) 43,599 F (16619 NW) BMI < 25 kg/m2 ≥ 1 metabolic abnormality WC, WHR ≥1 metabolic abnormality [HR = 1.26 (1.01–1.56)] Park et al. (2017) 
     WC ≥ 88 cm [HR = 1.58 (1.02–2.46)]  
 UK Biobank (UK) 149,928 NW (52,729 analyzed) BMI 18.5–24.9 kg/m2, BIA adiposity measures, WC, WHR BIA, WC, WHR Q5 vs. Q1 Arthur et al. (2020) 
     FMI [HR = 1.46 (1.20–1.78)]; TFMI [HR = 1.57 (1.20–1.80)];  
     WC [HR = 1.32 (1.09–1.60)]  
 Women's Health Initiative (USA) 3,327 F BMI 18.5–24.9 kg/m2 Upper quartiles fasting insulin Upper quartiles HOMA-IR – Q3+4 vs. Q1 Gunter et al. (2015) 
     Fasting insulin [HR = 2.06 (1.01–4.22)]  
     HOMA-IR [HR = 1.80 (0.88–3.70)]  
Breast cancer Nigerian Breast Cancer Study (Nigeria) 2,334 F (1,209 NW) BMI < 25 kg/m2 WHR top quartile (> 0.87) WC, WHR WHR Q4 vs. Q1 [OR = 2.81 (1.90–4.16)] Ogundiran et al. (2012) 
     WC > 81 cm [OR = 1.91 (1.39–2.61)]  
Endometrial cancer Alberta Cancer Registry (Canada) 1,477 F (406 NW) BMI < 25 kg/m2, MetS WC WC > 88 cm [OR = 5.09 (1.04–24.95)] Friedenreich et al. (2011) 
 UK Biobank (UK) 149,928 NW (77,738 analyzed) BMI 18.5–24.9 kg/m2, BIA adiposity measures, WC, WHR BIA, WC, WHR Trunk fat mass index Q5 vs. Q1 [HR = 1.72 (1.02–2.89)] Arthur et al. (2020) 
Colorectal cancer EPIC Study (Europe) 1,474 (790 M, 684 F; 523 NW) BMI < 25 kg/m2 C-peptide top tertile (>4.74 ng/mL) WC C-peptide T3: CRC [HR = 1.59 (1.10–2.28)]; Murphy et al. (2016) 
     Colon [HR = 1.49 (0.92–2.43)]; Rectal [HR = 1.82 (1.02–3.23)]  
     WC ≥ 80 cm F or ≥ 94 cm M:  
     CRC [HR = 1.35 (0.95–1.91)]; Colon [HR = 1.18 (0.74–1.88)];  
     Rectal [HR = 1.76 (1.01–3.05)]  
 Women's Health Initiative (USA) 21,170 F (5,175 NW) BMI 18.5–< 25 kg/m2, MetS WC CRC [HR = 1.65 (0.99–2.74)] Kabat et al. (2018) 
     Colon [HR = 1.79 (1.02–3.12)]  
 UK Biobank (UK) 149,928 NW (149,928 analyzed) BMI 18.5–24.9 kg/m2, BIA adiposity measures, WC, WHR BIA, WC, WHR Men only: Arthur et al. (2020) 
     CRC, trunk to leg fat mass Q5 vs. Q1 [HR = 1.63 (1.14–2.32)]  
     Colon, trunk to leg fat mass Q5 vs. Q1 [HR = 1.92 (1.20–3.07)]  
     Colon, WC Q5 vs. Q1 [HR = 1.68 (1.05–2.66)]  
     No significant associations among women  
Primary liver cancer (HCC and ICC) Liver Cancer Pooling Project (North America) 1,167,244 (707,281 F, 457,755 M; 338,070 NW) BMI 18.5–< 25 kg/m2 WC, WHR WC WC per 5-cm increase [HR = 1.14 (1.07–1.21)] Florio et al. (2020) 
Prostate cancer National Health Check-ups Database (Korea) 11,771,252 M (7,391,410 NW) BMI < 25 kg/m2, MetS WC HR = 1.14 (1.12–1.17) Kim et al. (2019) 
Cohort
Cancer TypeNameCharacteristicsMONW ParametersAdiposity MeasuresCancer risk, MONW vs. MHNWReference
Breast cancer, postmenopausal Women's Health Initiative (USA) 3,460 NW F BMI 18.5–24.9 kg/m2 Upper quartiles total fat mass (>22 kg), trunk fat mass (>9.3 kg) DXA Q4 vs. Q1 Iyengar et al. (2019) 
     Total fat mass [HR = 1.89 (1.21–2.95)]  
     Trunk fat mass [HR = 1.88 (1.18–2.98)]  
 The Sister Study (USA) 43,599 F (16619 NW) BMI < 25 kg/m2 ≥ 1 metabolic abnormality WC, WHR ≥1 metabolic abnormality [HR = 1.26 (1.01–1.56)] Park et al. (2017) 
     WC ≥ 88 cm [HR = 1.58 (1.02–2.46)]  
 UK Biobank (UK) 149,928 NW (52,729 analyzed) BMI 18.5–24.9 kg/m2, BIA adiposity measures, WC, WHR BIA, WC, WHR Q5 vs. Q1 Arthur et al. (2020) 
     FMI [HR = 1.46 (1.20–1.78)]; TFMI [HR = 1.57 (1.20–1.80)];  
     WC [HR = 1.32 (1.09–1.60)]  
 Women's Health Initiative (USA) 3,327 F BMI 18.5–24.9 kg/m2 Upper quartiles fasting insulin Upper quartiles HOMA-IR – Q3+4 vs. Q1 Gunter et al. (2015) 
     Fasting insulin [HR = 2.06 (1.01–4.22)]  
     HOMA-IR [HR = 1.80 (0.88–3.70)]  
Breast cancer Nigerian Breast Cancer Study (Nigeria) 2,334 F (1,209 NW) BMI < 25 kg/m2 WHR top quartile (> 0.87) WC, WHR WHR Q4 vs. Q1 [OR = 2.81 (1.90–4.16)] Ogundiran et al. (2012) 
     WC > 81 cm [OR = 1.91 (1.39–2.61)]  
Endometrial cancer Alberta Cancer Registry (Canada) 1,477 F (406 NW) BMI < 25 kg/m2, MetS WC WC > 88 cm [OR = 5.09 (1.04–24.95)] Friedenreich et al. (2011) 
 UK Biobank (UK) 149,928 NW (77,738 analyzed) BMI 18.5–24.9 kg/m2, BIA adiposity measures, WC, WHR BIA, WC, WHR Trunk fat mass index Q5 vs. Q1 [HR = 1.72 (1.02–2.89)] Arthur et al. (2020) 
Colorectal cancer EPIC Study (Europe) 1,474 (790 M, 684 F; 523 NW) BMI < 25 kg/m2 C-peptide top tertile (>4.74 ng/mL) WC C-peptide T3: CRC [HR = 1.59 (1.10–2.28)]; Murphy et al. (2016) 
     Colon [HR = 1.49 (0.92–2.43)]; Rectal [HR = 1.82 (1.02–3.23)]  
     WC ≥ 80 cm F or ≥ 94 cm M:  
     CRC [HR = 1.35 (0.95–1.91)]; Colon [HR = 1.18 (0.74–1.88)];  
     Rectal [HR = 1.76 (1.01–3.05)]  
 Women's Health Initiative (USA) 21,170 F (5,175 NW) BMI 18.5–< 25 kg/m2, MetS WC CRC [HR = 1.65 (0.99–2.74)] Kabat et al. (2018) 
     Colon [HR = 1.79 (1.02–3.12)]  
 UK Biobank (UK) 149,928 NW (149,928 analyzed) BMI 18.5–24.9 kg/m2, BIA adiposity measures, WC, WHR BIA, WC, WHR Men only: Arthur et al. (2020) 
     CRC, trunk to leg fat mass Q5 vs. Q1 [HR = 1.63 (1.14–2.32)]  
     Colon, trunk to leg fat mass Q5 vs. Q1 [HR = 1.92 (1.20–3.07)]  
     Colon, WC Q5 vs. Q1 [HR = 1.68 (1.05–2.66)]  
     No significant associations among women  
Primary liver cancer (HCC and ICC) Liver Cancer Pooling Project (North America) 1,167,244 (707,281 F, 457,755 M; 338,070 NW) BMI 18.5–< 25 kg/m2 WC, WHR WC WC per 5-cm increase [HR = 1.14 (1.07–1.21)] Florio et al. (2020) 
Prostate cancer National Health Check-ups Database (Korea) 11,771,252 M (7,391,410 NW) BMI < 25 kg/m2, MetS WC HR = 1.14 (1.12–1.17) Kim et al. (2019) 

