Next Article in Journal
Surgical Oncology in 2025: Challenges, Innovations, and the Road Ahead for Young Surgical Oncologists
Previous Article in Journal
Focal Therapy for Localized Prostate Cancer: A Case Series with Cost Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Insulin Resistance in Cancer

Jefferson Einstein Montgomery Hospital, 559 West Germantown Pike, East Norriton, PA 19403, USA
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2025, 32(9), 477; https://doi.org/10.3390/curroncol32090477
Submission received: 28 May 2025 / Revised: 13 August 2025 / Accepted: 15 August 2025 / Published: 25 August 2025

Simple Summary

Insulin resistance is a condition, common in obesity and type 2 diabetes, where the body’s cells do not respond properly to insulin, a hormone that controls blood sugar. This review explains how this metabolic problem is a significant risk factor for several major cancers, including those of the breast, liver, and endometrium. The high levels of insulin, chronic inflammation, and altered cellular energy usage associated with insulin resistance create an environment that can fuel the growth and spread of tumors. Since insulin resistance is a modifiable condition, understanding this link opens up new strategies for cancer prevention and treatment, emphasizing the importance of lifestyle interventions and metabolic health.

Abstract

Insulin resistance (IR) is a prevalent metabolic condition characterized by reduced cellular responsiveness to insulin and consequent hyperinsulinemia, and it is a key component of type 2 diabetes and metabolic syndrome. A growing body of evidence suggests IR is a critical accomplice in the pathogenesis of various cancers. This review synthesizes evidence on underlying molecular mechanisms, including the integrated roles of the insulin/IGF system, chronic inflammation, metabolic reprogramming, and mitochondrial dysfunction. This review proposes that metabolic dysregulation should be viewed as a modifiable oncogenic force. This perspective illuminates new pathways for understanding cancer development and offers promising avenues for prevention and therapeutic intervention.

1. Introduction

Insulin resistance (IR) is a pathological condition in which the body’s cells become less responsive to the action of insulin, leading to elevated blood insulin levels. Traditionally associated with type 2 diabetes, obesity, and metabolic syndrome, insulin resistance has gained significant attention for its potential role in cancer development and progression. Insulin, along with insulin-like growth factors (IGFs), is involved in regulating cell growth, metabolism, and survival. Given that cancer is fundamentally a disease of dysregulated cell growth, the potential connection between insulin resistance and cancer has become a focal point of research. This review will examine the evidence supporting the link between insulin resistance and cancer, explore the underlying pathophysiology, and discuss the clinical implications for cancer prevention and treatment.
The relationship between metabolic disorders and cancer has been known for over 100 years and represents a significant public health challenge [1,2]. A meta-analysis examining the relationship between T2DM and cancer risk revealed that diabetes was linked to a 10% higher relative risk (RR) of developing cancer (RR: 110; 95% confidence interval [CI]: 1.04–1.17) [3]. For non-sex-specific cancers, the overall cancer risk is estimated to be 1.15–1.17 times higher in individuals with diabetes relative to the general population [4]. The global burden of T2DM continues to rise at an alarming rate, with projections estimating that over 500 million people will be affected by 2030 [5,6]. While the incidence rates of some major cancers have stabilized or declined in recent years due to public health efforts [7,8,9], the increasing prevalence of metabolic disease presents a significant and ongoing public health concern with potential implications for future cancer rates.

2. Search Strategy

This narrative review is based on a literature search of the PubMed, MEDLINE, and Google Scholar databases for articles published up to July 2025. The search terms included “insulin resistance and cancer,” “hyperinsulinemia,” “IGF system,” “diabetes and cancer,” “obesity and cancer,” “inflammation,” “metformin,” “mitochondrial dysfunction,” and “Warburg effect,” in combination with specific cancer types such as “breast,” “colorectal,” “pancreatic,” “liver,” and “endometrial.” Foundational experimental studies, large epidemiological cohorts, meta-analyses, and clinical trials were prioritized for inclusion to provide a comprehensive overview of the topic.

3. Core Molecular Mechanisms Linking Insulin Resistance and Cancer

The link between IR and cancer is underpinned by several interconnected biological mechanisms that create a systemic environment rich in growth-promoting signals, inflammatory mediators, and metabolic substrates that can initiate and fuel tumor growth. These mechanisms do not operate in isolation but form a vicious cycle where metabolic dysregulation, inflammation, and oncogenic signaling reinforce one another.

