Letter to the Editor Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Apr 21, 2025; 31(15): 104574
Published online Apr 21, 2025. doi: 10.3748/wjg.v31.i15.104574
Prognostic value of the triglyceride-glucose index in advanced gastric cancer: A call for further exploration
Hong-Jie Meng, Yi Mao, De-Qing Zhao, Sheng-Guang Shi, Department of Gastrointestinal Surgery Ward, Zhuji People’s Hospital, Zhuji 311800, Zhejiang Province, China
ORCID number: Hong-Jie Meng (0009-0003-8794-6893).
Author contributions: Meng HJ conceptualization, writing, reviewing and editing; Mao Y methodology and formal analysis; Zhao DQ and Shi SG supervision and writing of the original draft; Meng HJ, Mao Y, Zhao DQ, and Shi SG participated in drafting the manuscript and have read it, and all authors thoroughly reviewed and endorsed the final manuscript.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Hong-Jie Meng, MD, Associate Chief Physician, Senior Researcher, Department of Gastrointestinal Surgery Ward, Zhuji People’s Hospital, No. 9 Jianmin Road, Taozhu Street, Zhuji 311800, Zhejiang Province, China. mhj7768@163.com
Received: December 25, 2024
Revised: March 9, 2025
Accepted: March 17, 2025
Published online: April 21, 2025
Processing time: 114 Days and 22.5 Hours

Abstract

Gastric cancer (GC) remains a leading cause of cancer-related mortality worldwide, necessitating the identification of reliable prognostic indicators to enhance treatment outcomes. Recent research has highlighted the triglyceride-glucose (TyG) index as a potential surrogate marker for insulin resistance, which may significantly influence the prognosis of patients undergoing immunotherapy combined with chemotherapy. In this context, the study by Yao et al demonstrates that a high TyG index correlates with improved overall survival and progression-free survival in advanced GC patients receiving sintilimab and chemotherapy. Specifically, patients in the high TyG group had a significantly longer median progression-free survival of 9.8 months [95% confidence interval (CI): 9.2-10.9] compared to 8.0 months (95%CI: 7.5-8.5) in the low TyG group (hazard ratio = 0.58, 95%CI: 0.43-0.79, P < 0.001). Similarly, the median overall survival was significantly longer in the high TyG group at 23.1 months (95%CI: 21.2-NA) vs 16.5 months (95%CI: 13.9-18.3) in the low TyG group (hazard ratio = 0.30, 95%CI: 0.21-0.42, P < 0.001). These findings underscore the strong prognostic potential of the TyG index in guiding treatment strategies for advanced GC. These findings underscore the need for further investigation into the TyG index’s role as a prognostic tool and its underlying mechanisms in influencing treatment efficacy. We advocate for additional multicenter studies to validate these results and explore the TyG index’s applicability across diverse patient populations, ultimately aiming to refine treatment strategies and improve patient outcomes in advanced GC.

Key Words: Advanced gastric cancer; Chemotherapy; Insulin resistance; Prognostic marker; Triglyceride-glucose index; Tumor microenvironment

Core Tip: The triglyceride-glucose (TyG) index, a surrogate marker for insulin resistance, has emerged as a potential prognostic factor in advanced gastric cancer. Recent findings suggest that a high TyG index correlates with improved overall survival and progression-free survival in patients undergoing immunotherapy combined with chemotherapy. This highlights the clinical relevance of insulin sensitivity in modulating treatment efficacy. Further multicenter studies are essential to validate its prognostic value across diverse populations and to explore underlying mechanisms. Integrating the TyG index into comprehensive prognostic models may optimize treatment strategies and improve patient outcomes in advanced gastric cancer.



TO THE EDITOR

We are writing to discuss the insightful article by Yao et al[1], recently published. This study presents a significant advancement in our understanding of prognostic factors in advanced gastric cancer (GC), particularly in the context of emerging immunotherapeutic strategies.

