Skip to main content

MINI REVIEW article

Front. Oncol., 10 May 2024
Sec. Cancer Immunity and Immunotherapy
This article is part of the Research Topic The Tumor Microenvironment and Immunotherapy for Head and Neck Tumors View all 7 articles

The role of neck adipose tissue in lymph node metastasis of head and neck cancer

Yiqi Pan&#x;Yiqi PanYing Xu&#x;Ying XuCui FanCui FanXiangwan MiaoXiangwan MiaoYilin ShenYilin ShenQuan WangQuan WangJichang WuJichang WuHaixia HuHaixia HuHao WangHao WangMingliang Xiang*Mingliang Xiang*Bin Ye*Bin Ye*
  • Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China

Previous studies indicated that adipose tissue significantly influences cancer invasion and lymphatic metastasis. However, the impact of neck adipose tissue (NAT) on lymph node metastasis associated with head and neck cancer remains ambiguous. Here, we systematically assess the classification and measurement criteria of NAT and evaluate the association of adipose tissue and cancer-associated adipocytes with head and neck cancer. We delve into the potential mechanisms by which NAT facilitate cervical lymph node metastasis in head and neck cancer, particularly through the secretion of adipokines such as leptin, adiponectin, and Interleukin-6. Our aim is to elucidate the role of NAT in the progression and metastasis of head and neck cancer, offering new insights into prevention and treatment.

1 Introduction

Cervical lymph nodes metastasis (LNM) is crucial in the clinical staging of head and neck cancer (HNC), and it serves as a vital indicator for assessing the progression and prognosis (1). Although adipose tissue (AT) is the predominant tissue surrounding cervical LNM, its relationship with LNM in HNC remains elusive. Previous studies have shown that breast cancer and prostate cancers are surrounded by abundant AT, forming a unique microenvironment between AT and cancer cells (2, 3). There exists a same crosstalk between cancer cells and adipocytes in HNC. This interplay continuously alters the tumor microenvironment, thus leading to the formation of specialized AT. AT can induce metabolic reprogramming in cancer, facilitating the uptake of free fatty acids and glycerol from adipocytes. This uptake serves as an energy source for oxidative phosphorylation in mitochondria, resulting in an “anti-Warburg effect” that enhances the invasion and metastasis of cancers such as breast and prostate cancers (24). AT releases higher levels of adipose-derived cytokines, such as leptin, Interleukin-6 (IL-6), and CC-chemokine ligand 5 (CCL5), promoting cancer proliferation and invasion (2). Esposito et al. identified a notable association between positive CCL5 staining in peritumoral adipocytes and LNM in breast cancer (5). Further, adipocytes in breast cancer rely on the fatty acid synthase ACSL3(acyl-CoA synthetase long-chain family member 3) to release MUFA (oleic acid), enabling cancer to resist ferroptosis (6). In prostate cancer, AT plays a role in recruiting immunosuppressive cells, modifying the extracellular matrix, supporting neovascularization, and inducing malignant tumor invasion (7). Yousuke Shimizu et al. discovered the existence of 2% LNM in the prostatic anterior fat pad of prostate cancer patients. Consequently, Urology guidelines recommend the routine removal of prostatic anterior fat pad during radical prostatectomy to minimize the risk of residual tumor tissue (8). It is evident that AT significantly influences LNM in some malignant tumors such as breast and prostate cancers.

Recent studies have identified an indirect link between AT and the invasion and metastasis of HNC (9, 10). Studies focusing on thyroid cancer (11), nasopharyngeal carcinoma (12), and oral squamous cell carcinoma (13) have demonstrated associations between body mass index (BMI) and incidence rate, aggressive pathological features, and unfavorable clinical outcomes. Lymphatic metastasis serves as a primary route for the local metastasis of HNC. AT, the principal energy source in the tumor microenvironment, facilitates lymphangiogenesis (14). Additionally, overwhelming evidence supports the notion that AT is an endocrine tissue that can secrete a variety of adipokines, such as leptin (15), adiponectin (16), and IL-6 (17), which also contribute to cancer invasion (15). Therefore, we analyzed and summarized the correlation between NAT and lymph node metastasis in HNC. We started by summarizing the current methodologies for quantifying NAT. Then, we tried to research the associations between NAT and LNM in HNC. Finally, we analyzed the mechanisms by which NAT might promote LNM. Our aim is to identify potential NAT risk factors for LNM in HNC, ultimately improving the prognosis of patients with HNC.

1.1 Definition, classification, and function of adipose tissue

AT, a specialized connective tissue, predominantly consists of adipocytes (18). Beyond adipocytes, it also comprises adipose-derived stem cells, preadipocytes, fibroblasts, lymphocytes, macrophages, and vascular endothelial cells (19). It is essential for mechanical support, thermoregulation, energy storage and release, appetite, and immune regulation (18). BMI is calculated by the formula: weight divided by height squared (kg/m²), which serves as an indirect indicator of overall adiposity (20).

