Open Access

Multi‑faceted role of cancer‑associated adipocytes in the tumor microenvironment (Review)

  • Authors:
    • Huihui Yao
    • Songbing He
  • View Affiliations

  • Published online on: October 20, 2021     https://doi.org/10.3892/mmr.2021.12506
  • Article Number: 866
  • Copyright: © Yao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Adipocytes are a type of stromal cell found in numerous different tissues that serve an active role in the tumor microenvironment. Cancer‑associated adipocytes (CAAs) display a malignant phenotype and are found at the invasive tumor front, which mediates the crosstalk network between adipocytes (the precursor cells that will become cancer‑associated adipocytes in the future) and cancer cells. The present review covers the mechanisms of adipocytes in the development of cancer, including metabolic reprogramming, chemotherapy resistance and adipokine regulation. Furthermore, the potential mechanisms involved in the adipocyte‑cancer cell cycle in various types of cancer, including breast, ovarian, colon and rectal cancer, are discussed. Deciphering the complex network of CAA‑cancer cell crosstalk will provide insights into tumor biology and optimize therapeutic strategies.

Introduction

Tumor initiation and development depend on epithelial tissue and are closely associated with the surrounding tumor microenvironment (TME). Although no consensus has been reached concerning the definition of the TME, which is mainly composed of tumor cells, peripheral immune and inflammatory cells, tumor-related fibroblasts, peripheral interstitial tissue, microvessels and various cytokines and chemokines, it has become an important research focus to understand tumor development and to identify target for cancer treatment (1). Metabolic transformation is not only a sign of cancer, but also a key goal of cancer treatment. Due to the influence of cancer cells, the physiological characteristics of the TME (such as metabolism, secretion and immunity, amongst others) are no longer regulated by the body, which has a far-reaching impact on the progress of cancer. Remodeling of the TME has therefore become a cancer treatment strategy. However, how changes in the TME metabolic levels affect cancer metabolism and behavior remains unclear (2,3). Moreover, whether adipocytes (cells abundant in the TME) play a key role in the malignant progression of tumors remains to be determined. Considering the relationship between tumor progression and obesity, adipocytes are considered to be essential components of the TME (4). Currently, from the available in vitro, in vivo and clinical data, it can be demonstrated that the characteristics of adipocyte-derived factors change during tumor progression and tumor cells can also significantly affect the surrounding adipocytes. When peritumoral adipocytes display an altered phenotype and specific biological features (such as the decrease in differentiation markers of mature adipocytes, production of a large number of fat-derived factors and the promotion of the metabolic reprogramming of cancer cells), they are termed cancer-associated adipocytes (CAAs) (5). CAAs are considered to serve an important role in the TME. Previous studies reported that adipocytes co-cultivated with cancer cells exhibited considerable morphological and functional alterations, including reduced lipid content and decreased adipocyte markers, such as adiponectin, leptin and fatty acid-binding protein (FABP)2 (68). Overexpression of interleukin (IL)-6, IL-1 and matrix metalloproteinase (MMP)-11 is also detected in activated adipocytes, which are also referred to as CAAs. Notably, in a tumor subset, growth and metastasis predominantly occur adjacent to adipocytes, for example, in breast cancer (BC), or at anatomical sites where cancer cells are close to the adipose tissue (AT), including in gastric, colorectal and ovarian cancer (9). Adipocytes participate in a highly complex inflammatory cycle that is regulated by tumor cells to promote tumor development. Moreover, the malignant functions of CAAs may amplify this cycle and therefore CAAs may serve as an obstacle to tumor treatment. A previous study has demonstrated that tumor drug resistance is an important driver of disease progression and could be a potential target for new therapies. The role of adipocytes in tumor drug resistance has been overlooked. CAAs cause drug resistance in oncological treatments, including chemotherapy, radiotherapy, hormone therapy and immunotherapy, which may lead to the persistence of tumor remnants and increase the risk of tumor recurrence (8). However, to the best of our knowledge the role of CAAs and how they evolve from adipocytes during tumor progression has previously not been described in detail. The present review has focused on the adipocyte-cancer cell circle in terms of metabolism, adipokines and drug resistance. The potential mechanisms underlying the dynamic communication between CAAs and numerous types of cancer, including BC, ovarian cancer and colorectal cancer (CRC), especially in the co-occurrence of obesity, which may result in neoteric therapy, have also been investigated.

Mature adipocytes and CAAs

AT comprises a highly complex and heterogeneous group of cellular components, mostly consisting of adipocytes. There are also numerous other types of stromal cells in AT, including endothelial cells, pericytes, macrophages and adipocyte progenitor cells. Both adipocytes and stromal vascular cells in AT contribute with several other factors towards tumor growth and maintenance (1012). AT is an important and complex tissue that regulates energy balance and is distributed in almost all compartments of the human body. Histologically, there are three main types of AT: i) White AT (WAT), which accounts for >95% of the fat mass; ii) brown AT, which accounts for 1–2% of the fat mass; and iii) beige AT, which is difficult to quantify as it is scattered under the skin near the spine and clavicle in adults and cannot be taken out as a whole. WAT is the most dynamic AT in the human body, accounting for 2–70% of the body weight (13). Mature adipocytes are the main cell type in WAT, containing a small number of mitochondria (14). Mature adipocytes function by secreting various adipocytokines, and are characterized by a round shape with only one large lipid droplet (15). Compared with mature adipocytes, CAAs possess an irregular morphology, with small lipid droplets and a small volume. Furthermore, tumor-associated adipocytes usually have some changes (such as differentiation markers), as later described. Following lipolysis, differentiation markers of mature adipocytes decrease, such as adipocyte p2 and FABP4 (16,17). In the presence of cancer cells, especially at the tumor invasive front, CAAs generate fibroblast-like cells named adipocyte-derived fibroblasts after undergoing a process via which adipocytes decompose their lipids into glycerol and free fatty acids, and adopting a phenotypic change. These adipocyte-derived fibroblasts, as part of the cancer-associated fibroblast (CAF) population, are also involved in the malignant progression of tumors. It has also been indicated that CAAs may represent an intermediate form through which CAFs can arise (18). Moreover, CAAs have also been found to produce a number of adipose-derived factors via endocrine and paracrine signaling pathways, including chemokine (C-C motif) ligand (CCL)2, CCL5, IL-6, tumor necrosis factor α (TNFα), vascular endothelial growth factor (VEGF) and leptin; however, lower levels of adiponectin are produced (5,6). In terms of metabolic changes, CAAs promote numerous catabolic processes releasing high-energy metabolites, including lactic acid, pyruvic acid, free fatty acids and ketone bodies (5). Furthermore, CAAs provide sufficient energy for cancer cells by secreting exogenous fatty acids, leading to the metabolic reprogramming of cancer cells (19). Tumor cell-derived signaling molecules induce the lipolysis of adipocytes and promote the evolution of adipocytes to CAAs (20). Fig. 1 displays the complex relationship among tumors, adipocytes and CAAs. Therefore, CAAs are thought to serve a paramount role in tumor invasion and progression via the endocrine and paracrine signaling pathways.

Distinctive role of CAAs in different types of cancer

CAAs in CRC

Obesity and CRC are both global health issues and epidemiological data has demonstrated that obesity is positively correlated with CRC (21). Obesity serves a direct and independent role in the development of CRC. Previous studies have reported that cancer cells can increase carnitine palmitoyltransferase 1A (CPT1A) expression by interacting with CAAs, which in turn upregulates mitochondrial fatty acid oxidation and enables cancer cells to tolerate a hypoxic environment (21,22). Furthermore, crosstalk between tumor resident adipocytes and CRC cells is considered to be a contributing factor in promoting cancer progression (23). Adipocyte-derived medium can directly promote CRC progression, which also further suggests that CAAs are a positive factor in carcinogenesis (24). CAAs serve a vital role in promoting inflammation and angiogenesis. By comparing inflammation and angiogenesis in lean and obese patients with or without CRC, it was determined that the plasma levels of proinflammatory and angiogenic factors were at least partially elevated in obese patients with CRC. This study therefore revealed that peritumoral visceral AT is crucial for the development of CRC (25). Furthermore, to avoid the influence of individual differences on the experiment, in vitro experiments were conducted to compare the inflammatory reactions adjacent to and distant from the tumor site in the same patient. It was confirmed that the difference was due to the special influence of the tumor environment rather than due to the differences between individuals. However, this experimental result has not been confirmed in vivo (26). Another study demonstrated that the average levels of proinflammatory cytokines IL-6, IL-4 and granulocyte-macrophage colony stimulating factor in patients with obesity and CRC, were significantly higher than those in lean patients with CRC and patients with obesity only. The levels of other pro-inflammatory markers in the plasma displayed different trends; for example, IL-8 was only upregulated in patients with CRC and not obesity, whereas interferon γ and TNFα were upregulated in patients with obesity and not CRC. Plasma levels of numerous proinflammatory cytokines, including IL-1α, IL-6, TNFα, CCL-2 and plasminogen activator inhibitor-1 were increased in both patients with obesity and patients with CRC; however, there was no significant difference between obese and lean patients. Plasma VEGF levels were also upregulated in response to both CRC and obesity, which is consistent with results reported in a previous study (27). It has also been reported that the expression of inducible nitric oxide synthase (iNOS) increases under inflammatory conditions, such as pancreatitis or obesity (28). Under inflammatory conditions, nitric oxide (NO) is produced by iNOS, an enzyme primarily expressed by macrophages and to a lesser extent by adipocytes. In previous studies, the concentrations of nitrite and nitrate in medium were used as indicators to evaluate the release of NO under inflammatory conditions (27,28). Consequently, the inflammatory characteristics of adipocytes away from and around the tumor site in both lean patients and patients with obesity and CRC were compared. The release of nitrite and nitrate in adipocytes around the tumor site was increased in lean patients with CRC compared with that in patients with obesity. However, adipocytes far from the tumor site did not show increased nitrite and nitrate secretions (29).

Previous studies have demonstrated that the TME has pro-inflammatory properties, which may contribute to the occurrence and progression of tumors (30). It can therefore be hypothesized that these pro-inflammatory factors in the TME may not only be due to increased macrophage infiltration as previously reported (31), but may also be derived from tumor-associated adipocytes. It has also been reported that the expression levels of cyclooxygenase-2 (COX-2) and peroxisome proliferator-activated receptor (PPAR)-γ are increased in the cancerous tissues of patients with CRC. Similarly, the gene expression of COX-2 and PPAR-γ is increased in tumor-associated adipocytes. A previous study has reported that the expression levels of COX-2 and PPAR-γ increase in the peritumoral tissues of patients with CRC. The gene expression levels of COX-2 and PPAR-γ are also increased in tumor-associated adipocytes (32).

Overall, the aforementioned studies indicate that the plasma levels of different pro-inflammatory and angiogenic factors in patients with CRC are increased, some of which may come from tumor-associated adipocytes. Tumor-associated adipocytes are considered a vital source of pro-inflammatory and angiogenic factors, which may affect the progress of tumor biology and the clinical prognosis of the patients. A recent study has reported that an imbalance of intestinal microbiota caused by obesity increases harmful microbiota and metabolites and decreases beneficial microbes (including Akkermansia muciniphila) and metabolites (short-chain fatty acids) (33). In bile acid metabolism, bile acids promote the progression of CRC, especially in obese patients. Bile acid-dependent inhibition of the farnesoid X receptor (FXR) can promote the occurrence of CRC, which provides a theoretical basis for FXR to become an antitumor target in CRC in the future (33). The potential mechanism of obesity-promoting CRC therefore needs to be explored further.

CAAs in BC

BC is one of the most common cancer types worldwide and is the second highest cause of cancer-related mortality in women (34). The TME is a heterogeneous ecosystem composed of infiltrating immune cells, mesenchymal support cells and a matrix that promotes tumor progression. Adipocytes are the main cellular component of the BC microenvironment. Recent evidence demonstrates that adipocytes promote tumor progression via the interaction and dynamic communication between tumor cells and adipocytes (35). AT serves a key role as an energy reservoir, in which endocrine cells can produce various bioactive substances (14). A recent study has determined that the characteristics of adipocyte-derived cytokines change during tumor progression, which has been confirmed in human BC samples (36). Furthermore, an increasing number of studies have confirmed that adipocytes adjacent to invasive cancer cells, namely CAAs, participate in BC progression (17,36). Moreover, inflammatory factors secreted by CAAs can influence the behavior of BC cells, changing the characteristics and phenotypes of BC cells to increase invasiveness (36). Therefore, it can be hypothesized that abnormal AT, especially that adjacent to BC, is a highly complex participant in the interaction between the tumor and microenvironment and is regulated by BC cells. This interaction may be amplified as a result of the malignant function of CAAs (16).