Abbreviations: MONW, metabolic obesity in the normal weight; MHNW, metabolically healthy normal weight; HCC, hepatocellular cancer; ICC, intrahepatic cholangiocarcinoma; EPIC, European Prospective Investigation into Cancer and Nutrition; NW, normal weight; F, female; M, male; BMI, body mass index; BIA, bioelectrical impedance analysis; WC, waist circumference; WHR, waist-to-hip ratio; HOMA-IR, homeostasis model assessment of insulin resistance; MetS, metabolic syndrome; DXA, dual energy X-ray absorptiometry; FMI, fat mass index; TFMI, trunk fat mass index; CRC, colorectal cancer.

Breast and endometrial cancers

A growing body of cancer research has focused on breast cancer risk in MONW women. Obesity increases the risk of estrogen receptor–positive (ER+) breast cancer in postmenopausal women and is associated with higher rates of relapse and worse survival outcomes (19, 24). Interestingly, MR studies have shown genetically predicted BMI to be inversely associated with breast cancer risk regardless of menopausal status, a surprising finding that contradicts robust evidence from observational studies linking obesity to increased risk of postmenopausal ER+ breast cancer. It is thought that genetically predicted BMI does not adequately account for the contribution of environmental and lifestyle factors to weight gain later in life, resulting in increased BMI and higher breast cancer risk among older women (20, 25).

Among normal weight women, elevated risks for breast cancer have been reported in association with excess body fat and metabolic abnormalities. Specifically, increased central adiposity has been associated with increased risk of postmenopausal breast cancer across ethnicities, although body composition measurement techniques and criteria for analysis have differed between studies (Table 1). Most have relied on operator-dependent techniques such as waist circumference (WC), waist-to-hip ratio (WHR), and skinfold thickness to approximate body composition (26–28). Consistent with these findings, a study of bioelectrical impedance analysis (BIA) of normal BMI UK Biobank subjects found that total body and trunk fat mass indices were positively associated with postmenopausal breast cancer risk (29). In another recent study, using dual-energy X-ray absorptiometry (DXA) to objectively quantify fat mass in a cohort of postmenopausal women with normal BMI, Iyengar and colleagues found that whole-body and trunk fat mass in the upper two quartiles were associated with significantly elevated risks for invasive ER+ breast cancer (7). Whether MONW patients are at increased risk for other subtypes of breast cancer remains to be investigated. Increased trunk fat in these groups was also associated with elevated levels of insulin and triglycerides (TG), and lower high-density lipoprotein cholesterol (HDL-C), reflecting the presence of metabolic dysfunction in these women (7). Research has also shown that, in the absence of documented excess body fat, normal weight women with one or more cardiometabolic abnormalities associated with metabolic syndrome (MetS) had a higher risk of developing postmenopausal breast cancer (28). These findings are consistent with an earlier study that demonstrated increased postmenopausal breast cancer risk among normal-BMI women with relatively elevated fasting insulin levels, while those with lower fasting insulin were not found to have increased risk regardless of weight status (30). Whether MONW also raises the risk of premenopausal breast cancer is less clear, with some studies reporting significant associations (26, 27, 31) while others have not (28). Consistent with the link between MONW and ER+ breast cancer, there is also emerging evidence of an increased risk of endometrial cancer, another hormonally driven cancer (29, 32).

Colorectal cancer

Observational and MR studies have demonstrated that many malignancies associated with obesity are of gastrointestinal origin, including colorectal, esophageal, liver, and pancreatic cancers, but limited research has been undertaken to understand them in the context of MONW (20, 33). Of these, colorectal cancer has been the best characterized in the MONW population (Table 1). While MONW was found to be associated with increased colorectal cancer risk in an analysis encompassing both men and women combined (8), a study using the Women's Health Initiative reported no significant associations among women (34). Another recent study reported positive associations between certain measures of central adiposity and risk of colorectal and colon cancer among men, but not among women (29). The evidence demonstrates an association between MONW and increased risk of colorectal cancer, with strong suggestion of sex dependence such that the association is stronger for males than females, and more specifically for colon than rectal cancer (29). Findings of sex differences have also been reported in the literature on obesity and colorectal cancer, with a stronger association between increased BMI and colorectal cancer reported in men than women (35).

Other malignancies

Although MONW has been found to increase the risk of nonalcoholic fatty liver disease and non-alcoholic steatohepatitis (36), investigation of its association with liver cancer, an obesity-associated malignancy, has been limited (37). One study found that central obesity, but not gluteofemoral size, conferred an increased risk of primary liver cancer in both overweight and normal weight individuals (38). MONW has also been shown to be associated with an increased risk of prostate cancer (9).

Associations between MONW and other obesity-related cancers have remained largely unexamined. Further research is needed to determine whether MONW is associated with higher risks of additional cancer types, including hematologic malignancies, and whether it affects clinical outcomes.

Pathogenesis of MONW

Similar to that of obesity, the pathogenesis of MONW is rooted in the accumulation of excess body fat, leading to a state of metabolic dysregulation and chronic subclinical inflammation that is characterized by abnormal lipidomic, endocrinologic, and immune pathways (6, 7, 39). The physiology of adipose tissue is complex. White adipose tissue (WAT), which acts as a primary site for fat storage, exists underneath the skin as subcutaneous adipose tissue (SAT) or surrounding organs within the abdominal cavity as visceral adipose tissue (VAT). The amount and distribution of these distinct fat depots have substantial clinical implications, as increased VAT confers significantly greater cardiometabolic risk than SAT (40). The observation that some MONW individuals develop metabolic abnormalities without excessive weight gain suggests that different patterns of adipogenesis may underlie MONW pathophysiology in some individuals, leading to the onset of metabolic dysfunction at a lower BMI than observed in their obese counterparts.

Both genetic predisposition and lifestyle choices have been correlated with increased body fat percentages (BF%). The search for the genetic underpinnings of metabolic obesity has uncovered several genetic loci that appear to be implicated in MONW (41). On the basis of genome-wide association study (GWAS) findings, Yaghootkar and colleagues analyzed genetic risk scores for insulin resistance and identified 11 risk alleles in association with abnormal metabolic profiles in conjunction with normal BMI, a lipodystrophy-like phenotype seen in MONW (42, 43). This phenotype is characterized by higher VAT to SAT ratio, increased hepatic steatosis, elevated levels of TG and alanine transaminase, and decreased HDL-C, sex hormone binding globulin (SHBG), and adiponectin (42). The cluster includes common genetic variants involving genes such as IRS1, known to increase the risk of diabetes and CVD despite decreased body fat and SAT, and PPARG, a crucial regulator of adipocyte differentiation (42). Clinically, these risk alleles are associated with lower BMI but higher risks for cardiometabolic diseases, including type II diabetes mellitus, coronary artery disease, hypertension, and carotid plaque burden (42, 44).