There are three principal types of AT, namely, white adipose tissue (WAT), brown adipose tissue (BAT), and beige adipose tissue. WAT is characterized by large unilocular lipid droplets and a limited number of mitochondria (21). WAT is the most abundant type in the adult neck (22). WAT primarily regulates the storage and release of energy to cater to the needs of various tissues (23). White adipocytes can be converted to thermogenic beige adipocytes following stimuli such as exercise or cold exposure, a process termed “browning of WAT” (24). Beige adipocytes are thermogenically active, bear a morphological and biochemical resemblance to BAT, and include multilocular lipid droplets and abundant mitochondria (25).

BAT is distinguished by small multilocular lipid droplets and a profusion of mitochondria rich in cytochrome content (21). A series of studies using 18F-FDG-PET/CT detection found that the most common location of BAT in the adult neck is in the frontal aspect, superficial and lateral to the sternocleidomastoid muscle, and supraclavicular regions (26, 27). Although BAT constitutes a minor portion of body mass, it is pivotal for non-shivering heat production during cold exposure (18). Recent studies have revealed intriguing links between BAT and cancer proliferation. Takahiro Seki et al. observed that cold environment was found to activated substantial BAT in the mice, leading to inhibited energy uptake and consequent tumor cell apoptosis (28).

2 Relationship between BMI, NAT and lymph node metastasis in HNC

2.1 Relationship between BMI, NAT and lymph node metastasis in thyroid cancer

NC can serve as a direct indicator of NAT accumulation around the respiratory tract or within the cervical subcutaneous AT layer, while BMI provides an indirect assessment of NAT. Many studies have delved into the prognostic value of NC and BMI in thyroid cancer. Excluding patients with a tumor size greater than 2 cm, Kim et al. found that male patients with lateral LNM had a notably larger NC compared with those without metastasis. Thus, NC emerged as a predictor of cervical LNM in male patients with thyroid cancer (29). A retrospective cohort study involving 796 patients diagnosed with early-stage papillary thyroid cancer found that when BMI was ≥18.5 kg/m², the average number of LNM in the central and lateral cervical regions increased proportionately with BMI. For overweight patients, the incidence of central and lateral cervical LNM was 55.3% and 37.9%, respectively. By contrast, these figures for the obese group were 55.9% and 45.3%, respectively (30). Similarly, Li et al. demonstrated that obese patients with papillary thyroid cancer had a significantly higher incidence of metastasis to the central and lateral lymph nodes compared with their normal-weight counterparts (31). By contrast, Zhao et al. found that their findings did not reveal any significant differences in cervical LNM across different BMI categories (32). Numerous studies have demonstrated that overall AT and NAT facilitate the process of LNM in thyroid cancer. However, disparities between individual studies arise from variations in sample size, ethnic background, and the inherent limitation of BMI (its inability to distinguish between muscle and AT or to quantify specific AT compartments). Although there is a strong correlation between BMI and NAT, there is insufficient evidence to directly replace association between BMI and HNC with that between NAT and HNC.

2.2 Relationship between BMI, NAT and other HNC

BMI serves as a significant indicator of NAT. In the context of oral cancer, Bao et al. analyzed BMI data from 1,395 oral cancer patients between 2007 and 2018. They observed that underweight patients exhibited inferior survival outcomes (HR = 1.585; 95% CI: 1.207–2.082) (13). Choi et al. employed contrast-enhanced CT scans of the neck (from the anterior superior of the hyoid to the third cervical spine and inferiorly to the first rib) in 79 patients with various HNC. Using the 3D slicer tool, they measured NAT volume changes both pre- and post-radiotherapy over one year. Their findings indicated that patients with low NAT volume before and after treatment had poorer overall survival rates. Further, significant weight loss during treatment was also linked to diminished overall and recurrence-free survival rates (33). An increased NAT volume appears to improve the prognosis of patients with HNC. This improvement might be attributed to the protective role of AT in helping patients withstand the side effects of radiotherapy and the nutritional challenges associated with cancer. Expanding on this, Huang et al. studied 400 stage III or IVa nasopharyngeal carcinoma patients. Their research probed the correlation between pretreatment BMI and clinical outcomes in patients undergoing chemoradiotherapy. The results revealed 5-year failure-free survival rates of 44%, 61%, 68%, and 73%, and 5-year overall survival rates of 51%, 68%, 80%, and 72% for underweight, normal weight, overweight, and obese groups, respectively (34). They postulated that an adequate volume of AT could potentially ameliorate the adverse effects of chemoradiotherapy in advanced nasopharyngeal carcinoma cases and counteract cachexia in cancer patients. Similarly, our research group has previously demonstrated that adipose tissue and lipid metabolism related factors exhibited a regulatory influence on the process of LNM and prognosis in patients with HNC (35, 36). However, given the limited research on the interplay between NAT and HNC, further studies are essential to ascertain the exact impact of NAT on prognosis and the potential mechanisms by which AT might promote cervical LNM in HNC.