In the microenvironment of BC, the expression and secretion profiles of CAA-mediated inflammatory factors are altered. The secretion of CCL2, CCL5 (36), IL-1β, IL-6 (35), TNFα, VEGF and leptin (37) is increased in the BC microenvironment, further promoting the proliferation, invasion and angiogenesis of tumor cells. Therefore, CAAs promote the tumorigenesis, invasion and metastasis of BC. Furthermore, CAAs in the treatment of BC such as chemotherapy, radiotherapy, hormone therapy and immunotherapy can result in tumor resistance, leading to the persistence of residual tumors and an increased risk of tumor recurrence (16). Moreover, when mature adipocytes transform into CAAs, tumor cells become metabolic parasites by ingesting metabolites such as ketones, pyruvate, fatty acids and lactic acid from CAAs. Consequently, tumor cells transition from relying on anaerobic glycolysis to preferentially using fatty acids for fatty acid oxidation to provide energy for themselves (3840). The regulatory mechanisms of CAAs in BC are complex, including adipokine secretion, metabolic reprogramming and extracellular matrix (ECM) remodeling. Several hypotheses have been proposed stating that obesity promotes the development and progression of BC (39,40). However, the underlying mechanisms remain unclear due to a lack of sufficient models to study the relationship between obesity and BC.

CAAs in ovarian cancer

The biological characteristics of ovarian cancer are different from those of other types of the cancer, as ovarian cancer can exhibit hematogenous metastasis, which is rare and often limited to the AT-rich omentum (41). Although experiments have confirmed that the adipocytes in the omentum are the key to inducing ovarian cancer cell omental metastasis, the specific mechanism remains unclear. According to recent studies, secretory acidic cysteine rich protein (SPARC) may serve a role in this process (42,43). SPARC is an ECM protein, which serves an important role in maintaining the homeostasis of the surrounding tissue environment. SPARC can serve a role in tissue remodeling, including in the regulation of adipocyte differentiation. SPARC can also inhibit adipocyte production, reduce the production of CAAs, interfere with the interaction between cancer cells and mesothelial cells, and normalize the TME (42). Previous studies have demonstrated that the protein and lipid metabolomics of ovarian cancer cells change significantly following coculture with adipocytes. Among them, fat chaperone protein FABP4 was induced in tumor cells by adipocytes, and makes the cancer cells more invasive and metastatic, which serves a key regulatory role in changes to lipid metabolism in cancer cells. Furthermore, targeting FABP4 can reduce the metastasis of cancer cells to the greater omentum and improves cell sensitivity to carboplatin (4244). Another study demonstrated that the selective metastasis of ovarian cancer to the greater omentum is caused by the CCL2 receptor, C-C chemokine receptor type (CCR)2, on ovarian cancer cells (44). CCL-2 is produced by the binding of omental adipocytes to homologous receptor CCR2 on ovarian cancer cells, which promotes cancer cell migration and omental metastasis by activating related signaling pathways such as the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway. Metformin can specifically inhibit the secretion of CCL-2 and therefore serves a role in the treatment of ovarian cancer (44). To help understand the cause of omental metastasis in ovarian cancer, a previous study has verified that primary omental adipocytes can induce the proliferation and invasion of ovarian cancer cells both in vivo and in vitro (45). CD36 is a transmembrane glycoprotein belonging to the class B scavenger receptor family. CD36 promotes the uptake of fatty acids and cholesterol and transmits intracellular signals to regulate fatty acid metabolism in tumor cells. Furthermore, CD36 serves a crucial role in the bioenergy adaptation of OVCA cells in an adipocyte-rich microenvironment. Omental adipocytes can reprogram tumor metabolism by upregulating oocyte CD36 expression (46). Functional studies have demonstrated that miR-21 combined with apoptotic protease activating factor-1, a new direct therapeutic target, could inhibit the apoptosis of ovarian cancer cells and induce chemotherapy resistance (47). Furthermore, evidence suggests that the tumor-promoting effect of CAAs results from both systemic and local effects of hormones involved in lipid hemostasis mediated by direct contact, paracrine factors or both (47). In the microenvironment, CAA will affect the transformation of the metabolic phenotype of ovarian cancer, which plays an important role in the occurrence and development of ovarian cancer (48).

Mechanistic insights linking CAAs with cancer

Although there is increasing evidence to suggest that CAAs are positively correlated with certain types of cancer, the underlying molecular mechanisms remain unclear. CAA induces abnormal metabolic reprogramming (among which lipids serve an unusual role in the process of tumor progression, as well as being a metabolic substrate), chemoresistance and the secretion of various adipokines, which may serve important roles in the complex process of carcinogenesis.

Metabolic reprogramming

Metabolic reprogramming to meet the bioenergetic and biosynthetic demands, and maintain redox balance and cell proliferation, is a hallmark of cancer (49). From benign growth to malignant and invasive lesions, the changes of cell metabolic requirements are complex and dynamic. With uncontrolled proliferation, cancer cells need increasing amounts of biomolecules to produce new sister cells. The metabolic substrates of these cancer cells change when they invade the surrounding stromal tissue and interact with new cell types. Furthermore, when tumors metastasize, cancer cells face a series of new metabolic challenges in different environments. Therefore, cancer cells need to adjust their metabolic programs to adapt to a constantly changing situation.

Adipocytes adapt to their own metabolic processes through dynamic interactions with tumors, which further supports the proliferation of tumor cells (50). Furthermore, to meet the extreme energy requirements of cancer cells, CAA metabolic reprogramming that occurs involves alterations in the metabolism of macronutrients, including carbohydrates, lipids and amino acids (51). In terms of glycolysis, the tricarboxylic acid cycle, amino acids, nucleotides and lipid metabolism, the metabolic processes in cancerous and healthy tissues are substantially different (39). As described by the ‘Warburg effect’, cancer cells prefer to produce adenosine triphosphate by glycolysis despite the presence of oxygen (52,53). However, in order to adapt to the environmental changes of hypoxia and acidosis, the metabolic pattern of cancer cells changes to rely on lipid metabolism, especially fatty acid oxidation (54). Furthermore, cancer exhibits heterogeneity in genetic and microenvironmental parameters that influence cell metabolism. For instance, tumor metabolism influences, and is influenced by, the metabolite composition of the TME, while the interorgan and intratumor microenvironments define the metabolic properties of tumor cells. The TME serves an irreplaceable role in tumor metabolism. The ‘reverse Warburg effect’ describes how glycolytic metabolism in the cancer-related matrix supports adjacent cancer cells. The catabolic products include lactate monocarboxylate, pyruvate and ketone body. The transfer of these catabolic products can induce metabolic coupling of interstitial carcinoma, inducing cancer cells to produce ATP, increase their proliferation and reduce cell death. The ‘reverse Warburg effect’ is also important when cancer cells utilize the energy generated from stromal cells in the TME (2). As one of the main components of the TME, tumor cells inevitably use free fatty acids and glycerol produced by fat cell catabolism as energy sources (55,56). Tumor lipid metabolism is regulated by genetic and epigenetic changes in the tumor cells, which affects the process of receiving lipid from CAAs as a metabolic substrate. Fatty acids can be released from lipid droplets via the adipose triglyceride lipase (ATGL)-dependent lipolysis pathway. ATGL expression in cancer cells is upregulated by the interaction between adipocytes and cancer cells (38). Through the co-culture of adipocytes and cancer cells, it has been determined that free fatty acids in adipocytes can be transferred to cancer cells. This process can upregulate CPT1A and other various protease expression levels acting on the electron transfer chain, which promotes fatty acid metabolism in cancer cells (57). Tumor cells can not only increase the expression of ATGL to accelerate the utilization of adipocyte-derived lipids, but also enhance the intracellular transport of fatty acids by increasing the expression of FABP5 (58). CD36 and fatty acid transporter-1 (FATP1) are mainly expressed in cancer cells adjacent to AT (5). Zaoui et al (59) reported that cancer cells could regulate their metabolic reprogramming via exogenous fatty acid uptake mediated by CD36 on the cell surface. Moreover, adipocyte-induced expression of CD36 and FATP1 promotes tumor progression by facilitating signal transduction in tumor cells (6062). Wang et al (63) reported the role of Janus kinase (JAK) and signal transducer and activator of transcription 3 (STAT3) signaling pathways in regulating lipid metabolism. Inhibition of the JAK/STAT3 signaling pathway inhibits the expression of numerous lipid metabolism genes, including CPT1B, which encodes a key enzyme of fatty acid oxidation (FAO). Adipocytes also affect immune cell metabolism. Adipocyte-derived leptin can change the metabolic pattern of CD8+ T cells via activation of STAT3-FAO and inhibiting glycolysis, leading to the downregulation of the effector function of CD8+ T cells (64).

CAA-derived lipid metabolism

Although previous studies have shown that lipids are mainly involved in the metabolic dynamics of the interaction between tumor cells and stromal cells, lipids can also play an additional role in the TME as a material source of cell membrane synthesis and an energy source of cancer cells (45,65,66). Certain tumor-associated stromal cells have been demonstrated to secrete autotoxin (ATX) (67). ATX hydrolyzes lysophosphatidylcholine to lysophosphatidic acid (LPA). LPA can be used as a signaling molecule in cancer cell mitosis and migration (68). Furthermore, adipose-derived stem cells and adipocytes in the TME can also produce ATX. Once cancer cells begin to invade surrounding tissues, inhibition of ATX has no significant effect on the growth of the primary tumor. However, disruption of the LPA/ATX axis helps to reduce cancer cell metastasis (6971). Moreover, studies on metabolic determinants supporting the metastatic potential of cancer cells have determined that lipid metabolism is a major promoter (72). It is now widely recognized that cancer cells with high metastatic potential often express CD36 and that mice fed with a high-fat diet exhibit an increase in the size and quantity of tumor lymph node metastases in a CD36-dependent manner (73). Moreover, numerous types of cancer, including melanoma, BC and prostate cancer, first pass through lymphatic vessels to the proximal lymph nodes, where lipid metabolism can be used as a driver for lymphangiogenesis (72,73). Furthermore, a previous study on the role of neutrophils in supporting lung colonization by metastatic BC cells, revealed that the destruction of leukotriene producing enzyme arachidonic acid 5-lipoxygenase, could significantly inhibit the pre-metastasis activity of neutrophils. These results suggested that lipid metabolism is not only involved in the initiation of cancer cell metastasis, but also in the colonization of peripheral metastasis sites (74).

ECM

In the TME, the ECM is composed of non-cellular polymer proteins and helper molecules. The ECM is an important component in the TME that determines cancer progression (75). Metastasis is the main cause of cancer-related mortality, but the metastasis of cancer cells into a new environment requires a cellular adaptation process. Cancer cells can achieve their own TME homeostasis via an interaction with the ECM. This functional adaptability needed in order to survive in a changing environment is called cancer cell phenotypic plasticity (76). ECM remodeling consists of two stages (the transformation from epithelial cells to mesenchymal cells, and the transformation from mesenchymal cells to epithelial cells) among which the transformation from epithelial to mesenchymal cells is established; however, there are few studies on this transformation process (77). The reason cancer cells can multiply indefinitely without being regulated by the body is that they can dedifferentiate and acquire stem cell-like characteristics. When the surrounding environment is relatively stable, cancer cells are also in a dormant state. However, in the case of external abnormalities, cancer cells can initiate TME remodeling and epithelial-mesenchymal transition (EMT), obtaining a stronger motor ability (77,78). The erosion and metastasis of malignant tumor is a dynamic and continuous process. Tumor cells first migrate from the primary site, invade the ECM, adhere to some molecules in the basement membrane and intercellular matrix, activate cells to synthesize and secrete various degrading enzymes, as well as assist tumor cells to enter blood vessels via the ECM. Then, tumor cells operate under the action of some factors, penetrate the blood vessel wall to the secondary site where they continue to proliferate and undergo metastasis. In short, abscission, adhesion, degradation, movement and proliferation occur through the whole process of malignant tumor erosion and metastasis. Therefore, it is not difficult to observed that the tumor will obtain a stronger metastatic ability when the tumor cells undergo the EMT process (77,78). However, a previous study has demonstrated that EMT is related to epithelial cell integration at the distal metastatic site of cancer cells (78). The reason why ECM has such a great impact on the TME remodeling of cancer cells remains to be fully elucidated.

