Open Access

Impact of obesity‑associated myeloid‑derived suppressor cells on cancer risk and progression (Review)

  • Authors:
    • Carlos Jiménez‑Cortegana
    • Cristian Gutiérrez‑García
    • Flora Sánchez‑Jiménez
    • Teresa Vilariño‑García
    • Rocio Flores‑Campos
    • Antonio Pérez‑Pérez
    • Carmen Garnacho
    • Maria L. Sánchez‑León
    • Daniel J. García‑Domínguez
    • Lourdes Hontecillas‑Prieto
    • Natalia Palazón‑Carrión
    • Luis De La Cruz‑Merino
    • Víctor Sánchez‑Margalet
  • View Affiliations

  • Published online on: June 27, 2024     https://doi.org/10.3892/ijo.2024.5667
  • Article Number: 79
  • Copyright: © Jiménez‑Cortegana 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

Obesity is a chronic disease caused by the accumulation of excessive adipose tissue. This disorder is characterized by chronic low‑grade inflammation, which promotes the release of proinflammatory mediators, including cytokines, chemokines and leptin. Simultaneously, chronic inflammation can predispose to cancer development, progression and metastasis. Proinflammatory molecules are involved in the recruitment of specific cell populations in the tumor microenvironment. These cell populations include myeloid‑derived suppressor cells (MDSCs), a heterogeneous, immature myeloid population with immunosuppressive abilities. Obesity‑associated MDSCs have been linked with tumor dissemination, progression and poor clinical outcomes. A comprehensive literature review was conducted to assess the impact of obesity‑associated MDSCs on cancer in both preclinical models and oncological patients with obesity. A secondary objective was to examine the key role that leptin, the most important proinflammatory mediator released by adipocytes, plays in MDSC‑driven immunosuppression Finally, an overview is provided of the different therapeutic approaches available to target MDSCs in the context of obesity‑related cancer.

1. Introduction

Obesity is a metabolic disease caused by the overaccumulation of subcutaneous or abdominal adipose tissue characterized by chronic low-grade inflammation (1). This status ultimately leads to the development of diseases such as cancer, cardiovascular disease, type 2 diabetes, hypertension, dyslipidemia and reproductive disorders (2-5).

Adipocytes release proinflammatory mediators such as tumor necrosis factor (TNF)-α, interferon (IFN), interleukin (IL)-1β and leptin, (6), which also play a key role in tumor dissemination and metastasis (7,8). Proinflammatory molecules are involved in the recruitment of suppressive cells, including myeloid-derived suppressor cells (MDSCs), in the tumor microenvironment (TME). MDSCs are a heterogeneous population of immature myeloid cells (IMCs) with potent immunosuppressive effects in cancer (9).

According to the literature, obesity and cancer create similar chronic inflammatory environments. Indeed, evidence has been provided of the impact of obesity-associated MDSCs on cancer progression. However, the mechanisms by which this occurs have remained to be fully elucidated. This paper addresses the relevant role that MDSCs play in the immune system. There is evidence supporting the protumorigenic role of MDSCs in obese murine models and patients. Some authors have described the mechanisms of action of obesity-associated MDSCs and suggest different therapeutic approaches targeting MDSCs in obesity-related cancer. The protective role of obesity-associated MDSCs in maintaining immune homeostasis is also briefly mentioned in this paper.

2. MDSCs

MDSCs are IMCs developed during myelopoiesis by a variety of cytokines, such as granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte colony-stimulating factor (G-CSF), IL-17 and TNF-α (10). In normal conditions, hematopoietic stem cells differentiate into common myeloid progenitors, which, in turn, differentiate into neutrophils, monocytes, dendritic cells (DCs) or macrophages. However, myelopoiesis is disrupted in pathological settings, thereby leading to abnormal development of mature myeloid cells. This leads to the accumulation of IMCs at specific sites, depending on the disease (11).

Two main MDSC subpopulations have been described in the literature, namely monocytic MDSCs (M-MDSCs) and granulocytic MDSCs (G-MDSCs). Both cell subsets were first identified in lymphoma BW-Sp3- and chicken ovalbumin-transfected EL-4 thymoma EG7-bearing mice as cell subfractions with different morphological, molecular and functional characteristics. MDSCs have been documented to resemble inflammatory monocytes (M-MDSCs) and immature neutrophil-like or low-density polymorphonuclear cells (G-MDSCs) (12). Characterization standards for MDSCs suggest defining mouse MDSCs as granulocytic marker 1 (Gr1)+CD11b+ cells expressing Ly6ChighLy6G- (M-MDSCs) or Ly6ClowLy6G+ (G-MDSCs), whereas human MDSCs are CD11b+human leukocyte antigen (HLA)-DRlow/- cells that express CD14+CD15- (M-MDSCs) or CD14-CD15+ (G-MDSCs), although G-MDSCs may also express CD66b (13). Other markers such as CD45 and CD33 have also been used to define human MDSCs (14,15). Of note, there is another MDSC subset known as 'early stage MDSCs', defined as immature Lin-(CD3, CD14, CD15, CD19, CD56) HLA-DR-CD33+ MDSCs (13).

The main role of MDSCs is to suppress immune responses, particularly T-cell responses, and it has been extensively described in a variety of settings (16-19). Of note, M-MDSCs mainly express nitric oxide synthase 2, whereas G-MDSCs express high levels of arginase type 1 (ARG1) (20,21). This means that each MDSC subset may use different mechanisms and effector molecules to inhibit immune responses. MDSC activity in cancer is as follows (Fig. 1):

MDSCs promote both the loss of the T-cell receptor (TCR)ζ-chain and cell cycle arrest in T cells. MDSCs exert this effect by taking up L-arginine and L-cysteine, which are essential amino acids for T-cell proliferation and expansion;

MDSCs release reactive oxygen species (ROS), such as hydrogen peroxide and peroxynitrite, to promote the loss of the TCRζ-chain;

MDSCs downregulate T-cell migration to draining lymph nodes and induce apoptosis via disintegrin and metalloproteinase domain-containing protein 17/CD62L interaction and galectin 9/T-cell immunoglobulin and mucin domain-containing protein 3 interactions;

MDSCs induce the development of M2 macrophages by releasing ARG1, IL-10 and transforming growth factor (TGF)-β;

MDSCs induce regulatory T cells (Tregs) by releasing ARG1, IL-10 and TFG-β and after CD40/CD40L and major histocompatibility complex class II/TCR interaction;

MDSCs prevent DC migration by downregulating DC maturation and antigen uptake via IL-10 and nitric oxide;

MDSCs inhibit the cytotoxic capacity of natural killer (NK) cells via TGF-β or indoleamine 2,3-dioxygenase;

MDSCs promote angiogenesis via vascular endothelial growth factor, which, in turn, promotes Treg and MDSC proliferation.

3. Obesity-associated MDSCs in preclinical tumor models

As shown in Table I, there is a growing body of evidence of the effects of MDSCs that link obesity and cancer. It has been demonstrated that the monocyte chemoattractant protein-1 [best known as C-C motif chemokine ligand 2 (CCL2)] is secreted by adipocytes to induce the migration of myeloid cells into tissues (22). In addition, CCL2 governs the migration of MDSCs into the TME after binding C-C motif chemokine receptor 2 (CCR2) (23). It was reported that CCL2 promotes breast cancer (BC) by stimulating aromatase gene expression in mammary adipose tissue and activating the signaling pathways involved in cytochrome P450 19A1 transcription (24,25). In this sense, CCL2 has been associated with high concentrations of visceral adipose tissue, MDSC recruitment and cancer progression in both renal adenocarcinoma-bearing mice (26) and a murine model of triple-negative BC with high concentrations of adipose tissue (27). CCR2-expressing MDSCs have also been documented to be highly infiltrated in mice with obesity-related renal tumors (28,29).

Table I

Impact of obesity-associated MDSCs in preclinical tumor models.

Table I

Impact of obesity-associated MDSCs in preclinical tumor models.

Type of cancerCountryMurine modelEffects on MDSCs(Refs.)
BreastUSABALB/c mice with 4T1 tumorsCCL2, which recruits MDSCs into the TME, was secreted by tumor-associated adipocytes, facilitating tumor progression. MDSC levels were higher in cancer-bearing mice, but a plasmid DNA encoding CCL2 trap reduced MDSCs within the TME.(27)
BreastUSAC57BL/6 mice with E0771 tumorsFas ligand-expressing G-MDSCs were promoted in obese cancer-bearing mice via the CXCL1/CXCR2 axis.(35)
BreastUSABALB/c and C57BL/6 mice with 4T1 tumorsMDSCs increased tumor progression in mice on an HFD compared to those on an LFD. Obesity and MDSC levels were associated with shorter overall survival and a significant elevated number of metastatic cells in the liver.(78)
BreastUSAC57BL/6J mice with E0771-luc tumorsObesity induced by an HFD increased tumor progression and MDSC levels in mice compared to those fed an LFD. Anti-PD1 inhibitor achieved both tumor regression and reduction of MDSCs.(118)
OvaryChinaC57BL/6J, BALB/c and OB/OB mice with ID8-Luc tumorsObesity increased the proportion of circulating MDSCs in obese mice compared to normal-weight mice.(33)
Obesity also promoted the expression of S100A8 and S100A9 by upregulating IL-6, promoting an increase of MDSCs, as well as tumor evasion and metastasis.
PancreaticUSAC57BL/6 and BALB/c mice with Panc.02 tumorsAdiposity was positively correlated with MDSCs and tumor growth. MDSCs inhibited effector T-cell responses, increased tumor growth and decreased survival in obese tumor-bearing mice.(34)
ProstateUSAC57BL/6 mice with RM1 tumorsG-MDSCs were decreased by two-fold in tumors upon CXCL1 silencing in both lean and obese mice.(37)
RenalUSABALB/c mice with Renca tumorsObesity not only induced early MDSC accumulation and intratumoral proinflammatory mediators, but also reduced the percentage of responder mice to immunotherapy.(26)
Specifically, IL-1β positively correlated with tumor size and tumor-infiltrating MDSCs.
RenalUSABALB/c mice with Renca tumorsMDSC levels were higher in tumor-bearing mice treated with an HFD compared to their lean counterparts.(28)
After treatment, MDSC levels frequently persisted, probably because MDSCs were developed during obesity, independently of the tumor status.
RenalUSABALB/c mice with Renca tumorsTumor-bearing mice fed an HFD had more tumor CCR2+ MDSCs than those fed an LFD.(29)
In obese mice, tumors had high levels of CCL2, but arginase levels remained unchanged.