3 Mechanism of adipose tissue promoting cervical lymph node metastasis of HNC

3.1 Lymphangiogenesis enhanced by adipose tissue

Lymphatic vessels in the human body play a significant role in lipid transport and absorption. Peter et al. reported that the expression of the fatty acid β-oxidation (FAO) pathway was markedly elevated in lymphatic vessels compared with other vessel types. Further investigation showed that by utilizing fatty acids for β-oxidation, lymphatic endothelial cells enhanced the expression of the lymphangiogenic factor-prox1, thus facilitating the formation of new lymphatic channels (37). The vascular endothelial growth factor-C (VEGF-C) stands out as a potent lymphangiogenic factor. Adipose-derived stem cells (ADSCs) are known to secrete growth factors and exosomes, thereby modulating the tumor microenvironment. A study carried out in 2018 unveiled that following VEGF-C treatment, ADSCs-secreted miR-132 was transferred to lymphatic endothelial cells via exosomes. The uptake of miR-132 by these cells stimulated their proliferation, migration, and formation of lymphatic channels. This discovery underscores the regulatory role of ADSCs exosomes in VEGF-C-mediated lymphangiogenesis (38). Collectively, these insights highlight the importance of AT in lymphatic vessel development and functionality. When primary tumors are present in the head and neck regions, NAT might facilitate lymphatic metastasis of these tumors by regulating lymphatic vessel.

3.2 Cancer-associated adipocytes

Adipocytes that interact with cancer cells are termed “cancer-associated adipocytes” (4). The idea that adipocytes might influence tumor progression was initially suggested by Spector et al. In 2003, Puneeth et al. discovered that adipocytes surrounding breast cancer tissues promoted tumor progression. This promotion was achieved by the secretion of collagen VI, which induced an anti-apoptotic transcriptional program and stabilized proto-oncogenes in tumor cells (39). Subsequently, Dirat et al. revealed that breast cancer cells exhibited increased invasiveness when co-cultivated with mature adipocytes. Further, the number of lung metastases was enhanced in mice injected with adipocytes co-cultivated with 4T1 tumor cells compared with mice injected with 4T1 cells alone. Intriguingly, when co-cultured with breast tumor cells, mature adipocytes showed a marked reduction in the number and size of lipid droplets and a decreased expression of adipocyte differentiation markers such as hormone-sensitive triglyceride lipase (HSL), resistin, and adiponectin. By contrast, upregulation of the expression of proinflammatory cytokines (e.g., IL6, IL1β, TNFα) and matrix remodeling proteins (e.g., MMP-11, PAI-1) was observed. In various solid tumors such as breast cancer (4), prostate cancer (7), melanoma (40), and colorectal cancer (41), the invasion of tumor cells into the surrounding AT is linked to a profound reduction of lipid in adipocytes. Nieman et al. noted that ovarian cancer preferentially metastasizes to the omentum, which is rich in adipocytes. They further found that co-culturing adipocytes with ovarian cancer cells led to a direct lipid transfer from adipocytes to the cancer cells. This process allowed cancer cells to utilize fatty acids by β-oxidation (42). In addition, cancer-associated adipocytes have been found to release high levels of cytokines and growth factors such as IL-6, CCL2, CCL5, IL1β, TNFα, and VEGF, which collectively contribute to enhanced tumor cell proliferation, invasion, and angiogenesis (43). Consequently, the interaction between cancer-associated adipocytes and cancer cells in the tumor microenvironment serves to bolster the survival, proliferation, and metastatic potential of the cancer through direct lipid exchange or adipokine secretion (44).

3.3 Adipokine in the tumor microenvironment promotes lymph node metastasis in HNC

3.3.1 Leptin

Leptin is a product encoded by the LEP gene on human chromosome 7. It is a 16 kDa adipokine synthesized and secreted by adipocytes, primarily playing a crucial role in regulating energy metabolism and promoting cell proliferation. Both leptin and its receptor are highly expressed in thyroid cancer, salivary gland carcinoma, oral squamous cell carcinoma, and laryngeal cancer. Further, the expression of leptin and its receptor is positively correlated with cancer invasiveness indicators including tumor size and LNM (4549). Cheng et al. assessed the levels of leptin and its receptor in 49 primary tumors and 15 LNM using immunohistochemistry. They discovered that leptin and its receptor were expressed in 37% and 51% of papillary thyroid carcinomas, respectively. The co-expression of leptin and its receptor in primary tumors was associated with a higher likelihood of LNM (50). Leptin can stimulate tumor cells invasion and inhibit tumor cells apoptosis. Eliane et al. found that in SCC-9 and SCC-4 oral squamous cell lines, leptin promoted the expression of genes related to angiogenesis and invasiveness such as E-cadherin, Col1A1, MMP2, and MMP9, thereby enhancing cell proliferation and invasiveness (49). Further, leptin can enhance the migration of thyroid cancer cells through the PI3K/AKT and MEK/ERK signaling pathways (51). Through an in vitro study, Shahab et al. determined that overexpression of the leptin receptor can inhibit apoptosis by upregulating BCl-XL and XIAP (anti-apoptotic genes) (47).

