Next Article in Journal
Challenges and Opportunities for Equity in US School Meal Programs: A Scoping Review of Qualitative Literature Regarding the COVID-19 Emergency
Next Article in Special Issue
Adherence to the Mediterranean Diet and Risk of Gastric Cancer: A Systematic Review and Meta-Analysis
Previous Article in Journal
Positive Regulation of Acetate in Adipocyte Differentiation and Lipid Deposition in Obese Mice
Previous Article in Special Issue
High Dietary Phosphorus Is Associated with Increased Breast Cancer Risk in a U.S. Cohort of Middle-Aged Women
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Treatments for Obesity: Implications for Cancer Prevention and Treatment

by
Carla Micaela Cuttica
1,*,
Irene Maria Briata
2 and
Andrea DeCensi
2,3
1
Endocrine Unit, E.O. Ospedali Galliera, 16128 Genoa, Italy
2
Division of Medical Oncology, E.O. Ospedali Galliera, 16128 Genoa, Italy
3
Wolfson Institute of Population Health, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AD, UK
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(17), 3737; https://doi.org/10.3390/nu15173737
Submission received: 8 August 2023 / Revised: 23 August 2023 / Accepted: 24 August 2023 / Published: 25 August 2023

Abstract

:
It is now established that obesity is related to a higher incidence of cancer during a lifespan. The effective treatment of obesity opens up new perspectives in the treatment of a relevant modifiable cancer risk factor. The present narrative review summarizes the correlations between weight loss in obesity and cancer. The current knowledge between obesity treatment and cancer was explored, highlighting the greatest potential for its use in the treatment of cancer in the clinical setting. Evidence for the effects of obesity therapy on proliferation, apoptosis, and response to chemotherapy is summarized. While more studies, including large, long-term clinical trials, are needed to adequately evaluate the relationship and durability between anti-obesity treatment and cancer, collaboration between oncologists and obesity treatment experts is increasingly important.

1. Introduction

The obesity pandemic is on the rise around the world; by 2030, it is expected that 1 in 5 women and 1 in 7 men will live with obesity, equal to over 1 billion people globally, producing a considerable impact on the number of years of life lost due to disease and premature death [1].
Obesity is a chronic, relapsing and multifactorial disease linked to excessive adiposity that predisposes individuals to other non-communicable diseases (NCDs) and medical complications, such as type 2 diabetes (T2DM), cardiovascular disease, chronic kidney disease, gallbladder disease, non-alcoholic fatty liver disease, gout, obstructive sleep apnea, osteoarthritis and cancer [2]. It is now known that obesity can increase the risk of different types of cancer, including cancers of the breast in postmenopausal women, colorectum, endometrium, kidney, liver, gallbladder, ovary, pancreas, gastric cardia, esophagus, thyroid, multiple myeloma and meningioma, and may worsen the survival of patients with cancer, especially for breast, bladder, colorectal, prostate and liver cancers. Obesity is estimated to overtake smoking as the main risk factor for cancer in many countries in the coming decades, making obesity treatment a new challenge in cancer control strategies [3,4]. Obesity often starts early in life and may influence the likelihood of developing one of the related NCDs in adulthood. Furer et al., in a large population-based cohort of 2.3 million Israeli adolescents, found that a high body mass index (BMI) in adolescence is positively associated with an increased risk of mortality after 10 years for all types of cancer in both sexes [5]. In addition, other authors have found an association between shorter survival and cumulative exposure to a higher BMI during early to mid-adulthood in patients with breast and colorectal cancer, irrespective of the presence of cardiometabolic disease prior to cancer diagnosis, pointing out how effective early life intervention strategies on BMI can become an important target of preventive intervention [6].
It is important to point out how reductive it is to talk about obesity based only on BMI. It is the excess of adiposity that gives rise to complications that compromise health and confer increased morbidity and mortality; therefore, the preferable term today is adiposity-based chronic disease (ABCD), which motivates the importance of excessive distribution of fat [7]. Adipose tissue distribution is of increasing interest due to its role in cancer development, as shown in a recent study of postmenopausal women with breast cancer with a normal BMI but high body fat evidenced with dual-energy X-ray absorptiometry (DXA) [8]. Adipose tissue can be classified into several different subtypes by function and anatomical location: white, brown and beige adipose tissue. Brown and beige adipose tissue deal with thermoregulation, whereas white adipose tissue is responsible for the energy storage of lipids (triacylglycerides) and is further divided into different compartments: subcutaneous adipose tissue and visceral adipose tissue. Visceral adipose tissue is more lipolytically active, contributing to the increase in free fatty acids and thus resulting in insulin resistance and to a more pro-cancer secretome than subcutaneous adipose tissue [9]. The accumulation of visceral adipose tissue in the body, in fact, contributes to an increase in tissue inflammation (through the infiltration of immune cells and the formation of crown-like structures by the macrophages that surround dead or dying adipocytes) and to an altered secretion of inflammatory factors that, in turn, are associated with the development of insulin resistance, metabolic syndrome and increased cancer risk and worse prognosis.
Furthermore, the importance of adipose tissue in tumorigenesis is supported by the finding that cold exposure produces a browning in adipose tissue that increases thermogenesis and glucose uptake while reducing glucose uptake, tumor growth, proliferation and hypoxia in cancer cells, making adipose tissue a possible target of cancer therapy by mediating tumor suppression through cold-impaired metabolism [10].
BMI alone does not distinguish between lean mass and fat mass, but it is inexpensive and widely used as is waist circumference, an indirect measure of abdominal obesity related to abdominal visceral fat. More accurate assessments of body fat distribution (e.g., bioelectrical impedance, DXA and magnetic resonance imaging) are expensive and limited in use in epidemiological studies [9].
Three mechanisms are mainly used to explain the relationship between obesity and cancer: sex hormone impaired metabolism, impaired insulin signaling and an excess of pro-inflammatory cytokines [3]. Weight gain in adulthood and increased body fat are known to increase the risk of endometrial cancer and the risk of hormone receptor-positive breast cancer in postmenopausal women in whom adipose tissue is the principal site for estrogen synthesis, making estrogen production a major contributor in women with obesity. Obesity is often characterized by hyperinsulinemia and insulin resistance, which are capable of stimulating the growth of cancer cells directly through insulin receptors and indirectly through insulin-like growth factor (IGF) receptors by inhibiting apoptosis and promoting cell growth, motility and invasion. Hyperinsulinemia also reduces sex hormone binding globulin levels (increasing free circulating estrogens that can promote estrogen-dependent tumors), reduces IGF-binding protein by increasing free IGF and increases the production of pro-inflammatory cytokines and adipokines, thus supporting the chronic low-grade inflammation implicated in the development of cancer, as illustrated elsewhere [3,9]. As far as adipokines are concerned, it is known that, in obesity, adiponectin decreases, and leptin increases. Leptin appears to be a pro-oncogenic factor: it is able to promote systemic inflammation, cell proliferation, angiogenesis and metastasis, also inhibiting apoptosis, immune surveillance and cell death; however, adiponectin has opposite effects. The imbalance between leptin and adiponectin plays an important role in the obesity–cancer relationship [9,11].
The recent novel adipokine circulating miRNAs, acting as epigenetic regulators, are capable of influencing adipocyte differentiation, white adipose tissue browning, lipid and glycemic homeostasis and insulin resistance with an essential role in obesity-associated inflammation, metabolic syndrome and predisposition to cancer. Moreover, miRNAs, extensively produced by adipose tissue in both paracrine and endocrine signaling, are able to participate in oncogenic and tumor suppressor molecules, modulating the cancer microenvironment, settlement, progression and invasion [12]. Gut hormone dysregulation is also involved in the pathogenesis of obesity, and emerging data hypothesize its implication in cancer development [3,13]. Gut hormones are secreted by entero-endocrine cells in the gastrointestinal tract; they react to the deficit or arrival of nutrients by acting as regulators of eating habits/appetite/satiety and regulators of energy homeostasis; they are also able to regulate insulin secretion. The dysregulation of gut hormones present in obesity is implicated in excessive nutrient intake, impaired insulin signaling leading to hyperinsulinemia and T2DM and chronic low-grade inflammation arising from adipose tissue, which are all mechanisms involved in the development of cancer [13].
The treatment of obesity, therefore, appears essential to reduce the risk of cancer.