3.1. The Insulin/IGF System: An Integrated Signaling Axis

The etiology of insulin resistance reflects a complex interplay between genetic and environmental factors, including a sedentary lifestyle and obesogenic diets [10,11,12]. This state leads to sustained hyperinsulinemia, which promotes carcinogenesis through its effects on the insulin/IGF system. The historical view of separate insulin and IGF systems is now considered obsolete; extensive evidence demonstrates an intimately integrated network [13].
This system’s key components include the insulin receptor (IR) and the IGF-1 receptor (IGF-1R). The IR exists in two isoforms: IR-A and IR-B. While IR-B is the primary metabolic isoform in adult tissues, IR-A is the predominant isoform in fetal tissues and is frequently overexpressed in cancer cells [14,15]. Crucially, IR-A not only binds insulin but also binds the potent mitogen IGF-2 with high affinity, making it a primary physiological receptor for IGF-2 [15,16]. The other IGF receptor, the IGF-2/mannose-6-phosphate receptor (IGF-2R), lacks a kinase domain and functions as a scavenger receptor that degrades IGF-2, thereby acting as a tumor suppressor [17].
In the state of IR-driven hyperinsulinemia,
  • Direct Mitogenic Signaling: Sustained hyperinsulinemia can directly activate the IR and, at high concentrations, the IGF-1R. Upon binding, these receptors activate two major signaling cascades:
    The Phosphatidylinositol 3-kinase (PI3K)/Akt Pathway: This pathway is pivotal, promoting glucose uptake, protein synthesis, and cell growth while strongly inhibiting apoptosis (programmed cell death) [18,19].
    The Ras/Mitogen-Activated Protein Kinase (MAPK) Pathway: This pathway primarily drives cell proliferation, migration, and differentiation, contributing to tumor expansion and metastasis [18].
  • Altered IGF-Axis Signaling: Hyperinsulinemia suppresses the hepatic production of IGF-binding proteins (IGFBP-1 and IGFBP-2), which may increase the bioavailability of IGF-1 [20,21]. However, the direct link between circulating IGF-1 levels and cancer risk is a subject of ongoing scientific debate. Bloodstream levels of IGF-1 have not been consistently reproducible as prognostic markers, and the clinical failure of all IGF-1R drug blockers has called into question the role of the IGF-1/IGF-1R axis as a primary driver of malignancy (as reviewed in [22,23]). A more scientifically supported view is that the potent cellular and tissue growth-promoting signals (e.g., via the PI3K and MAPK pathways) induced by hyperinsulinemia contribute to the benign proliferative stages of tumor development. These signals can create a permissive environment that synergizes with other intra-tumoral events necessary for the switch to a malignant phenotype. To date, no definitive evidence suggests that activation of the insulin or IGF-1 receptor by itself can trigger the “benign versus malignant transformation switch.” In contrast, a more direct role in tumor progression has been demonstrated for IGF-II, which is often secreted by cancer cells and acts as a key autocrine/paracrine factor in the tumor microenvironment, promoting the angiogenic switch and malignant progression [24].

3.2. Chronic Inflammation and Dysregulated Adipokines

Chronic low-grade inflammation, driven largely by obesity, is a hallmark of insulin resistance and plays a pivotal role in cancer development [24,25]. In this state, visceral adipose tissue produces an excess of pro-inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), which are consistently elevated in individuals with IR [26,27]. These cytokines activate inflammatory signaling pathways like nuclear factor kappa B (NF-κB), which promotes cell survival and proliferation [25]. This chronic inflammatory environment fosters carcinogenesis by
  • Generating reactive oxygen species (ROS) that cause DNA damage and genetic instability [28,29].
  • Promoting angiogenesis through factors like VEGF [30].
  • Suppressing anti-tumor immune surveillance [31].
This process is further mediated by adipokines, bioactive molecules secreted from adipose tissue. In obesity, there is a dysregulated secretion of these molecules, characterized by
  • Elevated Leptin: This pro-inflammatory adipokine promotes cancer cell proliferation, angiogenesis, and metastasis, partly by activating the PI3K/Akt and MAPK signaling pathways [32,33].
  • Reduced Adiponectin: This anti-inflammatory adipokine normally exerts anti-tumorigenic effects by activating AMPK, which inhibits cell proliferation and induces apoptosis. Its reduction in obesity removes this protective mechanism [34,35].
The imbalance between high leptin and low adiponectin, coupled with the systemic inflammation from IR, creates a pro-tumorigenic environment that is highly conducive to cancer initiation and progression [36].

3.3. Metabolic Reprogramming, Hypoxia, and Plasticity

Insulin resistance profoundly alters cellular metabolism, creating conditions that support tumorigenesis. Cancer cells exhibit metabolic reprogramming, most famously the Warburg effect, where they preferentially metabolize glucose through aerobic glycolysis rather than more efficient oxidative phosphorylation (OXPHOS). This metabolic shift, while less efficient for ATP production, rapidly generates the biosynthetic precursors (nucleotides, lipids, and amino acids) needed for fast proliferation [18,19]. The hyperglycemia and hyperinsulinemia characteristic of IR provide abundant glucose to fuel this process.
This altered metabolism is intimately linked to hypoxia. The rapid, disorganized growth of tumors often outstrips their blood supply, leading to hypoxic (low-oxygen) regions. Hypoxia activates master regulatory proteins called hypoxia-inducible factors (HIFs), particularly HIF-1α. HIFs reinforce the Warburg effect by upregulating glucose transporters and glycolytic enzymes, and they also promote angiogenesis and metastasis, contributing to a more aggressive cancer phenotype [19,37].
A critical emerging concept is metabolic plasticity. To survive therapeutic stress or nutrient deprivation, cancer cells can adapt their metabolism by shifting to alternative fuel sources. In insulin-resistant states, where circulating free fatty acids are elevated, cancer cells can increase their reliance on fatty acid oxidation. Furthermore, many cancers become dependent on glutamine metabolism to support biosynthesis and maintain redox balance [38]. This adaptability is a key mechanism of treatment resistance. As a result, targeting these metabolic escape routes, for example, with glutaminase inhibitors (e.g., Telaglenastat), has become a promising therapeutic strategy to overcome resistance in metabolically reprogrammed cancers [39,40,41].