[i] LFD, low-fat diet; HFD, high-fat diet; TME, tumor microenvironment; G-MDSC, granulocytic myeloid-derived suppressor cell; CCL, C-C motif chemokine ligand; CCR, C-C motif chemokine receptor; PD-1, programmed cell death 1.

Similarly, the macrophage inflammatory protein-1 γ (best known as CCL9) is upregulated in murine models with obesity (30) and is considered another important agent in the recruitment of MDSCs in the TME (31). CCL9 may induce CD11b+Gr1+ MDSC expansion after binding CCR1 (particularly in G-MDSCs from spleen and bone marrow) in oral squamous cell carcinoma (OSCC)-bearing mice after treatment with a high-fat diet (HFD). This diet boosted the immunosuppressive effects of MDSCs via intracellular fatty acid uptake (32).

In, thymus-expressed chemokine (best known as CCL25), together with GM-CSF, S100A8 and S100A9 (both upregulated by IL5 and IL-6), were indicated to be involved in MDSC recruitment and expansion in peripheral blood, the bone marrow and tumor tissue from obese ovarian cancer-bearing mice (33). The key role of IL-6 has also been demonstrated in a pancreatic cancer mouse model with HFD-induced obesity. The HFD led to G-MDSC and M-MDSC expansion, tumor growth and proliferation, and failure of T-cell responses (34).

MDSCs also accumulated in the TME of E0771 and Py8119 BC-bearing mice using an HFD via the growth-regulated α protein [C-X-C motif chemokine ligand 1 (CXCL1)]/CXCR2 signaling pathway. This axis was associated with the body mass index (BMI) and poor survival (35). Adipose tissue from obese patients with prostate cancer also mobilized IL-8 [known as CXCL8 and an important chemoattractant for MDSCs (36)] to recruit adipose stromal cells into the TME and induce tumor growth and progression. Of note, CXCL1 silencing in vivo promoted CD11b+Gr1+ MDSC depletion in obese and lean mice (37).

4. Obesity-associated MDSCs in cancer patients

It has been estimated that excess adipose tissue increases the risk of human cancer by 20%. The underlying mechanisms that link cancer to obesity need to be completely understood for adequate treatments to be developed (38). For these reasons, most of the studies focus on the long-term effectiveness of treatments (39) instead of the impact of immunosuppressive cells on obesity-related cancer. Only two studies analyzed MDSC-like cell populations in obese cancer patients (Table II).

Table II

Impact of obesity-associated MDSCs in cancer patients.

Table II

Impact of obesity-associated MDSCs in cancer patients.

Type ofcancerCountryGroupsResults(Refs.)
KidneyUSAGroup 1: Obese patients with kidney cancer;TANs from Group 1 were phenotypically and metabolically different from neutrophils of Group 2.(43)
Group 2: Healthy donorsTANs resembled MDSCs due to the high expression of specific markers, such as CD45, CD15 and arginase-1, and low expression of other markers, such as HLA-DR.
Obesity and adipose tissues were significantly associated with TANs, arginase 1 and disease progression.
OSCCChinaGroup 1: Obese patients with OSCC;Expression of CD33, a marker of human MDSCs, was significantly correlated with local adipocyte levels in Group 1.(32)
Group 2: Non-obese patients with OSCCBoth CD33 expression and high adipocyte levels were associated with poor overall survival and progression-free survival in Group 1.

[i] TAN, tumor-associated neutrophil; OSCC, oral squamous cell carcinoma; HLA, human leukocyte antigen; MDSC, myeloid-derived suppressor cell.

Peng et al (32) demonstrated that CD33+ cell expression in obese patients with OSCC was significantly correlated with local adipocyte levels and negatively associated with overall survival and progression-free survival. CD33 is a surface cell marker that has been widely used to identify MDSCs in obesity (40) and in different types of cancer (9,41,42). However, CD33 may not be appropriate to differentiate MDSCs from other myeloid cells such as DCs or macrophages in tissues (13). However, Peng et al (32) demonstrated that CD33+ cells were increased in cancer patients with obesity, similar to CD11b+Gr1+ MDSCs in a cancer murine model treated with an HFD.

In patients with renal cancer with obesity, Margaroli et al (43) found that blood immature-like neutrophils acquired an immunosuppressive phenotype (resembling MDSCs) upon migration to the tumor site. Of note, immature-like neutrophils, which were highly expressed in neutrophil elastase and programmed cell death-ligand 1 (PD-L1), along with ARG1 levels, were significantly associated with adipose tissue distribution and disease progression. In line with these results, MDSCs have been proven to express high levels of PD-L1 (44). In addition, arginase has been reported to induce MDSCs in obesity and obesity-related diseases (45), including cancer (46). Also, neutrophil elastase, which facilitates tumor-cell intravasation and metastasis (47), can be released by CD33+ MDSCs in human cancer (48).

Of note, obesity is an independent risk factor for the development of non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH)-related hepatocellular carcinomas (HCC) (49). MDSCs, together with T cells and tumor-associated macrophages, are highly infiltrated in patients with NAFLD-/NASH-related HCC (50). Certain mechanisms have been suggested to explain the activation and recruitment of MDSCs in these diseases. For instance, it has been shown that the release of IL-13 could activate MDSCs to promote tumor progression (51). MDSCs expressing PD-L1 and inducible T-cell co-stimulator have been suggested to interact with exhausted programmed cell death 1 (PD-1)+CD8+ T cells to induce immunosuppression in these patients (50). The m6A reader protein YTH N6-methyladenosine RNA binding protein F1 promoted the signaling axis between the enhancer of zeste homolog 2 protein and IL-6 to recruit MDSCs; as a result, CD8 T-mediated immune responses were inhibited (52). In a mouse model, it has been demonstrated that CD11b+ Ly6CIntLy6G+ MDSCs, but not M-MDSCs, can be recruited into the liver TME by hepatic cyclin-dependent kinase 20 (also known as CCRK) signaling through mTORC1-dependent G-CSF expression (53). In this case, CD11b+ Ly6CIntLy6G+ MDSCs positively correlated with tumor weight. Also, the administration of anti-Ly6G antibody significantly suppressed CCRK-induced MDSCs and reduced liver-related HCC tumorigenicity (53).

5. Mechanism of action of MDSCs in obesity-related cancer

Based on experimental evidence from obese cancer murine models and obese patients with cancer, the milieu produced by chronic inflammation during obesity is likely to increase MDSC recruitment and expansion. This environment promotes cancer development and increases cancer risk, as shown in Fig. 2.

Chronic inflammation occurs when the immune system is under permanent, non-resolving stimulation, which can lead to the development of several health conditions, including obesity (54). During this prolonged process, leukocytes and plasma proteins are continually recruited into affected tissues to promote vascularization and phagocytosis via proinflammatory signaling (55,56). Chronic inflammation also induces lymphocyte and macrophage recruitment (57-59).

Adipose tissue has been traditionally known for its capacity to store energy, but it is currently considered a bona fide immune organ due to its interactions with the immune system (60). Obesity may increase the risk of different conditions and diseases such as cancer, cardiovascular disease, type 2 diabetes, hypertension, dyslipidemia and reproductive disorders (2-5). These effects are induced by the release of a variety of proinflammatory mediators, including TNF-α, IFN, IL-1β, IL-6, leptin, resistin, CCL2, CXCL5 and prostaglandin E2, during adipocyte hypertrophy and hyperplasia [extensively reviewed in (61,62)]. Similarly, chronic inflammation can predispose to cancer development, tumor cell dissemination and metastasis (7,8) by promoting mutagenic DNA damage via ROS or reactive nitrogen species production (63). The resulting epigenetic alterations, such as DNA methylation or microRNA dysregulation (64), increase oxidative stress and mediate molecular pathways, including NF-κB or signal transducer and activator of transcription (STAT)3 signaling (65). These epigenetic alterations also induce the release of proinflammatory cytokines (e.g., TNF-α, IL-1β, IL-6 and IL-17) (66).

As mentioned above, cancer and obesity both create similar chronic inflammatory environments. In a cancer-related environment, MDSCs are recruited (67), thus leading to tumor growth, resistance to cancer therapies and poor outcomes (11,68,69). In obese patients, MDSCs are recruited (70) partly through the migration of inhibitory factor-related protein (MRP)-8 (best known as S100A8) and MRP-14 (S100A9), which regulate MDSC-mediated immunosuppression (40,71). Of note, lipid and fatty acid metabolisms have been associated with the modulation of MDSC activity (72). Adeshakin et al (73) reported that fatty acid transport protein 2 (FATP2) confers a protumorigenic environment. This effect is exerted by FATP2 by regulating MDSC and ROS activity and inducing the accumulation of lipids via the upregulation of arachidonic acid metabolism (73). Furthermore, the proto-oncogene serine/threonine-protein kinase Pim-1 has been reported to strongly correlate with fatty acid oxidation, and its targeting resulted in MDSC depletion from the TME (74).