2. Weight Loss and Cancer Risk and Prognosis

Intentional weight loss (including diet/exercise/surgery), especially if greater than 10%, is able to reverse the proinflammatory state linked to obesity, e.g., reducing the levels of C-reactive protein, tumor necrosis factor-α, interleukin-6 and the leptin-to-adiponectin ratio. Data are limited because adipokines and cytokines are difficult to measure due to their low concentrations; however, although further investigation is needed in this area, many studies suggest that weight loss among overweight or obese people is helpful in reducing cancer risk [3,9,14,15]. Moreover, it is known that subjects with obesity/ABCD at the time of cancer diagnosis have worse outcomes and poorer survival outcomes [3,16]. In breast cancer, obesity seems to be linked to a heightened risk of recurrence and overall mortality with an estimated risk that ranges from 35% to 40% [16,17]. Importantly, this correlation persists regardless of menopausal status, hormone receptor status or specific cancer subtypes [17,18]. Also, being overweight or obese appears to be linked with the occurrence of distant and delayed recurrences in patients with breast cancer [17].
The “obesity paradox” debate that has occurred between the association of high body weight and cancer survival benefits and increased tolerance during some anticancer therapies may be explained by the association of obesity with subtypes of less aggressive tumors and other confounding factors, such as smoking and body composition [16]. Intervention studies predominantly conducted in breast, endometrial and prostate cancer trials showed positive effects of intentional weight loss based on diet composition, caloric intake and amount and nature of physical activity [19,20,21,22]. It is also known that bariatric surgery, achieving significant and long-term weight loss, is effective in reducing the incidence of obesity-associated cancer and the cancer-related mortality [23].
Weight gain following a breast cancer diagnosis also seems to slightly elevate the risk of overall mortality, and the harmful impact was most pronounced when weight gain exceeded 10% [17]. Therefore, the evaluation of BMI at the time of diagnosis may be the most significant predictor of breast cancer prognosis [17], and strategies aimed to control that increase are crucial. A recent randomized trial provided evidence for the favorable impact of a weight loss intervention through diet, physical activity and behavioral change on outcomes among women with breast cancer [24].
In individuals with prostate cancer, an increase of 5 kg/m2 in BMI was found to correspond to a 9% higher risk of prostate cancer-specific mortality and a 3% higher risk of all-cause mortality [25]. Additionally, there is a moderate–consistent association between obesity and biochemical recurrence following radical prostatectomy [26]. Despite the role of obesity in prostate cancer, only a few prostate cancer guidelines recommend the adoption of a healthy lifestyle, and only 7.2% provide advice on reaching or maintaining a healthy weight [22].
It should be emphasized that weight management strategies in overweight and obese cancer survivors may also play an important role in preventing non-cancer deaths. Diabetic patients with cancer have reduced overall survival compared with non-diabetics, in particular, related to an increased risk of non-cancer deaths, primarily cardiovascular, which may be further increased by some cancer treatments, and weight loss programs have been shown to be effective in the remission of diabetes [27].

3. Metformin and Cancer

Many studies have documented an association between metformin, a biguanide for the treatment of T2DM, and reduced cancer incidence and mortality [28].
Among high-risk patients with operable breast cancer without diabetes, adding metformin versus a placebo to standard cancer treatment did not significantly improve invasive disease-free survival in a large phase III trial [29]. However, patients with ERBB2+ breast cancer in the metformin group vs. the placebo group had longer invasive disease–free survival and overall survival [29]. Moreover, in vitro studies support the efficacy of metformin in cancer therapy and prevention, and a review of epidemiological studies on metformin treatment showed a positive trend for benefit [30,31]. In a randomized placebo controlled presurgical trial, metformin was able to reduce breast cancer cell proliferation in women with insulin-resistance, suggesting a potential effect of the drug in this subgroup of patients [32,33].
Multiple mechanisms are involved: (1) metformin indirectly reduces tumor proliferation via insulin-lowering activity in subjects with hyperinsulinemia, involving the insulin/IGF-1 pathway, which is known to contribute to cell growth and proliferation by activating both phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/Akt/mTOR and the Ras/Raf/mitogen-activated protein kinase (MAPK) pathways; (2) metformin also indirectly affects cancer cells by modulating the immune response via an anti-inflammatory effect; (3) metformin acts directly on respiratory Complex I of the electron transport chain in the mitochondria of pre-neoplastic and neoplastic cells, reducing the energy consumption of cells; (4) metformin is able to inhibit the activation of matrix metalloproteinase-9, blocking the invasion of cancer cells, and to activate growth suppressants through phosphorylation of the retinoblastoma protein [34,35]. Metformin, acting through suppression of the mitochondrial electron transport chain complex I, reduces mitochondrial ATP, which increases the adenosine monophosphate (AMP): ATP ratio; as a result, AMP-activated protein kinase is activated, which inhibits hepatic lipogenesis and gluconeogenesis [36]. Metformin-induced activation of the AMPK pathway is able to reduce the tumor-promoting activity of insulin and inhibit mTOR, which is closely related to tumor cell proliferation, making metformin an intriguing molecule in cancer therapy [37,38]. Due to modest weight-loss effects, metformin is not approved as a weight-loss agent, but it is often used as an off-label drug in obese patients with prediabetes, insulin resistance, metabolic syndrome and polycystic ovary syndrome, especially in the absence of lifestyle changes or when other anti-obesity drugs cannot be used [39]. The Diabetes Prevention Study has shown that metformin is able to reduce the incidence of diabetes and produce a slight but persistent reduction in body weight (approximately 2–3 kg), BMI and waist circumference in subjects with a high risk of T2DM [40]. The metformin-induced weight change is not attributed to an increase in energy expenditure but rather to a reduction in caloric intake via appetite suppression. One hypothesis is that metformin, by suppressing the mitochondrial complex I of the electron transport chain, reduces mitochondrial production of ATP and diverts glucose to anaerobic respiration, inducing mild metabolic acidosis by the production of lactate (particularly in the postprandial period) and thus inducing appetite suppression [41]. Metformin crosses the blood–brain barrier, acting in areas associated with food–reward relationships and acting in the hypothalamus by reducing orexigenic peptides, neuropeptide Y and agouti-related protein, possibly by increasing the signal transducer and activator of transcription 3 (STAT3) that is identified as a key mediator of feeding [42]. Metformin is also capable of acting on the suppression of appetite, promoting, through a direct local gastro-intestinal action, the secretion of the anorexic intestinal incretin hormone glucagon-like peptide 1 (GLP-1), and also through the inhibition of dipeptidyl peptidase-IV (DPP-IV), an enzyme that degrades GLP-1 [43,44].

4. Obesity Therapy and Cancer Treatment and Prevention

Novel specific therapeutic strategies are currently present to address the global obesity epidemic and its comorbidities, and several promising drug targets are under study. The medications currently approved in Europe for the treatment of obesity are orlistat, naltrexone/bupropion, liraglutide and semaglutide and, in the United States, short-term phentermine and a combination of phentermine/topiramate (in 2020, the US Food and Drug Administration ordered the withdrawal of lorcaserin from the market since a drug safety clinical trial showed an increased risk of cancer [45,46].
Orlistat (tetrahydrolipstatin, a synthetic derivative of lipstatin produced by Streptomyces toxytricini) is a pancreatic and gastric lipase inhibitor, which reduces the absorption of dietary fat, developed as an anti-obesity drug. Orlistat inhibiting fatty acid synthase, a lipogenic enzyme that catalyzes fatty acid synthesis, has been shown to decrease proliferation and increase apoptosis in various tumor cell lines [47,48,49,50,51].
There are currently very few studies on the effects of the combination of naltrexone/bupropion, short-term phentermine and phentermine/topiramate focused on cancer effects in obesity. In an 8-week intervention study of the effects of the Mediterranean diet and naltrexone/bupropion therapy, a fixed-dose combination product containing naltrexone, an opioid antagonist, and bupropion, an aminoketone antidepressant, in breast cancer survivors overweight or obese did not show superior effects when compared to the Mediterranean diet alone [52,53].
Liraglutide and semaglutide are two GLP-1 receptor agonists (GLP-1RAs) formerly known as antidiabetic drugs for the treatment of T2DM and currently approved, in higher dosages, for chronic weight management in addition to diet and physical activity in patients with obesity (BMI ≥ 30 kg/m2) or who are overweight (BMI ≥ 27 kg/m2) with at least one weight-related comorbidity (such as hypertension, T2DM, dyslipidemia): liraglutide 3 mg subcutaneous daily (approved for obesity in adults and adolescents 12 years and older) and semaglutide 2.4 mg subcutaneous weekly [54,55,56,57].
GLP-1 is an incretin hormone secreted by gastrointestinal L cells in response to nutrients in the lumen; it accounts for up to 70% of insulin secretion in response to nutrient intake, and it also lowers glucagone secretion while minimizing hypoglycemia. GLP-1 is capable of suppressing appetite and delaying gastric emptying, both of which are responsible for its slimming effects [58]. GLP-1 inhibits food intake, mediating appetite suppression by directly stimulating POMC/CART neurons and indirectly inhibiting neurotransmission in neurons expressing neuropeptide Y and agouti-related peptide via GABA-dependent signaling [59]. GLP-1 half-life is very short (less than 2 min), and it is rapidly degraded by enzymes (DPP-IV and neutral endopeptidase -NEP). Semaglutide and liraglutide are modified long-acting analogues of native GLP-1 (Aib 8, Arg 34 and Arg 34 substitutions, respectively). With the addition of an albumin-binding C16 fatty acid side chain, the half-life of liraglutide is 13–15 h. The half-life of Semaglutide is 165 h, resulting from an amino acid replacement (preventing DPP-IV degradation) and the addition of a C18 fatty diacid. Additionally, semaglutide has positive effects on hedonic aspects and reward-related behaviors of food intake (e.g., reducing cravings and lowering altered food preference) [45,56,60]. However, they are costly and may have adverse effects in some individuals, the most common mainly at the beginning of the treatment cycle and consisting of gastrointestinal disturbances (including nausea, diarrhea, vomiting, constipation), which are mostly mild, transient and dose-dependent but may lead to discontinuation of treatment in some patients [58].
In addition to diet and physical activity, liraglutide is capable of producing at least 5–10% weight loss compared to the placebo, and semaglutide is capable of producing a mean reduction of 14.9% in body weight from baseline, semaglutide being more effective than liraglutide in weight loss (10% or more: 70.9% of participants vs. 25.6%, 15% or more: 55.6% vs. 12.0% and 20% or more: 38.5% vs. 6.0% respectively; all p < 0.001) [61,62,63].
GLP1-receptors (GLP-1Rs) are widely distributed in the body, and in addition to the effects mentioned above, they have multiple biological effects: cardioprotective, neuroprotective, reduction in neuroinflammation and fat deposition, appetite suppression and gastric emptying delay [64].
Notably, liraglutide compared to the placebo in patients at high risk of cardiovascular disease and without T2DM was able to significantly reduce visceral adipose tissue (−12.5% vs. placebo −1.6%) and levels of CRP (−38%) [65].
Semaglutide, on the other hand, appears to have positive effects on skeletal muscle; can significantly reduce body weight and intramuscular fat accumulation; promote muscle protein synthesis; increase the relative proportion of skeletal muscle and improve muscle function in obese mice by decreasing the levels of triglycerides, serum cholesterol, low-density lipoprotein, high-density lipoprotein, TNF-α, IL-6, IL-1β and fasting insulin resistance index (HOMA-IR) and significantly improving type I/type II muscle fiber ratio, total muscle fiber area, muscle fiber density, sarcomere length and mitochondrial number and area [66].
GLP1-RAs (receptor agonists) also show interesting perspectives on tumor development and prognosis. Meta-analyses and systematic review of clinical trials demonstrated that, in obese patients (with or without T2DM), GLP-1RAs do not increase the risk of breast cancer, pancreatic cancer, thyroid cancer and malignancies in general [67,68,69], although some authors underscore concerns about potential tumor-related adverse effects when combining GLP-1RAs with DPP-IV inhibitors [70].
Furthermore, there is still controversy regarding the possible association between GLP1-RAs and thyroid cancer risk: some authors showed correlations while others showed no correlation, also pointing out potential surveillance bias [71,72,73]. However, although there is no clear evidence of the development of c-cell malignancies, it should be noted that GLP-1RAs are currently not recommended in patients with family or personal history of medullary thyroid cancer, as well as multiple endocrine neoplasia syndrome type 2 [74,75]. Further data are expected in the coming years from targeted clinical trials [76].
Randomized clinical trials reported the incidence of malignant neoplasms in overweight or obese adults as 0.7% (3/407; basal cell carcinoma, breast cancer and papillary thyroid cancer) and 1.1% (6/535; breast neoplasms, endometrial adenocarcinoma, marginal zone lymphoma and melanoma) in the semaglutide group and 0.5% (1/204; invasive lobular breast carcinoma) and 0.4% (1/268; metastatic lung cancer) in the control group, respectively [77,78]. In a recent randomized clinical trial comparing liraglutide to semaglutide, malignancies occurred in 2.4% with semaglutide (3/126; basal cell carcinoma, clear cell renal cell carcinoma and invasive ductal breast carcinoma) in 2.4% with liraglutide (3/127; basal cell carcinoma, invasive ductal BC and invasive lobular breast cancer) and 1.2% with placebo (1/85; invasive ductal breast cancer) [63].