3.3.2 Adiponectin

Adiponectin is a primary adipokine secreted by AT that can also be produced by cardiomyocytes, skeletal muscle cells, and lymphocytes (52). Adiponectin belongs to the complement factor C1q-like protein superfamily. Adiponectin primarily functions in regulating glucose metabolism and stimulating FAO (53). Recently, a strong inverse correlation was shown between adiponectin levels and the incidence of various malignant tumors, such as colorectal cancer, breast cancer, prostate cancer, leukemia, and endometrial cancer. Adiponectin is also considered a potent anticancer factor that inhibits cancer growth. In endometrial cancer, adiponectin activates AMPK and downregulates Bcl-2 and MMP-9 expression, consequently inhibiting the invasion of tumor cells and promoting tumor cell apoptosis (54). However, research on the association between adiponectin and HNC is limited. Nicholas et al. found a significant independent negative correlation between circulating adiponectin levels and the risk of thyroid cancer (55). Cheng et al. determined that AdipoR1 was expressed in 27% of primary malignant tumors, while AdipoR2 was found in 47% of primary malignant tumors via immunohistochemical staining of 49 thyroid tumor samples and metastatic lymph nodes. In addition, negative expression of both adiponectin receptors was significantly correlated with extrathyroidal invasion, multicentricity, and higher TNM staging (56). Ersilia et al. discovered that adiponectin inhibited the proliferation of papillary thyroid cancer cell lines (BCPAP and K1) and anaplastic thyroid cancer cell lines (CAL62). Current research suggests that adiponectin exerts its effect by binding to its receptors and regulating AKT/mTOR/PI3K and MAPK signaling pathways, which are associated with cell proliferation and energy modulation, thereby inhibiting the activity and growth of thyroid cancer (16). Evidently, adiponectin serves as a protective adipokine against HNC. Therefore, reduced levels of adiponectin in obese individuals can potentially promote the onset of HNC.

3.3.3 Interleukin-6

IL-6 is a multifunctional cytokine that plays a crucial role in the broad biological activity of cancer cells. IL-6 is involved in immune modulation and tumorigenesis. In pathological conditions of obesity and cancer, IL-6 levels secreted by adipocytes are significantly increased (17). Nandita et al. found that serum IL-6 levels positively correlated with tumor size, extrathyroidal invasion, and distant metastasis in papillary thyroid carcinoma patients (17). In prostate cancer (57), breast cancer (58), ovarian cancer (59), non-small cell lung cancer (60), and endometrial cancer (61), IL-6 also had a correlation with clinical progression of the cancer. Numerous studies suggest that IL-6 can promote tumor invasion, inhibit tumor cell death, and facilitate tumor cell immune evasion through various mechanisms. IL-6 binds to a specific binding receptor on the cell membrane (IL-6R), leading to activation of the JAK/STAT3 pathway and promoting thyroid tumor cell invasion (62). Similarly, Mingyu et al. collected specimens from normal tissues, vocal cord leukoplakia, and HNC. they found that levels of IL-6 were higher than in normal epithelium. It was discovered that IL-6 transcriptionally activates xCT, a key amino acid antiporter, via the JAK2/STAT3 signaling pathway. The upregulation of xCT induces ferroptosis resistance and tumor progression, suggesting IL-6 as a novel oncogenic ferroptosis inhibitor (63).

3.3.4 Other substances

Extracellular vesicles serve as a critical conduit for communication between adipocytes and tumor cells. These vesicles transport proteins and fatty acids related to lipid metabolism. Once internalized by tumor cells, they enhance FAO (14). Adipocytes cultured in high-fat conditions exhibit an increased secretion of extracellular vesicles. The fatty acids from these vesicles accumulate in the lipid droplets of cancer cells and are subsequently released during fat autophagy, further driving FAO (14). Further, cancer cells can release extracellular vesicles that stimulate lipolysis in adipocytes (44). For example, extracellular vesicles from lung cancer cells were found to be enriched with IL-6, which triggers lipolysis in adipocytes by activating the STAT3 pathway (64). Beyond this, adipose tissue releases other adipokines including interleukin-8 (65), IGFBP-2 (66), β-hydroxybutyrate (67), CD36 (35), FASN (35) and IGF-1 (68), which can influence tumor invasion.

4 Conclusion

Given the association of breast cancer, prostate cancer, and other malignant tumors with surrounding AT, as well as studies correlating NC and BMI with LNM in HNC, it is evident that NAT foster cervical LNM in HNC. On the one hand, NAT can directly fuel tumors and nascent lymphatic vessels through FAO. In addition, it might secrete adipokines or extracellular vesicles that modulate tumor growth or alter the microenvironment around the malignant tumor, thereby influencing malignant tumor invasion and migration. On the other hand, HNC can induce the transformation of typical adipocytes in the neck to cancer-associated adipocytes. This can shift the metabolic expression patterns of HNC, creating a feedback loop that fosters growth, migration, and LNM.

Currently, the bulk of research centers on the association between BMI and HNC, with limited epidemiological and foundational studies directly linking NAT to HNC. There is no universally accepted methodology for quantifying NAT. Future research should prioritize investigating the connection between NAT and various HNC, delineate the alterations in submental NAT instigated by HNC and discern the role of NAT in the progression of HNC. Delving into the mechanisms by which NAT drives HNC and LNM could provide novel insights and strategies for the diagnosis and management of HNC.

Author contributions

YP: Writing – original draft, Writing – review & editing. YX: Writing – original draft. CF: Writing – review & editing. XM: Writing – review & editing. YS: Writing – review & editing. QW: Writing – review & editing. JW: Writing – review & editing. HH: Writing – review & editing. HW: Writing – review & editing. MX: Writing – review & editing. BY: Writing – original draft, Writing – review & editing.