6. Obesity-associated MDSCs as a protective factor in immune homeostasis

Although low levels of circulating MDSCs are found in healthy individuals, these cells play a major role (see section on MDSCs) in maintaining immune homeostasis. Thus, MDSCs limit the hyperactivation of pro-inflammatory cells and reduce tissue damage (75). In obesity conditions, maintaining immune homeostasis is crucial to avoid or prevent adipose tissue dysfunction (76). Obesity is associated with metabolic disorders such as diabetes, which is characterized by high glucose levels resulting from the inability of insulin to suppress hepatic glucose production (77). The role of MDSCs in these settings is scarcely known. To date, only a couple of studies have demonstrated the protective role of MDSCs in obesity-driven metabolic disorders. Xia et al (70) demonstrated that Gr-1+CD11b+ MDSCs are highly expressed in the peripheral tissue of obese mice. This high expression protected mice from inflammation and improved insulin sensitivity and glucose tolerance (70). The authors suggested that MDSCs from obese mice induced not only the apoptosis of cytotoxic CD8+ T cells, but also the polarization of M1 macrophages into their M2 phenotype, thereby exerting insulin-sensitizing effects (70). Similarly, Clements et al (78) reported that MDSCs from obese mice reduced inflammation and had protective effects against metabolic disorders. In parallel, MDSCs increased fat accumulation and the risk of tumor progression and metastasis (78).

7. Obesity-associated MDSCs and cancer: The potential role of leptin

Leptin is a non-glycosylated hormone consisting of 167 amino acids that has been described in leptin-deficient (ob/ob) and leptin receptor (LEPR)-deficient (db/db) mice (79,80) as the product of the 'obese gene' (81). Leptin has pleiotropic effects, and it is not only expressed in adipose tissue, which is considered the main source of leptin, but also in a variety of tissues such as the brain, kidney, liver and skeletal muscle (82). Leptin can also bind different LEPRs [extensively reviewed in (83)] to transduce activation signals into cells through the Janus kinase 2/STAT3, MAPK/ERK1/2 and/or PI3K/AKT signaling pathways (84).

In normal conditions, leptin expression is regulated to restore physiological functions and homeostasis. This hormone has a critical role during chronic inflammation (85) in different settings, such as obesity (86), fertility (87) and pregnancy (88), and when interacting with immune cells via proinflammatory mediators (89,90). Closely related to the role of obesity in cancer, leptin has also been suggested to be involved in all stages of tumorigenesis. Specifically, leptin binds LEPRs to induce a variety of processes, including systemic inflammation, promoting cancer stem cell phenotypes, epithelial-mesenchymal transition, antiapoptotic proteins, hypoxia and angiogenic factors. As a result, leptin enhances cancer cell survival, proliferation and migration, and inhibits T-cell responses (91-96). Hence, leptin correlates with cancer risk and poor survival rates in numerous types of cancer, including breast (97), esophageal (98) and colorectal cancer (99).

Leptin levels have been documented to be increased in cancer-bearing mice treated with an HFD. Similar conclusions have been drawn from cancer patients with obesity (26,34). Specifically, Koprivčić et al (100) demonstrated that women with malignant breast tumors exhibited significantly higher leptin levels (22.24±22.58 ng/ml), as compared to those with benign tumors (11.21±9.46 ng/ml). In addition, patients with triple-negative breast tumors (the most aggressive type of BC) showed higher leptin concentrations (36.11±17.95 ng/ml) than patients with other breast tumor subtypes (100). Lower concentrations of leptin have also been reported in invasive BC and breast carcinoma in situ (10.4±7.0 and 8.7±5.3 ng/ml, respectively), particularly in postmenopausal women. Of note, leptin concentrations were still higher in these patients, as compared to healthy donors (8.4±5.3 ng/ml) (101). Another study comparing leptin levels in patients with prostate cancer and a healthy cohort revealed serum leptin levels of 14.18 and 1.63 ng/ml, respectively (102). Of note, Tong et al (103) did not find any relationship between serum leptin levels and lung cancer; however, the authors found a significant association between leptin expression in tissue and lung cancer. In the same way, LEPR has been reported to be overexpressed in cancer tissue, as compared with normal mucosa (104).

In line with the previous results, leptin has been suggested to be involved in the induction and accumulation of MDSCs (78). Apart from circulating pro-inflammatory cytokines, adipose tissue also releases other pro-inflammatory mediators such as leptin. This adipokine, in turn, may partially elevate the concentration of those cytokines. Therefore, leptin is involved in a feedback loop that may induce the inflammatory milieu typically found in a variety of cancers (105,106). The pro-inflammatory mediators elevated by leptin are also inducers of both, MDSC differentiation and accumulation in the TME (107-109).

8. Therapeutic approaches to target MDSCs in obesity-related cancers

Based on previous studies, a high BMI increases the risk of cancer (110), as demonstrated in endometrial cancer (from 2.5- to 7.1-fold), esophageal adenocarcinoma (4.8-fold), colon, gastric, liver, gallbladder, kidney and pancreatic cancer (from 1.5- to 1.8-fold) or multiple myeloma (from 1.2- to 1.5-fold) (111). However, there are no specific recommendations for the management of obese patients with cancer because obesity may i) influence treatment selection; ii) increase treatment-related toxicity; and iii) promote surgical complications, including infections and perioperative mortality. A decade ago, guidelines recommended estimating chemotherapy doses in obese patients based on their body weight (112). Doses of immunotherapy and targeted therapies have also recently been recommended to be calculated as a function of patients' body weight (113). In this regard, MDSC-targeted therapies may be a promising approach to improve clinical response in obese patients with cancer. The main strategies for MDSC targeting include: i) MDSC depletion; ii) inhibition of MDSC-mediated immunosuppression; iii) blocking MDSC recruitment; and iv) promoting the differentiation of MDSCs into granulocytes, monocytes and dendritic cells (114).

MDSC depletion in mice is commonly induced using antibodies against Gr1, which is located on the surface of murine MDSCs (13). In cancer-bearing mice with diet-induced obesity, different anti-Gr1 therapies have been shown to reduce MDSC expression in the TME (32,34,35,78). Chemotherapeutic agents have also been used to deplete MDSCs (115); however, according to the American Society of Clinical Oncology guidelines, evidence remains limited regarding the toxicity and efficacy of chemotherapy in obese cancer patients (113). This is probably due to the dysregulation of pharmacokinetics and metabolism (116). Consequently, there are no studies reporting the effects of chemotherapy on MDSCs in obese patients with cancer.

The inhibition of MDSC-mediated immunosuppression and the blocking of MDSC recruitment are the most widely used strategies in cancer immunotherapy. In this strategy, the immune checkpoint PD-1/anti-PD-L1 axis is blocked in one of the treatment groups. Anti-PD-1 treatments have shown promising survival rates in obese patients with renal cell carcinoma (26); therefore, it could be a therapeutic approach to reduce MDSC concentrations and improve T-cell responses (117,118). Also, the IL-6 inhibitor LMT28 has been demonstrated to reduce ovarian tumor growth in murine models of obesity and may also be a potential therapy for obese patients with cancer (33). IL-6 is a crucial regulator of MDSC activity and a target for cancer immunotherapy (108). Chemokine-targeted therapies are also useful to deplete the TME of MDSCs. In this sense, CCR2+ MDSCs have been found in high proportion in renal cancer murine models treated with an HFD (29). CCR2 binds CCL2, which is secreted by adipose tissue to promote MDSC recruitment. These findings suggest that the CCR2/CCL2 signaling pathway may be a pivotal target for MDSC depletion (27). Similarly, anti-CXCR1 and anti-CXCR2 have been successfully used in patients with prostate cancer with obesity due to it having an involvement in the CXCR1/CXCL1 and CXCR2/CXCL8 axis; however, levels of MDSCs were not measured in this study (37). By contrast, SX-682, a CXCR1 and CXCR2 inhibitor, has been demonstrated to abrogate tumor trafficking of MDSCs in head and neck cancer murine models (119); thus, SX-682 may be another potential therapeutic approach to inhibit MDSC-mediated immunosuppression.

MDSC differentiation into mature non-suppressive cells has also been tested. For instance, cytosine-phosphate-guanosine oligodeoxynucleotides (CPG-ODNs) can stimulate plasmacytoid dendritic cells to produce IFN-α and, in turn, promote MDSC maturation in vitro (120). CPG-ODNs have been proven to improve the efficacy of adenovirus 5 carrying the TNF-related apoptosis-inducing ligand (Ad5-TRAIL/CpG) in obesity-related renal cell carcinoma (28). In this sense, TRAIL was demonstrated to potently induce tumor-cell apoptosis and the activation of tumor-specific cytotoxic T cells (121); furthermore, the addition of Ad5-TRAIL/CpG prolonged TRAIL production (122). Of note, TRAIL receptors can be considered potential targets to selectively inhibit MDSCs without them affecting mature myeloid or lymphoid cells (123,124).

Other therapeutic approaches to induce MDSC differentiation into mature cells in cancer patients include STAT3 inhibition (125) and the use of all-trans retinoic acid (ATRA) (126) or vitamin D3 (127). Of these, only STAT3 inhibitors have been successfully used in cancer mouse models of obesity (128,129). However, the impact of this therapy on MDSCs remains to be evaluated. ATRA therapy may be optimal to prevent obesity-related metabolic syndrome (130), although its role in obesity-related cancer has not yet been tested. Conversely, the clinical applications of vitamin D in obese patients remain controversial because of the confounding factors and limitations reported in numerous studies (131). Hence, the use of vitamin D still cannot be recommended for obese patients with cancer, despite the promising results achieved in the depletion of tumor MDSCs.

9. Conclusions and future perspectives

Based on the evidence provided by preclinical and clinical studies, obesity may be potentially associated with carcinogenesis. The chronic inflammatory environment promoted by obesity leads to MDSC recruitment, which has a key role in cancer development and progression.