3.4. Mitochondrial Dysfunction

Mitochondrial dysfunction is a critical mechanism linking insulin resistance and cancer, serving as both a consequence of IR and a driver of carcinogenesis [40,42]. In the insulin-resistant state, impaired cellular metabolism increases the burden on mitochondria, leading to several pro-tumorigenic effects:
  • Increased Oxidative Stress: Dysfunctional mitochondria generate excessive reactive oxygen species (ROS). This chronic oxidative stress damages mitochondrial and nuclear DNA, contributing to the genomic instability that drives cancer initiation [43,44,45].
  • Impaired Apoptosis: Mitochondria are central to programmed cell death (apoptosis). In cancer, this process is often dysregulated by the upregulation of anti-apoptotic proteins like Bcl-2, which prevents the release of cytochrome c and blocks cell death. This not only promotes tumor survival but is also a key mechanism of resistance to many chemotherapies [46,47,48]. Targeting this pathway with BH3 mimetics (e.g., Venetoclax) is a promising strategy to restore apoptotic sensitivity [49].
  • Pro-inflammatory Signaling: Damaged mitochondria release signals that activate inflammatory pathways, such as the NLRP3 inflammasome, further contributing to the chronic inflammatory state that fuels both IR and cancer progression [50,51].

4. Insulin Resistance and Specific Cancers

Epidemiological studies consistently link IR with an increased risk of several cancers. A summary of the malignancies with the strongest association is provided in Table 1. The relative risk from a large meta-analysis is detailed in Table 2 [52], and the cancers with the strongest link include
  • Endometrial Cancer: This cancer has one of the strongest associations with IR and obesity. Hyperinsulinemia directly promotes endometrial cell proliferation and indirectly increases risk by reducing sex hormone-binding globulin (SHBG), which increases the bioavailability of estrogens [53,54].
  • Liver and Pancreatic Cancers: IR is a pivotal factor in the pathogenesis of non-alcoholic fatty liver disease (NAFLD), which can progress to hepatocellular carcinoma (HCC) [55,56]. Chronic hyperinsulinemia is also a major risk factor for pancreatic cancer, potentially by promoting local inflammation and directly stimulating pancreatic cell proliferation [57,58].
  • Colorectal Cancer: High levels of insulin and IGF-1 are associated with an increased risk of colorectal adenomas and carcinomas by stimulating epithelial cell proliferation and inhibiting apoptosis [59,60].
  • Breast Cancer: Particularly in postmenopausal women, IR is linked to increased risk. The key drivers are hyperinsulinemia and increased IGF-1 bioavailability, activating the PI3K/Akt and MAPK pathways, along with interactions with estrogen signaling [61,62,63].

5. Therapeutic Strategies and Future Outlook

The mounting evidence linking IR to cancer development has opened new avenues for prevention and treatment strategies. Understanding this relationship provides a clear rationale for targeting metabolic health.

5.1. Lifestyle Interventions

Lifestyle modification remains the cornerstone of managing insulin resistance and reducing cancer risk.
  • Diet: Dietary patterns that are low in refined sugars and processed foods and high in fiber have been shown to significantly improve insulin sensitivity and reduce inflammatory markers [64,65]. Healthy fats, such as those found in the Mediterranean diet, also contribute to these benefits [66].
  • Physical Activity: Regular physical activity, including both aerobic and resistance training, improves insulin sensitivity, reduces visceral fat, and lowers systemic inflammation. A minimum of 150 min of moderate-intensity activity per week is associated with a reduced risk for multiple cancer types [67,68,69].
Interventions that combine both dietary changes and physical activity have been shown to produce superior outcomes for metabolic health [70].

5.2. Pharmacological Approaches

Beyond lifestyle, several pharmacological agents that target insulin resistance are under investigation for their role in oncology.
  • Metformin: This first-line drug for T2DM improves insulin sensitivity, primarily by activating AMP-activated protein kinase (AMPK), which in turn inhibits the pro-growth mTOR pathway [71]. By reducing systemic hyperinsulinemia and inflammation, metformin has shown promise in pre-clinical models and some clinical studies. For instance, one trial found that low-dose metformin could suppress the formation of colorectal aberrant crypt foci, suggesting a potential role in chemoprevention [72]. However, the clinical evidence is mixed, highlighting significant limitations. The large, phase III MA.32 trial, which evaluated metformin as an adjuvant therapy in early-stage breast cancer, found no significant improvement in invasive disease-free survival or overall survival compared to placebo [73]. These mixed results underscore the critical need to identify the specific patient populations and cancer types that are most likely to benefit from metformin therapy.
  • Targeting the insulin/IGF system: Given its central role in driving cell proliferation and survival, the insulin/IGF system is an attractive therapeutic target. Strategies have included IGF-1R monoclonal antibodies (e.g., Cixitumumab) and dual IR/IGF-1R tyrosine kinase inhibitors (e.g., Linsitinib) [74,75]. While promising in preclinical studies, these agents have shown only modest efficacy in clinical trials and face significant challenges, including
    Metabolic Side Effects: Cross-reactivity with the insulin receptor’s metabolic functions can lead to hyperglycemia, complicating treatment.
    Resistance: Tumors often develop resistance by activating alternative growth factor pathways, limiting the long-term effectiveness of these inhibitors [76].

5.3. Future Directions and Emerging Research

To overcome these limitations, future research must focus on a more nuanced and personalized approach.
  • Predictive Biomarkers: A key priority is the identification of biomarkers to predict which patients will respond to metabolically targeted therapies. This could include circulating markers of IR (e.g., C-peptide and HOMA-IR) or tumor-specific markers like the expression level of the IR-A isoform. Understanding how a patient’s metabolic state influences their response to immune checkpoint inhibitors is another critical area for future investigation.
  • Metabolic Plasticity as a Resistance Mechanism: Cancer cells can evade therapy by adapting their metabolism, for instance by shifting their reliance from glucose to alternative fuel sources like fatty acids or glutamine [38]. This metabolic plasticity is a key mechanism of treatment resistance. As a result, targeting these metabolic escape routes, for example, with glutaminase inhibitors (e.g., Telaglenastat), is a promising strategy to block this adaptation and overcome resistance [39,40].
  • High-Risk Populations: The IR–cancer link has profound implications for specific high-risk groups that warrant further study. This includes investigating the long-term cancer predisposition of newborns of mothers with gestational diabetes, who are exposed to hyperinsulinemia in utero. Similarly, rare tumors that ectopically secrete IGF-2, inducing severe hypoglycemia, represent a unique clinical model of IGF-driven malignancy and can provide valuable mechanistic insights.