Behavioral factors like lower physical activity levels have been associated with MONW (29, 45, 46). A limited number of studies that examined associations between dietary patterns and MONW have yielded inconsistent findings. Some studies have reported higher carbohydrate and red meat intake and lower fruit and vegetable consumption in MONW individuals compared with healthy controls (29, 47). However, others have reported no significant dietary differences (45, 48). These inconsistent findings likely reflect the challenge of capturing nuanced differences in eating habits given a limited sample size. Notably, many studies have reported small but significantly higher BMI in MONW groups compared with healthy normal BMI subjects (6, 45, 46, 48).

Mechanisms of carcinogenesis

Growing evidence from the obesity literature has uncovered complex oncogenic mechanisms in the setting of increased adiposity. Such mechanisms are likely applicable to MONW given its overlapping pathophysiology with obesity. Adipose tissue dysfunction and metabolic dysregulation, present in both obesity and MONW, are thought to stimulate carcinogenesis through both local and systemic effects (49, 50). While a detailed review of biological mechanisms is beyond the scope of the present article, we provide below an overview of important factors to highlight the ways in which adiposity contributes to the development and progression of cancer (Fig. 2).

Figure 2.

Local and systemic effects of metabolic obesity and mechanisms for cancer promotion. Abbreviations: CLS, crown-like structures; MCP-1, monocyte chemoattractant protein-1; SHBG, sex hormone binding globulin; CRP, C-reactive protein; IGF-1, insulin-like growth factor-1; IGFBP, insulin-like growth factor binding protein; FA, fatty acids.

Figure 2.

Local and systemic effects of metabolic obesity and mechanisms for cancer promotion. Abbreviations: CLS, crown-like structures; MCP-1, monocyte chemoattractant protein-1; SHBG, sex hormone binding globulin; CRP, C-reactive protein; IGF-1, insulin-like growth factor-1; IGFBP, insulin-like growth factor binding protein; FA, fatty acids.

Close modal

Insulin signaling

Insulin resistance is a characteristic feature of metabolic obesity. The resulting elevated levels of circulating insulin are associated with increased cancer risk, including breast, endometrial, colon, and urinary tract cancers (49, 51). MR analyses have further demonstrated genetically predicted higher fasting insulin levels to be associated with increased breast and endometrial cancer risk (25, 52). Insulin exerts mitogenic effects through a number of direct and indirect mechanisms. In addition to binding directly to the insulin receptor, it also increases the availability of insulin-like growth factor (IGF)-1 through upregulation of its synthesis and downregulation of IGF binding protein production (53). Binding of insulin and IGF-1 to their cell surface receptors, which have been shown to be overexpressed in human cancer cells, leads to activation of PI3K/Akt and Ras–Raf–MAPK, pathways known to regulate cell proliferation and play important roles in tumorigenesis and cancer progression (53–55). Insulin and IGF-1 have been shown to induce VEGF, an important mediator of angiogenesis (56). In addition, insulin has been shown to increase glucose oxidation and cell division in cell models of obesity-related tumors (colon, breast, and prostate cancers), while the same effects did not occur in tumors not associated with obesity (57). Elevated insulin levels also reduce hepatic SHBG synthesis, resulting in increased circulating levels of free sex steroid hormones that may additionally predispose to the development of hormone-dependent cancers (51).

Circulating adipokines

Higher leptin and lower adiponectin levels have been observed in metabolically obese individuals (49). Clinically, elevated leptin levels have been found by many studies to be associated with increased risks of postmenopausal breast, endometrial, and colorectal cancers, while others have demonstrated no significant associations (49, 51). Mechanistically, leptin has been shown to exert mitogenic and antiapoptotic effects via activation of the PI3K/Akt, MAPK/ERK, and JAK/STAT pathways in multiple cancer cell models, including breast, endometrial, colon, and androgen-independent prostate cancer (58–61). Leptin also induces VEGF and promotes angiogenesis (49). On the other hand, reduced adiponectin levels have been found to be associated with increased risk of the same cancers (62–64). Adiponectin has been shown to exert growth inhibitory, proapoptotic, anti-inflammatory, and antiangiogenic effects (65–68). However, contrary to findings from observational studies, recent MR analyses identified no associations to support a causal relationship between genetically determined levels of adiponectin or leptin and the risk of developing five obesity-related cancers (colorectal, pancreatic, renal cell carcinoma, ovarian, and endometrial), highlighting the need for further investigation of the complex roles adipokines may play in cancer biology (69).

Estrogen

The role of estrogen in the development of postmenopausal ER+ breast cancer and endometrial cancer is well-documented among obese women and supported by evidence from MR analyses (20, 70). Estrogen production occurs through aromatase-mediated conversion of androgens, a process that takes place primarily in the ovary in premenopausal women. Following menopause, adipose tissue acts as a significant site for estrogen production. In metabolically obese individuals, increased adiposity and insulin resistance contribute to elevated free circulating estrogen levels through increased peripheral conversion of androgen to estrogen and reduced blood levels of SHBG (49, 51). The tumor-promoting effects of estrogen are mediated by complex mechanisms and crosstalk between signaling pathways involving insulin/IGF-1, adipokines, and inflammatory mediators. Estrogen promotes cell proliferation and inhibits apoptosis via direct agonism of estrogen receptor-α, and stimulates angiogenesis via VEGF induction (71, 72).

Inflammation

Inflammation is well-documented to occur in obesity and is associated with an increased risk of cancer (51). Inflammatory markers such as C-reactive protein (CRP) and IL6 have been shown by MR studies to potentially increase the risk of colorectal cancer and liver cancer, respectively (20). Under physiologic conditions, adipocytes and stromal vascular cells in adipose tissue play key roles in lipid storage, energy metabolism, and immune function. In obesity, and presumably in MONW, adipocyte hypertrophy leads to the development of cell hypoxia, injury, and eventual adipocyte death, resulting in adipose tissue inflammation (73). Elevated circulating levels of high-sensitivity CRP (hsCRP) and IL6 occur in individuals with increased adiposity, including in postmenopausal women with MONW (7, 70, 74). Interactions between adipocytes and surrounding inflammatory cells are believed to play a role in promoting tumorigenesis. In particular, the presence of crown-like structures (CLS), a histologic biomarker of obesity-related inflammation consisting of macrophages surrounding dead or dying adipocytes, have been associated with an increased risk of breast cancer (ref. 75; Fig. 2). Notably, CLS of the breast are associated with MONW (73). In the breast, the presence of CLS is associated with increased levels of aromatase, the rate-limiting enzyme for estrogen biosynthesis. Proinflammatory mediators, including leptin, are known to induce aromatase (76).

Body composition measurements

Given the link between altered quantities and distribution of body fat and disease predisposition, the ability to detect increased adiposity in a normal weight individual is crucial for identifying those at risk of adverse health outcomes. Methods for measuring body composition range from anthropometric indices to more advanced technologies, such as BIA, and imaging modalities such as DXA, CT, and MRI (Fig. 3).

Figure 3.

Methods of body composition measurement and their diagnostic outputs. A, BIA with mock report of body composition data. B, DXA machine and a whole-body DXA scan. C, MRI machine and an axial abdominal MRI scan. D, CT Imager with axial abdominal CT scan. Abbreviations: BIA, bioelectrical impedance analysis; DXA, dual-energy X-ray absorptiometry.

Figure 3.

Methods of body composition measurement and their diagnostic outputs. A, BIA with mock report of body composition data. B, DXA machine and a whole-body DXA scan. C, MRI machine and an axial abdominal MRI scan. D, CT Imager with axial abdominal CT scan. Abbreviations: BIA, bioelectrical impedance analysis; DXA, dual-energy X-ray absorptiometry.

Close modal

While CT and MRI provide the most detailed body composition data, including accurate quantifications of SAT and VAT, the high cost of these modalities and the radiation exposure associated with CT imaging limit their use for routine screening. Anthropometric measurements, most commonly WC, WHR, and skinfold thickness, incur negligible cost, are radiation-free, and can be easily obtained during an outpatient visit, but offer only a rough approximation of adiposity status. While WC and WHR are better predictors of health outcomes than BMI (77), their efficacy is heavily dependent on the site of measurement and their precision is limited by inter-operator variability (78).