MDSC depletion in obesity-related tumors has been successfully evaluated and associated with improved survival rates and lower tumor progression. However, most of the results have been obtained in murine models. It is worth mentioning that Gr1 inhibitors can only be tested in murine models, since Gr1 is a typical marker of mouse MDSCs. Therefore, its translation into clinical practice must be further investigated. Specific treatments for oncological patients may include anti-PD-1, anti-IL-6, anti-CCL2, anti-CXCR1, anti-CXCR2 and Ad5-TRAIL/CpG. However, there are a variety of MDSC-targeted options that still need to be tested, such as ATRA, vitamin D or STAT3 inhibitors, anti-CCR5, anti-VEGFR and chemotherapeutic agents (114).

Special emphasis has been placed on the role of obesity as a protective factor in tumorigenesis (the so-called 'obesity paradox') (132,133), which has been documented in a variety of tumors, including lymphomas (134,135); acute myeloid leukemia (136); hepatocellular cancer (137); colorectal cancer (138,139); renal cell carcinoma (140-143); lung cancer (144-146); or melanoma (147-149). In this sense, certain studies have demonstrated the inhibitory effects of leptin in vitro (150,151). Evidence has also been provided of an inverse relationship with cancer risk and a positive association with improved outcomes (151,152), which has been termed the 'leptin paradox' (96).

The reasons for the positive association between high amounts of adipose tissue and improved response to cancer immunotherapies remain unknown (153). These phenomena could be explained by the fact that BMI estimation does not only consider adipose tissue but also protective muscle mass; consequently, the BMI should be dismissed as a tool for measuring adiposity (154). Finding optimal parameters may be difficult, since there are many variables that hinder the accurate measurement of adiposity, such as sex, ethnicity, age and pathophysiological group (155).

Cancer immunosuppression driven by obesity-associated MDSCs seems clear, but further research is needed, particularly in cancer patients. There are still various gaps of knowledge, including i) the mechanisms involved in the relationship between obesity, cancer and MDSCs; ii) the mechanisms by which obesity or leptin exert protective effects in certain types of cancer and the role of MDSCs in these settings; and iii) the efficacy and safety of other MDSC-targeted therapies.

Availability of data and materials

Not applicable.

Authors' contributions

CJC, CGG, FSJ, TVG, RFC, APP, CG, MLSL, DJGD, LHP and NPC contributed to the literature search. CJC and CGG wrote the manuscript draft. LdlCM and VSM were involved in the conceptualization of the study. All authors contributed to the revision of the manuscript. All authors have 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 interest

The authors declare that they have no competing interests.

Abbreviations:

Ad5-TRAIL/CpG

adenovirus 5-TNF-related apoptosis inducing ligand/cytosine-phosphate-guanosine

ARG1

arginase type 1

BC

breast cancer

BMI

body mass index

CCL

C-C motif chemokine ligand

CXCL

C-X-C chemokine ligand

DC

dendritic cell

G-CSF

granulocyte colony-stimulating factor

G-MDSC

granulocytic myeloid-derived suppressor cell

GM-CSF

granulocyte-macrophage colony stimulating factor

HFD

high-fat diet

IDO

indoleamine 2,3-dyoxygenase

IFN

interferon

IL

interleukin

IMC

immature myeloid cell

LEPR

leptin receptor

LFD

low-fat diet

M-MDSC

monocytic MDSC

MHC

major histocompatibility complex

MRP

migration inhibitory factor-related protein

RNS

reactive nitrogen species

OSCC

oral squamous cell carcinoma

ROS

reactive oxygen species

STAT

signal transducer and activator of transcription

TCR

T-cell receptor

TGF

transforming growth factor

TME

tumor microenvironment

TNF

tumor necrosis factor

Treg

T-regulatory cell

VEGF

vascular endothelial growth factor

Acknowledgements

The authors acknowledge Mrs María Palazón-Carrión (Oncology Service, Virgen Macarena University Hospital, School of Medicine, University of Seville, Seville, Spain) for project administration work.

Funding

The study was supported by PAIDI Fonds to Resarch Groups (CTS-151) Junta de Andalucía (Andalucía, Spain).

References

1 

Lee H, Lee IS and Choue R: Obesity, inflammation and diet. Pediatr Gastroenterol Hepatol Nutr. 16:143–152. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Ben-Shmuel S, Rostoker R, Scheinman EJ and LeRoith D: Metabolic Syndrome, type 2 diabetes, and cancer: Epidemiology and potential mechanisms. Handb Exp Pharmacol. 233:355–372. 2016. View Article : Google Scholar

3 

Jiang SZ, Lu W, Zong XF, Ruan HY and Liu Y: Obesity and hypertension. Exp Ther Med. 12:2395–2399. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Klop B, Elte JW and Cabezas MC: Dyslipidemia in obesity: Mechanisms and potential targets. Nutrients. 5:1218–1240. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Broughton DE and Moley KH: Obesity and female infertility: Potential mediators of obesity's impact. Fertil Steril. 107:840–847. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Sanchez-Pino MD, Gilmore LA, Ochoa AC and Brown JC: Obesity-Associated myeloid immunosuppressive cells, key players in cancer risk and response to immunotherapy. Obesity (Silver Spring). 29:944–953. 2021. View Article : Google Scholar : PubMed/NCBI

7 

Munn LL: Cancer and inflammation. Wiley Interdiscip Rev Syst Biol Med. 9: View Article : Google Scholar : 2017.

8 

Greten FR and Grivennikov SI: Inflammation and Cancer: Triggers, mechanisms, and consequences. Immunity. 51:27–41. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Jimenez-Cortegana C, Palazon-Carrion N, Martin Garcia-Sancho A, Nogales-Fer nandez E, Carnicero-Gonzalez F, Rios-Herranz E, de la Cruz-Vicente F, Rodríguez-García G, Fernández-Álvarez R, Rueda Dominguez A, et al: Circulating myeloid-derived suppressor cells and regulatory T cells as immunological biomarkers in refractory/relapsed diffuse large B-cell lymphoma: Translational results from the R2-GDP-GOTEL trial. J Immunother Cancer. 9:e0023232021. View Article : Google Scholar : PubMed/NCBI

10 

Karin N: The development and homing of myeloid-derived suppressor cells: From a two-stage model to a multistep narrative. Front Immunol. 11:5575862020. View Article : Google Scholar : PubMed/NCBI

11 

Law AMK, Valdes-Mora F and Gallego-Ortega D: Myeloid-Derived suppressor cells as a therapeutic target for cancer. Cells. 9:5612020. View Article : Google Scholar : PubMed/NCBI

12 

Movahedi K, Guilliams M, Van den Bossche J, Van den Bergh R, Gysemans C, Beschin A, De Baetselier P and Van Ginderachter JA: Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cell-suppressive activity. Blood. 111:4233–4244. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Bronte V, Brandau S, Chen SH, Colombo MP, Frey AB, Greten TF, Mandruzzato S, Murray PJ, Ochoa A, Ostrand-Rosenberg S, et al: Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun. 7:121502016. View Article : Google Scholar : PubMed/NCBI

14 

Jimenez-Cortegana C, Liro J, Palazon-Carrion N, Salamanca E, Sojo-Dorado J, de la Cruz-Merino L, Pascual Á, Rodríguez-Baño J and Sánchez-Margalet V: Increased blood monocytic myeloid derived suppressor cells but low regulatory T lymphocytes in patients with mild COVID-19. Viral Immunol. 34:639–645. 2021. View Article : Google Scholar : PubMed/NCBI

15 

Jimenez-Cortegana C, Sanchez - Martinez P M, Palazon-Carrion N, Nogales-Fernandez E, Henao-Carrasco F, Martin Garcia-Sancho A, Rueda A, Provencio M, de la Cruz-Merino L and Sánchez-Margalet V: Lower survival and increased circulating suppressor cells in patients with relapsed/refractory diffuse large B-Cell lymphoma with deficit of vitamin D Levels Using R-GDP Plus Lenalidomide (R2-GDP): Results from the R2-GDP-GOTEL Trial. Cancers (Basel). 13:46222021. View Article : Google Scholar : PubMed/NCBI

16 

Farshidpour M, Ahmed M, Junna S and Merchant JL: Myeloid-derived suppressor cells in gastrointestinal cancers: A systemic review. World J Gastrointest Oncol. 13:1–11. 2021. View Article : Google Scholar : PubMed/NCBI

17 

O'Connor MA, Rastad JL and Green WR: The role of myeloid-derived suppressor cells in viral infection. Viral Immunol. 30:82–97. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Yan L, Liang M, Yang T, Ji J, Jose Kumar Sreena GS, Hou X, Cao M and Feng Z: The immunoregulatory role of myeloid-derived suppressor cells in the pathogenesis of Rheumatoid arthritis. Front Immunol. 11:5683622020. View Article : Google Scholar : PubMed/NCBI

19 

Yang Z, Guo J, Weng L, Tang W, Jin S and Ma W: Myeloid-derived suppressor cells-new and exciting players in lung cancer. J Hematol Oncol. 13:102020. View Article : Google Scholar : PubMed/NCBI

20 

Youn JI, Nagaraj S, Collazo M and Gabrilovich DI: Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J Immunol. 181:5791–5802. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Talmadge JE and Gabrilovich DI: History of myeloid-derived suppressor cells. Nat Rev Cancer. 13:739–752. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Arner E, Mejhert N, Kulyte A, Balwierz PJ, Pachkov M, Cormont M, Lorente-Cebrián S, Ehrlund A, Laurencikiene J, Hedén P, et al: Adipose tissue microRNAs as regulators of CCL2 production in human obesity. Diabetes. 61:1986–1993. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Oo MW, Kawai H, Takabatake K, Tomida S, Eguchi T, Ono K, Shan Q, Ohara T, Yoshida S, Omori H, et al: Resident stroma-secreted chemokine CCL2 governs myeloid-derived suppressor cells in the tumor microenvironment. JCI Insight. 7:e1489602022. View Article : Google Scholar :

24 

Martinez-Chacon G, Yatkin E, Polari L, Deniz Dinc D, Peuhu E, Hartiala P, Saarinen N and Mäkelä S: CC chemokine ligand 2 (CCL2) stimulates aromatase gene expression in mammary adipose tissue. FASEB J. 35:e215362021. View Article : Google Scholar : PubMed/NCBI