Previous studies have, however, demonstrated that CAAs affect tumor ECM remodeling (75,79). CAA-induced TGF-β expression can inhibit AT angiogenesis. Inhibition of angiogenesis can lead to hypoxia and fibrosis in AT, thereby mediating the occurrence of the EMT in BC cells (79). Adipocytes extracted from AT around prostate tumors were found to highly express proteins that regulate ECM structure, including TNFα, osteopontin and MMP9 (80). CAAs can also promote the invasiveness of renal cell carcinoma cells via leptin (81). Furthermore, adipocytes secrete and process type VI collagen, which provides survival promoting signals in the early stages of breast tumor growth. The type VI collagen cleavage product, endothelin, also serves an important role in the subsequent occurrence and development of BC (82). Overall, these observations suggest that CAAs may act as proto-cancerous stromal cells to remodel the ECM and create a tumor tolerant environment.

Drug resistance

CAAs in the TME serve a dynamic and complex role in the drug response and promotion of tumor growth (83). Both adipocyte and tumor lipid metabolism can affect the tumor drug response. CAAs provide metabolic substrates, growth factors and cytokines to tumor cells. CAAs can transdifferentiate into other stromal cells to change the growth environment and adjust the drug response of the tumor. In a variety of solid and non-solid types of cancer, adipocyte and lipid metabolism-mediated chemotherapy resistance involves a host of complex mechanisms (83,84). To date, there are two types of drugs used for targeted cancer therapy: i) Drugs targeting cancer cells; and ii) drugs targeting cellular and molecular components in the TME. Among the drugs targeting the cellular and molecular components of the TME, targeting tumor vessels with anti-angiogenic drugs (AADs) has become key to the treatment of numerous types of cancer (84). These drugs are usually combined with conventional chemotherapy drugs (85). In the present review, mainly the role of CAAs and lipid metabolism in anti-vascular drug resistance is discussed. Epidemiological evidence suggests that patients with obesity have poorer clinical outcomes than non-obese patients who receive the same treatment (86,87). However, in general, obese patients with cancer are in the advanced stages at the point of diagnosis (8587). The drug resistance properties of advanced cancer are therefore the main reasons why it is difficult to treat.

Previous studies have demonstrated that BC, ovarian cancer and prostate cancer rich in AT grow faster and more aggressively than those with less AT (88,89). Research into the reasons for this phenomenon has revealed a number of results. Adipocyte volume is increased in patients with obesity, resulting in an increased distance between capillaries. Owing to the relatively low density of microvessels the blood perfusion in the AT of patients with obesity is insufficient (90). Therefore, in patients with obesity, AT undergoes mild hypoxia, which affects the cellular and molecular composition of the adipose microenvironment. These changes inevitably affect the drug response of tumors growing in the vicinity of obese AT and may limit the distribution of anticancer drugs. Although tumor tissue is rich in blood vessels, the growth of tumors in anoxic obese AT may be due to decreased blood vessels and poor perfusion of disordered, tortuous and leaky vessels in the tumor itself, which results in restricted drug perfusion (91). A previous study has determined that tumors implanted with the same gene at different sites exhibit different responses to AAD treatment, which provides evidence that the tissue environment outside the tumor serves an important role in AAD resistance (92). Implantation of pancreatic ductal adenocarcinoma and CRC tumors in AT results in intrinsic AAD resistance, whereas the same tumors growing in non-AT are sensitive to the same AAD treatment (92). Similarly, AAD-sensitive hepatocellular carcinoma (HCC) growing in steatotic livers is resistant to anti-angiogenic therapy, whereas HCC in non-steatotic livers remains sensitive. These preclinical findings are thought to be associated with AAD resistance and the AT environment (84,92). Unexpectedly, for the stromal component of tumors, the microvessels of almost fat-free tumors and fat-rich tumors are equally sensitive to AAD treatment (92). Tumor growth depends on active angiogenesis, and the inhibition of angiogenesis weakens tumor growth. However, tumors in the adipose environment can continue to grow into large masses, even with a small amount of intratumoral microvascular tissue. Furthermore, angiogenesis has been shown to inhibit tumor growth in non-ATs (93). Thus, the unexpected discovery of tumors growing in AT distinguishes anti-angiogenic responses from tumor growth. Adipose vessels seem to be dependent on VEGFs. For anti-VEGF targeted treatment, inhibiting VEGF leads to significant vascular degeneration (94,95). Therefore, tumors growing in AT experience more severe hypoxia than those growing in non-AT. AAD-induced vascular reduction and hypoxia can limit the supply of circulating glucose, resulting in an impaired glucose-dependent metabolism (96). To survive and proliferate, cancer cells must use alternative energy mechanisms to generate energy. AAD-triggered hypoxia results in the following three lipid metabolic processes: i) The release of free fatty acids and glycerol as metabolites from adipocytes; ii) cancer cells increase free fatty acid uptake by upregulating CD36; and iii) cancer cells undergo metabolic pathway reprogramming, which activates the β-oxidation pathway resulting in free fatty acid metabolism-dependent energy production, supporting tumor growth and metastasis (17,97). These processes are illustrated in Fig. 2. Based on these findings, targeting CAAs and lipid metabolism will provide an attractive approach for cancer therapy. Compared with the use of a single drug, the combination of AAD and β-oxidation inhibitors (for example, etomoxir, a CPT1 inhibitor) has greater anticancer effects in animal models of HCC grown in steatotic livers (92). Furthermore, clinical studies have demonstrated that obesity is negatively associated with clinical benefits, such as decreased sensitivity of chemotherapeutic drugs and poor prognosis, in patients treated with anti-angiogenic therapies (98). Hypoxia induced by anti-VEGF treatment results in the high expression of IL-6 and fibroblast growth factor 2 (FGF2) (99). Similarly, anti-VEGF treatment can induce high expression levels of IL-6 and FGF2 in patients with BC, which contribute to AAD resistance (98,99). Furthermore, the number of adipocytes in tumors was directly and positively correlated with adverse AAD reactions (100).

Adipokines

Adipocytes can secrete >600 metabolites, hormones and cytokines, collectively known as adipokines (5). Although large-scale Mendelian randomization analyses have been performed to assess the possible causal relationship between adipokine concentrations and the risk of obesity-related cancer, including CRC and ovarian cancer, the results have demonstrated that there is no causal relationship (101). Adipokines serve an active role in regulating a variety of biological mechanisms, including insulin secretion, fat distribution, inflammatory response and energy consumption (5). The crosstalk between adipocytes and BC cells favors tumor proliferation, survival and metastasis (102). In terms of adipokines, CAA secretes more chemokines, such as CCL2, CCL5, IL-1β, IL-6, TNFα, VEGF and leptin, compared with relatively normal adipocytes (37), which can promote tumor invasion and metastasis (40,103). CAAs can also promote tumor growth and metastasis, compared with distant mature adipocytes, through fewer physical barriers (such as thickness and distance) and more active adipokine secretions (5). Furthermore, in addition to the aforementioned adipokines, the interaction between CAAs and cancer cells involves a variety of specialized adipokines, including resistin, insulin-like growth factor (IGF)-1, hepatocyte growth factor, platelet-derived growth factor BB, IL-1, IGF binding protein-2 and granulocyte colony stimulating factor (104106). In the subsequent sections of the present review, focus is given to the emerging roles of CAA-derived leptin, adiponectin, IL-6, CCL2 and CCL5 adipokines in cancer. Understanding the mechanisms of CAA-derived adipokines is important for understanding the behavior of tumor cells and formulating new therapeutics (Table I).

Table I.

Examples of cancer associated adipocyte-derived adipocytokines.

Table I.

Examples of cancer associated adipocyte-derived adipocytokines.

FactorReceptorPathways/key genesFunction(Refs.)
LeptinOBRER signal, JAK/STAT3, PI3K/AKT, IL-1/IL-1R, VEGF/VEGFR, FAK and SRC-1/STAT3Proliferation, differentiation, metastasis, self-renewal and chemoresistance and angiogenesis(52,90,9398)
Adiponectin AdipoR1/AdipoR2ERK1/2, AKT, TNFα, IL-1β, NF-κB, IL-6, IL-8, STK11 and AMPK/ULK1, AMPK and PI3K/AKTNegatively regulating cancer cell growth, autophagy, inhibiting proliferation and invasion(5,101)
CCL2CCR2/4Notch1, CXCL12 and CLSInducing the activity of CSCs, inflammation and malignant progression(111114)
CCL5CCR5ERK, EMT, AKT/mTORInflammation, poor DFS and OS(119,120,123)
IL-6IL-6R/gp130NF-κB, STAT3, EMTMigration, proliferation, invasion and pro-inflammatory response(134,135,138)

[i] CCL, chemokine (C-C motif) ligand; CCR, C-C chemokine receptor type; OBR, leptin receptor; AdipoR, adiponectin receptor; IL-6R, IL-6 receptor; gp130, glycoprotein 130; ER, estrogen receptor; JAK, Janus kinase; IL-1R, IL-1 receptor; FAK, focal adhesion kinase; SRC-1, steroid receptor coactivator-1; STK11, serine/threonine kinase 11; AMPK, adenosine 5′-monophosphate-activated protein kinase; ULK1, Unc-51 like autophagy activating kinase; CXCL12, chemokine (C-x-C motif) ligand 12; CLS, crown-like structure; CSC, cancer stem cell; EMT, epithelial-mesenchymal transition; DFS, disease-free survival; OS, overall survival.

Leptin

Leptin is a hormone with a molecular weight of 16 kDa, encoded by the LEP gene on human chromosome 7, and is mainly synthesized and secreted by adipocytes (107). Leptin is also produced by cancer cells (108). Leptin mediates multiple biological functions, including proliferation, differentiation, inflammation and nutrient absorption via the leptin receptor (OBR) (107), serving a role in the development and proliferation of normal and malignant tissues. It has been reported that increased leptin serum levels are positively correlated with cancer risk and with increased plasma leptin levels in patients with cancer. High leptin levels are also correlated with a high cancer grade, advanced tumor stage and invasive cancer subtypes, such as in BC and CRC (107). Compared with distant tumor sites, the expression of leptin in AT at the infiltration front is higher (109). Furthermore, the production and secretion of leptin is higher in CAAs than that in mature adipocytes (107). Therefore, leptin is involved in the interaction between adipocytes and cancer cells (16). Leptin may regulate numerous aspects of the occurrence, development and metastasis of BC via autocrine, endocrine and paracrine signaling pathways (5,16). Leptin can activate the estrogen receptor, JAK/STAT3 and PI3K/AKT signaling pathways to promote the proliferation of BC cells (91). Leptin accelerates the cell cycle of BC cells by increasing the expression of cyclinD1 and cyclin-dependent kinase 2 (110). Moreover, leptin can upregulate the mRNA and protein expression levels of IL-1/IL-1 receptor (IL-1R), further promoting the expression of VEGF and VEGFR, which promote angiogenesis (111). The role of leptin in BC invasion and metastasis has been thoroughly researched. Wei et al (112) reported that leptin promoted the EMT of BC cells via two main mechanisms: i) By upregulating the expression of pyruvate kinase M2; and ii) by activating the PI3K/AKT signaling pathway (112). Leptin also activates focal adhesion kinase to enhance the secretion of ECM remodelers via the steroid receptor coactivator-1/STAT3 signaling pathway, which suggests that leptin is associated with the establishment of a more invasive phenotype in BC cells (113). Previous studies have demonstrated that tumor cell proliferation and tumor growth are significantly reduced in leptin-deficient tumors; however, leptin-deficient and leptin receptor-deficient mice are severely obese. Moreover, OBR expression levels are significantly increased in colon tumors compared with those in the normal epithelium. Colonic leptin signaling-mediated CRC growth mainly occurs via the OBR/STAT3 signaling pathway (114). A previous study has demonstrated that adipocyte-derived leptin and IL-6 promote the local invasion and metastasis of tumor cells, which occurs by activating procollagen-lysine and 2-oxoglutarate 5-dioxygenase (lysine hydroxylase) 2 when tumor cells are co-cultured with mature adipocytes. Furthermore, leptin may promote BC invasion and metastasis via the PI3K/AKT/activating transcription factor-2 signaling pathway in tumor cells (115,116). Leptin also regulates the function of immune cells. Leptin can promote the progression of BC by activating STAT3-FAO, inhibiting glycolysis and downregulating the effector function of CD8+ T cells (64).