6. Conclusions

Insulin resistance is a significant and modifiable risk factor for the development and progression of several major cancers. Its oncogenic influence is exerted through a complex and integrated network involving the insulin/IGF system, chronic inflammation, and profound metabolic reprogramming. As the global prevalence of metabolic disease continues to rise, a deeper understanding of this connection is crucial for developing effective prevention and therapeutic strategies. While lifestyle interventions are fundamental, the future of pharmacological treatment lies in personalized medicine. Significant research gaps remain, and addressing them will require a focus on three key areas: robust biomarker discovery to guide patient selection, the development of intelligent combination therapies that can overcome metabolic plasticity, and a better understanding of cancer risk in metabolically vulnerable populations. Successfully navigating these challenges will be critical to reducing the global cancer burden in our increasingly insulin-resistant world.

Author Contributions

Conceptualization, D.B., C.B. and S.M.; methodology, D.B., B.K.S. and A.J.; software, S.M., B.K.S. and C.B.; validation, D.B., C.B. and S.M.; formal analysis, B.K.S.; investigation, C.B. and S.M.; resources, D.B.; data curation, B.K.S. and K.Q.; writing—original draft preparation, A.J., B.K.S., C.B., S.M. and K.Q.; writing—review and editing, A.J., B.K.S. and D.B.; visualization, B.K.S.; supervision, D.B.; project administration, D.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Lucio, G.; Raffaele, P.; Maria, M.-L.A. Diabetes and the Risk of Pancreatic Cancer. N. Engl. J. Med. 1994, 331, 81–84. [Google Scholar] [CrossRef]
  2. Giovannucci, E.; Harlan, D.M.; Archer, M.C.; Bergenstal, R.M.; Gapstur, S.M.; Habel, L.A.; Pollak, M.; Regensteiner, J.G.; Yee, D. Diabetes and cancer: A consensus report. Diabetes Care 2010, 33, 1674–1685. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  3. Suh, S.; Kim, K.W. Diabetes and cancer: Cancer should be screened in routine diabetes assessment. Diabetes Metab. J. 2019, 43, 733–743. [Google Scholar] [CrossRef]
  4. Zhu, B.; Qu, S. The relationship between diabetes mellitus and cancers and its underlying mechanisms. Front. Endocrinol. 2022, 13, 800995. [Google Scholar] [CrossRef]
  5. Magliano, D.J.; Boyko, E.J. IDF Diabetes Atlas 10th edition scientific committee. In IDF Diabetes Atlas, 10th ed.; International Diabetes Federation: Brussels, Belgium, 2021. [Google Scholar] [PubMed]
  6. Saeedi, P.; Petersohn, I.; Salpea, P.; Malanda, B.; Karuranga, S.; Unwin, N.; Colagiuri, S.; Guariguata, L.; Motala, A.A.; Ogurtsova, K.; et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas, 9th edition. Diabetes Res. Clin. Pract. 2019, 157, 107843. [Google Scholar] [CrossRef]
  7. Siegel, R.L.; Kratzer, T.B.; Giaquinto, A.N.; Sung, H.; Jemal, A. Cancer statistics, 2025. CA Cancer J. Clin. 2025, 75, 10. [Google Scholar] [CrossRef] [PubMed]
  8. DeSantis, C.E.; Ma, J.; Gaudet, M.M.; Newman, L.A.; Miller, K.D.; Goding Sauer, A.; Jemal, A.; Siegel, R.L. Breast cancer statistics, 2019. CA Cancer J. Clin. 2019, 69, 438–451. [Google Scholar] [CrossRef] [PubMed]
  9. Pearson-Stuttard, J.; Zhou, B.; Kontis, V.; Bentham, J.; Gunter, M.J.; Ezzati, M. Worldwide burden of cancer attributable to diabetes and high body-mass index: A comparative risk assessment. Lancet Diabetes Endocrinol. 2018, 6, e6–e15. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  10. Pollak, M. Insulin and insulin-like growth factor signalling in neoplasia. Nat. Rev. Cancer 2008, 8, 915–928. [Google Scholar] [CrossRef] [PubMed]
  11. Belfiore, A.; Malaguarnera, R. Insulin receptor and cancer. Endocr. Relat. Cancer 2011, 18, R125–R147. [Google Scholar] [CrossRef] [PubMed]
  12. Cohen, D.H.; LeRoith, D. Obesity, type 2 diabetes, and cancer: The insulin and IGF connection. Endocr. Relat. Cancer 2012, 19, F27–F45. [Google Scholar] [CrossRef] [PubMed]
  13. Calle, E.E.; Kaaks, R. Overweight, obesity and cancer: Epidemiological evidence and proposed mechanisms. Nat. Rev. Cancer 2004, 4, 579–591. [Google Scholar] [CrossRef] [PubMed]
  14. Dankner, R.; Boffetta, P.; Balicer, R.D.; Boker, L.K.; Sadeh, M.; Berlin, A.; Olmer, L.; Goldfracht, M.; Freedman, L.S. Time-Dependent Risk of Cancer After a Diabetes Diagnosis in a Cohort of 2.3 Million Adults. Am. J. Epidemiol. 2016, 183, 1098–1106. [Google Scholar] [CrossRef] [PubMed]