25 

Friesenhengst A, Pribitzer-Winner T, Miedl H, Prostling K and Schreiber M: Elevated aromatase (CYP19A1) expression is associated with a poor survival of patients with estrogen receptor positive breast cancer. Horm Cancer. 9:128–138. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Boi SK, Orlandella RM, Gibson JT, Turbitt WJ, Wald G, Thomas L, Buchta Rosean C, Norris KE, Bing M, Bertrand L, et al: Obesity diminishes response to PD-1-based immunotherapies in renal cancer. J Immunother Cancer. 8:e0007252020. View Article : Google Scholar :

27 

Liu Y, Tiruthani K, Wang M, Zhou X, Qiu N, Xiong Y, Pecot CV, Liu R and Huang L: Tumor-targeted gene therapy with lipid nanoparticles inhibits tumor-associated adipocytes and remodels the immunosuppressive tumor microenvironment in triple-negative breast cancer. Nanoscale Horiz. 6:319–329. 2021. View Article : Google Scholar : PubMed/NCBI

28 

James BR, Anderson KG, Brincks EL, Kucaba TA, Norian LA, Masopust D and Griffith TS: CpG-mediated modulation of MDSC contributes to the efficacy of Ad5-TRAIL therapy against renal cell carcinoma. Cancer Immunol Immunother. 63:1213–1227. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Hale M, Itani F, Buchta CM, Wald G, Bing M and Norian LA: Obesity triggers enhanced MDSC accumulation in murine renal tumors via elevated local production of CCL2. PLoS One. 10:e01187842015. View Article : Google Scholar : PubMed/NCBI

30 

Jiao P, Chen Q, Shah S, Du J, Tao B, Tzameli I, Yan W and Xu H: Obesity-related upregulation of monocyte chemotactic factors in adipocytes: Involvement of nuclear factor-kappaB and c-Jun NH2-terminal kinase pathways. Diabetes. 58:104–115. 2009. View Article : Google Scholar :

31 

Li B, Zhang S, Huang N, Chen H, Wang P, Yang J and Li Z: CCL9/CCR1 induces myeloidderived suppressor cell recruitment to the spleen in a murine H22 orthotopic hepatoma model. Oncol Rep. 41:608–618. 2019.

32 

Peng J, Hu Q, Chen X, Wang C, Zhang J, Ren X, Wang Y, Tao X, Li H, Song M, et al: Diet-induced obesity accelerates oral carcinogenesis by recruitment and functional enhancement of myeloid-derived suppressor cells. Cell Death Dis. 12:9462021. View Article : Google Scholar : PubMed/NCBI

33 

Yang Q, Yu B, Kang J, Li A and Sun J: Obesity promotes tumor immune evasion in ovarian cancer through increased production of myeloid-derived suppressor cells via IL-6. Cancer Manag Res. 13:7355–7363. 2021. View Article : Google Scholar : PubMed/NCBI

34 

Turbitt WJ, Collins SD, Meng H and Rogers CJ: Increased adiposity enhances the accumulation of MDSCs in the tumor microenvironment and adipose tissue of pancreatic tumor-bearing mice and in immune organs of tumor-free hosts. Nutrients. 11:30122019. View Article : Google Scholar : PubMed/NCBI

35 

Gibson JT, Orlandella RM, Turbitt WJ, Behring M, Manne U, Sorge RE and Norian LA: Obesity-Associated myeloid-derived suppressor cells promote apoptosis of tumor-infiltrating CD8 T cells and immunotherapy resistance in breast cancer. Front Immunol. 11:5907942020. View Article : Google Scholar : PubMed/NCBI

36 

Alfaro C, Teijeira A, Onate C, Perez G, Sanmamed MF, Andueza MP, Alignani D, Labiano S, Azpilikueta A, Rodriguez-Paulete A, et al: Tumor-Produced interleukin-8 attracts human myeloid-derived suppressor cells and elicits extrusion of neutrophil extracellular traps (NETs). Clin Cancer Res. 22:3924–3936. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Zhang T, Tseng C, Zhang Y, Sirin O, Corn PG, Li-Ning-Tapia EM, Troncoso P, Davis J, Pettaway C, Ward J, et al: CXCL1 mediates obesity-associated adipose stromal cell trafficking and function in the tumour microenvironment. Nat Commun. 7:116742016. View Article : Google Scholar : PubMed/NCBI

38 

De Pergola G and Silvestris F: Obesity as a major risk factor for cancer. J Obes. 2013:2915462013. View Article : Google Scholar : PubMed/NCBI

39 

Ross KH, Gogineni K, Subhedar PD, Lin JY and McCullough LE: Obesity and cancer treatment efficacy: Existing challenges and opportunities. Cancer. 125:1588–1592. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Bao Y, Mo J, Ruan L and Li G: Increased monocytic CD14(+) HLADRlow/-myeloid-derived suppressor cells in obesity. Mol Med Rep. 11:2322–2328. 2015. View Article : Google Scholar

41 

Rudolph BM, Loquai C, Gerwe A, Bacher N, Steinbrink K, Grabbe S and Tuettenberg A: Increased frequencies of CD11b(+) CD33(+) CD14(+) HLA-DR(low) myeloid-derived suppressor cells are an early event in melanoma patients. Exp Dermatol. 23:202–204. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Verschoor CP, Johnstone J, Millar J, Dorrington MG, Habibagahi M, Lelic A, Loeb M, Bramson JL and Bowdish DM: Blood CD33(+)HLA-DR(-) myeloid-derived suppressor cells are increased with age and a history of cancer. J Leukoc Biol. 93:633–637. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Margaroli C, Cardenas MA, Jansen CS, Moon Reyes A, Hosseinzadeh F, Hong G, Zhang Y, Kissick H, Tirouvanziam R and Master VA: The immunosuppressive phenotype of tumor-infiltrating neutrophils is associated with obesity in kidney cancer patients. Oncoimmunology. 9:17477312020. View Article : Google Scholar : PubMed/NCBI

44 

Noman MZ, Desantis G, Janji B, Hasmim M, Karray S, Dessen P, Bronte V and Chouaib S: PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med. 211:781–790. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Hafida S, Mirshahi T and Nikolajczyk BS: The impact of bariatric surgery on inflammation: Quenching the fire of obesity? Curr Opin Endocrinol Diabetes Obes. 23:373–378. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Grzywa TM, Sosnowska A, Matryba P, Rydzynska Z, Jasinski M, Nowis D and Golab J: Myeloid cell-derived arginase in cancer immune response. Front Immunol. 11:9382020. View Article : Google Scholar : PubMed/NCBI

47 

Deryugina E, Carre A, Ardi V, Muramatsu T, Schmidt J, Pham C and Quigley JP: Neutrophil elastase facilitates tumor cell intravasation and early metastatic events. iScience. 23:1017992020. View Article : Google Scholar : PubMed/NCBI

48 

Lerman I, Garcia-Hernandez ML, Rangel-Moreno J, Chiriboga L, Pan C, Nastiuk KL, Krolewski JJ, Sen A and Hammes SR: Infiltrating myeloid cells exert protumorigenic actions via neutrophil elastase. Mol Cancer Res. 15:1138–1152. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Saitta C, Pollicino T and Raimondo G: Obesity and liver cancer. Ann Hepatol. 18:810–815. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Li M, Wang L, Cong L, Wong CC, Zhang X, Chen H, Zeng T, Li B, Jia X, Huo J, et al: Spatial proteomics of immune microenvironment in nonalcoholic steatohepatitis-associated hepatocellular carcinoma. Hepatology. 79:560–574. 2024. View Article : Google Scholar

51 

Ponziani FR, Bhoori S, Castelli C, Putignani L, Rivoltini L, Del Chierico F, Sanguinetti M, Morelli D, Paroni Sterbini F, Petito V, et al: Hepatocellular carcinoma is associated with gut microbiota profile and inflammation in nonalcoholic fatty liver disease. Hepatology. 69:107–120. 2019. View Article : Google Scholar

52 

Wang L, Zhu L, Liang C, Huang X, Liu Z, Huo J, Zhang Y, Zhang Y, Chen L, Xu H, et al: Targeting N6-methyladenosine reader YTHDF1 with siRNA boosts antitumor immunity in NASH-HCC by inhibiting EZH2-IL-6 axis. J Hepatol. 79:1185–1200. 2023. View Article : Google Scholar : PubMed/NCBI

53 

Sun H, Yang W, Tian Y, Zeng X, Zhou J, Mok MTS, Tang W, Feng Y, Xu L, Chan AWH, et al: An inflammatory-CCRK circuitry drives mTORC1-dependent metabolic and immunosuppressive reprogramming in obesity-associated hepatocellular carcinoma. Nat Commun. 9:52142018. View Article : Google Scholar : PubMed/NCBI

54 

Furman D, Campisi J, Verdin E, Carrera-Bastos P, Targ S, Franceschi C, Ferrucci L, Gilroy DW, Fasano A, Miller GW, et al: Chronic inflammation in the etiology of disease across the life span. Nat Med. 25:1822–1832. 2019. View Article : Google Scholar : PubMed/NCBI

55 

Muller WA: Getting leukocytes to the site of inflammation. Vet Pathol. 50:7–22. 2013. View Article : Google Scholar : PubMed/NCBI

56 

Klevebro S, Bjorkander S, Ekstrom S, Merid SK, Gruzieva O, Malarstig A, Johansson Å, Kull I, Bergström A and Melén E: Inflammation-related plasma protein levels and association with adiposity measurements in young adults. Sci Rep. 11:113912021. View Article : Google Scholar : PubMed/NCBI