Adiponectin

Adiponectin, a hormone with a molecular weight of ~30 kDa, is encoded by the ADIPOQ gene (117,118). Adiponectin serves a protective role in tumor progression by binding to adiponectin receptor 1 and adiponectin receptor 2. The decrease in adiponectin secretion in CAAs (108) suggests a relationship between adiponectin and its related signaling proteins in antitumor activity. Adiponectin negatively regulates the cancer cell proliferation by regulating inflammatory signaling molecules, such as extracellular signal-regulated kinase (ERK)1/2, AKT, TNFα, IL-1β, nuclear factor (NF)-κB, IL-6, IL-8 and CCL2 (118). Autophagy is a fundamental vacuolar lysosomal degradation process known to help prevent the accumulation of damaged proteins and organelles, recycle cytoplasmic components and maintain intracellular homeostasis (119). Adiponectin secreted by adipocytes is an effective inducer of cytotoxic autophagy. CAAs are functionally diverse and can induce adipocyte senescence via the adiponectin-dependent autophagy signaling pathway, thereby enhancing malignant behavior (120). Adenosine 5′-monophosphate (AMP)-activated protein kinase (AMPK) is an important energy sensor that can be regulated by cytokines, such as leptin and adiponectin, promoting autophagy, regulating cellular metabolism and maintaining energy balance (121). Chung et al (119) proposed that adiponectin can induce cancer autophagy and is involved in the regulation of the serine/threonine kinase 11 and AMPK/Unc-51 like autophagy activating kinase (ULK1) axes. ULK1 is an autophagy-initiating kinase. Under nutrient sufficient conditions, mTOR disrupts the interaction between ULK1 and AMPK by inhibiting the activation of ULK1 (122). Moreover, adiponectin can inhibit the growth and invasion of BC cells and induce apoptosis by triggering the AMPK signaling pathway and inhibiting the PI3K/AKT signaling pathway (107). In patients with obesity, this relationship uses the adiponectin to leptin ratio, compared with patients who are not obese, and poses a significant problem. Theriau et al (123) revealed that a low ratio of adiponectin to leptin in patients with obesity can rapidly induce MCF-7 BC cells to enter the cell cycle (123). Therefore, this increase in adiponectin levels and the decrease in the leptin-to-adiponectin ratio are related to a decrease in BC growth. In CRC, AT in the TME leads to systemic low-grade and subclinical inflammation, which is conducive to the carcinogenesis of various tissues in the body. However, AT with endocrine activity influences the risk of systemic carcinogenesis directly related to inflammation and by significantly reducing the expression of adipokines, especially adiponectin (124). Adiponectin and its receptors, can induce the activation of various mechanisms, thus exerting a cancer-preventive effect. Adiponectin participates in the AMPK signaling pathway to regulate the cell cycle by regulating the activity of various transcription factors, including p53, and has been reported to inhibit signaling pathways involved in proliferation, such as the Mtor (125), Wnt/GSK3β (126) and JAK/STAT signaling pathways (127). Furthermore, a previous study reported that adiponectin can inhibit cell proliferation (128). The low mRNA expression levels of COX-2 and high expression levels of T-cadherin in CRC HCT116 cells suggest that adiponectin may directly inhibit tumor growth via COX-2 (129).

CCL2

The chemokine CCL2 is encoded by the CCL2 gene and is also referred to as MCP-1 (130). In the TME, different cells, including cancer cells, endothelial cells and fibroblasts, can secrete CCL2 into the extracellular environment. CCL2 binds to G protein-coupled receptors CCR2 and CCR4, and acts as a chemoattractant to recruit CCR2-expressing immune cells to inflammatory regions (130). A previous study found that CCL2 expression levels increased in BC e0771 cells, which recruited more adipocytes and monocytes/macrophages (39). High expression levels of CCL2 are associated with decreased survival in patients with BC (131). Tsuyada et al (132) reported that BC cells secrete cytokines by activating the STAT3 promoter and consequently the STAT3 signaling pathway in fibroblasts, resulting in increased CCL2 expression and secretion. CCL2 can also induce Notch1 expression and downstream signaling pathways in BC cells, thereby inducing cancer stem cell (CSC) activity. Furthermore, CCL2 expression is significantly correlated with angiogenesis (133). CCL2 may also serve an important role in the crosstalk between adipocytes and macrophages. Studies have found that increased expression levels of CCL2 and IL-1β in AT induce macrophages to secrete chemokine (C-x-C motif) ligand 12 (CXCL12), which is associated with obesity (134,135). It has also been demonstrated that mammary epithelial cells surrounding AT recruit macrophages and form a crown-like structure by secreting CCL2, which is related to the malignant progression of BC. CCL2 has been proposed as a therapeutic target for metastatic BC (134136). In the presence of BC cells, adipocytes can revert to an immature proliferation phenotype, increasing the production of adipocyte-derived CCL2 and promoting cell migration through adipokines such as IL-6 and CCL2 (15,136). Furthermore, neutralization experiments using anti-IL-6 or CCL2 antibodies have demonstrated that the migration enhancing effect of CAA-conditioned media can be abolished (15). A recent study has determined that chronic inflammation induced by CCL2 can significantly promote tumor growth and connective tissue matrix formation via the early entry of macrophages and fibroblasts into the TME (137).

CCL5

Chemokine CCL5 has a molecular weight of 8 kDa and is located on chromosome 17q12; it is an effective chemokine that attracts leukocytes and is a multifunctional inflammatory mediator that can be expressed by BC cells (138). CCL5 is highly expressed in BC and can also be produced by a variety of cells, including mesenchymal stem cells (139). CCL5 overexpression is associated with ERK phosphorylation in tumor cells and with low 5-year disease-free survival and overall cancer survival in patients with early human epidermal growth factor receptor-2 (HER2)-positive BC (140). The abundance of CCL5 in the peritumoral AT of patients with triple negative BC (TNBC) is also associated with parallel low tumor metastasis and overall survival. Song et al (141) demonstrated that when adipocytes were cocultured with a TNBC cell line, adipocytes could enhance the EMT effect by secreting more CCL5. Karnoub et al (103) reported that BC cells stimulate the secretion of CCL5 and that paracrine CCL5 reversibly binds to CCR5 on the membrane surface of human BC MDA-MB-231 cells to enhance migration, invasion and metastasis (103). Another study demonstrated that when MDA-MB-231 TNBC cells were co-cultured with human adipocytes, CCL5 levels were increased in the surrounding tissues, resulting in the enhancement of MDA-MB-231 cell invasion and metastasis ability (36). Therefore, antagonizing CCL5 using specific CCL5 small molecule inhibitors can reduce the invasion of BC cells (36). Numerous studies have demonstrated that the CCL5/CCR5 axis is highly activated in TNBC and HER2-positive BC. The invasive ability of CCR5+ BC cells that responded to CCL5 was 40× higher compared with that of CCR5 cells that did not respond to CCR5 (142144). In the mTOR-dependent mechanism, MCF-7 cells overexpressing CCR5 have a stronger proliferation ability (145). Moreover, it has been shown that inhibition of CCL5/CCR5 signaling in endothelial cells leads to the defective activation of the AKT/mTOR signaling pathway and abnormal vascular and tumor growth in vitro and in vivo (36). Therefore, stromal cells may secrete CCL5 into the TME, where CCL5 may activate the AKT/mTOR signaling pathway to promote tumor metastasis by binding to CCR5. Moreover, CCL5 partially impairs triglyceride synthesis in adipocytes via its cognate receptors by downregulating the production of lipase and sterol regulatory element binding protein-1 (146). CCL5 also recruits macrophages (147) and T helper 1 and T helper 17 cells (148) in the TME, which can induce and maintain the striated structure of the matrix in the inflammatory microenvironment. This structure allows carcinogenic cell behavior and is destroyed by blocking CCL5 in vitro during matrix deposition (149). Therefore, CCL5 could be a potential target for BC precision therapy, but its specific mechanism requires further investigation.

IL-6

The level of IL-6 secreted by adipocytes is significantly increased under the pathological conditions of obesity and cancer. IL-6 is a cytokine involved in multiple biological activities, including hematopoiesis, immune regulation and tumorigenesis (150). Following coculture with BC cells, the expression and secretion of IL-6 in adipocytes increases (151). Among the proinflammatory cytokines involved in obesity-related inflammation, IL-6 has been determined to be the most important in CRC pathogenesis (15). IL-6 serves an important role in CRC, which may be related to the expression of the IL-1β/IL-6 network in the TME. The significance of this finding stems from the regulation of IL-6 synthesis by IL-1β and its synergistic pro-inflammatory effects. Lee et al (152) demonstrated that mouse 3T3-L1 adipocytes indirectly cocultured with BC cells upregulated the expression of IL-6 and pentraxin 3, which is consistent with the overexpression of IL-6 observed in CAAs of human BC tissue (152). Furthermore, IL-6 acts as an independent poor prognostic factor for overall survival and is associated with poor 5-year disease-free survival in patients with steroid-refractory metastatic BC (153). IL-6 promotes tumor cell proliferation, survival and angiogenesis by regulating the JAK/STAT3 signaling pathway (154). In HER2-positive BC, IL-6 can also induce the generation and maintenance of BC CSCs via the NF-κB and STAT3 signaling pathways to promote tumor progression (155). IL-6 also regulates the self-renewal of BC CSCs and promotes the survival and proliferation of stem cells when the Notch, Wnt and TGF-β signaling pathways are activated (156). Nickel et al (157) demonstrated that the migration ability of TNBC cells was significantly increased and the secretion of IL-6 was increased following coculture with adipocytes (157). Moreover, adipocyte-derived IL-6 can also enhance the invasive behavior of BC cells and induce the EMT phenotype (158). IL-6-induced BC metastasis is reversed when the IL-6 receptor (IL-6R) is blocked by an anti-IL-6R antibody (159,160). Blocking IL-6 signaling in BC cells alters the expression of EMT regulatory genes, disrupts the stability of focal adhesions and decreases the mobility of cells (161).

Conclusions

CAAs significantly affect tumor growth, metastasis and drug sensitivity via multiple mechanisms, including paracrine, juxtacrine and endocrine signaling, metabolites and metabolic reprogramming. This effect is greatest in tumors that are closely related to adipocytes, such as BC, CRC and ovarian cancer. Therefore, targeting CAAs and lipid metabolism would be a potential approach for cancer treatment. Furthermore, adipocytes are excellent candidates for altering tumor behavior via heterotypic signal transduction processes that secrete adipokines, such as hormones, growth factors, cytokines and other molecules. In the TME, the expression and secretion profiles of inflammatory mediators in adipocytes are altered. The secretion of chemokines, such as CCL5, CCL2, IL-6 and leptin, further promotes the proliferation, invasion and angiogenesis of tumor cells.

In summary, the present review mainly focused on the role of CAAs in cancer. The link between CAAs and cancer is important, and great progress has been made to elucidate the underlying biological mechanisms of CAA and the pathogenesis of cancer. CAAs induce leptin, IL-6, TNFα, CCL2 and CCL5 expression and disturb gut microbiota and bile acid homeostasis. These changes promote carcinogenesis, mediated by downstream signaling pathways. Based on the current understanding of this mechanism, several promising methods can be proposed for CAA in the treatment of tumors. However, several challenges remain for CAA identification. Certain CAA-related genes and protein markers have been identified, but there are no uniform criteria for the authentication of CAAs. An increased understanding of the link between CAA risk factors and carcinogenic processes will help develop more promising therapeutic targets and approaches for CAA-related cancer treatment in the future.