SIGNIFICANCE OF THE TYG INDEX

The triglyceride-glucose (TyG) index is calculated using fasting triglyceride and fasting glucose levels, following the formula: TyG = ln [fasting triglycerides (mg/dL) × fasting glucose (mg/dL)/2], serves as a surrogate marker for insulin resistance[2]. Insulin resistance has been implicated in various malignancies, including GC, where it may promote tumorigenesis through several mechanisms such as enhanced cellular proliferation, increased angiogenesis, and immune evasion[3]. The findings from Yao et al[1] indicating that a high TyG index correlates with improved overall survival (OS) and progression-free survival in patients undergoing treatment with sintilimab and chemotherapy are particularly noteworthy. Specifically, their study reported that patients with a high TyG index (≥ 1.79) had a significantly longer median progression-free survival of 9.8 months [95% confidence interval (CI): 9.2-10.9] compared to 8.0 months (95%CI: 7.5-8.5) in the low TyG index group (< 1.79) [hazard ratio (HR) = 0.58, 95%CI: 0.43-0.79, P < 0.001]. The median OS was also significantly extended in the high TyG index group, reaching 23.1 months (95%CI: 21.2-NA) compared to 16.5 months (95%CI: 13.9-18.3) in the low TyG index group (HR = 0.30, 95%CI: 0.21-0.42, P < 0.001). In multivariate analysis, the TyG index was identified as an independent prognostic factor for OS (HR = 0.36, 95%CI: 0.24-0.55, P < 0.001), further reinforcing its predictive value. These results suggest that metabolic status, as reflected by the TyG index, plays a crucial role in shaping treatment outcomes in advanced GC. This relationship suggests that insulin sensitivity may play a role in modulating the efficacy of immunotherapy, potentially influencing treatment outcomes[1,3].

CLINICAL IMPLICATIONS

The TyG index has shown significant prognostic value in advanced GC, with Yao et al[1] reporting a notable survival benefit in patients with a high TyG index (median OS: 23.1 months vs 16.5 months, HR = 0.30, P < 0.001)[2]. These findings suggest that metabolic profiling could serve as an additional stratification tool in clinical decision-making, allowing for more tailored treatment strategies. Integrating the TyG index into prognostic models alongside established biomarkers, such as programmed death-ligand 1 (PD-L1) expression and electrocorticography performance status, may refine patient selection for intensive therapeutic regimens, particularly in combination with immunotherapy[4,5].

Given the mechanistic links between insulin resistance and immunotherapy response, the TyG index may also help predict immune checkpoint inhibitor efficacy. Insulin resistance, through the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway, promotes tumor growth and alters the tumor microenvironment, contributing to immune suppression and metabolic competition. Patients with a high TyG index, indicative of metabolic dysregulation, may benefit from adjunctive metabolic interventions (e.g., metformin or mTOR inhibitors) to enhance their responsiveness to immunotherapy[1]. Additionally, the predictive role of the TyG index in identifying patients at risk of immune exhaustion due to metabolic competition warrants further investigation[6,7].

Future research should explore whether metabolic modulation strategies could synergize with immune checkpoint blockade to improve patient outcomes. For example, patients with a high TyG index could be prioritized for intensive therapeutic regimens or innovative treatment approaches, while those with lower indices might benefit from closer monitoring or alternative therapeutic options. Such stratification could ultimately contribute to enhanced survival outcomes and improved quality of life for affected patients[1,8,9].

MECHANISTIC INSIGHTS

This study paves the way for deeper investigation into the biological mechanisms connecting insulin resistance with cancer progression and treatment response[10,11]. Insulin resistance is known to activate multiple oncogenic signaling pathways, notably the PI3K/Akt/mTOR pathway[12]. This pathway regulates cellular metabolism, proliferation, and survival, and its dysregulation is implicated in cancer progression and therapeutic resistance. Persistent insulin resistance leads to chronic activation of PI3K/Akt signaling, promoting tumor growth and impairing the efficacy of immune checkpoint inhibitors by fostering an immunosuppressive environment[13]. Hyperactivation of mTOR can also reduce T-cell function, limiting the anti-tumor immune response[14,15]. Given the intricate interplay between metabolic dysfunction and GC progression, we summarize the key metabolic factors associated with the TyG index and their respective effects in Table 1.