Funding

The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This study was supported by grants from the National Natural Science Foundation of China (grant No.82101212, 82101209, 82301296, 82301297), and Science and Technology Commission of Shanghai Municipality (grant No.23ZR1440200, 21ZR1440200).

Acknowledgments

This is a short text to acknowledge the contributions of specific colleagues, institutions, or agencies that aided the efforts of the authors.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

The handling editor JH declared a shared parent affiliation with the author(s) at the time of review.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Zhang X, Rui M, Lin C, Li Z, Wei D, Han R, et al. The association between body mass index and efficacy of pembrolizumab as second-line therapy in patients with recurrent/metastatic head and neck squamous cell carcinoma. Cancer Med. (2023) 12:2702–12. doi: 10.1002/cam4.5152

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Kothari C, Diorio C, Durocher F. The importance of breast adipose tissue in breast cancer. Int J Mol Sci. (2020) 21:5760. doi: 10.3390/ijms21165760

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Feng S, Lou K, Luo C, Zou J, Zou X, Zhang G. Obesity-related cross-talk between prostate cancer and peripheral fat: potential role of exosomes. Cancers (Basel). (2022) 14:5077. doi: 10.3390/cancers14205077

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Dirat B, Bochet L, Dabek M, Daviaud D, Dauvillier S, Majed B, et al. Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Res. (2011) 71:2455–65. doi: 10.1158/0008-5472.Can-10-3323

PubMed Abstract | CrossRef Full Text | Google Scholar

5. D'Esposito V, Liguoro D, Ambrosio MR, Collina F, Cantile M, Spinelli R, et al. Adipose microenvironment promotes triple negative breast cancer cell invasiveness and dissemination by producing Ccl5. Oncotarget. (2016) 7:24495–509. doi: 10.18632/oncotarget.8336

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Xie Y, Wang B, Zhao Y, Tao Z, Wang Y, Chen G, et al. Mammary adipocytes protect triple-negative breast cancer cells from ferroptosis. J Hematol Oncol. (2022) 15:72. doi: 10.1186/s13045-022-01297-1

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Saha A, Kolonin MG, DiGiovanni J. Obesity and prostate cancer - microenvironmental roles of adipose tissue. Nat Rev Urol. (2023) 20:579–96. doi: 10.1038/s41585-023-00764-9

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Shimizu Y, Ogawa K, Kida K, Tsuchihashi K, Kanamaru S, Ishihara M, et al. [Lymph node metastasis to the prostatic anterior fat pad in the patients treated with prostatectomy]. Hinyokika Kiyo. (2018) 64:359–63. doi: 10.14989/ActaUrolJap_64_9_359

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Dieringer P, Klass E, Caine B, Smith-Gagen J. Associations between body mass and papillary thyroid cancer stage and tumor size: A population-based study. J Cancer Res Clin Oncol. (2015) 141:93–8. doi: 10.1007/s00432-014-1792-2

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Fattouh M, Chang GY, Ow TJ, Shifteh K, Rosenblatt G, Patel VM, et al. Association between pretreatment obesity, sarcopenia, and survival in patients with head and neck cancer. Head Neck. (2019) 41:707–14. doi: 10.1002/hed.25420

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Economides A, Giannakou K, Mamais I, Economides PA, Papageorgis P. Association between aggressive clinicopathologic features of papillary thyroid carcinoma and body mass index: A systematic review and meta-analysis. Front Endocrinol (Lausanne). (2021) 12:692879. doi: 10.3389/fendo.2021.692879

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Li W, Shen LJ, Chen T, Sun XQ, Zhang Y, Wu M, et al. Overweight/obese status associates with favorable outcome in patients with metastatic nasopharyngeal carcinoma: A 10-year retrospective study. Chin J Cancer. (2016) 35:75. doi: 10.1186/s40880-016-0139-6

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Bao X, Liu F, Lin J, Chen Q, Chen L, Chen F, et al. Nutritional assessment and prognosis of oral cancer patients: A large-scale prospective study. BMC Cancer. (2020) 20:146. doi: 10.1186/s12885-020-6604-2

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Clement E, Lazar I, Attané C, Carrié L, Dauvillier S, Ducoux-Petit M, et al. Adipocyte extracellular vesicles carry enzymes and fatty acids that stimulate mitochondrial metabolism and remodeling in tumor cells. EMBO J. (2020) 39:e102525. doi: 10.15252/embj.2019102525

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Herrera-Vargas AK, García-Rodríguez E, Olea-Flores M, Mendoza-Catalán MA, Flores-Alfaro E, Navarro-Tito N. Pro-angiogenic activity and vasculogenic mimicry in the tumor microenvironment by leptin in cancer. Cytokine Growth Factor Rev. (2021) 62:23–41. doi: 10.1016/j.cytogfr.2021.10.006

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Nigro E, Orlandella FM, Polito R, Mariniello RM, Monaco ML, Mallardo M, et al. Adiponectin and leptin exert antagonizing effects on proliferation and motility of papillary thyroid cancer cell lines. J Physiol Biochem. (2021) 77:237–48. doi: 10.1007/s13105-021-00789-x