Acknowledgements

Not applicable.

Funding

The present review was supported by the National Science Foundation (grant no. BK20191172), the Project of Gusu Medical Key Talent (grant no. GSWS2020005) and the Project of New Pharmaceutics and Medical Apparatuses (grant no. SLJ2021007).

Availability of data and materials

Not applicable.

Authors' contributions

The concept, design and drafting of the review were performed by HY and SH. Data collection, involving searching, analyzing and summarizing the literature, was carried out by HY and manuscript revision was performed by SH. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CAA

cancer-associated adipocyte

TME

tumor microenvironment

FABP

fatty acid-binding protein

IL

interleukin

MMP

matrix metalloproteinase

AT

adipose tissue

WAT

white adipose tissue

CAF

cancer-associated fibroblast

CCL

chemokine (C-C motif) ligand

TNFα

tumor necrosis factor α

VEGF

vascular endothelial growth factor

CRC

colorectal cancer

CCL-2

monocyte chemotactic protein 1

iNOS

inducible nitric oxide synthase

NO

nitric oxide

COX-2

cyclooxygenase-2

PPAR

peroxisome proliferator-activated receptor

FXR

farnesoid X receptor

BC

breast cancer

ECM

extracellular matrix

miR

microRNA

ATGL

adipose triglyceride lipase

CPT1A

carnitine palmitoyltransferase 1A

FATP1

fatty acid transporter-1

JAK

Janus kinase

STAT3

signal transducer and activator of transcription 3

FAO

fatty acid oxidation

ATX

autotoxin

LPA

lysophosphatidic acid

AAD

antiangiogenic drug

HCC

hepatocellular carcinoma

FGF2

fibroblast growth factor 2

OBR

leptin receptor

PI3K

phosphatidylinositol 3-kinase

AKT

protein kinase B

ERK

extracellular signal-regulated kinase

NF-κB

nuclear factor-κB

AMPK

adenosine 5′-monophosphate-activated protein kinase

mTOR

mammalian target of rapamycin

TNBC

triple-negative BC

HER2

human epidermal growth factor receptor-2

CSC

cancer stem cell

References

1 

Laplane L, Duluc D, Larmonier N, Pradeu T and Bikfalvi A: The multiple layers of the tumor environment. Trends Cancer. 4:802–809. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Elia I and Haigis MC: Metabolites and the tumour microenvironment: From cellular mechanisms to systemic metabolism. Nat Metab. 3:21–32. 2021. View Article : Google Scholar : PubMed/NCBI

3 

Chen Q, Liu G, Liu S, Su H, Wang Y, Li J and Luo C: Remodeling the tumor microenvironment with emerging nanotherapeutics. Trends Pharmacol Sci. 39:59–74. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Lazar I, Clement E, Attane C, Muller C and Nieto L: A new role for extracellular vesicles: How small vesicles can feed tumors' big appetite. J Lipid Res. 59:1793–1804. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Wu Q, Li B, Li Z, Li J and Sun S and Sun S: Cancer-associated adipocytes: Key players in breast cancer progression. J Hematol Oncol. 12:952019. View Article : Google Scholar : PubMed/NCBI

6 

Dirat B, Bochet L, Dabek M, Daviaud D, Dauvillier S, Majed B, Wang YY, Meulle A, Salles B, Le Gonidec S, et al: Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Res. 71:2455–2465. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Vaupel H, Schmidberger A and Mayer A: The Warburg effect: Essential part of metabolic reprogramming and central contributor to cancer progression. Int J Radiat Biol. 95:912–919. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Hoxhaj G and Manning BD: The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism. Nat Rev Cancer. 20:74–88. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Nieman KM, Romero IL, Van Houten B and Lengyel E: Adipose tissue and adipocytes support tumorigenesis and metastasis. Biochim Biophys Acta. 1831:1533–1541. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Iyengar P, Combs TP, Shah SJ, Gouon-Evans V, Pollard JW, Albanese C, Flanagan L, Tenniswood MP, Guha C, Lisanti MP, et al: Adipocyte-secreted factors synergistically promote mammary tumorigenesis through induction of anti-apoptotic transcriptional programs and proto-oncogene stabilization. Oncogene. 22:6408–6423. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Park J, Morley TS, Kim M, Clegg DJ and Scherer PE: Obesity and cancer-mechanisms underlying tumour progression and recurrence. Nat Rev Endocrinol. 10:455–465. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Donohoe CL, Lysaght J, O'sullivan J and Reynolds JV: Emerging concepts linking obesity with the hallmarks of cancer. Trends Endocrinol Metab. 28:46–62. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Kahn CR, Wang G and Lee KY: Altered adipose tissue and adipocyte function in the pathogenesis of metabolic syndrome. J Clin Invest. 129:3990–4000. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Scheja L and Heeren J: The endocrine function of adipose tissues in health and cardiometabolic disease. Nat Rev Endocrinol. 15:507–524. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Fujisaki K, Fujimoto H, Sangai T, Nagashima T, Sakakibara M, Shiina N, Kuroda M, Aoyagi Y and Miyazaki M: Cancer-mediated adipose reversion promotes cancer cell migration via IL-6 and MCP-1. Breast Cancer Res Treat. 150:255–263. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Zhao C, Wu M, Zeng N, Xiong M, Hu W, Lv W, Yi Y, Zhang Q and Wu Y: Cancer-associated adipocytes: Emerging supporters in breast cancer. J Exp Clin Cancer Res. 39:1562020. View Article : Google Scholar : PubMed/NCBI

17 

Choi J, Cha YJ and Koo JS: Adipocyte biology in breast cancer: From silent bystander to active facilitator. Prog Lipid Res. 69:11–20. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Rybinska I, Agresti R, Trapani A, Tagliabue E and Triulzi T: Adipocytes in breast cancer, the thick and the thin. Cells. 9:5602020. View Article : Google Scholar : PubMed/NCBI

19 

Pérez-Escuredo J, Van Hée VF, Sboarina M, Falces J, Payen VL, Pellerin L and Sonveaux P: Monocarboxylate transporters in the brain and in cancer. Biochim Biophys Acta. 1863:2481–2497. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Yang E, Wang X, Gong Z, Yu M, Wu H and Zhang D: Exosome-mediated metabolic reprogramming: The emerging role in tumor microenvironment remodeling and its influence on cancer progression. Signal Transduct Target Ther. 5:2422020. View Article : Google Scholar : PubMed/NCBI

21 

Xiong X, Wen YA, Fairchild R, Zaytseva YY, Weiss HL, Evers BM and Gao T: Upregulation of CPT1A is essential for the tumor-promoting effect of adipocytes in colon cancer. Cell Death Dis. 11:7362020. View Article : Google Scholar : PubMed/NCBI

22 

Schlaepfer IR, Rider L, Rodrigues LU, Gijón MA, Pac CT, Romero L, Cimic A, Sirintrapun SJ, Glodé LM, Eckel RH and Cramer SD: Lipid catabolism via CPT1 as a therapeutic target for prostate cancer. Mol Cancer Ther. 13:2361–2371. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Tabuso M, Homer-Vanniasinkam S, Adya R and Arasaradnam RP: Role of tissue microenvironment resident adipocytes in colon cancer. World J Gastroenterol. 23:5829–5835. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Ko JH, Um JY, Lee SG, Yang WM, Sethi G and Ahn KS: Conditioned media from adipocytes promote proliferation, migration, and invasion in melanoma and colorectal cancer cells. J Cell Physiol. 234:18249–18261. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Fontana L, Eagon JC, Trujillo ME, Scherer PE and Klein S: Visceral fat adipokine secretion is associated with systemic inflammation in obese humans. Diabetes. 56:1010–1013. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Catalán V, Gómez-Ambrosi J, Rodríguez A, Ramírez B, Silva C, Rotellar F, Hernández-Lizoain JL, Baixauli J, Valentí V, Pardo F, et al: Up-regulation of the novel proinflammatory adipokines lipocalin-2, chitinase-3 like-1 and osteopontin as well as angiogenic-related factors in visceral adipose tissue of patients with colon cancer. J Nutr Biochem. 22:634–641. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Peterson JE, Zurakowski D, Italiano JE Jr, Michel LV, Connors S, Oenick M, D'Amato RJ, Klement GL and Folkman J: VEGF, PF4 and PDGF are elevated in platelets of colorectal cancer patients. Angiogenesis. 15:265–273. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Lumeng CN, Deyoung SM, Bodzin JL and Saltiel AR: Increased inflammatory properties of adipose tissue macrophages recruited during diet-induced obesity. Diabetes. 56:16–23. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA and Chen H: Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. 112:1821–1830. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Harvey AE, Lashinger LM and Hursting SD: The growing challenge of obesity and cancer: An inflammatory issue. Ann N Y Acad Sci. 1229:45–52. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Wagner M, Bjerkvig R, Wiig H, Melero-Martin JM, Lin RZ, Klagsbrun M and Dudley AC: Inflamed tumor-associated adipose tissue is a depot for macrophages that stimulate tumor growth and angiogenesis. Angiogenesis. 15:481–495. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Amor S, Iglesias-de la Cruz MC, Ferrero E, García-Villar O, Barrios V, Fernandez N, Monge L, García-Villalón AL and Granado M: Peritumoral adipose tissue as a source of inflammatory and angiogenic factors in colorectal cancer. Int J Colorectal Dis. 31:365–375. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Ye P, Xi Y, Huang Z and Xu P: Linking obesity with colorectal cancer: Epidemiology and mechanistic insights. Cancers (Basel). 12:14082020. View Article : Google Scholar : PubMed/NCBI

34 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Lapeire L, Hendrix A, Lambein K, Van Bockstal M, Braems G, Van Den Broecke R, Limame R, Mestdagh P, Vandesompele J, Vanhove C, et al: Cancer-associated adipose tissue promotes breast cancer progression by paracrine oncostatin M and Jak/STAT3 signaling. Cancer Res. 74:6806–6819. 2014. View Article : Google Scholar : PubMed/NCBI

36 

D'Esposito V, Liguoro D, Ambrosio MR, Collina F, Cantile M, Spinelli R, Raciti GA, Miele C, Valentino R, Campiglia P, et al: Adipose microenvironment promotes triple negative breast cancer cell invasiveness and dissemination by producing CCL5. Oncotarget. 7:24495–24509. 2016. View Article : Google Scholar : PubMed/NCBI

37 

De Palma M, Biziato D and Petrova TV: Microenvironmental regulation of tumour angiogenesis. Nat Rev Cancer. 17:457–474. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Wang YY, Attané C, Milhas D, Dirat B, Dauvillier S, Guerard A, Gilhodes J, Lazar I, Alet N, Laurent V, et al: Mammary adipocytes stimulate breast cancer invasion through metabolic remodeling of tumor cells. JCI Insight. 2:e874892017. View Article : Google Scholar : PubMed/NCBI

39 

Attane C, Milhas D, Hoy AJ and Muller C: Metabolic remodeling induced by adipocytes: A new Achille heels in invasive breast cancer? Curr Med Chem. 27:3984–4001. 2020. View Article : Google Scholar : PubMed/NCBI

40 

Bussard KM, Mutkus L, Stumpf K, Gomez-Manzano C and Marini FC: Tumor associated stromal cells as key contributors to the tumor microenvironment. Breast Cancer Res. 18:842016. View Article : Google Scholar : PubMed/NCBI

41 

Lengyel E: Ovarian cancer development and metastasis. Am J Pathol. 177:1053–1064. 2010. View Article : Google Scholar : PubMed/NCBI

42 

John B, Naczki C, Patel C, Ghoneum A, Qasem S, Salih Z and Said N: Regulation of the bi-directional cross-talk between ovarian cancer cells and adipocytes by SPARC. Oncogene. 38:4366–4383. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Mukherjee A, Chiang CY, Daifotis HA, Nieman KM, Fahrmann JF, Lastra RR, Romero IL, Fiehn O and Lengyel E: Adipocyte-induced FABP4 expression in ovarian cancer cells promotes metastasis and mediates carboplatin resistance. Cancer Res. 80:1748–1761. 2020. View Article : Google Scholar : PubMed/NCBI