57 

Ellulu MS, Patimah I, Khaza'ai H, Rahmat A and Abed Y: Obesity and inflammation: The linking mechanism and the complications. Arch Med Sci. 13:851–863. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Sakai Y and Kobayashi M: Lymphocyte 'homing' and chronic inflammation. Pathol Int. 65:344–354. 2015. View Article : Google Scholar : PubMed/NCBI

59 

Ingersoll MA, Platt AM, Potteaux S and Randolph GJ: Monocyte trafficking in acute and chronic inflammation. Trends Immunol. 32:470–477. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Wensveen FM, Valentic S, Sestan M, Wensveen TT and Polic B: Interactions between adipose tissue and the immune system in health and malnutrition. Semin Immunol. 27:322–333. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Kumar DP, Koka S, Li C and Rajagopal S: Inflammatory mediators in obesity. Mediators Inflamm. 2019:94818192019. View Article : Google Scholar : PubMed/NCBI

62 

Howe LR, Subbaramaiah K, Hudis CA and Dannenberg AJ: Molecular pathways: Adipose inflammation as a mediator of obesity-associated cancer. Clin Cancer Res. 19:6074–6083. 2013. View Article : Google Scholar : PubMed/NCBI

63 

Kawanishi S, Ohnishi S, Ma N, Hiraku Y and Murata M: Crosstalk between DNA damage and inflammation in the multiple steps of carcinogenesis. Int J Mol Sci. 18:18082017. View Article : Google Scholar : PubMed/NCBI

64 

Murata M: Inflammation and cancer. Environ Health Prev Med. 23:502018. View Article : Google Scholar : PubMed/NCBI

65 

Fan Y, Mao R and Yang J: NF-ĸB and STAT3 signaling pathways collaboratively link inflammation to cancer. Protein Cell. 4:176–185. 2013. View Article : Google Scholar : PubMed/NCBI

66 

Del Prete A, Allavena P, Santoro G, Fumarulo R, Corsi MM and Mantovani A: Molecular pathways in cancer-related inflammation. Biochem Med (Zagreb). 21:264–275. 2011. View Article : Google Scholar : PubMed/NCBI

67 

Yang Y, Li C, Liu T, Dai X and Bazhin AV: Myeloid-Derived suppressor cells in tumors: From mechanisms to antigen specificity and microenvironmental regulation. Front Immunol. 11:13712020. View Article : Google Scholar : PubMed/NCBI

68 

Ma P, Beatty PL, McKolanis J, Brand R, Schoen RE and Finn OJ: Circulating myeloid derived suppressor cells (MDSC) that accumulate in premalignancy share phenotypic and functional characteristics with MDSC in cancer. Front Immunol. 10:14012019. View Article : Google Scholar : PubMed/NCBI

69 

Veglia F, Sanseviero E and Gabrilovich DI: Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 21:485–498. 2021. View Article : Google Scholar : PubMed/NCBI

70 

Xia S, Sha H, Yang L, Ji Y, Ostrand-Rosenberg S and Qi L: Gr-1+ CD11b+ myeloid-derived suppressor cells suppress inflammation and promote insulin sensitivity in obesity. J Biol Chem. 286:23591–23599. 2011. View Article : Google Scholar : PubMed/NCBI

71 

Srikrishna G: S100A8 and S100A9: New insights into their roles in malignancy. J Innate Immun. 4:31–40. 2012. View Article : Google Scholar :

72 

Siddiqui S and Glauben R: Fatty acid metabolism in myeloid-derived suppressor cells and tumor-associated macrophages: Key factor in cancer immune evasion. Cancers (Basel). 14:2502022. View Article : Google Scholar : PubMed/NCBI

73 

Adeshakin AO, Liu W, Adeshakin FO, Afolabi LO, Zhang M, Zhang G, Wang L, Li Z, Lin L, Cao Q, et al: Regulation of ROS in myeloid-derived suppressor cells through targeting fatty acid transport protein 2 enhanced anti-PD-L1 tumor immunotherapy. Cell Immunol. 362:1042862021. View Article : Google Scholar : PubMed/NCBI

74 

Xin G, Chen Y, Topchyan P, Kasmani MY, Burns R, Volberding PJ, Wu X, Cohn A, Chen Y, Lin CW, et al: Targeting PIM1-Mediated metabolism in myeloid suppressor cells to treat cancer. Cancer Immunol Res. 9:454–469. 2021. View Article : Google Scholar : PubMed/NCBI

75 

Sanchez-Pino MD, Dean MJ and Ochoa AC: Myeloid-derived suppressor cells (MDSC): When good intentions go awry. Cell Immunol. 362:1043022021. View Article : Google Scholar : PubMed/NCBI

76 

Zhen Y, Shu W, Hou X and Wang Y: Innate immune system orchestrates metabolic homeostasis and dysfunction in visceral adipose tissue during obesity. Front Immunol. 12:7028352021. View Article : Google Scholar : PubMed/NCBI

77 

Klein S, Gastaldelli A, Yki-Jarvinen H and Scherer PE: Why does obesity cause diabetes? Cell Metab. 34:11–20. 2022. View Article : Google Scholar : PubMed/NCBI

78 

Clements VK, Long T, Long R, Figley C, Smith DMC and Ostrand-Rosenberg S: Frontline Science: High fat diet and leptin promote tumor progression by inducing myeloid-derived suppressor cells. J Leukoc Biol. 103:395–407. 2018. View Article : Google Scholar : PubMed/NCBI

79 

Ingalls AM, Dickie MM and Snell GD: Obese, a new mutation in the house mouse. J Hered. 41:317–318. 1950. View Article : Google Scholar : PubMed/NCBI

80 

Hummel KP, Dickie MM and Coleman DL: Diabetes, a new mutation in the mouse. Science. 153:1127–1278. 1966. View Article : Google Scholar : PubMed/NCBI

81 

Zhang Y, Proenca R, Maffei M, Barone M, Leopold L and Friedman JM: Positional cloning of the mouse obese gene and its human homologue. Nature. 372:425–432. 1994. View Article : Google Scholar : PubMed/NCBI

82 

Munzberg H and Heymsfield SB: New insights into the regulation of leptin gene expression. Cell Metab. 29:1013–1014. 2019. View Article : Google Scholar : PubMed/NCBI

83 

Gorska E, Popko K, Stelmaszczyk-Emmel A, Ciepiela O, Kucharska A and Wasik M: Leptin receptors. Eur J Med Res. 15(Suppl 2): S50–S54. 2010. View Article : Google Scholar

84 

Park HK and Ahima RS: Leptin signaling. F1000Prime Rep. 6:732014. View Article : Google Scholar : PubMed/NCBI

85 

Perez-Perez A, Sanchez-Jimenez F, Vilarino-Garcia T and Sanchez-Margalet V: Role of leptin in inflammation and vice versa. Int J Mol Sci. 21:58872020. View Article : Google Scholar : PubMed/NCBI

86 

Obradovic M, Sudar-Milovanovic E, Soskic S, Essack M, Arya S, Stewart AJ, Gojobori T and Isenovic ER: Leptin and Obesity: Role and Clinical Implication. Front Endocrinol (Lausanne). 12:5858872021. View Article : Google Scholar : PubMed/NCBI

87 

Vilarino-Garcia T, Perez-Perez A, Santamaria-Lopez E, Prados N, Fernandez-Sanchez M and Sanchez-Margalet V: Sam68 mediates leptin signaling and action in human granulosa cells: Possible role in leptin resistance in PCOS. Endocr Connect. 9:479–488. 2020. View Article : Google Scholar : PubMed/NCBI

88 

Perez-Perez A, Toro A, Vilarino-Garcia T, Maymo J, Guadix P, Duenas JL, Fernández-Sánchez M, Varone C and Sánchez-Margalet V: Leptin action in normal and pathological pregnancies. J Cell Mol Med. 22:716–727. 2018. View Article : Google Scholar :

89 

Fernandez-Riejos P, Najib S, Santos-Alvarez J, Martin-Romero C, Perez-Perez A, Gonzalez-Yanes C and Sánchez-Margalet V: Role of leptin in the activation of immune cells. Mediators Inflamm. 2010:5683432010. View Article : Google Scholar : PubMed/NCBI

90 

Perez-Perez A, Vilarino-Garcia T, Fernandez-Riejos P, Martin-Gonzalez J, Segura-Egea JJ and Sanchez-Margalet V: Role of leptin as a link between metabolism and the immune system. Cytokine Growth Factor Rev. 35:71–84. 2017. View Article : Google Scholar : PubMed/NCBI

91 

Dutta D, Ghosh S, Pandit K, Mukhopadhyay P and Chowdhury S: Leptin and cancer: Pathogenesis and modulation. Indian J Endocrinol Metab. 16(Suppl 3): S596–S600. 2012. View Article : Google Scholar

92 

Ando S and Catalano S: The multifactorial role of leptin in driving the breast cancer microenvironment. Nat Rev Endocrinol. 8:263–275. 2011. View Article : Google Scholar : PubMed/NCBI

93 

Feldman DE, Chen C, Punj V, Tsukamoto H and Machida K: Pluripotency factor-mediated expression of the leptin receptor (OB-R) links obesity to oncogenesis through tumor-initiating stem cells. Proc Natl Acad Sci USA. 109:829–834. 2012. View Article : Google Scholar :

94 

Ghasemi A, Saeidi J, Azimi-Nejad M and Hashemy SI: Leptin-induced signaling pathways in cancer cell migration and invasion. Cell Oncol (Dordr). 42:243–260. 2019. View Article : Google Scholar : PubMed/NCBI

95 

Ray A and Cleary MP: The potential role of leptin in tumor invasion and metastasis. Cytokine Growth Factor Rev. 38:80–97. 2017. View Article : Google Scholar : PubMed/NCBI

96 

Jimenez-Cortegana C, Lopez-Saavedra A, Sanchez-Jimenez F, Perez-Perez A, Castineiras J, Virizuela-Echaburu JA, de la Cruz-Merino L and Sánchez-Margalet V: Leptin, both bad and good actor in cancer. Biomolecules. 11:9132021. View Article : Google Scholar : PubMed/NCBI

97 

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

98 

Greer KB, Falk GW, Bednarchik B, Li L and Chak A: Associations of serum adiponectin and leptin with barrett's esophagus. Clin Gastroenterol Hepatol. 13:2265–2272. 2015. View Article : Google Scholar : PubMed/NCBI

99 

Li C, Quan J, Wei R, Zhao Z, Guan X, Liu Z, Zou S, Wang X and Jiang Z: Leptin overexpression as a poor prognostic factor for colorectal cancer. Biomed Res Int. 2020:75325142020.PubMed/NCBI

100 

Koprivčić I, Marjanovic K, Matic A, Tolusic Levak M, Lovric I, Pauzar B, Erić I and Wertheimer V: Serum leptin level in breast cancer. Acta Clin Croat. 61:79–85. 2022.