  15. Frasca, F.; Pandini, G.; Scalia, P.; Sciacca, L.; Mineo, R.; Costantino, A.; Goldfine, I.D.; Belfiore, A.; Vigneri, R. Insulin receptor isoform A, a newly recognized, high-affinity insulin-like growth factor II receptor in fetal and cancer cells. Mol. Cell Biol. 1999, 19, 3278–3288. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  16. Belfiore, A. The role of insulin receptor isoforms and hybrid insulin/IGF-I receptors in human cancer. Curr. Pharm. Des. 2007, 13, 671–686. [Google Scholar] [CrossRef] [PubMed]
  17. Louvi, A.; Accili, D.; Efstratiadis, A. Growth-promoting interaction of IGF-II with the insulin receptor during mouse embryonic development. Dev. Biol. 1997, 189, 33–48. [Google Scholar] [CrossRef] [PubMed]
  18. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [PubMed]
  19. Nieman, K.M.; Romero, I.L.; Van Houten, B.; Lengyel, E. Adipose tissue and adipocytes support tumorigenesis and metastasis. Biochim. Biophys. Acta 2013, 1831, 1533–1541. [Google Scholar] [CrossRef]
  20. Lipscombe, L.L.; Goodwin, P.J.; Zinman, B.; McLaughlin, J.R.; Hux, J.E. The impact of diabetes on survival following breast cancer. Breast Cancer Res. Treat. 2008, 109, 389–395. [Google Scholar] [CrossRef] [PubMed]
  21. Vigneri, P.; Frasca, F.; Sciacca, L.; Pandini, G.; Vigneri, R. Diabetes and cancer. Endocr. Relat. Cancer 2009, 16, 1103–1123. [Google Scholar] [CrossRef] [PubMed]
  22. Baserga, R. The decline and fall of the IGF-I receptor. J. Cell Physiol. 2013, 228, 675–679. [Google Scholar] [CrossRef] [PubMed]
  23. Scalia, P.; Marino, I.R.; Asero, S.; Pandini, G.; Grimberg, A.; El-Deiry, W.S.; Williams, S.J. Autocrine IGF-II-Associated Cancers: From a Rare Paraneoplastic Event to a Hallmark in Malignancy. Biomedicines 2023, 12, 40. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  24. Christofori, G.; Naik, P.; Hanahan, D. A second signal supplied by insulin-like growth factor II in oncogene-induced tumorigenesis. Nature 1994, 369, 414–418. [Google Scholar] [CrossRef] [PubMed]
  25. Karin, M. Nuclear factor-kappaB in cancer development and progression. Nature 2006, 441, 431–436. [Google Scholar] [CrossRef] [PubMed]
  26. Park, J.; Morley, T.S.; Kim, M.; Clegg, D.J.; Scherer, P.E. Obesity and cancer—Mechanisms underlying tumour progression and recurrence. Nat. Rev. Endocrinol. 2014, 10, 455–465. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  27. Wang, X.; Bao, W.; Liu, J.; Ouyang, Y.Y.; Wang, D.; Rong, S.; Xiao, X.; Shan, Z.L.; Zhang, Y.; Yao, P.; et al. Inflammatory markers and risk of type 2 diabetes: A systematic review and meta-analysis. Diabetes Care 2013, 36, 166–175. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  28. Reuter, S.; Gupta, S.C.; Chaturvedi, M.M.; Aggarwal, B.B. Oxidative stress, inflammation, and cancer: How are they linked? Free Radic. Biol. Med. 2010, 49, 1603–1616. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  29. Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  30. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related inflammation. Nature 2008, 454, 436–444. [Google Scholar] [CrossRef] [PubMed]
  31. Crusz, S.M.; Balkwill, F.R. Inflammation and cancer: Advances and new agents. Nat. Rev. Clin. Oncol. 2015, 12, 584–596. [Google Scholar] [CrossRef] [PubMed]
  32. Garofalo, C.; Surmacz, E. Leptin and cancer. J. Cell Physiol. 2006, 207, 12–22. [Google Scholar] [CrossRef] [PubMed]
  33. Endo, H.; Hosono, K.; Uchiyama, T.; Sakai, E.; Sugiyama, M.; Takahashi, H.; Nakajima, N.; Wada, K.; Takeda, K.; Nakagama, H.; et al. Leptin acts as a growth factor for colorectal tumours at stages subsequent to tumour initiation in murine colon carcinogenesis. Gut 2011, 60, 1363–1371. [Google Scholar] [CrossRef] [PubMed]
  34. Saxena, N.K.; Sharma, D. Metastasis suppression by adiponectin: LKB1 rises up to the challenge. Cell Adhes. Migr. 2010, 4, 358–362. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  35. Dalamaga, M.; Diakopoulos, K.N.; Mantzoros, C.S. The role of adiponectin in cancer: A review of current evidence. Endocr. Rev. 2012, 33, 547–594. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  36. Housa, D.; Housová, J.; Vernerová, Z.; Haluzík, M. Adipocytokines and cancer. Physiol. Res. 2006, 55, 233–244. [Google Scholar] [CrossRef] [PubMed]
  37. Nieto, M.A.; Huang, R.Y.; Jackson, R.A.; Thiery, J.P. EMT: 2016. Cell 2016, 166, 21–45. [Google Scholar] [CrossRef] [PubMed]