44 

Sun C, Li X, Guo E, Li N, Zhou B, Lu H, Huang J, Xia M, Shan W, Wang B, et al: MCP-1/CCR-2 axis in adipocytes and cancer cell respectively facilitates ovarian cancer peritoneal metastasis. Oncogene. 39:1681–1695. 2020. View Article : Google Scholar : PubMed/NCBI

45 

Nieman KM, Kenny HA, Penicka CV, Ladanyi A, Buell-Gutbrod R, Zillhardt MR, Romero IL, Carey MS, Mills GB, Hotamisligil GS, et al: Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat Med. 17:1498–1503. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Coburn CT, Knapp FF Jr, Febbraio M, Beets AL, Silverstein RL and Abumrad NA: Defective uptake and utilization of long chain fatty acids in muscle and adipose tissues of CD36 knockout mice. J Biol Chem. 275:32523–32529. 2000. View Article : Google Scholar : PubMed/NCBI

47 

Au Yeung CL, Co NN, Tsuruga T, Yeung TL, Kwan SY, Leung CS, Li Y, Lu ES, Kwan K, Wong KK, et al: Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat Commun. 7:111502016. View Article : Google Scholar : PubMed/NCBI

48 

Renehan AG, Tyson M, Egger M, Heller RF and Zwahlen M: Body-mass index and incidence of cancer: A systematic review and meta-analysis of prospective observational studies. Lancet. 371:569–578. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Sun X, Wang M, Wang M, Yu X, Guo J, Sun T, Li X, Yao L, Dong H and Xu Y: Metabolic reprogramming in triple-negative breast cancer. Front Oncol. 10:4282020. View Article : Google Scholar : PubMed/NCBI

50 

Santander AM, Lopez-Ocejo O, Casas O, Agostini T, Sanchez L, Lamas-Basulto E, Carrio R, Cleary MP, Gonzalez-Perez RR and Torroella-Kouri M: Paracrine interactions between adipocytes and tumor cells recruit and modify macrophages to the mammary tumor microenvironment: The role of obesity and inflammation in breast adipose tissue. Cancers (Basel). 7:143–178. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Cairns RA, Harris IS and Mak TW: Regulation of cancer cell metabolism. Nat Rev Cancer. 11:85–95. 2011. View Article : Google Scholar : PubMed/NCBI

52 

Warburg O: On the origin of cancer cells. Science. 123:309–314. 1956. View Article : Google Scholar : PubMed/NCBI

53 

Vander Heiden MG, Cantley LC and Thompson CB: Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science. 324:1029–1033. 2009. View Article : Google Scholar : PubMed/NCBI

54 

Corn KC, Windham MA and Rafat M: Lipids in the tumor microenvironment: From cancer progression to treatment. Prog Lipid Res. 80:1010552020. View Article : Google Scholar : PubMed/NCBI

55 

Pallegar NK and Christian SL: Adipocytes in the tumour microenvironment. Adv Exp Med Biol. 1234:1–13. 2020. View Article : Google Scholar : PubMed/NCBI

56 

Dias AS, Almeida CR, Helguero LA and Duarte IF: Metabolic crosstalk in the breast cancer microenvironment. Eur J Cancer. 121:154–171. 2019. View Article : Google Scholar : PubMed/NCBI

57 

Balaban S, Shearer RF, Lee LS, van Geldermalsen M, Schreuder M, Shtein HC, Cairns R, Thomas KC, Fazakerley DJ, Grewal T, et al: Adipocyte lipolysis links obesity to breast cancer growth: Adipocyte-derived fatty acids drive breast cancer cell proliferation and migration. Cancer Metab. 5:12017. View Article : Google Scholar : PubMed/NCBI

58 

Yang D, Li Y, Xing L, Tan Y, Sun J, Zeng B, Xiang T, Tan J, Ren G and Wang Y: Utilization of adipocyte-derived lipids and enhanced intracellular trafficking of fatty acids contribute to breast cancer progression. Cell Commun Signal. 16:322018. View Article : Google Scholar : PubMed/NCBI

59 

Zaoui M, Morel M, Ferrand N, Fellahi S, Bastard JP, Lamazière A, Larsen AK, Béréziat V, Atlan M and Sabbah M: Breast-associated adipocytes secretome induce fatty acid uptake and invasiveness in breast cancer cells via CD36 independently of body mass index, menopausal status and mammary density. Cancers (Basel). 11:20122019. View Article : Google Scholar : PubMed/NCBI

60 

Ladanyi A, Mukherjee A, Kenny HA, Johnson A, Mitra AK, Sundaresan S, Nieman KM, Pascual G, Benitah SA, Montag A, et al: Adipocyte-induced CD36 expression drives ovarian cancer progression and metastasis. Oncogene. 37:2285–2301. 2018. View Article : Google Scholar : PubMed/NCBI

61 

Zhang M, Di Martino JS, Bowman RL, Campbell NR, Baksh SC, Simon-Vermot T, Kim IS, Haldeman P, Mondal C, Yong-Gonzales V, et al: Adipocyte-derived lipids mediate melanoma progression via FATP proteins. Cancer Discov. 8:1006–1025. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Lopes-Coelho F, Andre S, Felix A and Serpa J: Breast cancer metabolic cross-talk: Fibroblasts are hubs and breast cancer cells are gatherers of lipids. Mol Cell Endocrinol. 462:93–106. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Wang T, Fahrmann JF, Lee H, Li YJ, Tripathi SC, Yue C, Zhang C, Lifshitz V, Song J, Yuan Y, et al: JAK/STAT3-regulated fatty acid β-oxidation is critical for breast cancer stem cell self-renewal and chemoresistance. Cell Metab. 27:136–150.e5. 2018. View Article : Google Scholar : PubMed/NCBI

64 

Zhang C, Yue C, Herrmann A, Song J, Egelston C, Wang T, Zhang Z, Li W, Lee H, Aftabizadeh M, et al: STAT3 activation-induced fatty acid oxidation in CD8+ T effector cells is critical for obesity-promoted breast tumor growth. Cell Metab. 31:148–161.e5. 2020. View Article : Google Scholar : PubMed/NCBI

65 

Cawthorn WP, Scheller EL, Learman BS, Parlee SD, Simon BR, Mori H, Ning X, Bree AJ, Schell B, Broome DT, et al: Bone marrow adipose tissue is an endocrine organ that contributes to increased circulating adiponectin during caloric restriction. Cell Metab. 20:368–375. 2014. View Article : Google Scholar : PubMed/NCBI

66 

Ye H, Adane B, Khan N, Sullivan T, Minhajuddin M, Gasparetto M, Stevens B, Pei S, Balys M, Ashton JM, et al: Leukemic stem cells evade chemotherapy by metabolic adaptation to an adipose tissue niche. Cell Stem Cell. 19:23–37. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Auciello FR, Bulusu V, Oon C, Tait-Mulder J, Berry M, Bhattacharyya S, Tumanov S, Allen-Petersen BL, Link J, Kendsersky ND, et al: A stromal lysolipid-autotaxin signaling axis promotes pancreatic tumor progression. Cancer Discov. 9:617–627. 2019. View Article : Google Scholar : PubMed/NCBI

68 

Benesch MG, Tang X, Maeda T, Ohhata A, Zhao YY, Kok BP, Dewald J, Hitt M, Curtis JM, McMullen TP and Brindley DN: Inhibition of autotaxin delays breast tumor growth and lung metastasis in mice. FASEB J. 28:2655–2666. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Yang L, Yu X and Yang Y: Autotaxin upregulated by STAT3 activation contributes to invasion in pancreatic neuroendocrine neoplasms. Endocr Connect. 7:1299–1307. 2018. View Article : Google Scholar : PubMed/NCBI

70 

Azare J, Doane A, Leslie K, Chang Q, Berishaj M, Nnoli J, Mark K, Al-Ahmadie H, Gerald W, Hassimi M, et al: Stat3 mediates expression of autotaxin in breast cancer. PLoS One. 6:e278512011. View Article : Google Scholar : PubMed/NCBI

71 

Schmid R, Wolf K, Robering JW, Strauß S, Strissel PL, Strick R, Rübner M, Fasching PA, Horch RE, Kremer AE, et al: ADSCs and adipocytes are the main producers in the autotaxin-lysophosphatidic acid axis of breast cancer and healthy mammary tissue in vitro. BMC Cancer. 18:12732018. View Article : Google Scholar : PubMed/NCBI

72 

Corbet C and Feron O: Emerging roles of lipid metabolism in cancer progression. Curr Opin Clin Nutr Metab Care. 20:254–260. 2017. View Article : Google Scholar : PubMed/NCBI

73 

Pascual G, Avgustinova A, Mejetta S, Martín M, Castellanos A, Attolini CS, Berenguer A, Prats N, Toll A, Hueto JA, et al: Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature. 541:41–45. 2017. View Article : Google Scholar : PubMed/NCBI

74 

Wculek SK and Malanchi I: Neutrophils support lung colonization of metastasis-initiating breast cancer cells. Nature. 528:413–417. 2015. View Article : Google Scholar : PubMed/NCBI

75 

Poltavets V, Kochetkova M, Pitson SM and Samuel MS: The role of the extracellular matrix and its molecular and cellular regulators in cancer cell plasticity. Front Oncol. 8:4312018. View Article : Google Scholar : PubMed/NCBI

76 

Bacac M and Stamenkovic I: Metastatic cancer cell. Annu Rev Pathol. 3:221–247. 2008. View Article : Google Scholar : PubMed/NCBI

77 

Quail DF and Joyce JA: Microenvironmental regulation of tumor progression and metastasis. Nat Med. 19:1423–1437. 2013. View Article : Google Scholar : PubMed/NCBI

78 

Tsai JH and Yang J: Epithelial-mesenchymal plasticity in carcinoma metastasis. Genes Dev. 27:2192–2206. 2013. View Article : Google Scholar : PubMed/NCBI

79 

Ribeiro RJ, Monteiro CP, Cunha VF, Azevedo AS, Oliveira MJ, Monteiro R, Fraga AM, Príncipe P, Lobato C, Lobo F, et al: Tumor cell-educated periprostatic adipose tissue acquires an aggressive cancer-promoting secretory profile. Cell Physiol Biochem. 29:233–240. 2012. View Article : Google Scholar : PubMed/NCBI

80 

Lee SG, Kim JS, Kim HJ, Schlaepfer DD, Kim IS and Nam JO: Endothelial angiogenic activity and adipose angiogenesis is controlled by extracellular matrix protein TGFBI. Sci Rep. 11:96442021. View Article : Google Scholar : PubMed/NCBI

81 

Campo-Verde-Arbocco F, López-Laur JD, Romeo LR, Giorlando N, Bruna FA, Contador DE, López-Fontana G, Santiano FE, Sasso CV, Zyla LE, et al: Human renal adipose tissue induces the invasion and progression of renal cell carcinoma. Oncotarget. 8:94223–94234. 2017. View Article : Google Scholar : PubMed/NCBI

82 

Iyengar P, Espina V, Williams TW, Lin Y, Berry D, Jelicks LA, Lee H, Temple K, Graves R, Pollard J, et al: Adipocyte-derived collagen VI affects early mammary tumor progression in vivo, demonstrating a critical interaction in the tumor/stroma microenvironment. J Clin Invest. 115:1163–1176. 2005. View Article : Google Scholar : PubMed/NCBI

83 

Bochet L, Meulle A, Imbert S, Salles B, Valet P and Muller C: Cancer-associated adipocytes promotes breast tumor radioresistance. Biochem Biophys Res Commun. 411:102–106. 2011. View Article : Google Scholar : PubMed/NCBI

84 

Tennant DA, Durán RV and Gottlieb E: Targeting metabolic transformation for cancer therapy. Nat Rev Cancer. 10:267–277. 2010. View Article : Google Scholar : PubMed/NCBI