More objective modalities for body composition measurement include BIA and DXA. BIA is a radiation-free technology that estimates total BF% and fat-free mass by differentiating between nonconductive (fat) and conductive tissues (muscle and bone). BIA measurements are low-cost and can be conveniently performed at the point of care, although the equipment may cost more than $5,000. The reliability of BIA measurements is affected by shifts in water-electrolyte balance, which limits BIA's use in individuals with altered volume status (79).

DXA, readily available at many facilities for routine osteoporosis screening, utilizes X-rays emitted at two energy levels to quantify fat and lean tissue. If equipped with the appropriate computer software, DXA can reliably provide body composition measurements on both whole-body and regional levels, including total fat%, trunk fat%, total fat mass, and trunk fat mass (79). While DXA software can estimate VAT, its ability to discriminate between different types of fat (i.e., visceral, subcutaneous and intramuscular) is less precise than that of CT or MRI (79). However, the radiation exposure from DXA is minimal compared with that from CT. The effective radiation dose from one whole-body DXA scan is under 10 microSieverts, about the same as background radiation at sea level for one day (80). A whole-body scan may cost up to several hundred dollars and would require referral to a facility with DXA equipped to assess body composition.

Blood-based Biomarkers

Lipid and glycemic profiles

Although the presence of dyslipidemia and insulin resistance is well-documented in MONW individuals, the search for a sensitive, specific, and clinically feasible test to identify MONW remains a challenge. For research purposes, studies have frequently relied on components of MetS as indices of cardiometabolic health. Wildman and colleagues proposed the addition of CRP and insulin resistance to the MetS criteria in the evaluation of metabolic obesity, defined as the presence of two or more of these components (81).

The sensitivity and specificity of these criteria are not well characterized and may depend on the ethnicity, sex, and for women, the menopausal status of the individual. Current evidence demonstrates that a relative increase in central adiposity among normal BMI postmenopausal women is associated with higher risks for cardiovascular disease and breast cancer (6, 7). While those with relatively high levels of central adiposity were found to have altered lipid profiles, glycemic traits, and CRP levels, only a subset would meet the thresholds defined by MetS and Wildman's criteria (6, 7). Whether these existing criteria can effectively identify normal BMI individuals at risk for adverse long-term health outcomes requires further investigation.

Inflammatory markers and adipokines

In MONW, a proinflammatory state similar to that seen in obesity has been observed. Elevated blood levels of numerous inflammatory biomarkers, including CRP and hsCRP (6, 7, 82, 83), IL6 (6, 7, 84), TNFα (84), complement C3 (82), white blood cell count (7), and ferritin (85), have been reported in MONW individuals across ethnicities. CRP, IL6, and C3 have additionally been shown to be positively correlated with excess central adiposity in MONW individuals (6, 7, 82, 83).

Alterations in plasma adipokine levels have also been observed in MONW individuals, including decreased adiponectin (6, 7, 84, 86, 87) and increased leptin (6, 7, 84). While the precise criteria for defining MONW vary across studies, changes in adiponectin and leptin levels have been consistently associated with increased adiposity.

Lifestyle and pharmacologic interventions

While studies have demonstrated associations between behavioral risk factors and adverse health outcomes in MONW individuals (45–47), clinical trials of interventions within this population remain limited. There is emerging evidence that weight reduction may be effective in improving body composition and metabolic function in nonobese individuals (88). A noncontrolled trial of 11 MONW East Asian subjects showed a diet-induced 5% weight loss led to significant decreases in total fat mass, VAT and SAT volumes, and intrahepatic fat (89). Weight reduction was also associated with improvements in lipid and metabolic profiles, with decreases in total cholesterol, TG, and LDL-C, and increases in insulin sensitivity and postprandial insulin clearance rate (89).

Pharmacologic agents are increasingly prescribed for obese patients who are unable to achieve adequate weight loss through lifestyle changes alone. However, little is known about their efficacy in reducing body fat or mitigating adverse health outcomes in the MONW population. Furthermore, the safety profile and therapeutic index would need to be established in this patient population.

Defining the problem

MONW is gaining recognition as a clinically significant condition associated with a growing number of adverse health outcomes, including cancer. Raising awareness among both clinicians and patients about the negative health impacts of MONW should be made a public health priority in areas with relatively high prevalence. Additional efforts to educate the public about metabolic obesity and increased cancer risk, a lesser-known health consequence than CVD, may be beneficial. While MONW has been found to be associated with higher risks of developing postmenopausal breast, colorectal, primary liver, and endometrial cancers (Table 1), further research is needed to evaluate MONW as a potential risk factor for other malignancies associated with obesity, such as esophageal adenocarcinoma, renal cell carcinoma, and multiple myeloma, among others. MONW has been found to correlate with increased risk of all-cancer mortality (22, 23), but there are currently no data on whether MONW affects treatment response, recurrence, or cancer-specific survival. Additional research is needed to characterize the full scope of MONW's potential effects on cancer outcomes and determine possible benefits for additional management considerations for patients with cancer with MONW, such as tight glycemic control.

In all likelihood, the mechanisms underlying obesity-related tumor development will be directly relevant to MONW-associated cancers. Evidence from GWAS studies points to a possible genetic basis for the MONW phenotype. Further investigation of the genetic underpinnings of MONW may elucidate its pathophysiology and mechanisms of cancer development. It is conceivable that a greater understanding of mechanism will provide the basis for risk-reducing pharmacological interventions that may complement behavioral approaches.

Challenges in diagnosis

Despite its serious health consequences, MONW remains under-recognized in the clinical setting. The standard practice of relying upon BMI for risk stratification continues to provide false reassurance when evaluating normal weight patients, even though clinicians are aware of BMI's limitations. The heterogeneity of definitions used in the study of MONW has made diagnosing the condition a challenge. Although criteria differ across studies, they can be broadly classified into those that define MONW by increased adiposity and those that are based on indices of metabolic dysfunction. Existing evidence demonstrates these two elements of MONW to be highly correlated; therefore, the presence of increased central adiposity or cardiometabolic abnormality in a normal BMI individual should raise clinical suspicion for MONW.

A standardized definition for MONW is needed to more accurately determine the prevalence of the condition. Further research is required to determine ethnicity- and sex-specific cut-off points for metabolic and inflammatory biomarkers as well as DXA- and BIA-derived measures of adiposity that will help to establish a diagnosis of MONW. Observational cohorts such as the UK Biobank and the Women's Health Initiative have contributed valuable baseline data on body composition, blood analytes, and clinical parameters from large samples of normal weight individuals. However, most MONW studies using such data have relied on intracohort comparisons of adiposity (e.g., top vs. bottom quartiles) to identify associations between indices of MONW and cancer risk. Such analyses result in cohort-specific cut-off points with limited generalizability.

While observational studies have led to the identification of blood biomarkers of MONW such as insulin and CRP, new “omics”-based technologies may vastly expand the scope of novel biomarker discovery to include genomic, epigenomic, transcriptomic, proteomic, metabolomic, and microbiota analyses. Analyses of existing observational cohort biobanks may prove useful in this discovery effort.

Ideally, large-scale prospective cohort studies would enable the development of a common definition for MONW. A possible next step to moving forward with establishing a consensus definition of MONW may be to convene a panel of experts to outline standardized diagnostic criteria for MONW. Precedents for the use of expert panels in establishing clinical criteria include BMI classifications for obesity and NCEP ATP III criteria for metabolic syndrome.

Beyond the lack of standardized criteria for diagnosing MONW, additional unmet challenges limit the widespread recognition of MONW in the clinical setting. The costs associated with DXA will be prohibitive for many in the absence of insurance reimbursement for testing, though BIA offers a viable cost-effective alternative. In addition, given the current absence of effective interventions to treat MONW, further research is needed to identify useful therapeutic approaches. In the absence of proven interventions, simply making the diagnosis of MONW brings little clinical benefit.