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Kobawala TP, Trivedi TI, Gajjar KK, Patel DH, Patel GH, Ghosh NR. Significance of interleukin-6 in papillary thyroid carcinoma. J Thyroid Res. (2016) 2016:6178921. doi: 10.1155/2016/6178921

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Barthelemy J, Bogard G, Wolowczuk I. Beyond energy balance regulation: the underestimated role of adipose tissues in host defense against pathogens. Front Immunol. (2023) 14:1083191. doi: 10.3389/fimmu.2023.1083191

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Sacca PA, Calvo JC. Periprostatic adipose tissue microenvironment: metabolic and hormonal pathways during prostate cancer progression. Front Endocrinol (Lausanne). (2022) 13:863027. doi: 10.3389/fendo.2022.863027

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Hobday S, Armache M, Paquin R, Nurimba M, Baddour K, Linder D, et al. The body mass index paradox in head and neck cancer: A systematic review and meta-analysis. Nutr Cancer. (2023) 75:48–60. doi: 10.1080/01635581.2022.2102659

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Álvarez-Artime A, García-Soler B, Sainz RM, Mayo JC. Emerging roles for browning of white adipose tissue in prostate cancer Malignant behaviour. Int J Mol Sci. (2021) 22:5560. doi: 10.3390/ijms22115560

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Trim WV, Lynch L. Immune and non-immune functions of adipose tissue leukocytes. Nat Rev Immunol. (2022) 22:371–86. doi: 10.1038/s41577-021-00635-7

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Cancel M, Pouillot W, Mahéo K, Fontaine A, Crottès D, Fromont G. Interplay between prostate cancer and adipose microenvironment: A complex and flexible scenario. Int J Mol Sci. (2022) 23:10762. doi: 10.3390/ijms231810762

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Cheng L, Wang J, Dai H, Duan Y, An Y, Shi L, et al. Brown and beige adipose tissue: A novel therapeutic strategy for obesity and type 2 diabetes mellitus. Adipocyte. (2021) 10:48–65. doi: 10.1080/21623945.2020.1870060

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Cohen P, Kajimura S. The cellular and functional complexity of thermogenic fat. Nat Rev Mol Cell Biol. (2021) 22:393–409. doi: 10.1038/s41580-021-00350-0

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Cypess AM, Lehman S, Williams G, Tal I, Rodman D, Goldfine AB, et al. Identification and importance of brown adipose tissue in adult humans. N Engl J Med. (2009) 360:1509–17. doi: 10.1056/NEJMoa0810780

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Zhang F, Hao G, Shao M, Nham K, An Y, Wang Q, et al. An adipose tissue atlas: an image-guided identification of human-like bat and beige depots in rodents. Cell Metab. (2018) 27:252–62.e3. doi: 10.1016/j.cmet.2017.12.004

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Seki T, Yang Y, Sun X, Lim S, Xie S, Guo Z, et al. Brown-fat-mediated tumour suppression by cold-altered global metabolism. Nature. (2022) 608:421–8. doi: 10.1038/s41586-022-05030-3

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Kim MR, Kim SS, Huh JE, Lee BJ, Lee JC, Jeon YK, et al. Neck circumference correlates with tumor size and lateral lymph node metastasis in men with small papillary thyroid carcinoma. Korean J Intern Med. (2013) 28:62–71. doi: 10.3904/kjim.2013.28.1.62

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Wu C, Wang L, Chen W, Zou S, Yang A. Associations between body mass index and lymph node metastases of patients with papillary thyroid cancer: A retrospective study. Med (Baltimore). (2017) 96:e6202. doi: 10.1097/md.0000000000006202

CrossRef Full Text | Google Scholar

31. Li C, Dionigi G, Liang N, Guan H, Sun H. The relationship between body mass index and different regional patterns of lymph node involvement in papillary thyroid cancers. Front Oncol. (2021) 11:767245. doi: 10.3389/fonc.2021.767245

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Zhao S, Jia X, Fan X, Zhao L, Pang P, Wang Y, et al. Association of obesity with the clinicopathological features of thyroid cancer in a large, operative population: A retrospective case-control study. Med (Baltimore). (2019) 98:e18213. doi: 10.1097/md.0000000000018213

CrossRef Full Text | Google Scholar

33. Choi Y, Ahn KJ, Jang J, Shin NY, Jung SL, Kim BS, et al. Prognostic value of computed tomography-based volumetric body composition analysis in patients with head and neck cancer: feasibility study. Head Neck. (2020) 42:2614–25. doi: 10.1002/hed.26310

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Huang PY, Wang CT, Cao KJ, Guo X, Guo L, Mo HY, et al. Pretreatment body mass index as an independent prognostic factor in patients with locoregionally advanced nasopharyngeal carcinoma treated with chemoradiotherapy: findings from a randomised trial. Eur J Cancer. (2013) 49:1923–31. doi: 10.1016/j.ejca.2013.01.027

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Miao X, Wang H, Fan C, Song Q, Ding R, Wu J, et al. Enhancing prognostic accuracy in head and neck squamous cell carcinoma chemotherapy via a lipid metabolism-related clustered polygenic model. Cancer Cell Int. (2023) 23:164. doi: 10.1186/s12935-023-03014-5