101 

Wu MH, Chou YC, Chou WY, Hsu GC, Chu CH, Yu CP, Yu JC and Sun CA: Circulating levels of leptin, adiposity and breast cancer risk. Br J Cancer. 100:578–582. 2009. View Article : Google Scholar : PubMed/NCBI

102 

Singh SK, Grifson JJ, Mavuduru RS, Agarwal MM, Mandal AK and Jha V: Serum leptin: A marker of prostate cancer irrespective of obesity. Cancer Biomark. 7:11–15. 2010. View Article : Google Scholar : PubMed/NCBI

103 

Tong X, Ma Y, Zhou Q, He J, Peng B, Liu S, Yan Z, Yang X and Fan H: Serum and tissue leptin in lung cancer: A meta-analysis. Oncotarget. 8:19699–19711. 2017. View Article : Google Scholar : PubMed/NCBI

104 

Chludzinska-Kasperuk S, Lewko J, Sierzantowicz R, Krajewska-Kulak E and Reszec-Gielazyn J: The effect of serum leptin concentration and leptin receptor expression on colorectal cancer. Int J Environ Res Public Health. 20:49512023. View Article : Google Scholar : PubMed/NCBI

105 

Inacio Pinto N, Carnier J, Oyama LM, Otoch JP, Alcantara PS, Tokeshi F and Nascimento CM: Cancer as a proinflammatory environment: Metastasis and cachexia. Mediators Inflamm. 2015:7910602015. View Article : Google Scholar : PubMed/NCBI

106 

Ostrand-Rosenberg S: Myeloid derived-suppressor cells: Their role in cancer and obesity. Curr Opin Immunol. 51:68–75. 2018. View Article : Google Scholar : PubMed/NCBI

107 

Zhao X, Rong L, Zhao X, Li X, Liu X, Deng J, Wu H, Xu X, Erben U, Wu P, et al: TNF signaling drives myeloid-derived suppressor cell accumulation. J Clin Invest. 122:4094–4104. 2012. View Article : Google Scholar : PubMed/NCBI

108 

Weber R, Groth C, Lasser S, Arkhypov I, Petrova V, Altevogt P, Utikal J and Umansky V: IL-6 as a major regulator of MDSC activity and possible target for cancer immunotherapy. Cell Immunol. 359:1042542021. View Article : Google Scholar

109 

Elkabets M, Ribeiro VS, Dinarello CA, Ostrand-Rosenberg S, Di Santo P, Apte RN and Vosshenrich CA: IL-1β regulates a novel myeloid-derived suppressor cell subset that impairs NK cell development and function. Eur J Immunol. 40:3347–3357. 2010. View Article : Google Scholar : PubMed/NCBI

110 

Avgerinos KI, Spyrou N, Mantzoros CS and Dalamaga M: Obesity and cancer risk: Emerging biological mechanisms and perspectives. Metabolism. 92:121–135. 2019. View Article : Google Scholar

111 

Lauby-Secretan B, Scoccianti C, Loomis D, Grosse Y, Bianchini F and Straif K; International Agency for Research on Cancer Handbook Working Group: Body Fatness and Cancer-Viewpoint of the IARC Working Group. N Engl J Med. 375:794–798. 2016. View Article : Google Scholar : PubMed/NCBI

112 

Griggs JJ, Mangu PB, Anderson H, Balaban EP, Dignam JJ, Hryniuk WM, Morrison VA, Pini TM, Runowicz CD, Rosner GL, et al: Appropriate chemotherapy dosing for obese adult patients with cancer: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 30:1553–1561. 2012. View Article : Google Scholar : PubMed/NCBI

113 

Griggs JJ, Bohlke K, Balaban EP, Dignam JJ, Hall ET, Harvey RD, Hecht DP, Klute KA, Morrison VA, Pini TM, et al: Appropriate systemic therapy dosing for obese adult patients with cancer: ASCO Guideline Update. J Clin Oncol. 39:2037–2048. 2021. View Article : Google Scholar : PubMed/NCBI

114 

De Cicco P, Ercolano G and Ianaro A: The new Era of cancer immunotherapy: Targeting myeloid-derived suppressor cells to overcome immune evasion. Front Immunol. 11:16802020. View Article : Google Scholar : PubMed/NCBI

115 

Wang Y, Jia A, Bi Y, Wang Y, Yang Q, Cao Y, Li Y and Liu G: Targeting myeloid-derived suppressor cells in cancer immunotherapy. Cancers (Basel). 12:26262020. View Article : Google Scholar : PubMed/NCBI

116 

Horowitz NS and Wright AA: Impact of obesity on chemotherapy management and outcomes in women with gynecologic malignancies. Gynecol Oncol. 138:201–206. 2015. View Article : Google Scholar : PubMed/NCBI

117 

Li X, Zhong J, Deng X, Guo X, Lu Y, Lin J, Huang X and Wang C: Targeting myeloid-derived suppressor cells to enhance the antitumor efficacy of immune checkpoint blockade therapy. Front Immunol. 12:7541962021. View Article : Google Scholar

118 

Pingili AK, Chaib M, Sipe LM, Miller EJ, Teng B, Sharma R, Asemota S, Al Abdallah Q, Mims TS, Marion TN, et al: Immune checkpoint blockade reprograms systemic immune landscape and tumor microenvironment in obesity-associated breast cancer. Cell Rep. 35:1092852021. View Article : Google Scholar : PubMed/NCBI

119 

Greene S, Robbins Y, Mydlarz WK, Huynh AP, Schmitt NC, Friedman J, Horn LA, Palena C, Schlom J, Maeda DY, et al: Inhibition of MDSC Trafficking with SX-682, a CXCR1/2 Inhibitor, Enhances NK-cell immunotherapy in head and neck cancer models. Clin Cancer Res. 26:1420–1431. 2020. View Article : Google Scholar

120 

Zoglmeier C, Bauer H, Noerenberg D, Wedekind G, Bittner P, Sandholzer N, Rapp M, Anz D, Endres S and Bourquin C: CpG blocks immunosuppression by myeloid-derived suppressor cells in tumor-bearing mice. Clin Cancer Res. 17:1765–1775. 2011. View Article : Google Scholar : PubMed/NCBI

121 

VanOosten RL and Griffith TS: Activation of tumor-specific CD8+ T Cells after intratumoral Ad5-TRAIL/CpG oligodeoxynucleotide combination therapy. Cancer Res. 67:11980–11990. 2007. View Article : Google Scholar : PubMed/NCBI

122 

Griffith TS and Broghammer EL: Suppression of tumor growth following intralesional therapy with TRAIL recombinant adenovirus. Mol Ther. 4:257–266. 2001. View Article : Google Scholar : PubMed/NCBI

123 

Condamine T, Kumar V, Ramachandran IR, Youn JI, Celis E, Finnberg N, El-Deiry WS, Winograd R, Vonderheide RH, English NR, et al: ER stress regulates myeloid-derived suppressor cell fate through TRAIL-R-mediated apoptosis. J Clin Invest. 124:2626–2639. 2014. View Article : Google Scholar : PubMed/NCBI

124 

Dominguez GA, Condamine T, Mony S, Hashimoto A, Wang F, Liu Q, Forero A, Bendell J, Witt R, Hockstein N, et al: Selective targeting of myeloid-derived suppressor cells in cancer patients using DS-8273a, an Agonistic TRAIL-R2 Antibody. Clin Cancer Res. 23:2942–2950. 2017. View Article : Google Scholar :

125 

Zou S, Tong Q, Liu B, Huang W, Tian Y and Fu X: Targeting STAT3 in Cancer Immunotherapy. Mol Cancer. 19:1452020. View Article : Google Scholar : PubMed/NCBI

126 

Nefedova Y, Fishman M, Sherman S, Wang X, Beg AA and Gabrilovich DI: Mechanism of all-trans retinoic acid effect on tumor-associated myeloid-derived suppressor cells. Cancer Res. 67:11021–11028. 2007. View Article : Google Scholar : PubMed/NCBI

127 

Chen PT, Hsieh CC, Wu CT, Yen TC, Lin PY, Chen WC and Chen MF: 1α,25-Dihydroxyvitamin D3 inhibits esophageal squamous cell carcinoma progression by Reducing IL6 Signaling. Mol Cancer Ther. 14:1365–1375. 2015. View Article : Google Scholar : PubMed/NCBI

128 

Chang CC, Wu MJ, Yang JY, Camarillo IG and Chang CJ: Leptin-STAT3-G9a signaling promotes obesity-mediated breast cancer progression. Cancer Res. 75:2375–2386. 2015. View Article : Google Scholar : PubMed/NCBI

129 

Park JW, Han CR, Zhao L, Willingham MC and Cheng SY: Inhibition of STAT3 activity delays obesity-induced thyroid carcinogenesis in a mouse model. Endocr Relat Cancer. 23:53–63. 2016. View Article : Google Scholar

130 

Berry DC and Noy N: All-trans-retinoic acid represses obesity and insulin resistance by activating both peroxisome proliferation-activated receptor beta/delta and retinoic acid receptor. Mol Cell Biol. 29:3286–3296. 2009. View Article : Google Scholar : PubMed/NCBI