4.1. GLP-1 Receptor Agonists and Breast Cancer

Experimental studies investigated the effects of GLP-1 RAs on breast cancer. GLP-1R is expressed in human breast cancer tissue and cell lines. By inhibiting nuclear translocation of NF-κB and target gene expression, GLP-1RA exenadin-4 dose-dependently reduced the growth of breast cancer cell lines in vitro as well as in vivo in transplanted athymic nude mice. In contrast, a DPP-IV inhibitor (linagliptin) did not affect breast cancer cell proliferation, suggesting that GLP-1 might attenuate cell proliferation through GLP-1R activation [79].
Liraglutide in breast cancer cell lines is able to modulate epigenetics, reducing cell viability, migration, DNA methyltransferase activity, decreasing the DNA methylation profile for CDH1, ESR1 and ADAM33 gene promoter regions and, consequently, increasing their expression. Furthermore, liraglutide and the combination treatment of liraglutide and paclitaxel or methotrexate was shown to be effective in reducing tumor growth in in vivo studies by modulating CDH1 and ADAM33 gene expression in mice, suggesting liraglutide as a possible treatment for breast cancer [80].
Liraglutide, in a dose-dependent manner, was able to reduce proliferation and increase apoptosis in human breast cancer cell line MCF 7 cells compared with the control group by inhibiting the expression of miRNA-27a, which subsequently increases the expression of adenosine monophosphate-activated protein kinase (AMPKα2 protein), the authors suggest the experimental basis for clinical breast cancer treatment strategies [81].
Liraglutide has been shown to have anti-proliferative effects on MCF-7 human breast cancer cells cultured in obese adipose tissue-derived stem cells-conditioned medium (ADSCs-CM -the multipotent mesenchymal lineage implicated in breast cancer development, invasion and metastasis, by the secretion of cytokines, adipokines, interleukins, tumor necrosis factor-alpha and growth factors that promote growth, migration and invasion of breast cancer cells). The drug was able to decrease the levels of inflammatory mediators, suppress the pro-proliferative effects of leptin and enhance the anti-proliferative effects of adiponectin in ADSCs-CMs. Therefore, the authors suggest that liraglutide could mitigate breast cancer cell growth in obese subjects [82].
However, liraglutide at higher concentrations in an approximate toxicological context in human triple negative breast cancer cells (MDA-MB-231 and MDA-MB-468) and in transplanted tumors was shown to be able to promote progression through the signal NOX4/ROS/VEGF pathway after GLP-1R activation, suggesting attention to dosages and breast cancer phenotype [83].
In the SCALE trials evaluating the effect of liraglutide in the treatment of obesity, a non-significant higher incidence of breast cancer was observed in the treatment group (15 vs. 3 events; incidence, 4.36 vs. 1.80 events for 1000 person-years) in women who had greater weight loss in the first year of treatment and thus partially attributed to detection bias due to facilitation of mass detection during breast exams [61]. In the UK Clinical Practice Research Datalink national database, the use of GLP-1RAs was not associated with an increased risk of breast cancer over an average follow-up of 3.5 years [84]. A recent systematic review and meta-analysis assessing the incidence of breast cancer in subjects treated with GLP-1RAs found no increased risk [67].

4.2. GLP-1 Receptor Agonists and Prostate Cancer

Interesting perspectives between GLP1-RAs use and prostate cancer are emerging. The randomized, double-blind, controlled LEADER study (Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results) in a secondary outcome analysis showed a lower incidence of prostate cancer in the liraglutide group (n = 26) compared to the placebo (n = 47) with HR 0.54 (95% CI: 0.34–0.88) [85]. Lu et al., in a large UK Clinical Practice Research Datalink patient cohort (2007 to 2019), found that GLP-1RAs were associated with a reduced risk of prostate cancer compared with sulfonylurea use (rates incidence = 156.4 vs. 232.0 per 100,000 person-years, respectively, HR = 0.65, 95% CI = 0.43, 0.99) [86].
Interestingly, GLP-1Rs are expressed in human prostate cancer tissue obtained by radical prostatectomy from patients with nondiabetic prostate cancer; they are co-located with P504S/α-methylacyl-CoA racemase, a cytoplasmic protein identified as a sensitive and specific positive marker for prostate carcinoma. GLP1-RA Exenedin-4 (Ex-4), in a dose-dependent manner, significantly reduced prostate cancer proliferation in in vitro cell lines, androgen dependent LNCap (the strongest effect) and androgen independent PC3 and DU145 in accordance with the abundant expression of GLP-1R in LNCap cells. Indeed, this anti-proliferative effect was abolished by GLP-1R antagonist or GLP-1R knockdown. Ex-4 attenuated in vivo prostate cancer growth induced by transplantation of LNCap cells into athymic mice, significantly reducing tumor expression of P504S, Ki67 and phosphorylated ERK-MAPK mediated by the cAMP-PKA pathway [87].
Some authors, using the GLP-1RA Ex-4, observed, via the PI3K/AKT/mTOR pathway, a reduction in the resistance of prostate cancer cells to the androgen-receptor inhibitor enzalutamide with a greater effect observed in advanced cancers. The increased chemosensitivity of cancer cells could be due to indirect inhibition of the tumor migration, invasion and growth [88].
In human prostate cancer cells (PC3 and LNCap), Ex-4 is able to promote the anti-proliferative effects of radiation via the activation of AMPK phosphorylation and the subsequent inhibition of phosphorylated mTOR, cyclin B and p34cdc2 protein kinase activation [89].
By binding with GLP-1R, liraglutide also significantly inhibited cell proliferation and induced cell apoptosis in LNCap prostate cancer cell lines by regulating the P38 pathway [90]. Liraglutide, in combination with Docetaxel, a first-line chemotherapy agent in metastatic castration-resistant prostate carcinoma, caused cell cycle arrest in LNCaP and induced apoptosis via the ERK/MAPK and AKT/PI3K pathways, thus suggesting new therapeutic strategies for lower therapeutic doses of docetaxel and, therefore, its resistance and toxicity [91]. Forced overexpression of GLP-1R using a lentiviral vector (ALVA-41-GLP-1R cells) was able to attenuate prostate cancer cell proliferation by inhibiting cell cycle progression in vitro and in vivo; the authors concluded that GLP-1R activation could be a potential therapy for prostate cancer [92].