Future directions in treatment

Treatment strategies studied in the context of obesity, including lifestyle interventions and pharmacotherapies, should be tested in the MONW population. Among healthy nonobese subjects, daily caloric restriction has been found to reduce CVD risk as well as VAT, SAT, and intramyocellular lipid content (88). A small trial in MONW individuals has shown diet-induced weight loss to result in a reduction in body fat and improvement in clinical parameters (89). However, there are currently no data from larger-scale long-term studies to verify these findings in MONW subjects, though a trial evaluating the effects of caloric restriction has been initiated (ClinicalTrials.gov Identifier: NCT03239782). Many dietary approaches have been found to improve body composition in obese subjects, including low-energy, low-fat, low-carbohydrate, high-protein, ketogenic, and whole food plant-based diets (90–92). Intermittent fasting, a caloric restriction paradigm comprising variants of alternating fasting and feeding periods, has recently gained recognition for its potential efficacy in reducing multiple cardiometabolic risk factors, including lowering blood pressure and improving insulin resistance (93, 94). Whether these benefits are translatable to MONW remains to be tested.

Higher muscle mass is known to be protective against CVD and cancer mortality (95, 96). Therefore, the preservation of total-body lean mass should be prioritized in any MONW therapy. Dietary interventions may be designed to maximize muscle retention, such as increasing protein intake and maintaining a slower rate of weight reduction (90). Studies in overweight and obese populations have shown that exercise in conjunction with dietary changes can increase loss of fat mass, with resistance training resulting in higher preservation of lean mass than aerobic training (97). High-intensity interval training and medium-intensity continuous training have also been found to reduce whole-body fat mass without significant loss of lean mass or changes in body weight (98).

Medications that induce weight loss in obese individuals have yet to be studied in MONW cohorts. Oral antihyperglycemic agents like metformin, sodium-coupled glucose transporter 2 inhibitors, and glucagon-like peptide-1 receptor agonists have been shown to significantly reduce body fat mass but with mixed data on their effects on lean mass reduction (99–101). FDA-approved weight loss medications may be effective, but their significant side effect profiles may outweigh any potential benefits for the MONW population.

MONW is an underdiagnosed condition that significantly increases cancer risk among individuals with normal BMI. Increased awareness of MONW among both healthcare providers and the at-risk demographic is necessary if the long-term morbidity and mortality associated with it are to be reduced. Importantly, future research is needed to establish standardized diagnostic criteria and measure the effectiveness of different lifestyle and pharmacologic interventions in improving body composition and mitigating adverse health outcomes in MONW populations, including the development of MONW-associated cancers. Hopefully, increased awareness of MONW and its complications will lead to an enhanced effort to develop interventions to effectively reduce the risk of preventable life-threatening diseases.

No disclosures were reported.

This work was supported by the Breast Cancer Research Foundation (BCRF-19–034, to A.J. Dannenberg; BCRF-19–140, to T.E. Rohan), and NCI U54 CA210184 (to A.J. Dannenberg). B. Liu conducted this research as part of the Area of Concentration (AOC) Program of the Weill Cornell MD Curriculum.