  38. DeBerardinis, R.J.; Chandel, N.S. Fundamentals of cancer metabolism. Sci. Adv. 2016, 2, e1600200. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  39. Schulte, M.L.; Fu, A.; Zhao, P.; Li, J.; Geng, L.; Smith, S.T.; Kondo, J.; Coffey, R.J.; Johnson, M.O.; Rathmell, J.C.; et al. Pharmacological blockade of ASCT2-dependent glutamine transport leads to antitumor efficacy in preclinical models. Nat. Med. 2018, 24, 194–202. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  40. Stine, Z.E.; Walton, Z.E.; Altman, B.J.; Hsieh, A.L.; Dang, C.V. MYC, Metabolism, and Cancer. Cancer Discov. 2015, 5, 1024–1039. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  41. Jin, J.; Byun, J.K.; Choi, Y.K.; Park, K.G. Targeting glutamine metabolism as a therapeutic strategy for cancer. Exp. Mol. Med. 2023, 55, 706–715. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  42. Lowell, B.B.; Shulman, G.I. Mitochondrial dysfunction and type 2 diabetes. Science 2005, 307, 384–387. [Google Scholar] [CrossRef] [PubMed]
  43. Murphy, M.P. How mitochondria produce reactive oxygen species. Biochem. J. 2009, 417, 1–13. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  44. Valko, M.; Rhodes, C.J.; Moncol, J.; Izakovic, M.; Mazur, M. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem. Biol. Interact. 2006, 160, 1–40. [Google Scholar] [CrossRef] [PubMed]
  45. Ott, M.; Gogvadze, V.; Orrenius, S.; Zhivotovsky, B. Mitochondria, oxidative stress and cell death. Apoptosis 2007, 12, 913–922. [Google Scholar] [CrossRef] [PubMed]
  46. Youle, R.J.; Strasser, A. The BCL-2 protein family: Opposing activities that mediate cell death. Nat. Rev. Mol. Cell Biol. 2008, 9, 47–59. [Google Scholar] [CrossRef] [PubMed]
  47. Delbridge, A.R.; Grabow, S.; Strasser, A.; Vaux, D.L. Thirty years of BCL-2: Translating cell death discoveries into novel cancer therapies. Nat. Rev. Cancer 2016, 16, 99–109. [Google Scholar] [CrossRef] [PubMed]
  48. Singh, R.; Letai, A.; Sarosiek, K. Regulation of apoptosis in health and disease: The balancing act of BCL-2 family proteins. Nat. Rev. Mol. Cell Biol. 2019, 20, 175–193. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  49. Montero, J.; Sarosiek, K.A.; DeAngelo, J.D.; Maertens, O.; Ryan, J.; Ercan, D.; Piao, H.; Horowitz, N.S.; Berkowitz, R.S.; Matulonis, U.; et al. Drug-induced death signaling strategy rapidly predicts cancer response to chemotherapy. Cell 2015, 160, 977–989. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  50. Tschopp, J.; Schroder, K. NLRP3 inflammasome activation: The convergence of multiple signalling pathways on ROS production? Nat. Rev. Immunol. 2010, 10, 210–215. [Google Scholar] [CrossRef] [PubMed]
  51. Zhou, R.; Yazdi, A.S.; Menu, P.; Tschopp, J. A role for mitochondria in NLRP3 inflammasome activation. Nature 2011, 469, 221–225. [Google Scholar] [CrossRef] [PubMed]
  52. Arcidiacono, B.; Iiritano, S.; Nocera, A.; Possidente, K.; Nevolo, M.T.; Ventura, V.; Foti, D.; Chiefari, E.; Brunetti, A. Insulin resistance and cancer risk: An overview of the pathogenetic mechanisms. J. Diabetes Res. 2012, 2012, 789174. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  53. Hernandez, A.V.; Pasupuleti, V.; Benites-Zapata, V.A.; Thota, P.; Deshpande, A.; Perez-Lopez, F.R. Insulin resistance and endometrial cancer risk: A systematic review and meta-analysis. Eur. J. Cancer 2015, 51, 2747–2758. [Google Scholar] [CrossRef] [PubMed]
  54. Mu, N.; Zhu, Y.; Wang, Y.; Zhang, H.; Xue, F. Insulin resistance: A significant risk factor of endometrial cancer. Gynecol. Oncol. 2012, 125, 751–757. [Google Scholar] [CrossRef] [PubMed]
  55. Fujii, H.; Kawada, N.; Japan Study Group Of Nafld Jsg-Nafld. The Role of Insulin Resistance and Diabetes in Nonalcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2020, 21, 3863. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  56. Chiefari, E.; Mirabelli, M.; La Vignera, S.; Tanyolaç, S.; Foti, D.P.; Aversa, A.; Brunetti, A. Insulin Resistance and Cancer: In Search for a Causal Link. Int. J. Mol. Sci. 2021, 22, 11137. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  57. Wolpin, B.M.; Bao, Y.; Qian, Z.R.; Wu, C.; Kraft, P.; Ogino, S.; Stampfer, M.J.; Sato, K.; Ma, J.; Buring, J.E.; et al. Hyperglycemia, insulin resistance, impaired pancreatic β-cell function, and risk of pancreatic cancer. J. Natl. Cancer Inst. 2013, 105, 1027–1035. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  58. Zhang, A.M.Y.; Xia, Y.H.; Lin, J.S.H.; Chu, K.H.; Wang, W.C.K.; Ruiter, T.J.J.; Yang, J.C.C.; Chen, N.; Chhuor, J.; Patil, S.; et al. Hyperinsulinemia acts via acinar insulin receptors to initiate pancreatic cancer by increasing digestive enzyme production and inflammation. Cell Metab. 2023, 35, 2119–2135. [Google Scholar] [CrossRef] [PubMed]