4.3. GLP-1 Receptor Agonists and Pancreatic Cancer

Recent meta-analyses and reviews evaluating the association between GLP1-RAs and pancreatic cancer have found no significant associations in the use of incretin-based therapies and increased risk of pancreatic cancer [93,94]. On the contrary, it should be considered that obesity could be a risk factor for the development of pancreatic carcinoma [95]. Indeed, preclinical experimental data have shown beneficial effects of GLP1-RAs on pancreatic cancer cell lines. Zhao et al. [96], in human pancreatic tumor tissues, showed lower levels or lack of expression of GLP-1R than in pancreatic tissues adjacent to the tumor. GLP-1R negative expression was more frequent in advanced tumors and was associated with a poor prognosis. GLP-1R activation by liraglutide produces an antitumor effect on human pancreatic cancer cells through inhibition of the PI3K/AKT pathway, suggesting beneficial effects of GLP-1-based therapies in this tumor [96]. GLP-1RAs could also activate cAMP, subsequently inhibiting AKT and ERK1/2 signaling pathways and causing apoptosis and inhibition of proliferation in a human pancreatic cancer cell line in vitro and reducing xenograft pancreatic tumor growth in vivo, contributing to address the long-term safety issues of GLP-1-based therapies [97]. In addition, liraglutide, which regulates the NF-kB signaling pathway and the downstream ATP-binding cassette subfamily G member 2 (ABCG2), showed significant antiproliferative and pro-apoptotic effects in gemcitabine-resistant human pancreatic cancer cells resistant to various drugs, increasing the chemosensitivity in both in vitro and in vivo experiments [98].

4.4. GLP-1 Receptor Agonists and Gynecological Cancers

On gynecological cancers, GLP-1RAs exert an inhibitory effect on cervical cancer growth in T2DM; treatment with the antidiabetic GLP1-RAs attenuated hyperglycemia might promote cancer growth by increasing proteasome alpha subunit 2 (PSMA2) expression [99].
Upregulation of GLP1-R is able to block the growth of endometrial cancer cells (Ishikawa and RL95-2) by activating the cAMP/PKA signaling pathway [100]. Furthermore, liraglutide was able to suppress the progression of endometrial cancer in human Ishikawa endometrial cancer cells. GLP-1R expression is associated with estrogen and progesterone receptors positive status. Higher GLP-1R expression may be associated with a better prognosis in patients with endometrial cancer, and the use of liraglutide to target autophagy in endometrial cancer cells may be a potential new treatment for endometrial cancer [101].
GLP-1R is expressed in both human ovarian cancer tissues and cell lines. GLP1-R activation by Ex-4 is able to produce antitumor effects in human ovarian cancer cells by reducing growth, migration and invasion and promoting apoptosis through inhibition of the PI3K/Akt extension. GLP-1 activation is also able to attenuate tumor formation by ovarian cancer cells in vivo in nude mice [102].
It is known that tumor metastases are facilitated by the remodeling of the extracellular matrix at the tumor site and also through an alteration of the balance between metalloproteinases (MMPs), a family of proteolytic enzymes that contribute to the breakdown of extracellular matrix components, and the tissue inhibitors of metalloproteinases (TIMPs), promoting tumor growth, invasion and metastasis. MMP production also enhances the angiogenic response by VEGF expression potentially affecting the metastatic potential of cancer cells. In SKOV-3 and CAOV-3 human ovarian cancer cells, GLP1-RA (Ex-4) was able to reduce the expression of the key metalloproteinases MMP-2 and MMP-9, to modulate their inhibitors TIMP-1 and TIMP-2, to inhibit migration and induce apoptosis by reducing the production of adhesion molecules and inhibiting apoptosis in TNF-α-stimulated endothelial cells [103,104].

4.5. GLP-1 Receptor Agonists and Colorectal Cancer

In CT26 colon cancer cells, GLP-1R activation reduces cell growth and survival, increasing intracellular cAMP levels and inhibiting the activity of glycogen synthase kinase 3 and ERK1/2, a member of the mitogen activated protein kinase family; moreover, GLP-1R activation augmented apoptosis is induced by irinotecan, a topoisomerase I inhibitor [105].
By inhibiting the PI3K/Akt/mTOR signaling pathway, liraglutide could block the cell cycle, reducing cell proliferation, migration and invasion and promoting apoptosis in colorectal cancer cell lines. Liraglutide reduced cyclin D1, an important protein in the cell cycle that regulates the transition from G1 to S phase in the cell proliferation cycle, and MMP-11, one of the matrix metalloproteinases, which is the main enzyme that causes degradation of the basement membrane and of the extracellular matrix [106].
GLP-1R also appears to play a role in the inhibition of glioma cell survival, migration, proliferation and invasion in a GLP-1R/SIRT3 pathway-dependent manner [107].

5. Conclusions

It is now widely known that overweight and obesity are risk factors for cancer development and prognosis. Therefore, the prevention and treatment of obesity appears to be of increasing value as a treatment of a modifiable factor causing cancer. Extensive ongoing research about the effects of obesity treatment, especially focused on GLP1-RAs, on occurrence and development of cancer have highlighted the increased potential for their use in the clinical setting of cancer treatment, first reducing obesity/ABCD and, second, influencing cancer cells by altering proliferation, apoptosis, extracellular matrix remodeling and the response to chemotherapy. Further studies, including large, long term clinical trials are needed to properly evaluate the relationship between anti-obesity treatment and cancer risk and prognosis, and the long-term efficacy of these agents on weight control versus lifestyle or diet changes. These studies require strong collaboration between oncologists and experts in the treatment of obesity.