1.
Rothman
KJ
. 
BMI-related errors in the measurement of obesity
.
Int J Obes
2008
;
32
:
S56
9
.
2.
Appropriate body-mass index for asian populations and its implications for policy and intervention strategies
.
Lancet
2004
;
363
:
157
63
.
3.
Ruderman
NB
,
Schneider
SH
,
Berchtold
P
. 
The “metabolically-obese,” normal-weight individual
.
Am J Clin Nutr
1981
;
34
:
1617
21
.
4.
Stefan
N
,
Schick
F
,
Häring
H-U
. 
Causes, characteristics, and consequences of metabolically unhealthy normal weight in humans
.
Cell Metab
2017
;
26
:
292
300
.
5.
Franco
LP
,
Morais
CC
,
Cominetti
C
. 
Normal-weight obesity syndrome: diagnosis, prevalence, and clinical implications
.
Nutr Rev
2016
;
74
:
558
70
.
6.
Chen
G-C
,
Arthur
R
,
Iyengar
NM
,
Kamensky
V
,
Xue
X
,
Wassertheil-Smoller
S
, et al
Association between regional body fat and cardiovascular disease risk among postmenopausal women with normal body mass index
.
Eur Heart J
2019
;
40
:
2849
55
.
7.
Iyengar
NM
,
Arthur
R
,
Manson
JE
,
Chlebowski
RT
,
Kroenke
CH
,
Peterson
L
, et al
Association of body fat and risk of breast cancer in postmenopausal women with normal body mass index: a secondary analysis of a randomized clinical trial and observational study
.
JAMA Oncol
2019
;
5
:
155
63
.
8.
Murphy
N
,
Cross
AJ
,
Abubakar
M
,
Jenab
M
,
Aleksandrova
K
,
Boutron-Ruault
M-C
, et al
A nested case-control study of metabolically defined body size phenotypes and risk of colorectal cancer in the european prospective investigation into cancer and nutrition (EPIC)
.
PLoS Med
2016
;
13
:
e1001988
.
9.
Kim
JW
,
Ahn
ST
,
Oh
MM
,
Moon
DG
,
Han
K
,
Park
HS
. 
Incidence of prostate cancer according to metabolic health status: a nationwide cohort study
.
J Korean Med Sci
2019
;
34
:
e49
.
10.
Marques-Vidal
P
,
Pécoud
A
,
Hayoz
D
,
Paccaud
F
,
Mooser
V
,
Waeber
G
, et al
Prevalence of normal weight obesity in Switzerland: effect of various definitions
.
Eur J Nutr
2008
;
47
:
251
.
11.
Kim
MK
,
Han
K
,
Kwon
H-S
,
Song
K-H
,
Yim
HW
,
Lee
W-C
, et al
Normal weight obesity in Korean adults
.
Clin Endocrinol (Oxf)
2014
;
80
:
214
20
.
12.
Wulan
SN
,
Westerterp
KR
,
Plasqui
G
. 
Ethnic differences in body composition and the associated metabolic profile: A comparative study between Asians and Caucasians
.
Maturitas
2010
;
65
:
315
9
.
13.
Rush
EC
,
Freitas
I
,
Plank
LD
. 
Body size, body composition and fat distribution: comparative analysis of European, Maori, Pacific Island and Asian Indian adults
.
Br J Nutr
2009
;
102
:
632
41
.
14.
Romero-Corral
A
,
Somers
VK
,
Sierra-Johnson
J
,
Korenfeld
Y
,
Boarin
S
,
Korinek
J
, et al
Normal weight obesity: a risk factor for cardiometabolic dysregulation and cardiovascular mortality
.
Eur Heart J
2009
;
31
:
737
46
.
15.
Marques-Vidal
P
,
Pécoud
A
,
Hayoz
D
,
Paccaud
F
,
Mooser
V
,
Waeber
G
, et al
Normal weight obesity: relationship with lipids, glycaemic status, liver enzymes and inflammation
.
Nutr Metab Cardiovasc Dis
2010
;
20
:
669
75
.
16.
De Lorenzo
A
,
Martinoli
R
,
Vaia
F
,
Di Renzo
L
. 
Normal weight obese (NWO) women: An evaluation of a candidate new syndrome
.
Nutr Metab Cardiovasc Dis
2006
;
16
:
513
23
.
17.
Guerrero-Romero
F
,
Rodriguez-Moran
M
. 
Serum magnesium in the metabolically-obese normal-weight and healthy-obese subjects
.
Eur J Intern Med
2013
;
24
:
639
43
.
18.
Lauby-Secretan
B
,
Scoccianti
C
,
Loomis
D
,
Grosse
Y
,
Bianchini
F
,
Straif
K
. 
Body fatness and cancer—viewpoint of the iarc working group
.
N Engl J Med
2016
;
375
:
794
8
.
19.
Calle
EE
,
Rodriguez
C
,
Walker-Thurmond
K
,
Thun
MJ
. 
Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults
.
N Engl J Med
2003
;
348
:
1625
38
.
20.
Pierce
BL
,
Kraft
P
,
Zhang
C
. 
Mendelian randomization studies of cancer risk: a literature review
.
Curr Epidemiol Rep
2018/05/18 ed.
2018
;
5
:
184
96
.
21.
Sahakyan
KR
,
Somers
VK
,
Rodriguez-Escudero
JP
,
Hodge
DO
,
Carter
RE
,
Sochor
O
, et al
Normal-weight central obesity: implications for total and cardiovascular mortality
.
Ann Intern Med
2015
;
163
:
827
35
.
22.
Zhang
C
,
Rexrode
KM
,
van Dam
RM
,
Li
TY
,
Hu
FB
. 
Abdominal obesity and the risk of all-cause, cardiovascular, and cancer mortality
.
Circulation
2008
;
117
:
1658
67
.
23.
Sun
Y
,
Liu
B
,
Snetselaar
LG
,
Wallace
RB
,
Caan
BJ
,
Rohan
TE
, et al
Association of normal-weight central obesity with all-cause and cause-specific mortality among postmenopausal women
.
JAMA Netw Open
2019
;
2
:
e197337–
.
24.
Protani
M
,
Coory
M
,
Martin
JH
. 
Effect of obesity on survival of women with breast cancer: systematic review and meta-analysis
.
Breast Cancer Res Treat
2010
;
123
:
627
35
.
25.
Shu
X
,
Wu
L
,
Khankari
NK
,
Shu
X-O
,
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
.
26.
Ogundiran
TO
,
Huo
D
,
Adenipekun
A
,
Campbell
O
,
Oyesegun
R
,
Akang
E
, et al
Body fat distribution and breast cancer risk: findings from the Nigerian breast cancer study
.
Cancer Causes Control
2012
;
23
:
565
74
.
27.
Ronco
A
,
Boeing
H
,
Schulz
M
,
Schulze
M
,
Pischon
T
. 
A case-control study on fat-to-muscle ratio and risk of breast cancer
.
Nutr Cancer
2009
;
61
:
466
74
.
28.
Park
Y-MM
,
White
AJ
,
Nichols
HB
,
O'Brien
KM
,
Weinberg
CR
,
Sandler
DP
. 
The association between metabolic health, obesity phenotype and the risk of breast cancer
.
Int J Cancer
2017
;
140
:
2657
66
.
29.
Arthur
RS
,
Dannenberg
AJ
,
Kim
M
,
Rohan
TE
. 
The association of body fat composition with risk of breast, endometrial, ovarian and colorectal cancers among normal weight participants in the UK Biobank
.
Br J Cancer
2020
.
30.
Gunter
MJ
,
Xie
X
,
Xue
X
,
Kabat
GC
,
Rohan
TE
,
Wassertheil-Smoller
S
, et al
Breast cancer risk in metabolically healthy but overweight postmenopausal women
.
Cancer Res
2015
;
75
:
270
.
31.
His
M
,
Biessy
C
,
Torres-Mejía
G
,
Ángeles-Llerenas
A
,
Alvarado-Cabrero
I
,
Sánchez
GI
, et al
Anthropometry, body shape in early-life and risk of premenopausal breast cancer among Latin American women: results from the PRECAMA study
.
Sci Rep
2020
;
10
:
2294
.
32.
Friedenreich
CM
,
Biel
RK
,
Lau
DCW
,
Csizmadi
I
,
Courneya
KS
,
Magliocco
AM
, et al
Case–control study of the metabolic syndrome and metabolic risk factors for endometrial cancer
.
Cancer Epidemiol Biomark Amp Prev
2011
;
20
:
2384
.
33.
Calle
EE
,
Kaaks
R
. 
Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms
.
Nat Rev Cancer
2004
;
4
:
579
91
.
34.
Kabat
GC
,
Kim
MY
,
Stefanick
M
,
Ho
GYF
,
Lane
DS
,
Odegaard
AO
, et al
Metabolic obesity phenotypes and risk of colorectal cancer in postmenopausal women
.
Int J Cancer
2018
;
143
:
543
51
.
35.
Abar
L
,
Vieira
AR
,
Aune
D
,
Sobiecki
JG
,
Vingeliene
S
,
Polemiti
E
, et al
Height and body fatness and colorectal cancer risk: an update of the WCRF–AICR systematic review of published prospective studies
.
Eur J Nutr
2018
;
57
:
1701
20
.
36.
Tobari
M
,
Hashimoto
E
,
Taniai
M
,
Ikarashi