  59. Lu, C.C.; Chu, P.Y.; Hsia, S.M.; Wu, C.H.; Tung, Y.T.; Yen, G.C. Insulin induction instigates cell proliferation and metastasis in human colorectal cancer cells. Int. J. Oncol. 2017, 50, 736–744. [Google Scholar] [CrossRef] [PubMed]
  60. Teng, J.A.; Wu, S.G.; Chen, J.X.; Li, Q.; Peng, F.; Zhu, Z.; Qin, J.; He, Z.Y. The Activation of ERK1/2 and JNK MAPK Signaling by Insulin/IGF-1 Is Responsible for the Development of Colon Cancer with Type 2 Diabetes Mellitus. PLoS ONE 2016, 11, e0149822. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  61. Rose, D.P.; Vona-Davis, L. The cellular and molecular mechanisms by which insulin influences breast cancer risk and progression. Endocr. Relat. Cancer 2012, 19, R225–R241. [Google Scholar] [CrossRef] [PubMed]
  62. Gunter, M.J.; Hoover, D.R.; Yu, H.; Wassertheil-Smoller, S.; Rohan, T.E.; Manson, J.E.; Li, J.; Ho, G.Y.; Xue, X.; Anderson, G.L.; et al. Insulin, insulin-like growth factor-I, and risk of breast cancer in postmenopausal women. J. Natl. Cancer Inst. 2009, 101, 48–60. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  63. Novosyadlyy, R.; Lann, D.E.; Vijayakumar, A.; Rowzee, A.; Lazzarino, D.A.; Fierz, Y.; Carboni, J.M.; Gottardis, M.M.; Pennisi, P.A.; Molinolo, A.A.; et al. Insulin-mediated acceleration of breast cancer development and progression in a nonobese model of type 2 diabetes. Cancer Res. 2010, 70, 741–751. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  64. Augustin, L.S.A.; Kendall, C.W.C.; Jenkins, D.J.A.; Willett, W.C.; Astrup, A.; Barclay, A.W.; Björck, I.; Brand-Miller, J.C.; Brighenti, F.; Buyken, A.E.; et al. Glycemic index, glycemic load and glycemic response: An International Scientific Consensus Summit from the International Carbohydrate Quality Consortium (ICQC). Nutr, Metab, Cardiovasc. Dis. 2015, 25, 795–815. [Google Scholar] [CrossRef] [PubMed]
  65. McRae, M.P. Dietary Fiber Intake and Type 2 Diabetes Mellitus: An Umbrella Review of Meta-analyses. J. Chiropr. Med. 2018, 17, 44–53. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  66. Imamura, F.; Micha, R.; Wu, J.H.; de Oliveira Otto, M.C.; Otite, F.O.; Abioye, A.I.; Mozaffarian, D. Effects of Saturated Fat, Polyunsaturated Fat, Monounsaturated Fat, and Carbohydrate on Glucose-Insulin Homeostasis: A Systematic Review and Meta-analysis of Randomised Controlled Feeding Trials. PLoS Med. 2016, 13, e1002087. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  67. Colberg, S.R.; Sigal, R.J.; Yardley, J.E.; Riddell, M.C.; Dunstan, D.W.; Dempsey, P.C.; Horton, E.S.; Castorino, K.; Tate, D.F. Physical Activity/Exercise and Diabetes: A Position Statement of the American Diabetes Association. Diabetes Care 2016, 39, 2065–2079. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  68. Moore, S.C.; Lee, I.M.; Weiderpass, E.; Campbell, P.T.; Sampson, J.N.; Kitahara, C.M.; Keadle, S.K.; Arem, H.; Berrington de Gonzalez, A.; Hartge, P.; et al. Association of Leisure-Time Physical Activity with Risk of 26 Types of Cancer in 1.44 Million Adults. JAMA Int. Med. 2016, 176, 816–825. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  69. Pedersen, B.K.; Saltin, B. Exercise as medicine—Evidence for prescribing exercise as therapy in 26 different chronic diseases. Scand. J. Med. Sci. Sport 2015, 25, 1–72. [Google Scholar] [CrossRef] [PubMed]
  70. Strasser, B.; Pesta, D. Resistance training for diabetes prevention and therapy: Experimental findings and molecular mechanisms. Biomed. Res. Int. 2013, 2013, 805217. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  71. Pernicova, I.; Korbonits, M. Metformin--mode of action and clinical implications for diabetes and cancer. Nat. Rev. Endocrinol. 2014, 10, 143–156. [Google Scholar] [CrossRef] [PubMed]
  72. Hosono, K.; Endo, H.; Takahashi, H.; Sugiyama, M.; Sakai, E.; Uchiyama, T.; Suzuki, K.; Iida, H.; Sakamoto, Y.; Yoneda, K.; et al. Metformin suppresses colorectal aberrant crypt foci in a short-term clinical trial. Cancer Prev. Res. 2010, 3, 1077–1083. [Google Scholar] [CrossRef] [PubMed]