131 

Karampela I, Sakelliou A, Vallianou N, Christodoulatos GS, Magkos F and Dalamaga M: Vitamin D and Obesity: Current evidence and controversies. Curr Obes Rep. 10:162–180. 2021. View Article : Google Scholar : PubMed/NCBI

132 

Lennon H, Sperrin M, Badrick E and Renehan AG: The obesity paradox in cancer: A review. Curr Oncol Rep. 18:562016. View Article : Google Scholar : PubMed/NCBI

133 

Lee DH and Giovannucci EL: The obesity paradox in cancer: Epidemiologic insights and perspectives. Curr Nutr Rep. 8:175–181. 2019. View Article : Google Scholar : PubMed/NCBI

134 

Weiss L, Melchardt T, Habringer S, Boekstegers A, Hufnagl C, Neureiter D, Hopfinger G, Greil R and Egle A: Increased body mass index is associated with improved overall survival in diffuse large B-cell lymphoma. Ann Oncol. 25:171–176. 2014. View Article : Google Scholar

135 

Stevenson JKR, Qiao Y, Chan KKW, Beca J, Isaranuwatchai W, Guo H, Schwartz D, Arias J, Gavura S, Dai WF, et al: Improved survival in overweight and obese patients with aggressive B-cell lymphoma treated with rituximab-containing chemotherapy for curative intent. Leuk Lymphoma. 60:1399–1408. 2019. View Article : Google Scholar

136 

Brunner AM, Sadrzadeh H, Feng Y, Drapkin BJ, Ballen KK, Attar EC, Amrein PC, McAfee SL, Chen YB, Neuberg DS and Fathi AT: Association between baseline body mass index and overall survival among patients over age 60 with acute myeloid leukemia. Am J Hematol. 88:642–646. 2013. View Article : Google Scholar : PubMed/NCBI

137 

Tsang NM, Pai PC, Chuang CC, Chuang WC, Tseng CK, Chang KP, Yen TC, Lin JD and Chang JT: Overweight and obesity predict better overall survival rates in cancer patients with distant metastases. Cancer Med. 5:665–675. 2016. View Article : Google Scholar : PubMed/NCBI

138 

Schlesinger S, Siegert S, Koch M, Walter J, Heits N, Hinz S, Jacobs G, Hampe J, Schafmayer C and Nöthlings U: Postdiagnosis body mass index and risk of mortality in colorectal cancer survivors: A prospective study and meta-analysis. Cancer Causes Control. 25:1407–1418. 2014. View Article : Google Scholar : PubMed/NCBI

139 

Amptoulach S, Gross G and Kalaitzakis E: Differential impact of obesity and diabetes mellitus on survival after liver resection for colorectal cancer metastases. J Surg Res. 199:378–385. 2015. View Article : Google Scholar : PubMed/NCBI

140 

Parker AS, Lohse CM, Cheville JC, Thiel DD, Leibovich BC and Blute ML: Greater body mass index is associated with better pathologic features and improved outcome among patients treated surgically for clear cell renal cell carcinoma. Urology. 68:741–746. 2006. View Article : Google Scholar : PubMed/NCBI

141 

Waalkes S, Merseburger AS, Kramer MW, Herrmann TR, Wegener G, Rustemeier J, Hofmann R, Schrader M, Kuczyk MA and Schrader AJ: Obesity is associated with improved survival in patients with organ-confined clear-cell kidney cancer. Cancer Causes Control. 21:1905–1910. 2010. View Article : Google Scholar : PubMed/NCBI

142 

Hakimi AA, Furberg H, Zabor EC, Jacobsen A, Schultz N, Ciriello G, Mikklineni N, Fiegoli B, Kim PH, Voss MH, et al: An epidemiologic and genomic investigation into the obesity paradox in renal cell carcinoma. J Natl Cancer Inst. 105:1862–1870. 2013. View Article : Google Scholar : PubMed/NCBI

143 

Albiges L, Hakimi AA, Xie W, McKay RR, Simantov R, Lin X, Lee JL, Rini BI, Srinivas S, Bjarnason GA, et al: Body mass index and metastatic renal cell carcinoma: Clinical and biological correlations. J Clin Oncol. 34:3655–3663. 2016. View Article : Google Scholar : PubMed/NCBI

144 

Lam VK, Bentzen SM, Mohindra P, Nichols EM, Bhooshan N, Vyfhuis M, Scilla KA, Feigenberg SJ, Edelman MJ and Feliciano JL: Obesity is associated with long-term improved survival in definitively treated locally advanced non-small cell lung cancer (NSCLC). Lung Cancer. 104:52–57. 2017. View Article : Google Scholar : PubMed/NCBI

145 

Shepshelovich D, Xu W, Lu L, Fares A, Yang P, Christiani D, Zhang J, Shiraishi K, Ryan BM, Chen C, et al: Body Mass Index (BMI), BMI change, and overall survival in patients with SCLC and NSCLC: A pooled analysis of the International lung cancer consortium. J Thorac Oncol. 14:1594–1607. 2019. View Article : Google Scholar : PubMed/NCBI

146 

Ardesch FH, Ruiter R, Mulder M, Lahousse L, Stricker BHC and Kiefte-de Jong JC: The obesity paradox in lung cancer: Associations with body size versus body shape. Front Oncol. 10:5911102020. View Article : Google Scholar : PubMed/NCBI

147 

Hayes AJ and Larkin J: BMI and outcomes in melanoma: More evidence for the obesity paradox. Lancet Oncol. 19:269–270. 2018. View Article : Google Scholar : PubMed/NCBI

148 

McQuade JL, Daniel CR, Hess KR, Mak C, Wang DY, Rai RR, Park JJ, Haydu LE, Spencer C, Wongchenko M, et al: Association of body-mass index and outcomes in patients with metastatic melanoma treated with targeted therapy, immunotherapy, or chemotherapy: A retrospective, multicohort analysis. Lancet Oncol. 19:310–322. 2018. View Article : Google Scholar : PubMed/NCBI

149 

Smith LK, Arabi S, Lelliott EJ, McArthur GA and Sheppard KE: Obesity and the impact on cutaneous melanoma: Friend or Foe? Cancers (Basel). 12:15832020. View Article : Google Scholar : PubMed/NCBI

150 

Somasundar P, Yu AK, Vona-Davis L and McFadden DW: Differential effects of leptin on cancer in vitro. J Surg Res. 113:50–55. 2013. View Article : Google Scholar

151 

Thompson KJ, Lau KN, Johnson S, Martinie JB, Iannitti DA, McKillop IH and Sindram D: Leptin inhibits hepatocellular carcinoma proliferation via p38-MAPK-dependent signalling. HPB (Oxford). 13:225–233. 2011. View Article : Google Scholar : PubMed/NCBI

152 

Paik SS, Jang SM, Jang KS, Lee KH, Choi D and Jang SJ: Leptin expression correlates with favorable clinicopathologic phenotype and better prognosis in colorectal adenocarcinoma. Ann Surg Oncol. 16:297–303. 2009. View Article : Google Scholar

153 

Murphy WJ and Longo DL: The surprisingly positive association between obesity and cancer immunotherapy efficacy. JAMA. 321:1247–1248. 2019. View Article : Google Scholar : PubMed/NCBI

154 

Cespedes Feliciano EM, Kroenke CH and Caan BJ: The obesity paradox in cancer: How important is muscle? Annu Rev Nutr. 38:357–379. 2018. View Article : Google Scholar : PubMed/NCBI

155 

Donini LM, Pinto A, Giusti AM, Lenzi A and Poggiogalle E: Obesity or BMI Paradox? Beneath the Tip of the Iceberg. Front Nutr. 7:532020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2024
Volume 65 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jiménez‑Cortegana C, Gutiérrez‑García C, Sánchez‑Jiménez F, Vilariño‑García T, Flores‑Campos R, Pérez‑Pérez A, Garnacho C, Sánchez‑León ML, García‑Domínguez DJ, Hontecillas‑Prieto L, Hontecillas‑Prieto L, et al: Impact of obesity‑associated myeloid‑derived suppressor cells on cancer risk and progression (Review). Int J Oncol 65: 79, 2024
APA
Jiménez‑Cortegana, C., Gutiérrez‑García, C., Sánchez‑Jiménez, F., Vilariño‑García, T., Flores‑Campos, R., Pérez‑Pérez, A. ... Sánchez‑Margalet, V. (2024). Impact of obesity‑associated myeloid‑derived suppressor cells on cancer risk and progression (Review). International Journal of Oncology, 65, 79. https://doi.org/10.3892/ijo.2024.5667
MLA
Jiménez‑Cortegana, C., Gutiérrez‑García, C., Sánchez‑Jiménez, F., Vilariño‑García, T., Flores‑Campos, R., Pérez‑Pérez, A., Garnacho, C., Sánchez‑León, M. L., García‑Domínguez, D. J., Hontecillas‑Prieto, L., Palazón‑Carrión, N., De La Cruz‑Merino, L., Sánchez‑Margalet, V."Impact of obesity‑associated myeloid‑derived suppressor cells on cancer risk and progression (Review)". International Journal of Oncology 65.2 (2024): 79.
Chicago
Jiménez‑Cortegana, C., Gutiérrez‑García, C., Sánchez‑Jiménez, F., Vilariño‑García, T., Flores‑Campos, R., Pérez‑Pérez, A., Garnacho, C., Sánchez‑León, M. L., García‑Domínguez, D. J., Hontecillas‑Prieto, L., Palazón‑Carrión, N., De La Cruz‑Merino, L., Sánchez‑Margalet, V."Impact of obesity‑associated myeloid‑derived suppressor cells on cancer risk and progression (Review)". International Journal of Oncology 65, no. 2 (2024): 79. https://doi.org/10.3892/ijo.2024.5667