85 

Ueda S, Saeki T, Osaki A, Yamane T and Kuji I: Bevacizumab induces acute hypoxia and cancer progression in patients with refractory breast cancer: Multimodal functional imaging and multiplex cytokine analysis. Clin Cancer Res. 23:5769–5778. 2017. View Article : Google Scholar : PubMed/NCBI

86 

Levitsky A, Brismar K, Hafström I, Hambardzumyan K, Lourdudoss C, van Vollenhoven RF and Saevarsdottir S: Obesity is a strong predictor of worse clinical outcomes and treatment responses in early rheumatoid arthritis: Results from the SWEFOT trial. RMD Open. 3:e0004582017. View Article : Google Scholar : PubMed/NCBI

87 

Castillo JJ, Mulkey F, Geyer S, Kolitz JE, Blum W, Powell BL, George SL, Larson RA and Stone RM: Relationship between obesity and clinical outcome in adults with acute myeloid leukemia: A pooled analysis from four CALGB (alliance) clinical trials. Am J Hematol. 91:199–204. 2016. View Article : Google Scholar : PubMed/NCBI

88 

Laurent V, Guérard A, Mazerolles C, Le Gonidec S, Toulet A, Nieto L, Zaidi F, Majed B, Garandeau D, Socrier Y, et al: Periprostatic adipocytes act as a driving force for prostate cancer progression in obesity. Nat Commun. 7:102302016. View Article : Google Scholar : PubMed/NCBI

89 

Osman MA and Hennessy BT: Obesity correlation with metastases development and response to first-line metastatic chemotherapy in breast cancer. Clin Med Insights Oncol. 9:105–112. 2015. View Article : Google Scholar : PubMed/NCBI

90 

Engin A: Obesity-associated breast cancer: Analysis of risk factors. Adv Exp Med Biol. 960:571–606. 2017. View Article : Google Scholar : PubMed/NCBI

91 

Cao Y: Tumor angiogenesis and molecular targets for therapy. Front Biosci (Landmark Ed). 14:3962–3973. 2009. View Article : Google Scholar : PubMed/NCBI

92 

Iwamoto H, Abe M, Yang Y, Cui D, Seki T, Nakamura M, Hosaka K, Lim S, Wu J, He X, et al: Cancer lipid metabolism confers antiangiogenic drug resistance. Cell Metab. 28:104–117.e5. 2018. View Article : Google Scholar : PubMed/NCBI

93 

Hedlund EM, Yang X, Zhang Y, Yang Y, Shibuya M, Zhong W, Sun B, Liu Y, Hosaka K and Cao Y: Tumor cell-derived placental growth factor sensitizes antiangiogenic and antitumor effects of anti-VEGF drugs. Proc Natl Acad Sci USA. 110:654–659. 2013. View Article : Google Scholar : PubMed/NCBI

94 

Honek J, Seki T, Iwamoto H, Fischer C, Li J, Lim S, Samani NJ, Zang J and Cao Y: Modulation of age-related insulin sensitivity by VEGF-dependent vascular plasticity in adipose tissues. Proc Natl Acad Sci USA. 111:14906–14911. 2014. View Article : Google Scholar : PubMed/NCBI

95 

Yang Y, Zhang Y, Iwamoto H, Hosaka K, Seki T, Andersson P, Lim S, Fischer C, Nakamura M, Abe M, et al: Discontinuation of anti-VEGF cancer therapy promotes metastasis through a liver revascularization mechanism. Nat Commun. 7:126802016. View Article : Google Scholar : PubMed/NCBI

96 

Levine AJ and Puzio-Kuter AM: The control of the metabolic switch in cancers by oncogenes and tumor suppressor genes. Science. 330:1340–1344. 2010. View Article : Google Scholar : PubMed/NCBI

97 

Park JH, Vithayathil S, Kumar S, Sung PL, Dobrolecki LE, Putluri V, Bhat VB, Bhowmik SK, Gupta V, Arora K, et al: Fatty acid oxidation-driven src links mitochondrial energy reprogramming and oncogenic properties in triple-negative breast cancer. Cell Rep. 14:2154–2165. 2016. View Article : Google Scholar : PubMed/NCBI

98 

Trédan O, Lacroix-Triki M, Guiu S, Mouret-Reynier MA, Barrière J, Bidard FC, Braccini AL, Mir O, Villanueva C and Barthélémy P: Angiogenesis and tumor microenvironment: Bevacizumab in the breast cancer model. Target Oncol. 10:189–198. 2015. View Article : Google Scholar : PubMed/NCBI

99 

Incio J, Ligibel JA, McManus DT, Suboj P, Jung K, Kawaguchi K, Pinter M, Babykutty S, Chin SM, Vardam TD, et al: Obesity promotes resistance to anti-VEGF therapy in breast cancer by up-regulating IL-6 and potentially FGF-2. Sci Transl Med. 10:eaag09452018. View Article : Google Scholar : PubMed/NCBI

100 

Casanovas O, Hicklin DJ, Bergers G and Hanahan D: Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell. 8:299–309. 2005. View Article : Google Scholar : PubMed/NCBI

101 

Dimou NL, Papadimitriou N, Mariosa D, Johansson M, Brennan P, Peters U, Chanock SJ, Purdue M, Bishop DT, Gago-Dominquez M, et al: Circulating adipokine concentrations and risk of five obesity-related cancers: A mendelian randomization study. Int J Cancer. 148:1625–1636. 2021. View Article : Google Scholar : PubMed/NCBI

102 

Cha YJ, Kim ES and Koo JS: Tumor-associated macrophages and crown-like structures in adipose tissue in breast cancer. Breast Cancer Res Treat. 170:15–25. 2018. View Article : Google Scholar : PubMed/NCBI

103 

Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, Richardson AL, Polyak K, Tubo R and Weinberg RA: Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 449:557–563. 2007. View Article : Google Scholar : PubMed/NCBI

104 

Lee JO, Kim N, Lee HJ, Lee YW, Kim SJ, Park SH and Kim HS: Resistin, a fat-derived secretory factor, promotes metastasis of MDA-MB-231 human breast cancer cells through ERM activation. Sci Rep. 6:189232016. View Article : Google Scholar : PubMed/NCBI

105 

Xiong Y, Russell DL, McDonald LT, Cowart LA and LaRue AC: Hematopoietic stem cell-derived adipocytes promote tumor growth and cancer cell migration. Int J Cancer Res Mol Mech. 3:102017.PubMed/NCBI

106 

Wang C, Gao C, Meng K, Qiao H and Wang Y: Human adipocytes stimulate invasion of breast cancer MCF-7 cells by secreting IGFBP-2. PLoS One. 10:e01193482015. View Article : Google Scholar : PubMed/NCBI

107 

Sánchez-Jiménez F, Pérez-Pérez A, de la Cruz-Merino L and Sánchez-Margalet V: Obesity and breast cancer: Role of leptin. Front Oncol. 9:5962019. View Article : Google Scholar : PubMed/NCBI

108 

Ando S, Barone I, Giordano C, Bonofiglio D and Catalano S: The multifaceted mechanism of leptin signaling within tumor microenvironment in driving breast cancer growth and progression. Front Oncol. 4:3402014.PubMed/NCBI

109 

Gnerlich JL, Yao KA, Fitchev PS, Goldschmidt RA, Bond MC, Cornwell M and Crawford SE: Peritumoral expression of adipokines and fatty acids in breast cancer. Ann Surg Oncol. 20 (Suppl 3):S731–S738. 2013. View Article : Google Scholar : PubMed/NCBI

110 

Okumura M, Yamamoto M, Sakuma H, Kojima T, Maruyama T, Jamali M, Cooper DR and Yasuda K: Leptin and high glucose stimulate cell proliferation in MCF-7 human breast cancer cells: Reciprocal involvement of PKC-alpha and PPAR expression. Biochim Biophys Acta. 1592:107–116. 2002. View Article : Google Scholar : PubMed/NCBI

111 

Zhou W, Guo S and Gonzalez-Perez RR: Leptin pro-angiogenic signature in breast cancer is linked to IL-1 signalling. Br J Cancer. 104:128–137. 2011. View Article : Google Scholar : PubMed/NCBI

112 

Wei L, Li K, Pang X, Guo B, Su M, Huang Y, Wang N, Ji F, Zhong C, Yang J, et al: Leptin promotes epithelial-mesenchymal transition of breast cancer via the upregulation of pyruvate kinase M2. J Exp Clin Cancer Res. 35:1662016. View Article : Google Scholar : PubMed/NCBI

113 

Juárez-Cruz JC, Zuñiga-Eulogio MD, Olea-Flores M, Castañeda-Saucedo E, Mendoza-Catalán MÁ, Ortuño-Pineda C, Moreno-Godínez ME, Villegas-Comonfort S, Padilla-Benavides T and Navarro-Tito N: Leptin induces cell migration and invasion in a FAK-Src-dependent manner in breast cancer cells. Endocr Connect. 8:1539–1552. 2019. View Article : Google Scholar : PubMed/NCBI

114 

Endo H, Hosono K, Uchiyama T, Sakai E, Sugiyama M, Takahashi H, Nakajima N, Wada K, Takeda K, Nakagama H and Nakajima A: Leptin acts as a growth factor for colorectal tumours at stages subsequent to tumour initiation in murine colon carcinogenesis. Gut. 60:1363–1371. 2011. View Article : Google Scholar : PubMed/NCBI

115 

He JY, Wei XH, Li SJ, Liu Y, Hu HL, Li ZZ, Kuang XH, Wang L, Shi X, Yuan ST and Sun L: Adipocyte-derived IL-6 and leptin promote breast cancer metastasis via upregulation of Lysyl Hydroxylase-2 expression. Cell Commun Signal. 16:1002018. View Article : Google Scholar : PubMed/NCBI

116 

Li K, Wei L, Huang Y, Wu Y, Su M, Pang X, Wang N, Ji F, Zhong C and Chen T: Leptin promotes breast cancer cell migration and invasion via IL-18 expression and secretion. Int J Oncol. 48:2479–2487. 2016. View Article : Google Scholar : PubMed/NCBI

117 

Maroni P: Leptin, adiponectin, and Sam68 in bone metastasis from breast cancer. Int J Mol Sci. 21:10512020. View Article : Google Scholar : PubMed/NCBI

118 

Sultana R, Kataki AC, Borthakur BB, Basumatary TK and Bose S: Imbalance in leptin-adiponectin levels and leptin receptor expression as chief contributors to triple negative breast cancer progression in Northeast India. Gene. 621:51–58. 2017. View Article : Google Scholar : PubMed/NCBI

119 

Chung SJ, Nagaraju GP, Nagalingam A, Muniraj N, Kuppusamy P, Walker A, Woo J, Győrffy B, Gabrielson E, Saxena NK and Sharma D: ADIPOQ/adiponectin induces cytotoxic autophagy in breast cancer cells through STK11/LKB1-mediated activation of the AMPK-ULK1 axis. Autophagy. 13:1386–1403. 2017. View Article : Google Scholar : PubMed/NCBI

120 

Wu Q, Li B and Sun S and Sun S: Unraveling adipocytes and cancer links: Is there a role for senescence? Front Cell Dev Biol. 8:2822020. View Article : Google Scholar : PubMed/NCBI

121 

Hardie DG: AMP-activated/SNF1 protein kinases: Conserved guardians of cellular energy. Nat Rev Mol Cell Biol. 8:774–785. 2007. View Article : Google Scholar : PubMed/NCBI

122 

Kim J, Kundu M, Viollet B and Guan KL: AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. 13:132–141. 2011. View Article : Google Scholar : PubMed/NCBI

123 

Theriau CF, Sauvé OS, Beaudoin MS, Wright DC and Connor MK: Proliferative endocrine effects of adipose tissue from obese animals on MCF7 cells are ameliorated by resveratrol supplementation. PLoS One. 12:e01838972017. View Article : Google Scholar : PubMed/NCBI

124 

Tae CH, Kim SE, Jung SA, Joo YH, Shim KN, Jung HK, Kim TH, Cho MS, Kim KH and Kim JS: Involvement of adiponectin in early stage of colorectal carcinogenesis. BMC Cancer. 14:8112014. View Article : Google Scholar : PubMed/NCBI