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Miao X, Wang B, Chen K, Ding R, Wu J, Pan Y, et al. Perspectives of lipid metabolism reprogramming in head and neck squamous cell carcinoma: an overview. Front Oncol. (2022) 12:1008361. doi: 10.3389/fonc.2022.1008361

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Wong BW, Wang X, Zecchin A, Thienpont B, Cornelissen I, Kalucka J, et al. The role of fatty acid B-oxidation in lymphangiogenesis. Nature. (2017) 542:49–54. doi: 10.1038/nature21028

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Wang X, Wang H, Cao J, Ye C. Exosomes from adipose-derived stem cells promotes vegf-C-dependent lymphangiogenesis by regulating mirna-132/tgf-B Pathway. Cell Physiol Biochem. (2018) 49:160–71. doi: 10.1159/000492851

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Iyengar P, Combs TP, Shah SJ, Gouon-Evans V, Pollard JW, Albanese C, et al. Adipocyte-secreted factors synergistically promote mammary tumorigenesis through induction of anti-apoptotic transcriptional programs and proto-oncogene stabilization. Oncogene. (2003) 22:6408–23. doi: 10.1038/sj.onc.1206737

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Kwan HY, Fu X, Liu B, Chao X, Chan CL, Cao H, et al. Subcutaneous adipocytes promote melanoma cell growth by activating the akt signaling pathway: role of palmitic acid. J Biol Chem. (2014) 289:30525–37. doi: 10.1074/jbc.M114.593210

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Olszańska J, Pietraszek-Gremplewicz K, Domagalski M, Nowak D. Mutual impact of adipocytes and colorectal cancer cells growing in co-culture conditions. Cell Commun Signal. (2023) 21:130. doi: 10.1186/s12964-023-01155-8

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Nieman KM, Kenny HA, Penicka CV, Ladanyi A, Buell-Gutbrod R, Zillhardt MR, et al. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat Med. (2011) 17:1498–503. doi: 10.1038/nm.2492

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Wu Q, Li B, Li Z, Li J, Sun S, Sun S. Cancer-associated adipocytes: key players in breast cancer progression. J Hematol Oncol. (2019) 12:95. doi: 10.1186/s13045-019-0778-6

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Dumas JF, Brisson L. Interaction between adipose tissue and cancer cells: role for cancer progression. Cancer Metastasis Rev. (2021) 40:31–46. doi: 10.1007/s10555-020-09934-2

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Zhang GA, Hou S, Han S, Zhou J, Wang X, Cui W. Clinicopathological implications of leptin and leptin receptor expression in papillary thyroid cancer. Oncol Lett. (2013) 5:797–800. doi: 10.3892/ol.2013.1125

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Zhang KY, Liu CY, Hua L, Wang SL, Li J. Clinical evaluation of salivary carbohydrate antigen 125 and leptin in controls and parotid tumours. Oral Dis. (2016) 22:630–8. doi: 10.1111/odi.12505

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Uddin S, Bavi P, Siraj AK, Ahmed M, Al-Rasheed M, Hussain AR, et al. Leptin-R and its association with pi3k/akt signaling pathway in papillary thyroid carcinoma. Endocr Relat Cancer. (2010) 17:191–202. doi: 10.1677/erc-09-0153

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Hussain SR, Naqvi H, Gupta S, Mahdi AA, Kumari P, Waseem M, et al. A study on oncogenic role of leptin and leptin receptor in oral squamous cell. Tumour Biol. (2015) 36:6515–23. doi: 10.1007/s13277-015-3342-1

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Sobrinho Santos EM, Guimarães TA, Santos HO, Cangussu LMB, de Jesus SF, Fraga CAC, et al. Leptin acts on neoplastic behavior and expression levels of genes related to hypoxia, angiogenesis, and invasiveness in oral squamous cell carcinoma. Tumour Biol. (2017) 39:1010428317699130. doi: 10.1177/1010428317699130

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Cheng SP, Chi CW, Tzen CY, Yang TL, Lee JJ, Liu TP, et al. Clinicopathologic significance of leptin and leptin receptor expressions in papillary thyroid carcinoma. Surgery. (2010) 147:847–53. doi: 10.1016/j.surg.2009.11.004

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Cheng SP, Yin PH, Hsu YC, Chang YC, Huang SY, Lee JJ, et al. Leptin enhances migration of human papillary thyroid cancer cells through the pi3k/akt and mek/erk signaling pathways. Oncol Rep. (2011) 26:1265–71. doi: 10.3892/or.2011.1388

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Zhou Y, Yang Y, Zhou T, Li B, Wang Z. Adiponectin and thyroid cancer: insight into the association between adiponectin and obesity. Aging Dis. (2021) 12:597–613. doi: 10.14336/ad.2020.0919

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Fang H, Judd RL. Adiponectin regulation and function. Compr Physiol. (2018) 8:1031–63. doi: 10.1002/cphy.c170046

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Ray I, Meira LB, Michael A, Ellis PE. Adipocytokines and disease progression in endometrial cancer: A systematic review. Cancer Metastasis Rev. (2022) 41:211–42. doi: 10.1007/s10555-021-10002-6