Author Contributions

Conceptualization, C.M.C. and A.D.; methodology and data curation, C.M.C. and I.M.B.; writing—original draft preparation, C.M.C.; writing—review and editing, I.M.B. and A.D.; supervision, A.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank the staff of the Oncology and Endocrinology Unit for their remarkable work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Global Obesity Observator. Obesity Atlas 2022. Available online: https://data.worldobesity.org/publications/?cat=15 (accessed on 27 February 2023).
  2. Bray, G.A.; Kim, K.K.; Wilding, J.P.H. Obesity: A chronic relapsing progressive disease process. A position statement of the World Obesity Federation. Obes. Rev. 2017, 18, 715–723. [Google Scholar] [CrossRef]
  3. WHO European Regional Obesity Report 2022. Available online: https://apps.who.int/iris/bitstream/handle/10665/353747/9789289057738-eng.pdf (accessed on 27 February 2023).
  4. 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, 794–798. [Google Scholar] [CrossRef]
  5. Furer, A.; Afek, A.; Sommer, A.; Keinan-Boker, L.; Derazne, E.; Levi, Z.; Tzur, D.; Tiosano, S.; Shina, A.; Glick, Y.; et al. Adolescent obesity and midlife cancer risk: A population-based cohort study of 2·3million adolescents in Israel. Lancet Diabetes Endocrinol. 2020, 8, 216–225. [Google Scholar] [CrossRef] [PubMed]
  6. Kohls, M.; Freisling, H.; Charvat, H.; Soerjomataram, I.; Viallon, V.; Davila Batista, V.; Kaaks, R.; Turzanski-Fortner, R.; Aleksandrova, K.; Schulze, M.B.; et al. Impact of cumulative body mass index and cardiometabolic diseases on survival among patients with colorectal and breast cancer: A multi centre cohort study. BMC Cancer 2022, 22, 546. [Google Scholar] [CrossRef] [PubMed]
  7. Mechanick, J.I.; Hurley, D.L.; Garvey, W.T. Adiposity-Based Chronic Disease as a new diagnostic term: The American Association of Clinical Endocrinologists and American College of Endocrinology position statement. Endocr. Pract. 2017, 23, 372–378. [Google Scholar] [CrossRef] [PubMed]
  8. Iyengar, N.M.; Arthur, R.; Manson, J.E.; Chlebowski, R.T.; Kroenke, C.H.; Peterson, L.; Cheng, T.-Y.D.; Feliciano, E.C.; Lane, D.; Luo, J.; 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, 155–163. [Google Scholar] [CrossRef] [PubMed]
  9. Absence of Excess Body Fatness. IARC Handb. Cancer Prev. 2018, 16, 1–646. Available online: http://publications.iarc (accessed on 27 February 2023).
  10. Seki, T.; Yang, Y.; Sun, X.; Lim, S.; Xie, S.; Guo, Z.; Xiong, W.; Kuroda, M.; Sakaue, H.; Hosaka, K.; et al. Brown-fat-mediated tumour suppression by cold-altered global metabolism. Nature 2022, 608, 421–428. [Google Scholar] [CrossRef]
  11. Harris, B.H.L.; Macaulay, V.M.; Harris, D.A.; Klenerman, P.; Karpe, F.; Lord, S.R.; Harris, A.L.; Buffa, F.M. Obesity: A perfect storm for carcinogenesis. Cancer Metastasis Rev. 2022, 41, 491–515. [Google Scholar] [CrossRef]
  12. Simões, H.G.; Corrêa, L.H.; Magalhães, K.G. The Impact of Adipose Tissue-Derived miRNAs in Metabolic Syndrome, Obesity, and Cancer. Front. Endocrinol. 2020, 11, 563816. [Google Scholar] [CrossRef]
  13. Guzzardi, M.A.; Pugliese, G.; Bottiglieri, F.; Pelosini, C.; Muscogiuri, G.; Barrea, L.; Savastano, S.; Colao, A. Obesity-related gut hormones and cancer: Novel insight into the pathophysiology. Internat. J. Obes. 2021, 45, 1886–1898. [Google Scholar] [CrossRef]
  14. Friedenreich, C.M.; Ryder-Burbidge, C.; McNeil, J. Physical activity, obesity and sedentary behavior in cancer aetiology: Epidemiologic evidence and biologic mechanisms. Mol. Oncol. 2021, 15, 790–800. [Google Scholar] [CrossRef] [PubMed]
  15. Luo, J.; Hendryx, M.; Manson, J.E.; Figueiredo, J.C.; LeBlanc, E.S.; Barrington, W.; Rohan, T.E.; Howard, B.V.; Reding, K.; Ho, G.Y.; et al. Intentional weight loss and obesity-related cancer risk. JNCI Cancer Spectr. 2019, 3, pkz054. [Google Scholar] [CrossRef] [PubMed]
  16. Anderson, A.S.; Martin, R.M.; Renehan, A.G.; Cade, J.; Copson, E.R.; Cross, A.J.; Grimmett, C.; Keaver, L.; King, A.; Riboli, E.; et al. On behalf of the UK NIHR Cancer and Nutrition Collaboration (Population Health Stream) Cancer survivorship, excess body fatness and weight-loss intervention—Where are we in 2020? Br. J. Cancer 2021, 124, 1057–1065. [Google Scholar] [CrossRef]
  17. Jiralerspong, S.; Goodwin, P.J. Obesity and Breast Cancer Prognosis: Evidence, Challenges, and Opportunities. J. Clin. Oncol. 2016, 34, 4203–4216. [Google Scholar] [CrossRef]
  18. Lohmann, A.E.; Soldera, S.V.; Pimentel, I.; Ribnikar, D.; Ennis, M.; Amir, E.; Goodwin, P.J. Association of Obesity with Breast Cancer Outcome in Relation to Cancer Subtypes: A Meta-Analysis. J Natl Cancer Inst. 2021, 113, 1465–1475. [Google Scholar] [CrossRef] [PubMed]
  19. Cava, E.; Marzullo, P.; Farinelli, D.; Gennari, A.; Saggia, C.; Riso, S.; Prodam, F. Breast Cancer Diet “BCD”: A Review of Healthy Dietary Patterns to Prevent Breast Cancer Recurrence and Reduce Mortality. Nutrients 2022, 14, 476. [Google Scholar] [CrossRef]
  20. Lake, B.; Damery, S.; Jolly, K. Effectiveness of weight loss interventions in breast cancer survivors: A systematic review of reviews. BMJ Open 2022, 12, e062288. [Google Scholar] [CrossRef]
  21. Kitson, S.; Ryan, N.; MacKintosh, M.L.; Edmondson, R.; Duffy, J.M.; Crosbie, E.J. Interventions for weight reduction in obesity to improve survival in women with endometrial cancer. Cochrane Database Syst. Rev. 2018, 1, CD012513. [Google Scholar] [CrossRef]
  22. Rivera-Izquierdo, M.; Martínez-Ruiz, V.; Jiménez-Moleón, J.J. Recommendations on Weight Loss and Healthy Lifestyle in Prostate Cancer Clinical Guidelines: A Systematic Review. Int. J. Environ. Res. Public Health 2022, 19, 1452. [Google Scholar] [CrossRef]
  23. Aminian, A.; Wilson, R.; Al-Kurd, A.; Tu, C.; Milinovich, A.; Kroh, M.; Rosenthal, R.J.; Brethauer, S.A.; Schauer, P.R.; Kattan, M.W.; et al. Association of Bariatric Surgery with Cancer Risk and Mortality in Adults with Obesity. JAMA 2022, 327, 2423–2433. [Google Scholar] [CrossRef] [PubMed]
  24. Goodwin, P.J.; Segal, R.J.; Vallis, M.; Ligibel, J.A.; Pond, G.R.; Robidoux, A.; Findlay, B.; Gralow, J.R.; Mukherjee, S.D.; Levine, M.; et al. The LISA randomized trial of a weight loss intervention in postmenopausal breast cancer. NPJ Breast Cancer 2020, 21, 6. [Google Scholar] [CrossRef] [PubMed]
  25. Rivera-Izquierdo, M.; Pérez de Rojas, J.; Martínez-Ruiz, V.; Pérez-Gómez, B.; Sánchez, M.J.; Khan, K.S.; Jiménez-Moleón, J.J. Obesity as a Risk Factor for Prostate Cancer Mortality: A Systematic Review and Dose-Response Meta-Analysis of 280,199 Patients. Cancers 2021, 13, 4169. [Google Scholar] [CrossRef]
  26. Rivera-Izquierdo, M.; Pérez de Rojas, J.; Martínez-Ruiz, V.; Arrabal-Polo, M.Á.; Pérez-Gómez, B.; Jiménez-Moleón, J.J. Obesity and biochemical recurrence in clinically localised prostate cancer: A systematic review and meta-analysis of 86,490 patients. Prostate Cancer Prostatic Dis. 2022, 25, 411–421. [Google Scholar] [CrossRef] [PubMed]
  27. Nathan, D.M.; Barrett Connor, E.; Crandall, J.P.; Edelstein, S.L.; Goldberg, R.B.; Horton, E.S.; Knowler, W.C.; Mather, K.J.; Orchard, T.J.; Pi-Sunyer, X.; et al. Long-term effects of lifestyle intervention or metformin on diabetes development and microvascular complications over 15-year follow-up: The Diabetes Prevention Program Outcomes Study Diabetes Prevention Program Research Group. Lancet Diabetes Endocrinol. 2015, 3, 866–875. [Google Scholar] [CrossRef]