125 

Li W, Saud SM, Young MR, Chen G and Hua B: Targeting AMPK for cancer prevention and treatment. Oncotarget. 6:7365–7378. 2015. View Article : Google Scholar : PubMed/NCBI

126 

Park SY, Kim D and Kee SH: Metformin-activated AMPK regulates β-catenin to reduce cell proliferation in colon carcinoma RKO cells. Oncol Lett. 17:2695–2702. 2019.PubMed/NCBI

127 

Rutherford C, Speirs C, Williams JJ, Ewart MA, Mancini SJ, Hawley SA, Delles C, Viollet B, Costa-Pereira AP, Baillie GS, et al: Phosphorylation of Janus kinase 1 (JAK1) by AMP-activated protein kinase (AMPK) links energy sensing to anti-inflammatory signaling. Sci Signal. 9:ra1092016. View Article : Google Scholar : PubMed/NCBI

128 

Waldner MJ, Foersch S and Neurath MF: Interleukin-6-a key regulator of colorectal cancer development. Int J Biol Sci. 8:1248–1253. 2012. View Article : Google Scholar : PubMed/NCBI

129 

Semaan J, Pinon A, Rioux B, Hassan L, Limami Y, Pouget C, Fagnère C, Sol V, Diab-Assaf M, Simon A and Liagre B: Resistance to 3-HTMC-induced apoptosis through activation of PI3K/Akt, MEK/ERK, and p38/COX-2/PGE2 pathways in human HT-29 and HCT116 colorectal cancer cells. J Cell Biochem. 117:2875–2885. 2016. View Article : Google Scholar : PubMed/NCBI

130 

Yoshimura T: The chemokine MCP-1 (CCL2) in the host interaction with cancer: A foe or ally? Cell Mol Immunol. 15:335–345. 2018. View Article : Google Scholar : PubMed/NCBI

131 

Bonapace L, Coissieux MM, Wyckoff J, Mertz KD, Varga Z, Junt T and Bentires-Alj M: Cessation of CCL2 inhibition accelerates breast cancer metastasis by promoting angiogenesis. Nature. 515:130–133. 2014. View Article : Google Scholar : PubMed/NCBI

132 

Tsuyada A, Chow A, Wu J, Somlo G, Chu P, Loera S, Luu T, Li AX, Wu X, Ye W, et al: CCL2 mediates cross-talk between cancer cells and stromal fibroblasts that regulates breast cancer stem cells. Cancer Res. 72:2768–2779. 2012. View Article : Google Scholar : PubMed/NCBI

133 

Saji H, Koike M, Yamori T, Saji S, Seiki M, Matsushima K and Toi M: Significant correlation of monocyte chemoattractant protein-1 expression with neovascularization and progression of breast carcinoma. Cancer. 92:1085–1091. 2001. View Article : Google Scholar : PubMed/NCBI

134 

Dommel S and Blüher M: Does C-C motif chemokine ligand 2 (CCL2) link obesity to a pro-inflammatory state? Int J Mol Sci. 22:15002021. View Article : Google Scholar : PubMed/NCBI

135 

Faria SS, Corrêa LH, Heyn GS, de Sant'Ana LP, Almeida RDN and Magalhães KG: Obesity and breast cancer: The role of crown-like structures in breast adipose tissue in tumor progression, prognosis, and therapy. J Breast Cancer. 23:233–245. 2020. View Article : Google Scholar : PubMed/NCBI

136 

Hsieh CC and Huang YS: Aspirin breaks the crosstalk between 3T3-L1 adipocytes and 4T1 breast cancer cells by regulating cytokine production. PLoS One. 11:e01471612016. View Article : Google Scholar : PubMed/NCBI

137 

Kuziel G, Thompson V, D'Amato JV and Arendt LM: Stromal CCL2 signaling promotes mammary tumor fibrosis through recruitment of myeloid-lineage cells. Cancers (Basel). 12:20832020. View Article : Google Scholar : PubMed/NCBI

138 

Kranjc MK, Novak M, Pestell RG and Lah TT: Cytokine CCL5 and receptor CCR5 axis in glioblastoma multiforme. Radiol Oncol. 53:397–406. 2019. View Article : Google Scholar : PubMed/NCBI

139 

Suárez-Nájera LE, Chanona-Pérez JJ, Valdivia-Flores A, Marrero-Rodríguez D, Salcedo-Vargas M, García-Ruiz DI and Castro-Reyes MA: Morphometric study of adipocytes on breast cancer by means of photonic microscopy and image analysis. Microsc Res Tech. 81:240–249. 2018. View Article : Google Scholar : PubMed/NCBI

140 

Zazo S, González-Alonso P, Martín-Aparicio E, Chamizo C, Luque M, Sanz-Álvarez M, Mínguez P, Gómez-López G, Cristóbal I, Caramés C, et al: Autocrine CCL5 effect mediates trastuzumab resistance by ERK pathway activation in HER2-positive breast cancer. Mol Cancer Ther. 19:1696–1707. 2020.PubMed/NCBI

141 

Song X, Zhou X, Qin Y, Yang J, Wang Y, Sun Z, Yu K, Zhang S and Liu S: Emodin inhibits epithelial-mesenchymal transition and metastasis of triple negative breast cancer via antagonism of CC-chemokine ligand 5 secreted from adipocytes. Int J Mol Med. 42:579–588. 2018.PubMed/NCBI

142 

Gao D, Rahbar R and Fish EN: CCL5 activation of CCR5 regulates cell metabolism to enhance proliferation of breast cancer cells. Open Biol. 6:1601222016. View Article : Google Scholar : PubMed/NCBI

143 

Sax MJ, Gasch C, Athota VR, Freeman R, Rasighaemi P, Westcott DE, Day CJ, Nikolic I, Elsworth B, Wei M, et al: Cancer cell CCL5 mediates bone marrow independent angiogenesis in breast cancer. Oncotarget. 7:85437–85449. 2016. View Article : Google Scholar : PubMed/NCBI

144 

Velasco-Velazquez M and Pestell RG: The CCL5/CCR5 axis promotes metastasis in basal breast cancer. Oncoimmunology. 2:e236602013. View Article : Google Scholar : PubMed/NCBI

145 

Velasco-Velázquez M, Jiao X, De La Fuente M, Pestell TG, Ertel A, Lisanti MP and Pestell RG: CCR5 antagonist blocks metastasis of basal breast cancer cells. Cancer Res. 72:3839–3850. 2012. View Article : Google Scholar : PubMed/NCBI

146 

Kim EJ, Kim YK, Kim S, Kim JE, Tian YD, Doh EJ, Lee DH and Chung JH: Adipochemokines induced by ultraviolet irradiation contribute to impaired fat metabolism in subcutaneous fat cells. Br J Dermatol. 178:492–501. 2018. View Article : Google Scholar : PubMed/NCBI

147 

Keophiphath M, Rouault C, Divoux A, Clement K and Lacasa D: CCL5 promotes macrophage recruitment and survival in human adipose tissue. Arterioscler Thromb Vasc Biol. 30:39–45. 2010. View Article : Google Scholar : PubMed/NCBI

148 

Shao L, Feng B, Zhang Y, Zhou H, Ji W and Min W: The role of adipose-derived inflammatory cytokines in type 1 diabetes. Adipocyte. 5:270–274. 2016. View Article : Google Scholar : PubMed/NCBI

149 

Brett E, Sauter M, Timmins É, Azimzadeh O, Rosemann M, Merl-Pham J, Hauck SM, Nelson PJ, Becker KF, Schunn I, et al: Oncogenic linear collagen VI of invasive breast cancer is induced by CCL5. J Clin Med. 9:9912020. View Article : Google Scholar : PubMed/NCBI

150 

Guo Y, Xu F, Lu T, Duan Z and Zhang Z: Interleukin-6 signaling pathway in targeted therapy for cancer. Cancer Treat Rev. 38:904–910. 2012. View Article : Google Scholar : PubMed/NCBI

151 

Kim HS, Jung M, Choi SK, Woo J, Piao YJ, Hwang EH, Kim H, Kim SJ and Moon WK: IL-6-mediated cross-talk between human preadipocytes and ductal carcinoma in situ in breast cancer progression. J Exp Clin Cancer Res. 37:2002018. View Article : Google Scholar : PubMed/NCBI

152 

Lee J, Hong BS, Ryu HS, Lee HB, Lee M, Park IA, Kim J, Han W, Noh DY and Moon HG: Transition into inflammatory cancer-associated adipocytes in breast cancer microenvironment requires microRNA regulatory mechanism. PLoS One. 12:e01741262017. View Article : Google Scholar : PubMed/NCBI

153 

Bachelot T, Ray-Coquard I, Menetrier-Caux C, Rastkha M, Duc A and Blay JY: Prognostic value of serum levels of interleukin 6 and of serum and plasma levels of vascular endothelial growth factor in hormone-refractory metastatic breast cancer patients. Br J Cancer. 88:1721–1726. 2003. View Article : Google Scholar : PubMed/NCBI

154 

Deng T, Lyon CJ, Bergin S, Caligiuri MA and Hsueh WA: Obesity, inflammation, and cancer. Annu Rev Pathol. 11:421–449. 2016. View Article : Google Scholar : PubMed/NCBI

155 

Liu S, Lee JS, Jie C, Park MH, Iwakura Y, Patel Y, Soni M, Reisman D and Chen H: HER2 overexpression triggers an IL1α proinflammatory circuit to drive tumorigenesis and promote chemotherapy resistance. Cancer Res. 78:2040–2051. 2018. View Article : Google Scholar : PubMed/NCBI

156 

Banerjee K and Resat H: Constitutive activation of STAT3 in breast cancer cells: A review. Int J Cancer. 138:2570–2578. 2016. View Article : Google Scholar : PubMed/NCBI

157 

Nickel A, Blücher C, Kadri OA, Schwagarus N, Müller S, Schaab M, Thiery J, Burkhardt R and Stadler SC: Adipocytes induce distinct gene expression profiles in mammary tumor cells and enhance inflammatory signaling in invasive breast cancer cells. Sci Rep. 8:94822018. View Article : Google Scholar : PubMed/NCBI

158 

Gyamfi J, Lee YH, Eom M and Choi J: Interleukin-6/STAT3 signalling regulates adipocyte induced epithelial-mesenchymal transition in breast cancer cells. Sci Rep. 8:88592018. View Article : Google Scholar : PubMed/NCBI

159 

Jin K, Pandey NB and Popel AS: Simultaneous blockade of IL-6 and CCL5 signaling for synergistic inhibition of triple-negative breast cancer growth and metastasis. Breast Cancer Res. 20:542018. View Article : Google Scholar : PubMed/NCBI

160 

Guo C, Chen Y, Gao W, Chang A, Ye Y, Shen W, Luo Y, Yang S, Sun P, Xiang R and Li N: Liposomal nanoparticles carrying anti-IL6R antibody to the tumour microenvironment inhibit metastasis in two molecular subtypes of breast cancer mouse models. Theranostics. 7:775–788. 2017. View Article : Google Scholar : PubMed/NCBI

161 

Gyamfi J, Lee YH, Min BS and Choi J: Niclosamide reverses adipocyte induced epithelial-mesenchymal transition in breast cancer cells via suppression of the interleukin-6/STAT3 signalling axis. Sci Rep. 9:113362019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2021
Volume 24 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yao H and Yao H: Multi‑faceted role of cancer‑associated adipocytes in the tumor microenvironment (Review). Mol Med Rep 24: 866, 2021
APA
Yao, H., & Yao, H. (2021). Multi‑faceted role of cancer‑associated adipocytes in the tumor microenvironment (Review). Molecular Medicine Reports, 24, 866. https://doi.org/10.3892/mmr.2021.12506
MLA
Yao, H., He, S."Multi‑faceted role of cancer‑associated adipocytes in the tumor microenvironment (Review)". Molecular Medicine Reports 24.6 (2021): 866.
Chicago
Yao, H., He, S."Multi‑faceted role of cancer‑associated adipocytes in the tumor microenvironment (Review)". Molecular Medicine Reports 24, no. 6 (2021): 866. https://doi.org/10.3892/mmr.2021.12506