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Mitsiades N, Pazaitou-Panayiotou K, Aronis KN, Moon HS, Chamberland JP, Liu X, et al. Circulating adiponectin is inversely associated with risk of thyroid cancer: in vivo and in vitro studies. J Clin Endocrinol Metab. (2011) 96:E2023–8. doi: 10.1210/jc.2010-1908

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Cheng SP, Liu CL, Hsu YC, Chang YC, Huang SY, Lee JJ. Expression and biologic significance of adiponectin receptors in papillary thyroid carcinoma. Cell Biochem Biophys. (2013) 65:203–10. doi: 10.1007/s12013-012-9419-1

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Nguyen DP, Li J, Tewari AK. Inflammation and prostate cancer: the role of interleukin 6 (Il-6). BJU Int. (2014) 113:986–92. doi: 10.1111/bju.12452

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Fujisaki K, Fujimoto H, Sangai T, Nagashima T, Sakakibara M, Shiina N, et al. Cancer-mediated adipose reversion promotes cancer cell migration via il-6 and mcp-1. Breast Cancer Res Treat. (2015) 150:255–63. doi: 10.1007/s10549-015-3318-2

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Cardenas C, Montagna MK, Pitruzzello M, Lima E, Mor G, Alvero AB. Adipocyte microenvironment promotes bcl(Xl) expression and confers chemoresistance in ovarian cancer cells. Apoptosis. (2017) 22:558–69. doi: 10.1007/s10495-016-1339-x

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Songür N, Kuru B, Kalkan F, Ozdilekcan C, Cakmak H, Hizel N. Serum interleukin-6 levels correlate with malnutrition and survival in patients with advanced non-small cell lung cancer. Tumori. (2004) 90:196–200. doi: 10.1177/030089160409000207

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Bellone S, Watts K, Cane S, Palmieri M, Cannon MJ, Burnett A, et al. High serum levels of interleukin-6 in endometrial carcinoma are associated with uterine serous papillary histology, a highly aggressive and chemotherapy-resistant variant of endometrial cancer. Gynecol Oncol. (2005) 98:92–8. doi: 10.1016/j.ygyno.2005.03.016

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Grivennikov S, Karin M. Autocrine il-6 signaling: A key event in tumorigenesis? Cancer Cell. (2008) 13:7–9. doi: 10.1016/j.ccr.2007.12.020

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Li M, Jin S, Zhang Z, Ma H, Yang X. Interleukin-6 facilitates tumor progression by inducing ferroptosis resistance in head and neck squamous cell carcinoma. Cancer Lett. (2022) 527:28–40. doi: 10.1016/j.canlet.2021.12.011

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Hu W, Ru Z, Zhou Y, Xiao W, Sun R, Zhang S, et al. Lung cancer-derived extracellular vesicles induced myotube atrophy and adipocyte lipolysis via the extracellular il-6-mediated stat3 pathway. Biochim Biophys Acta Mol Cell Biol Lipids. (2019) 1864:1091–102. doi: 10.1016/j.bbalip.2019.04.006

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Vairaktaris E, Yapijakis C, Serefoglou Z, Derka S, Vassiliou S, Nkenke E, et al. The interleukin-8 (-251a/T) polymorphism is associated with increased risk for oral squamous cell carcinoma. Eur J Surg Oncol. (2007) 33:504–7. doi: 10.1016/j.ejso.2006.11.002

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Wang C, Gao C, Meng K, Qiao H, Wang Y. Human adipocytes stimulate invasion of breast cancer mcf-7 cells by secreting igfbp-2. PloS One. (2015) 10:e0119348. doi: 10.1371/journal.pone.0119348

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Huang CK, Chang PH, Kuo WH, Chen CL, Jeng YM, Chang KJ, et al. Adipocytes promote Malignant growth of breast tumours with monocarboxylate transporter 2 expression via B-hydroxybutyrate. Nat Commun. (2017) 8:14706. doi: 10.1038/ncomms14706

PubMed Abstract | CrossRef Full Text | Google Scholar

68. D'Esposito V, Passaretti F, Hammarstedt A, Liguoro D, Terracciano D, Molea G, et al. Adipocyte-released insulin-like growth factor-1 is regulated by glucose and fatty acids and controls breast cancer cell growth in vitro. Diabetologia. (2012) 55:2811–22. doi: 10.1007/s00125-012-2629-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: adipose tissue, head and neck cancer, lymph node metastasis, tumor microenvironment, lymphangiogenesis

Citation: Pan Y, Xu Y, Fan C, Miao X, Shen Y, Wang Q, Wu J, Hu H, Wang H, Xiang M and Ye B (2024) The role of neck adipose tissue in lymph node metastasis of head and neck cancer. Front. Oncol. 14:1390824. doi: 10.3389/fonc.2024.1390824

Received: 24 February 2024; Accepted: 22 April 2024;
Published: 10 May 2024.

Edited by:

Jingzhou Hu, Shanghai Jiao Tong University, China

Reviewed by:

Yanming Liu, Zhejiang University, China

Copyright © 2024 Pan, Xu, Fan, Miao, Shen, Wang, Wu, Hu, Wang, Xiang and Ye. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Bin Ye, yb12237@rjh.com.cn; Mingliang Xiang, mingliangxiang@163.com

These authors have contributed equally to this work and share first authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.