  73. Goodwin, P.J.; Chen, B.E.; Gelmon, K.A.; Whelan, T.J.; Ennis, M.; Lemieux, J.; Ligibel, J.A.; Hershman, D.L.; Mayer, I.A.; Hobday, T.J.; et al. Effect of Metformin vs Placebo on Invasive Disease-Free Survival in Patients with Breast Cancer: The MA.32 Randomized Clinical Trial. JAMA 2022, 327, 1963–1973. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  74. Lerario, A.M.; Worden, F.P.; Ramm, C.A.; Hesseltine, E.A.; Stadler, W.M.; Else, T.; Shah, M.H.; Agamah, E.; Rao, K.; Hammer, G.D. The combination of insulin-like growth factor receptor 1 (IGF1R) antibody cixutumumab and mitotane as a first-line therapy for patients with recurrent/metastatic adrenocortical carcinoma: A multi-institutional NCI-sponsored trial. Horm. Cancer 2014, 5, 232–239. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  75. Mayer, I.A.; Arteaga, C.L. The PI3K/AKT Pathway as a Target for Cancer Treatment. Annu. Rev. Med. 2016, 67, 11–28. [Google Scholar] [CrossRef] [PubMed]
  76. Wright, S.C.E.; Vasilevski, N.; Serra, V.; Rodon, J.; Eichhorn, P.J.A. Mechanisms of Resistance to PI3K Inhibitors in Cancer: Adaptive Responses, Drug Tolerance and Cellular Plasticity. Cancers 2021, 13, 1538. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
Table 1. Summary of cancers with a strong association with insulin resistance.
Table 1. Summary of cancers with a strong association with insulin resistance.
Cancer TypeKey Associated MechanismsStrength of Epidemiological Evidence
Endometrial CancerHyperinsulinemia, Increased Bioavailable Estrogens (reduced SHBG), Chronic InflammationVery Strong
Liver Cancer (HCC)NAFLD/NASH Progression, Chronic Inflammation, Direct Mitogenic Effects of Insulin/IGF SystemVery Strong
Pancreatic CancerHyperinsulinemia, Local Pancreatic Inflammation, Direct Mitogenic EffectsStrong
Colorectal CancerHyperinsulinemia, IGF-1 Signaling, Chronic InflammationStrong
Breast Cancer (Postmenopausal)Hyperinsulinemia, IGF-1 Signaling, Increased Bioavailable Estrogens, InflammationStrong
Table 2. Relative risk of the association between T2DM and cancer. Adapted from “Insulin Resistance and Cancer Risk: An Overview of the Pathogenetic Mechanisms” by Arcidiacono, B. et al., 2012, Journal of Diabetes Research, PMC3372318. Licensed under CC BY 3.0. [52].
Table 2. Relative risk of the association between T2DM and cancer. Adapted from “Insulin Resistance and Cancer Risk: An Overview of the Pathogenetic Mechanisms” by Arcidiacono, B. et al., 2012, Journal of Diabetes Research, PMC3372318. Licensed under CC BY 3.0. [52].
CancerNumber (n) of Examined StudiesRelative Risk (CI 95%)
LiverCase–control (n = 13)2.50 (1.80–3.50)
Cohort (n = 7)2.51 (1.90–3.20)
Cohort (n = 18)2.01 (1.61–2.51)
EndometriumCase–control (n = 13)2.22 (1.80–2.74)
Cohort (n = 3)1.62 (1.21–2.16)
PancreasCase–control (n = 17)1.94 (1.53–2.36)
Cohort (n = 19)1.73 (1.59–1.88)
Cohort (n = 3)1.80 (1.50–2.10)
Cohort (n = 35)1.94 (1.66–2.27)
KidneyCase–control (n = 9)1.42 (1.06–1.91)
Biliary TractCase–control (n = 8) and cohort (n = 13)1.43 (1.18–1.72)
Case–control (n = 10) and cohort (n = 5)1.60 (1.38–1.87)
BladderCase–control (n = 7)1.37 (1.04–1.80)
Cohort (n = 3)1.43 (1.18–1.74)
ColorectalCase–control (n = 6)1.36 (1.23–1.50)
Cohort (n = 9)1.29 (1.16–1.43)
Case–control + cohort (n = 14)1.38 (1.26–1.51)
EsophagusCase–control (n = 6) and cohort (n = 11)1.30 (1.12–1.50)
Non-Hodgkin’s LymphomaCase–control (n = 5)1.12 (0.95–1.31)
Cohort (n = 11)1.41 (1.07–1.88)
Cohort (n = 10)1.18 (0.99–1.42)
Cohort (n = 3)1.79 (1.30–2.47)
BreastCase–control (n = 5)1.18 (1.05–1.32)
Cohort (n = 15)1.20 (1.11–1.30)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Subedi, B.K.; Bhimineni, C.; Modi, S.; Jahanshahi, A.; Quiza, K.; Bitetto, D. The Role of Insulin Resistance in Cancer. Curr. Oncol. 2025, 32, 477. https://doi.org/10.3390/curroncol32090477

AMA Style

Subedi BK, Bhimineni C, Modi S, Jahanshahi A, Quiza K, Bitetto D. The Role of Insulin Resistance in Cancer. Current Oncology. 2025; 32(9):477. https://doi.org/10.3390/curroncol32090477

Chicago/Turabian Style

Subedi, Bal Krishna, Charishma Bhimineni, Shivani Modi, Atousa Jahanshahi, Katherine Quiza, and Daniel Bitetto. 2025. "The Role of Insulin Resistance in Cancer" Current Oncology 32, no. 9: 477. https://doi.org/10.3390/curroncol32090477

APA Style

Subedi, B. K., Bhimineni, C., Modi, S., Jahanshahi, A., Quiza, K., & Bitetto, D. (2025). The Role of Insulin Resistance in Cancer. Current Oncology, 32(9), 477. https://doi.org/10.3390/curroncol32090477

Article Metrics

Back to TopTop