Table 1 The Role of the triglyceride-glucose index in metabolic dysfunction and gastric cancer progression.
Metabolic factor
Associated changes
Impact on gastric cancer
Insulin resistance↑ PI3K/Akt/mTOR activation, ↑ growth signals, ↓ apoptosisPromotes tumor proliferation and therapy resistance
Lipotoxicity↑ Free fatty acids, ↑ IL-6/TNF-α, ↑ inflammationEnhances pro-tumor immune suppression
Glucotoxicity↑ Oxidative stress, ↑ DNA damage, ↑ chronic inflammationFacilitates tumor progression and metastasis
Altered TME↑ Metabolic competition (glucose depletion), ↓ T-cell functionReduces efficacy of immunotherapy
Hyperlipidemia and obesity↑ Systemic inflammation, ↑ lipid metabolism dysfunctionAlters immune response, may influence prognosis

Additionally, insulin resistance influences the tumor microenvironment through multiple mechanisms. First, it enhances systemic and local inflammation via upregulation of pro-inflammatory cytokines (e.g., interleukin-6, tumor necrosis factor-α), which can create an immunosuppressive niche[10,16,17]. Second, the altered metabolic landscape associated with insulin resistance leads to increased competition for glucose between tumor cells and immune cells, particularly T cells. Since effector T cells rely heavily on glycolysis for energy, metabolic competition in an insulin-resistant state may result in T-cell exhaustion and impaired anti-tumor immunity. Finally, increased circulating insulin levels can stimulate the expression of PD-L1 on tumor and immune cells, potentially affecting the response to programmed death 1/PD-L1 blockade therapies[18,19]. Future research should focus on unraveling these intricate pathways and exploring their interactions with immunotherapeutic agents, such as sintilimab, to enhance our understanding and improve therapeutic outcomes.

NEED FOR MULTICENTER STUDIES

While the study by Yao et al[1] presents compelling evidence, the importance of multicenter studies cannot be overstated[20]. Such investigations are crucial for improving the generalizability of the findings across diverse populations and clinical settings[21]. Differences in genetic backgrounds, lifestyle factors, and comorbidities can profoundly impact both insulin resistance and cancer biology. Consequently, validating the TyG index as a prognostic tool in heterogeneous cohorts will be indispensable for confirming its applicability and reliability in routine clinical practice.

FUTURE DIRECTIONS

Future investigations should aim to integrate the TyG index with other biomarkers and clinical parameters to construct a more comprehensive prognostic model. Such models could include factors such as tumor staging, histological subtypes, and molecular characteristics, offering a more nuanced and holistic perspective on patient prognosis.

CONCLUSION

In conclusion, I commend Yao et al[1] for their valuable contribution to advancing our understanding of prognostic markers in advanced GC. The TyG index emerges as a promising tool for improving patient stratification and optimizing treatment strategies in this challenging clinical domain. Further research into the utility of this biomarker and its potential incorporation into routine clinical practice is essential to enhance outcomes for patients with advanced GC.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade A, Grade B, Grade B, Grade B, Grade B

Novelty: Grade A, Grade B, Grade B, Grade B, Grade B

Creativity or Innovation: Grade A, Grade B, Grade B, Grade B, Grade B

Scientific Significance: Grade A, Grade A, Grade B, Grade B, Grade B

P-Reviewer: Govindarajan KK; Li ZP; Wang SB S-Editor: Bai Y L-Editor: A P-Editor: Zhao S