  28. Gandini, S.; Puntoni, M.; Heckman-Stoddard, B.M.; Dunn, B.K.; Ford, L.; DeCensi, A.; Szabo, E. Metformin and cancer risk and mortality: A systematic review and meta-analysis taking into account biases and confounders. Cancer Prev. Res. 2014, 7, 867–885. [Google Scholar] [CrossRef]
  29. Goodwin, P.J.; Chen, B.E.; Gelmon, K.A.; Whelan, T.J.; Ennis, M.; Lemieux, J.; Ligibel, J.A.; Hershman, D.L.; Mayer, I.A.; Hobday, T.J.; et al. Effect of Metformin vs Placebo on Invasive Disease-Free Survival in Patients with Breast Cancer: The MA.32 Randomized Clinical Trial. JAMA 2022, 327, 1963–1973. [Google Scholar] [CrossRef]
  30. Palazzolo, G.; Mollica, H.; Lusi, V.; Rutigliani, M.; Di Francesco, M.; Pereira, R.C.; Filauro, M.; Paleari, L.; DeCensi, A.; Decuzzi, P. Modulating the Distant Spreading of Patient-Derived Colorectal Cancer Cells via Aspirin and Metformin. Transl. Oncol. 2020, 13, 100760. [Google Scholar] [CrossRef]
  31. Ahn, H.K.; Lee, Y.H.; Koo, K.C. Current Status and Application of Metformin for Prostate Cancer: A Comprehensive Review. Int. J. Mol. Sci. 2020, 12, 8540. [Google Scholar] [CrossRef]
  32. Bonanni, B.; Puntoni, M.; Cazzaniga, M.; Pruneri, G.; Serrano, D.; Guerrieri-Gonzaga, A.; Gennari, A.; Trabacca, M.S.; Galimberti, V.; Veronesi, P.; et al. Dual effect of metformin on breast cancer proliferation in a randomized presurgical trial. J. Clin. Oncol. 2012, 30, 2593–2600. [Google Scholar] [CrossRef]
  33. DeCensi, A.; Puntoni, M.; Guerrieri-Gonzaga, A.; Cazzaniga, M.; Serrano, D.; Lazzeroni, M.; Vingiani, A.; Gentilini, O.; Petrera, M.; Viale, G.; et al. Effect of Metformin on Breast Ductal Carcinoma In Situ Proliferation in a Randomized Presurgical Trial. Cancer Prev. Res. 2015, 8, 888–894. [Google Scholar] [CrossRef]
  34. Heckman-Stoddard, B.M.; DeCensi, A.; Sahasrabuddhe, V.V.; Ford, L.G. Repurposing metformin for the prevention of cancer and cancer recurrence. Diabetologia 2017, 60, 1639–1647. [Google Scholar] [CrossRef] [PubMed]
  35. Cameron, A.R.; Morrison, V.L.; Levin, D.; Mohan, M.; Forteath, C.; Beall, C.; McNeilly, A.D.; Balfour, D.J.K.; Savinko, T.; Wong, A.K.F.; et al. Anti-inflammatory effects of metformin irrespective of diabetes status. Circ. Res. 2016, 119, 652–665. [Google Scholar] [CrossRef] [PubMed]
  36. Zhou, G.; Myers, R.; Li, Y.; Chen, Y.; Shen, X.; Fenyk-Melody, J.; Wu, M.; Ventre, J.; Doebber, T.; Fujii, N.; et al. Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Investig. 2001, 108, 1167–1174. [Google Scholar] [CrossRef]
  37. Saxton, R.A.; Sabatini, D.M. mTOR Signaling in Growth, Metabolism, and Disease. Cell 2017, 9, 960–976. [Google Scholar] [CrossRef] [PubMed]
  38. Wen, J.; Yi, Z.; Chen, Y.; Huang, J.; Mao, X.; Zhang, L.; Zeng, Y.; Cheng, Q.; Ye, W.; Liu, Z.; et al. Efficacy of metformin therapy in patients with cancer: A meta analysis of 22 randomised controlled trials. BMC Med. 2022, 20, 402. [Google Scholar] [CrossRef]
  39. Garvey, W.T.; Mechanick, J.I.; Brett, E.M.; Garber, A.J.; Hurley, D.L.; Jastreboff, A.M.; Nadolsky, K.; Pessah-Pollack, R.; Plodkowski, R. American Association of Clinical Endocrinologists and American College of Endocrinology Comprehensive Clinical Practice Guidelines for Medical Care of Patients with Obesity. Endocr. Pract. 2016, 22 (Suppl. S3), 1–203. [Google Scholar] [CrossRef] [PubMed]
  40. Bray, G.A.; Edelstein, S.L.; Crandall, J.P.; Aroda, V.R.; Franks, P.W.; Fujimoto, W.; Horton, E.; Jeffries, S.; Montez, M.; Mudaliar, S.; et al. Long-term safety, tolerability, and weight loss associated with metformin in the Diabetes Prevention Program Outcomes Study. Diabetes Care 2012, 35, 731–737. [Google Scholar] [CrossRef]
  41. Yerevanian, A.; Soukas, A.A. Metformin: Mechanisms in Human Obesity and Weight Loss. Curr. Obes. Rep. 2019, 8, 156–164. [Google Scholar] [CrossRef]
  42. Huang, Y.C.; Hsu, C.C.; Lin, W.C.; Yin, T.K.; Huang, C.W.; Wang, P.W.; Chang, H.H.; Chiu, N.T. Effects of metformin on the cerebral metabolic changes in type 2 diabetic patients. Sci. World J. 2014, 2014, 694326. [Google Scholar] [CrossRef]
  43. Buse, J.B.; DeFronzo, R.A.; Rosenstock, J.; Kim, T.; Burns, C.; Skare, S.; Baron, A.; Fineman, M. The Primary Glucose-Lowering Effect of Metformin Resides in the Gut, Not the Circulation: Results from Short-term Pharmacokinetic and 12-Week Dose-Ranging Studies. Diabetes Care 2016, 39, 198–205. [Google Scholar] [CrossRef] [PubMed]
  44. Lindsay, J.R.; Duffy, N.A.; McKillop, A.M.; Ardill, J.; O’Harte, F.P.M.; Flatt, P.R.; Bell, P.M. Inhibition of dipeptidyl peptidase IV activity by oral metformin in type 2 diabetes. Diabetic Med. 2005, 22, 654–657. [Google Scholar] [CrossRef] [PubMed]
  45. Angelidi, A.M.; Belanger, M.J.; Kokkinos, A.; Koliaki, C.C.; Mantzoros, C.S. Novel Noninvasive Approaches to the Treatment of Obesity: From Pharmacotherapy to Gene Therapy. Endocr. Rev. 2022, 43, 507–557. [Google Scholar] [CrossRef]
  46. Tak, Y.J.; Lee, S.Y. Long-Term Efficacy and Safety of Anti-Obesity Treatment: Where Do We Stand? Curr. Obes. Rep. 2021, 10, 14–30. [Google Scholar] [CrossRef] [PubMed]
  47. Sokolowska, E.; Presler, M.; Goyke, E.; Milczarek, R.; Swierczynski, J.; Sledzinski, T. Orlistat Reduces Proliferation and Enhances Apoptosis in Human Pancreatic Cancer Cells (PANC-1). Anticancer Res. 2017, 37, 6321–6327. [Google Scholar] [CrossRef] [PubMed]
  48. Menendez, J.A.; Vellon, L.; Lupu, R. Antitumoral actions of the anti-obesity drug orlistat (XenicalTM) in breast cancer cells: Blockade of cell cycle progression, promotion of apoptotic cell death and PEA3-mediated transcriptional repression of Her2/neu (erbB-2) oncogene. Ann. Oncol. 2005, 16, 1253–1267. [Google Scholar] [CrossRef]
  49. Czumaj, A.; Zabielska, J.; Pakiet, A.; Mika, A.; Rostkowska, O.; Makarewicz, W.; Kobiela, J.; Sledzinski, T.; Stelmanska, E. In Vivo Effectiveness of Orlistat in the Suppression of Human Colorectal Cancer Cell Proliferation. Anticancer Res. 2019, 39, 3815–3822. [Google Scholar] [CrossRef]
  50. Cao, B.; Deng, H.; Cui, H.; Zhao, R.; Li, H.; Wei, B.; Chen, L. Knockdown of PGM1 enhances anticancer effects of orlistat in gastric cancer under glucose deprivation. Cancer Cell Int. 2021, 21, 481–495. [Google Scholar] [CrossRef]
  51. Shueng, P.W.; Chan, H.W.; Lin, W.C.; Kuo, D.Y.; Chuang, H.Y. Orlistat Resensitizes Sorafenib-Resistance in Hepatocellular Carcinoma Cells through Modulating Metabolism. Int. J. Mol. Sci. 2022, 23, 6501. [Google Scholar] [CrossRef]
  52. Shi, Q.; Wang, Y.; Hao, Q.; Vandvik, P.O.; Guyatt, G.; Li, J.; Chen, Z.; Xu, S.; Shen, Y.; Sun, F. Pharmacotherapy for adults with overweight and obesity: A systematic review and network meta-analysis of randomised controlled trials. Lancet 2022, 399, 259–269. [Google Scholar] [CrossRef]
  53. Cho, A.-R.; Choi, W.-J.; Kwon, Y.-J.; Sun Lee, H.; Ahn, S.G.; Lee, J.-W. Mediterranean Diet and Naltrexone/Bupropion Treatment for Weight Loss in Overweight and Obese Breast Cancer Survivors and Non-Cancer Participants: A Pilot Randomized Controlled Trial. Diabetes Metab. Syndr. Obes. 2020, 13, 3325–3335. [Google Scholar] [CrossRef]
  54. Available online: https://www.ema.europa.eu/en/documents/product-information/saxenda-epar-product-information_en.pdf (accessed on 7 August 2023).
  55. Available online: https://www.ema.europa.eu/en/documents/overview/wegovy-epar-medicine-overview_en.pdf (accessed on 7 August 2023).
  56. Knudsen, L.B.; Lau, J. The discovery and development of liraglutide and semaglutide. Front. Endocrinol. 2019, 10, 155. [Google Scholar] [CrossRef] [PubMed]