Y
,
Kodama
K
,
Kogiso
T
, et al
Characteristics of non-alcoholic steatohepatitis among lean patients in Japan: Not uncommon and not always benign
.
J Gastroenterol Hepatol
2019
;
34
:
1404
10
.
37.
Sun
B
,
Karin
M
. 
Obesity, inflammation, and liver cancer
.
J Hepatol
2012
;
56
:
704
13
.
38.
Florio
AA
,
Campbell
PT
,
Zhang
X
,
Zeleniuch-Jacquotte
A
,
Wactawski-Wende
J
,
Smith-Warner
SA
, et al
Abdominal and gluteofemoral size and risk of liver cancer: The liver cancer pooling project
.
Int J Cancer
2020
;
147
:
675
85
.
39.
Oliveros
E
,
Somers
VK
,
Sochor
O
,
Goel
K
,
Lopez-Jimenez
F
. 
The concept of normal weight obesity
.
Obes Obes Paradox Cardiovasc Dis
2014
;
56
:
426
33
.
40.
Cornier
M-A
,
Després
J-P
,
Davis
N
,
Grossniklaus
DA
,
Klein
S
,
Lamarche
B
, et al
Assessing adiposity
.
Circulation
2011
;
124
:
1996
2019
.
41.
Huang
LO
,
Loos
RJF
,
Kilpeläinen
TO
. 
Evidence of genetic predisposition for metabolically healthy obesity and metabolically obese normal weight
.
Physiol Genomics
2017
;
50
:
169
78
.
42.
Yaghootkar
H
,
Scott
RA
,
White
CC
,
Zhang
W
,
Speliotes
E
,
Munroe
PB
, et al
Genetic evidence for a normal-weight “metabolically obese” phenotype linking insulin resistance, hypertension, coronary artery disease, and type 2 diabetes
.
Diabetes
2014
;
63
:
4369
.
43.
Kilpeläinen
TO
,
Zillikens
MC
,
Stančákova
A
,
Finucane
FM
,
Ried
JS
,
Langenberg
C
, et al
Genetic variation near IRS1 associates with reduced adiposity and an impaired metabolic profile
.
Nat Genet
2011
;
43
:
753
60
.
44.
Yaghootkar
H
,
Lotta
LA
,
Tyrrell
J
,
Smit
RAJ
,
Jones
SE
,
Donnelly
L
, et al
Genetic evidence for a link between favorable adiposity and lower risk of type 2 diabetes, hypertension, and heart disease
.
Diabetes
2016
;
65
:
2448
60
.
45.
Conus
F
,
Allison
DB
,
Rabasa-Lhoret
R
,
St-Onge
M
,
St-Pierre
DH
,
Tremblay-Lebeau
A
, et al
Metabolic and behavioral characteristics of metabolically obese but normal-weight women
.
J Clin Endocrinol Metab
2004
;
89
:
5013
20
.
46.
Eckel
N
,
Mühlenbruch
K
,
Meidtner
K
,
Boeing
H
,
Stefan
N
,
Schulze
MB
. 
Characterization of metabolically unhealthy normal-weight individuals: Risk factors and their associations with type 2 diabetes
.
Metabolism
2015
;
64
:
862
71
.
47.
Choi
J
,
Se-Young
O
,
Lee
D
,
Tak
S
,
Hong
M
,
Park
SM
, et al
Characteristics of diet patterns in metabolically obese, normal weight adults (korean national health and nutrition examination survey III, 2005)
.
Nutr Metab Cardiovasc Dis
2012
;
22
:
567
74
.
48.
Dvorak
RV
,
DeNino
WF
,
Ades
PA
,
Poehlman
ET
. 
Phenotypic characteristics associated with insulin resistance in metabolically obese but normal-weight young women
.
Diabetes
1999
;
48
:
2210
.
49.
Hursting
SD
,
DiGiovanni
J
,
Dannenberg
AJ
,
Azrad
M
,
LeRoith
D
,
Demark-Wahnefried
W
, et al
Obesity, energy balance, and cancer: new opportunities for prevention
.
Cancer Prev Res (Phila Pa)
2012
;
5
:
1260
.
50.
Quail
DF
,
Dannenberg
AJ
. 
The obese adipose tissue microenvironment in cancer development and progression
.
Nat Rev Endocrinol
2019
;
15
:
139
54
.
51.
Gucalp
A
,
Iyengar
NM
,
Hudis
CA
,
Dannenberg
AJ
. 
Targeting obesity-related adipose tissue dysfunction to prevent cancer development and progression
.
Mol Targets Cancer Prev
2016
;
43
:
154
60
.
52.
Nead
KT
,
Sharp
SJ
,
Thompson
DJ
,
Painter
JN
,
Savage
DB
,
Semple
RK
, et al
Evidence of a causal association between insulinemia and endometrial cancer: a mendelian randomization analysis
.
J Natl Cancer Inst
2015
;
107
:
djv178
.
53.
Pollak
M
. 
Insulin and insulin-like growth factor signalling in neoplasia
.
Nat Rev Cancer
2008
;
8
:
915
28
.
54.
Wong
K-K
,
Engelman
JA
,
Cantley
LC
. 
Targeting the PI3K signaling pathway in cancer
.
Genet Cell Mech Oncog
2010
;
20
:
87
90
.
55.
Zoncu
R
,
Efeyan
A
,
Sabatini
DM
. 
mTOR: from growth signal integration to cancer, diabetes and ageing
.
Nat Rev Mol Cell Biol
2011
;
12
:
21
35
.
56.
Miele
C
,
Rochford
JJ
,
Filippa
N
,
Giorgetti-Peraldi
S
,
Van Obberghen
E
. 
Insulin and insulin-like growth factor-I induce vascular endothelial growth factor mRNA expression via different signaling pathways
.
J Biol Chem
2000
;
275
:
21695
702
.
57.
Rabin-Court
A
,
Rodrigues
MR
,
Zhang
X-M
,
Perry
RJ
. 
Obesity-associated, but not obesity-independent, tumors respond to insulin by increasing mitochondrial glucose oxidation
.
PLoS One
2019
;
14
:
e0218126
.
58.
Jardé
T
,
Caldefie-Chézet
F
,
Damez
M
,
Mishellany
F
,
Penault-Llorca
F
,
Guillot
J
, et al
Leptin and leptin receptor involvement in cancer development: a study on human primary breast carcinoma
.
Oncol Rep
2008
;
19
:
905
11
.
59.
Sharma
D
,
Saxena
NK
,
Vertino
PM
,
Anania
FA
. 
Leptin promotes the proliferative response and invasiveness in human endometrial cancer cells by activating multiple signal-transduction pathways
.
Endocr-Relat Cancer Endocr Relat Cancer
2006
;
13
:
629
40
.
60.
Hoda
MR
,
Keely
SJ
,
Bertelsen
LS
,
Junger
WG
,
Dharmasena
D
,
Barrett
KE
. 
Leptin acts as a mitogenic and antiapoptotic factor for colonic cancer cells
.
BJS Br J Surg
2007
;
94
:
346
54
.
61.
Onuma
M
,
Bub
JD
,
Rummel
TL
,
Iwamoto
Y
. 
Prostate cancer cell-adipocyte interaction: leptin mediates androgen-independent prostate cancer cell proliferation through c-jun NH2-terminal kinase
.
J Biol Chem
2003
;
278
:
42660
7
.
62.
Miyoshi
Y
,
Funahashi
T
,
Kihara
S
,
Taguchi
T
,
Tamaki
Y
,
Matsuzawa
Y
, et al
Association of serum adiponectin levels with breast cancer risk
.
Clin Cancer Res
2003
;
9
:
5699
.
63.
Soliman
PT
,
Wu
D
,
Tortolero-Luna
G
,
Schmeler
KM
,
Slomovitz
BM
,
Bray
MS
, et al
Association between adiponectin, insulin resistance, and endometrial cancer
.
Cancer
2006
;
106
:
2376
81
.
64.
XU
XT
,
XU
Q
,
TONG
JL
,
ZHU
MM
,
HUANG
ML
,
RAN
ZH
, et al
Meta-analysis: Circulating adiponectin levels and risk of colorectal cancer and adenoma
.
J Dig Dis
2011
;
12
:
234
44
.
65.
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
.
66.
Cong
Li
,
Gasser
J
,
Zhao
J
,
Yang
B
,
Li
F
,
Zhao
AZ
. 
Human adiponectin inhibits cell growth and induces apoptosis in human endometrial carcinoma cells, HEC-1-A and RL95–2
.
Endocr Relat Cancer
2007
;
14
:
713
20
.
67.
Kim
AY
,
Lee
YS
,
Kim
KH
,
Lee
JH
,
Lee
HK
,
Jang
S-H
, et al
Adiponectin represses colon cancer cell proliferation via AdipoR1- and -R2-mediated AMPK activation
.
Mol Endocrinol
2010
;
24
:
1441
52
.
68.
Bub
JD
,
Miyazaki
T
,
Iwamoto
Y
. 
Adiponectin as a growth inhibitor in prostate cancer cells
.
Biochem Biophys Res Commun
2006
;
340
:
1158
66
.
69.
Dimou
NL
,
Papadimitriou
N
,
Mariosa
D
,
Johansson
M
,
Brennan
P
,
Peters
U
, et al
Circulating adipokine concentrations and risk of five obesity-related cancers: A Mendelian randomization study
.
Int J Cancer
2021
;
148
:
1625
36
.
70.
Iyengar
NM
,
Hudis
CA
,
Dannenberg
AJ
. 
Obesity and cancer: local and systemic mechanisms
.
Annu Rev Med