References
1.  Yao ZY, Ma X, Cui YZ, Liu J, Han ZX, Song J. Impact of triglyceride-glucose index on the long-term prognosis of advanced gastric cancer patients receiving immunotherapy combined with chemotherapy. World J Gastroenterol. 2025;31:102249.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Reference Citation Analysis (8)]
2.  Cai C, Chen C, Lin X, Zhang H, Shi M, Chen X, Chen W, Chen D. An analysis of the relationship of triglyceride glucose index with gastric cancer prognosis: A retrospective study. Cancer Med. 2024;13:e6837.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
3.  Okamura T, Hashimoto Y, Hamaguchi M, Obora A, Kojima T, Fukui M. Triglyceride-glucose index (TyG index) is a predictor of incident colorectal cancer: a population-based longitudinal study. BMC Endocr Disord. 2020;20:113.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in RCA: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
4.  Tao S, Yu L, Li J, Xie Z, Huang L, Yang D, Tan Y, Zhang W, Huang X, Xue T. Prognostic value of triglyceride-glucose index in patients with chronic coronary syndrome undergoing percutaneous coronary intervention. Cardiovasc Diabetol. 2023;22:322.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
5.  Xu J, Xu W, Chen G, Hu Q, Jiang J. Association of TyG index with prehypertension or hypertension: a retrospective study in Japanese normoglycemia subjects. Front Endocrinol (Lausanne). 2023;14:1288693.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
6.  Liu W, Wang Y, Luo J, Liu M, Luo Z. Pleiotropic Effects of Metformin on the Antitumor Efficiency of Immune Checkpoint Inhibitors. Front Immunol. 2020;11:586760.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in RCA: 19]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
7.  Eikawa S, Nishida M, Mizukami S, Yamazaki C, Nakayama E, Udono H. Immune-mediated antitumor effect by type 2 diabetes drug, metformin. Proc Natl Acad Sci U S A. 2015;112:1809-1814.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 339]  [Cited by in RCA: 419]  [Article Influence: 41.9]  [Reference Citation Analysis (0)]
8.  Leone RD, Powell JD. Fueling the Revolution: Targeting Metabolism to Enhance Immunotherapy. Cancer Immunol Res. 2021;9:255-260.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 24]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
9.  Li Y, Tang J, Jiang J, Chen Z. Metabolic checkpoints and novel approaches for immunotherapy against cancer. Int J Cancer. 2022;150:195-207.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
10.  Zhan S, Wang L, Wang W, Li R. Insulin resistance in NSCLC: unraveling the link between development, diagnosis, and treatment. Front Endocrinol (Lausanne). 2024;15:1328960.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
11.  Leitner BP, Siebel S, Akingbesote ND, Zhang X, Perry RJ. Insulin and cancer: a tangled web. Biochem J. 2022;479:583-607.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in RCA: 23]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
12.  Zhang YY, Li YJ, Xue CD, Li S, Gao ZN, Qin KR. Effects of T2DM on cancer progression: pivotal precipitating factors and underlying mechanisms. Front Endocrinol (Lausanne). 2024;15:1396022.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
13.  Nepstad I, Hatfield KJ, Grønningsæter IS, Aasebø E, Hernandez-Valladares M, Hagen KM, Rye KP, Berven FS, Selheim F, Reikvam H, Bruserud Ø. Effects of insulin and pathway inhibitors on the PI3K-Akt-mTOR phosphorylation profile in acute myeloid leukemia cells. Signal Transduct Target Ther. 2019;4:20.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in RCA: 49]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
14.  Rascio F, Spadaccino F, Rocchetti MT, Castellano G, Stallone G, Netti GS, Ranieri E. The Pathogenic Role of PI3K/AKT Pathway in Cancer Onset and Drug Resistance: An Updated Review. Cancers (Basel). 2021;13:3949.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in RCA: 206]  [Article Influence: 51.5]  [Reference Citation Analysis (0)]
15.  He Y, Sun MM, Zhang GG, Yang J, Chen KS, Xu WW, Li B. Targeting PI3K/Akt signal transduction for cancer therapy. Signal Transduct Target Ther. 2021;6:425.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in RCA: 718]  [Article Influence: 179.5]  [Reference Citation Analysis (0)]
16.  Mani N, Andrews D, Obeng RC. Modulation of T cell function and survival by the tumor microenvironment. Front Cell Dev Biol. 2023;11:1191774.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
17.  Liu X, Hoft DF, Peng G. Tumor microenvironment metabolites directing T cell differentiation and function. Trends Immunol. 2022;43:132-147.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in RCA: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
18.  Pellegrino M, Secli V, D'Amico S, Petrilli LL, Caforio M, Folgiero V, Tumino N, Vacca P, Vinci M, Fruci D, de Billy E. Manipulating the tumor immune microenvironment to improve cancer immunotherapy: IGF1R, a promising target. Front Immunol. 2024;15:1356321.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
19.  Nojima I, Eikawa S, Tomonobu N, Hada Y, Kajitani N, Teshigawara S, Miyamoto S, Tone A, Uchida HA, Nakatsuka A, Eguchi J, Shikata K, Udono H, Wada J. Dysfunction of CD8 + PD-1 + T cells in type 2 diabetes caused by the impairment of metabolism-immune axis. Sci Rep. 2020;10:14928.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in RCA: 34]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
20.  Pan K, Nelson RA, Wactawski-Wende J, Lee DJ, Manson JE, Aragaki AK, Mortimer JE, Phillips LS, Rohan T, Ho GYF, Saquib N, Shadyab AH, Nassir R, Rhee JJ, Hurria A, Chlebowski RT. Insulin Resistance and Cancer-Specific and All-Cause Mortality in Postmenopausal Women: The Women's Health Initiative. J Natl Cancer Inst. 2020;112:170-178.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in RCA: 35]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
21.  Seifirad S, Alquran L. The bigger, the better? When multicenter clinical trials and meta-analyses do not work. Curr Med Res Opin. 2021;37:321-326.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]