  57. Guo, X.; Zhou, Z.; Lyu, X.; Xu, H.; Zhu, H.; Pan, H.; Wang, L.; Yang, H.; Gong, F. The Antiobesity Effect and Safety of GLP-1 Receptor Agonist in Overweight/Obese Patients Without Diabetes: A Systematic Review and Meta-Analysis. Horm. Metab. Res. 2022, 54, 458–471. [Google Scholar] [CrossRef] [PubMed]
  58. Drucker, D.J. Mechanisms of Action and Therapeutic Application of Glucagon-like Peptide-1. Cell Metab. 2018, 27, 740–756. [Google Scholar] [CrossRef]
  59. Secher, A.; Jelsing, J.; Baquero, A.F.; Hecksher-Sørensen, J.; Cowley, M.A.; Dalbøge, L.S.; Hansen, G.; Grove, K.L.; Pyke, C.; Raun, K.; et al. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J. Clin. Investig. 2014, 124, 4473–4488. [Google Scholar] [CrossRef]
  60. Blundell, J.; Finlayson, G.; Axelsen, M.B.; Flint, A.; Gibbons, C.; Kvist, T.; Hjerpsted, J.B. Effects of once-weekly semaglutide on appetite, energy intake, control of eating, food preference and body weight in subjects with obesity. Diabetes Obes. Metab. 2017, 19, 1242–1251. [Google Scholar] [CrossRef]
  61. Pi-Sunyer, X.; Astrup, A.; Fujioka, K.; Greenway, F.; Halpern, A.; Krempf, M.; Lau, D.C.; le Roux, C.W.; Ortiz, R.V.; Jensen, C.B.; et al. SCALE Obesity and Prediabetes NN8022-1839 Study Group. A Randomized, Controlled Trial of 3.0 mg of Liraglutide in Weight Management. N. Engl. J. Med. 2015, 373, 11–22. [Google Scholar] [CrossRef]
  62. Wilding, J.P.H.; Batterham, R.L.; Calanna, S.; Davies, M.; Van Gaal, L.F.; Lingvay, I.; McGowan, B.M.; Rosenstock, J.; Tran, M.; Wadden, T.A.; et al. STEP 1 Study Group Once-Weekly Semaglutide in Adults with Overweight or Obesity. N. Engl. J. Med. 2021, 384, 989–1002. [Google Scholar] [CrossRef]
  63. Rubino, D.M.; Greenway, F.L.; Khalid, U.; O’Neil, P.M.; Rosenstock, J.; Sørrig, R.; Wadden, T.A.; Wizert, A.; Garvey, W.T. STEP 8 Investigators Clinical Trial. Effect of Weekly Subcutaneous Semaglutide vs Daily Liraglutide on Body Weight in Adults With Overweight or Obesity Without Diabetes: The STEP 8 Randomized Clinical Trial. JAMA 2022, 327, 138–150. [Google Scholar] [CrossRef] [PubMed]
  64. McLean, B.A.; Wong, C.K.; Campbell, J.E.; Hodson, D.J.; Trapp, S.; Drucker, D.J. Revisiting the Complexity of GLP-1 Action from Sites of Synthesis to Receptor Activation. Endocr. Rev. 2021, 42, 101–132. [Google Scholar] [CrossRef]
  65. Neeland, I.J.; Marso, S.P.; Ayers, C.R.; Lewis, B.; Oslica, R.; Francis, W.; Rodder, S.; Pandey, A.; Joshi, P.H. Effects of liraglutide on visceral and ectopic fat in adults with overweight and obesity at high cardiovascular risk: A randomised, double-blind, placebo-controlled, clinical trial. Lancet Diabetes Endocrinol. 2021, 9, 595–605. [Google Scholar] [CrossRef] [PubMed]
  66. Ren, Q.; Chen, S.; Chen, X.; Niu, S.; Yue, L.; Pan, X.; Li, Z.; Chen, X. An Effective Glucagon-Like Peptide-1 Receptor Agonists, Semaglutide, Improves Sarcopenic Obesity in Obese Mice by Modulating Skeletal Muscle Metabolism. Drug Des. Devel. Ther. 2022, 16, 3723–3735. [Google Scholar] [CrossRef] [PubMed]
  67. Piccoli, G.F.; Mesquita, L.A.; Stein, C.; Aziz, M.; Zoldan, M.; Degobi, N.A.H.; Spiazzi, B.F.; Lopes, G.L., Jr.; Colpani, V.; Gerchman, F. Do GLP-1 Receptor Agonists Increase the Risk of Breast Cancer? A Systematic Review and Meta-Analysis. J. Clin. Endocrinol. Metab. 2021, 106, 912–921. [Google Scholar] [CrossRef]
  68. Hu, W.; Song, R.; Cheng, R.; Liu, C.; Guo, R.; Tang, W.; Zhang, J.; Zhao, Q.; Li, X.; Liu, J. Use of GLP-1 Receptor Agonists and Occurrence of Thyroid Disorders: A Meta-Analysis of Randomized Controlled Trials. Front. Endocrinol. 2022, 13, 927859. [Google Scholar] [CrossRef]
  69. Cao, C.; Yang, S.; Zhou, Z. GLP-1 receptor agonists and risk of cancer in type 2 diabetes: An updated meta-analysis of randomized controlled trials. Endocrine 2019, 66, 157–165. [Google Scholar] [CrossRef]
  70. Yang, Z.; Lv, Y.; Yu, M.; Mei, M.; Xiang, L.; Zhao, S.; Li, R. GLP-1 receptor agonist-associated tumor adverse events: A real-world study from 2004 to 2021 based on FAERS. Front. Pharmacol. 2022, 13, 925377. [Google Scholar] [CrossRef] [PubMed]
  71. Wang, J.; Kim, C.H. Differential Risk of Cancer Associated with Glucagon-like Peptide-1 Receptor Agonists: Analysis of Real-world Databases. Endocr. Res. 2022, 47, 18–25. [Google Scholar] [CrossRef]
  72. Funch, D.; Mortimer, K.; Ziyadeh, N.J.; Seeger, J.D.; Zhou, L.; Ng, E.; Ross, D.; Major-Pedersen, A.; Bosch-Traberg, H.; Gydesen, H. Risk of Thyroid Cancer Associated with Use of Liraglutide and Other Antidiabetic Drugs in a US Commercially Insured Population. Diabetes Metab. Syndr. Obes. 2021, 14, 2619–2629. [Google Scholar] [CrossRef]
  73. Gallo, M. Thyroid safety in patients treated with liraglutide. J. Endocrinol. Investig. 2013, 36, 140–145. [Google Scholar] [CrossRef]
  74. Hegedüs, L.; Sherman, S.I.; Tuttle, R.M.; von Scholten, B.J.; Rasmussen, S.; Karsbøl, J.D.; Daniels, G.H. LEADER Publication Committee on behalf of the LEADER Trial Investigators No Evidence of Increase in Calcitonin Concentrations or Development of C-Cell Malignancy in Response to Liraglutide for Up to 5 Years in the LEADER Trial. Diabetes Care 2018, 41, 620–622. [Google Scholar] [CrossRef]
  75. Smits, M.M.; Van Raalte, D.H. Safety of Semaglutide. Erratum in Corrigendum: Safety of Semaglutide. Front. Endocrinol. 2021, 12, 645563. [Google Scholar] [CrossRef]
  76. An Active Surveillance Program for Cases of Medullary Thyroid Carcinoma (MTC). Available online: https://clinicaltrials.gov/ct2/show/NCT01511393 (accessed on 7 August 2023).
  77. Wadden, T.A.; Bailey, T.S.; Billings, L.; Davies, M.; Frias, J.P.; Koroleva, A.; Lingvay, I.; O’Neil, P.M.; Rubino, D.M.; Skovgaard, D. Effect of subcutaneous semaglutide vs placebo as an adjunct to intensive behavioral therapy on body weight in adults with overweight or obesity: The STEP 3 randomized clinical trial. JAMA 2021, 325, 1403–1413. [Google Scholar] [CrossRef]
  78. Rubino, D.; Abrahamsson, N.; Davies, M.; Hesse, D.; Greenway, F.L.; Jensen, C.; Lingvay, I.; Mosenzon, O.; Rosenstock, J.; Rubioet, M.A. Effect of continued weekly subcutaneous semaglutide vs placebo on weight loss maintenance in adults with overweight or obesity: The STEP 4 randomized clinical trial. JAMA 2021, 325, 1414–1425. [Google Scholar] [CrossRef]
  79. Iwaya, C.; Nomiyama, T.; Komatsu, S.; Kawanami, T.; Tsutsumi, Y.; Hamaguchi, Y.; Horikawa, T.; Yoshinaga, Y.; Yamashita, S.; Tanaka, T. Exendin-4, a Glucagonlike Peptide-1 Receptor Agonist, Attenuates Breast Cancer Growth by Inhibiting NF-kb Activation. Endocrinology 2017, 158, 4218–4232. [Google Scholar] [CrossRef] [PubMed]
  80. Chequin, A.; Costa, L.E.; de Campos, F.F.; Moncada, A.D.B.; de Lima, L.T.F.; Sledz, R.L.; Picheth, G.F.; Adami, E.R.; Acco, A.; Gonçalves, M.B.; et al. Antitumoral activity of liraglutide, a new DNMT inhibitor in breast cancer cells in vitro and in vivo. Chem. Biol. Interact. 2021, 1, 349:109641. [Google Scholar] [CrossRef]
  81. Zhao, W.; Zhang, X.; Zhou, Z.; Sun, B.; Gu, W.; Liu, J.; Zhang, H. Liraglutide inhibits the proliferation and promotes the apoptosis of MCF-7 human breast cancer cells through downregulation of microRNA-27a expression. Mol. Med. Rep. 2018, 17, 5202–5212. [Google Scholar] [CrossRef]
  82. Alanteet, A.A.; Attia, H.A.; Shaheen, S.; Alfayez, M.; Alshanawani, B. Anti-Proliferative Activity of Glucagon-Like Peptide-1 Receptor Agonist on Obesity-Associated Breast Cancer: The Impact on Modulating Adipokines’ Expression in Adipocytes and Cancer Cells. Dose Response. 2021, 19, 1559325821995651. [Google Scholar] [CrossRef]