2015
;
66
:
297
309
.
71.
Yager
JD
,
Davidson
NE
. 
Estrogen carcinogenesis in breast cancer
.
N Engl J Med
2006
;
354
:
270
82
.
72.
Péqueux
C
,
Raymond-Letron
I
,
Blacher
S
,
Boudou
F
,
Adlanmerini
M
,
Fouque
M-J
, et al
Stromal estrogen receptor-α promotes tumor growth by normalizing an increased angiogenesis
.
Cancer Res
2012
;
72
:
3010
9
.
73.
Iyengar
N
,
Brown
K
,
Zhou
X
,
Gucalp
A
,
Subbaramaiah
K
,
Giri
D
, et al
Metabolic obesity, adipose inflammation and elevated breast aromatase in women with normal body mass index
.
Cancer Prev Res
2017
;
10
.
74.
Brestoff
JR
,
Artis
D
. 
Immune regulation of metabolic homeostasis in health and disease
.
Cell
2015
;
161
:
146
60
.
75.
Carter
JM
,
Hoskin
TL
,
Pena
MA
,
Brahmbhatt
R
,
Winham
SJ
,
Frost
MH
, et al
Macrophagic “crown-like structures” are associated with an increased risk of breast cancer in benign breast disease
.
Cancer Prev Res
2018
;
11
:
113
9
.
76.
Catalano
S
,
Marsico
S
,
Giordano
C
,
Mauro
L
,
Rizza
P
,
Panno
ML
, et al
Leptin enhances, via AP-1, expression of aromatase in the MCF-7 cell line
.
J Biol Chem
2003
;
278
:
28668
76
.
77.
Dobbelsteyn
C
,
Joffres
M
,
MacLean
D
,
Flowerdew
G
.,
The Canadian Heart Health Surveys Research Group
. 
A comparative evaluation of waist circumference, waist-to-hip ratio and body mass index as indicators of cardiovascular risk factors. the canadian heart health surveys
.
Int J Obes
2001
;
25
:
652
61
.
78.
Bosy-Westphal
A
,
Booke
C-A
,
Blöcker
T
,
Kossel
E
,
Goele
K
,
Later
W
, et al
Measurement site for waist circumference affects its accuracy as an index of visceral and abdominal subcutaneous fat in a caucasian population
.
J Nutr
2010
;
140
:
954
61
.
79.
Ceniccola
GD
,
Castro
MG
,
Piovacari
SMF
,
Horie
LM
,
Corrêa
FG
,
Barrere
APN
, et al
Current technologies in body composition assessment: advantages and disadvantages
.
Nutrition
2019
;
62
:
25
31
.
80.
Shepherd
JA
,
Ng
BK
,
Sommer
MJ
,
Heymsfield
SB
. 
Body composition by DXA
.
Bone
2017
;
104
:
101
5
.
81.
Wildman
RP
,
Muntner
P
,
Reynolds
K
,
McGinn
AP
,
Rajpathak
S
,
Wylie-Rosett
J
, et al
The obese without cardiometabolic risk factor clustering and the normal weight with cardiometabolic risk factor clustering: prevalence and correlates of 2 phenotypes among the US population (NHANES 1999–2004)
.
Arch Intern Med
2008
;
168
:
1617
24
.
82.
Karkhaneh
M
,
Qorbani
M
,
Mohajeri-Tehrani
MR
,
Hoseini
S
. 
Association of serum complement C3 with metabolic syndrome components in normal weight obese women
.
J Diabetes Metab Disord
2017
;
16
:
49
49
.
83.
Bednarek-Tupikowska
G
,
Zdrojowy-Wełna
A
,
Stachowska
B
,
Kuliczkowska-Płaksej
J
,
Matczak-Giemza
M
,
Kubicka
E
, et al
Accumulation of abdominal fat in relation to selected proinflammatory cytokines concentrations in non-obese wrocław inhabitants. accumul abdom fat relat sel proinflammatory cytokines conc non-obese wroc inhabitants
.
Endokrynologia
2014
;
65
:
449
55
.
84.
Hyun
YJ
,
Koh
SJ
,
Chae
JS
,
Kim
JY
,
Kim
OY
,
Lim
HH
, et al
Atherogenecity of LDL and unfavorable adipokine profile in metabolically obese, normal-weight woman
.
Obesity
2008
;
16
:
784
9
.
85.
Kim
J-W
,
Kim
DH
,
Roh
YK
,
Ju
SY
,
Nam
H-Y
,
Nam
G-E
, et al
Serum ferritin levels are positively associated with metabolically obese normal weight: a nationwide population-based study
.
Medicine
2015
;
94
:
e2335
.
86.
Katsuki
A
,
Suematsu
M
,
Gabazza
EC
,
Murashima
S
,
Nakatani
K
,
Togashi
K
, et al
Decreased high–molecular weight adiponectin–to–total adiponectin ratio in sera is associated with insulin resistance in Japanese metabolically obese, normal-weight men with normal glucose tolerance
.
Diabetes Care
2006
;
29
:
2327
.
87.
Zachariah Justin
P
,
Rene
Q
,
Nelson Kerrie
P
,
Zhaoyang
T
,
Keaney John
F
,
Sullivan Lisa
M
, et al
Prospective relation of circulating adipokines to incident metabolic syndrome: the framingham heart study
.
J Am Heart Assoc
2017
;
6
:
e004974
.
88.
Most
J
,
Gilmore
LA
,
Smith
SR
,
Han
H
,
Ravussin
E
,
Redman
LM
. 
Significant improvement in cardiometabolic health in healthy nonobese individuals during caloric restriction-induced weight loss and weight loss maintenance
.
Am J Physiol Endocrinol Metab
2017
;
314
:
E396
405
.
89.
Chooi
YC
,
Ding
C
,
Chan
Z
,
Choo
J
,
Sadananthan
SA
,
Michael
N
, et al
Moderate weight loss improves body composition and metabolic function in metabolically unhealthy lean subjects
.
Obesity
2018
;
26
:
1000
7
.
90.
Aragon
AA
,
Schoenfeld
BJ
,
Wildman
R
,
Kleiner
S
,
VanDusseldorp
T
,
Taylor
L
, et al
International society of sports nutrition position stand: diets and body composition
.
J Int Soc Sports Nutr
2017
;
14
:
16
.
91.
Gershuni
VM
,
Yan
SL
,
Medici
V
. 
Nutritional ketosis for weight management and reversal of metabolic syndrome
.
Curr Nutr Rep
2018
;
7
:
97
106
.
92.
Kahleova
H
,
Fleeman
R
,
Hlozkova
A
,
Holubkov
R
,
Barnard
ND
. 
A plant-based diet in overweight individuals in a 16-week randomized clinical trial: metabolic benefits of plant protein
.
Nutr Diabetes
2018
;
8
:
58
58
.
93.
Gabel
K
,
Varady
KA
. 
Current research: effect of time restricted eating on weight and cardiometabolic health
.
J Physiol
2020
.
94.
de Cabo
R
,
Mattson
MP
. 
Effects of intermittent fasting on health, aging, and disease
.
N Engl J Med
2019
;
381
:
2541
51
.
95.
Caan
BJ
,
Cespedes Feliciano
EM
,
Kroenke
CH
. 
The importance of body composition in explaining the overweight paradox in cancer-counterpoint
.
Cancer Res
2018
;
78
:
1906
12
.
96.
Srikanthan
P
,
Horwich
TB
,
Tseng
CH
. 
Relation of muscle mass and fat mass to cardiovascular disease mortality
.
Am J Cardiol
2016
;
117
:
1355
60
.
97.
Beavers
KM
,
Ambrosius
WT
,
Rejeski
WJ
,
Burdette
JH
,
Walkup
MP
,
Sheedy
JL
, et al
Effect of exercise type during intentional weight loss on body composition in older adults with obesity
.
Obesity
2017
;
25
:
1823
9
.
98.
Wewege
M
,
van den Berg
R
,
Ward
RE
,
Keech
A
. 
The effects of high-intensity interval training vs. moderate-intensity continuous training on body composition in overweight and obese adults: a systematic review and meta-analysis
.
Obes Rev
2017
;
18
:
635
46
.
99.
Feng
W-H
,
Bi
Y
,
Li
P
,
Yin
T-T
,
Gao
C-X
,
Shen
S-M
, et al
Effects of liraglutide, metformin and gliclazide on body composition in patients with both type 2 diabetes and non-alcoholic fatty liver disease: a randomized trial
.
J Diabetes Investig
2019
;
10
:
399
407
.
100.
Yanovski
JA
,
Krakoff
J
,
Salaita
CG
,
McDuffie
JR
,
Kozlosky
M
,
Sebring
NG
, et al
Effects of metformin on body weight and body composition in obese insulin-resistant children
.
Diabetes
2011
;
60
:
477
.
101.
Sargeant
JA
,
Henson
J
,
King
JA
,
Yates
T
,
Khunti
K
,
Davies
MJ
. 
A review of the effects of glucagon-like peptide-1 receptor agonists and sodium-glucose cotransporter 2 inhibitors on lean body mass in humans
.
Endocrinol Metab Seoul Korea
2019
;
34
:
247
62
.