  83. Liu, Z.Z.; Duan, X.X.; Yuan, M.C.; Yu, J.; Han, X.; Lan, L.; Liu, N.-B.W.; Wang, Y.; Qin, J.F. Glucagon-like peptide-1 receptor activation by liraglutide promotes breast cancer through NOX4/ROS/VEGF pathway. Life Sci. 2022, 294, 120370. [Google Scholar] [CrossRef] [PubMed]
  84. Hicks, B.M.; Yin, H.; Yu, O.H.Y.; Pollak, M.N.; Platt, R.W.; Azoulay, L. Glucagon-like peptide-1 analogues and risk of breast cancer in women with type 2 diabetes: Population based cohort study using the UK Clinical Practice Research Datalink. BMJ 2016, 355, i5340. [Google Scholar] [CrossRef]
  85. Nauck, M.A.; Jensen, T.J.; Rosenkilde, C.; Calanna, S.; Buse, J.B. Neoplasms Reported with Liraglutide or Placebo in People with Type 2 Diabetes: Results from the LEADER Randomized Trial. Diabetes Care 2018, 41, 1663–1671. [Google Scholar] [CrossRef] [PubMed]
  86. Lu, S.; Yin, H.; Yu, O.H.Y.; Azoulay, L. Incretin-Based Drugs and the Incidence of Prostate Cancer Among Patients with Type 2 Diabetes. Epidemiology 2022, 33, 563–571. [Google Scholar] [CrossRef]
  87. Nomiyama, T.; Kawanami, T.; Irie, S.; Hamaguchi, Y.; Terawaki, Y.; Murase, K.; Tsutsumi, Y.; Nagaishi, R.; Tanabe, M.; Morinaga, H.; et al. Exendin-4, a GLP-1 Receptor Agonist, Attenuates Prostate Cancer Growth. Diabetes 2014, 63, 3891–3905. [Google Scholar] [CrossRef]
  88. He, W.; Shao, Y.; Yi, Y.; Huang, W.; Feng, G.; Li, J. Exendin-4 Enhances the Sensitivity of Prostate Cancer to Enzalutamide by Targeting Akt Activation. Prostate 2020, 80, 367–375. [Google Scholar] [CrossRef]
  89. He, W.; Li, J. Exendin-4 enhances radiation response of prostate cancer. Prostate 2018, 78, 1125–1133. [Google Scholar] [CrossRef] [PubMed]
  90. Li, X.-N.; Bu, H.-M.; Ma, X.-H.; Lu, S.; Zhao, S.; Cui, Y.-L.; Sun, J. Glucagon-like Peptide-1 Analogues Inhibit Proliferation and Increase Apoptosis of Human Prostate Cancer Cells in vitro. Exp. Clin. Endocrinol. Diabetes 2017, 125, 91–97. [Google Scholar] [CrossRef] [PubMed]
  91. Eftekhari, S.; Montazeri, H.; Tarighi, P. Synergistic Anti-Tumor Effects of Liraglutide, a Glucagon-Like Peptide-1 Receptor Agonist, Along with Docetaxel on LNCaP Prostate Cancer Cell Line. Eur. J. Pharmacol. 2020, 878, 173102. [Google Scholar] [CrossRef]
  92. Shigeoka, T.; Nomiyama, T.; Kawanami, T.; Hamaguchi, Y.; Horikawa, T.; Tanaka, T.; Irie, S.; Motonaga, R.; Hamanoue, N.; Tanabe, M.; et al. Activation of overexpressed glucagon-like peptide-1 receptor attenuates prostate cancer growth by inhibiting cell cycle Progression. J. Diabetes Investig. 2020, 11, 1137–1149. [Google Scholar] [CrossRef]
  93. Cao, C.; Yang, S.; Zhou, Z. GLP-1 Receptor Agonists and Pancreatic Safety Concerns in Type 2 Diabetic Patients: Data from Cardiovascular Outcome Trials. Endocrine 2020, 68, 518–525. [Google Scholar] [CrossRef] [PubMed]
  94. Suryadevara, V.; Roy, A.; Sahoo, J.; Kamalanathan, S.; Naik, D.; Mohan, P.; Kalayarasan, R. Incretin based therapy and pancreatic cancer: Realising the reality. World, J. Gastroenterol. 2022, 28, 2881–2889. [Google Scholar] [CrossRef]
  95. Pothuraju, R.; Rachagani, S.; Junker, W.M.; Chaudhary, S.; Saraswathi, V.; Kaur, S.; Batra, S.K. Pancreatic cancer associated with obesity and diabetes: An alternative approach for its targeting. J. Exp. Clin. Cancer Res. 2018, 37, 319–333. [Google Scholar] [CrossRef]
  96. Zhao, H.; Wang, L.; Wei, R.; Xiu, D.; Tao, M.; Ke, J.; Liu, Y.; Yang, J.; Hong, T. Activation of GlucagonLike Peptide-1 Receptor Inhibits Tumourigenicity and Metastasis of Human Pancreatic Cancer Cells via PI3K/Akt Pathway. Diabetes Obes. Metab. 2014, 16, 850–860. [Google Scholar] [CrossRef]
  97. Zhao, H.; Wei, R.; Wang, L.; Tian, Q.; Tao, M.; Ke, J.; Liu, Y.; Hou, W.; Zhang, L.; Yang, J.; et al. Activation of GlucagonLike Peptide-1 Receptor Inhibits Growth and Promotes Apoptosis of Human Pancreatic Cancer Cells in a cAMP-Dependent Manner. Am. J. Physiol. Endocrinol. Metab. 2014, 306, E1431–E1441. [Google Scholar] [CrossRef]
  98. Zhao, H.J.; Jiang, X.; Hu, L.J.; Yang, L.; Deng, L.D.; Wang, Y.P.; Ren, Z.-P. Activation of GLP-1 Receptor Enhances the Chemosensitivity of Pancreatic Cancer Cells. J. Mol. Endocrinol. 2020, 64, 103–113. [Google Scholar] [CrossRef]
  99. Mao, D.; Cao, H.; Shi, M.; Wang, C.C.; Kwong, J.; Li, J.J.X.; Hou, Y.; Ming, X.; Lee, H.M.; Tian, X.Y.; et al. Increased CoExpression of PSMA2 and GLP-1 Receptor in Cervical Cancer Models in Type 2 Diabetes Attenuated by Exendin-4: A Translational Case-Control Study. EBioMedicine 2021, 65, 103242. [Google Scholar] [CrossRef]
  100. Li, W.; Gu, Y.; Liu, S.; Ruan, F.; Lv, W. GLP1R inhibits the progression of endometrial carcinoma through activation of cAMP/PKA pathway. J. Clin. Lab. Anal. 2022, 36, e24604. [Google Scholar] [CrossRef]
  101. Kanda, R.; Hiraike, H.; Wada-Hiraike, O.; Ichinose, T.; Nagasaka, K.; Sasajima, Y.; Ryo, E.; Fujii, T.; Osuga, Y.; Ayabe, T. Expression of the Glucagon-Like Peptide-1 Receptor and Its Role in Regulating Autophagy in Endometrial Cancer. BMC Cancer 2018, 18, 657. [Google Scholar] [CrossRef]
  102. He, W.; Yu, S.; Wang, L.; He, M.; Cao, X.; Li, Y.; Xiao, H. Exendin-4 Inhibits Growth and Augments Apoptosis of Ovarian Cancer Cells. Mol. Cell. Endocrinol. 2016, 436, 240–249. [Google Scholar] [CrossRef]
  103. Ebrahem, Q.; Chaurasia, S.S.; Vasanji, A.; Qi, J.H.; Klenotic, P.A.; Cutler, A.; Asosingh, K.; Erzurum, S.; Anand-Apte, B. Cross-talk between vascular endothelial growth factor and matrix metalloproteinases in the induction of neovascularization in vivo. Am. J. Pathol. 2010, 176, 496–503. [Google Scholar] [CrossRef] [PubMed]
  104. Kosowska, A.; Gallego-Colon, E.; Garczorz, W.; Kłych-Ratuszny, A.; Aghdam, M.R.F.; Woz Niak, M.; Witek, A.; Wróblewska-Czech, A.; Cygal, A.; Wojnar, J.; et al. Exenatide Modulates Tumor-Endothelial Cell Interactions in Human Ovarian Cancer Cells. Endocr. Connect. 2017, 6, 856–865. [Google Scholar] [CrossRef] [PubMed]
  105. Koehler, J.A.; Kain, T.; Drucker, D.J. Glucagon-like peptide-1 receptor activation inhibits growth and augments apoptosis in murine CT26 colon cancer cells. Endocrinology 2011, 152, 3362–3372. [Google Scholar] [CrossRef] [PubMed]
  106. Tong, G.; Peng, T.; Chen, Y.; Sha, L.; Dai, H.; Xiang, Y.; Zou, Z.; He, H.; Wang, S. Effects of GLP-1 Receptor Agonists on Biological Behavior of Colorectal Cancer Cells by Regulating PI3K/AKT/mTOR Signaling Pathway. Front. Pharmacol. 2022, 13, 901559. [Google Scholar] [CrossRef] [PubMed]
  107. Nie, Z.J.; Zhang, Y.G.; Chang, Y.H.; Li, Q.Y.; Zhang, Y.L. Exendin-4 Inhibits Glioma Cell Migration, Invasion and Epithelial-to-Mesenchymal Transition Through GLP-1R/Sirt3 Pathway. BioMed. Pharmacother. 2018, 106, 1364–1369. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cuttica, C.M.; Briata, I.M.; DeCensi, A. Novel Treatments for Obesity: Implications for Cancer Prevention and Treatment. Nutrients 2023, 15, 3737. https://doi.org/10.3390/nu15173737

AMA Style

Cuttica CM, Briata IM, DeCensi A. Novel Treatments for Obesity: Implications for Cancer Prevention and Treatment. Nutrients. 2023; 15(17):3737. https://doi.org/10.3390/nu15173737

Chicago/Turabian Style

Cuttica, Carla Micaela, Irene Maria Briata, and Andrea DeCensi. 2023. "Novel Treatments for Obesity: Implications for Cancer Prevention and Treatment" Nutrients 15, no. 17: 3737. https://doi.org/10.3390/nu15173737

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

Article Metrics

Back to TopTop