Available via license: CC BY-NC-ND 4.0
Content may be subject to copyright.
Vol.:(0123456789)
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
Discover Medicine
Review
Insights onmetabolic connections andinterplay betweencancer
anddiabetes: role ofmulti‑target drugs
AngelineJulius1· SureshMalakondaiah1· RaghuBabuPothireddy2
Received: 24 December 2024 / Accepted: 17 March 2025
© The Author(s) 2025 OPEN
Abstract
Cancer is recognized as a leading cause of death globally, imposing signicant health burdens. Traditional cancer treat-
ments encompass chemotherapy, surgery, and radiotherapy. Chemotherapy employs cytotoxic chemicals either alone or
in combination. However, these therapies can adversely aect normal cells and are hindered by drug resistance. Explora-
tion of alternative therapeutic approaches such as use of antidiabetic drugs for cancer treatment to rule out challenges
in current therapy is much needed. Antidiabetic medications like sulfonylureas, biguanides, and thiazolidinediones have
demonstrated benecial eects and are being repurposed for cancer management. The review discusses mechanisms
underlying their anticancer properties linked to metabolic factors common to both diseases, including hyperglycemia,
hyperinsulinemia, inammation, oxidative stress, and obesity. Nevertheless, certain antidiabetic drugs may pose risks
for developing cancer, particularly pancreatic cancer. Despite the concerns, the overall benecial impact of these agents
in cancer treatment outweighs their potential drawbacks. The review highlights the metabolic connections between
cancer and diabetes, as well as the mechanistic actions of antidiabetic drugs on cancer.
Keywords Hyperglycemia· Inammation· Oxidative stress· Multi-target drugs· Anti-diabetic drugs
1 Introduction
Cancer and diabetes are among the most prevalent and challenging diseases of the twenty-rst century, signicantly
contributing to global morbidity and mortality. Both conditions are multifaceted in nature, characterized by intricate
pathophysiological mechanisms involving metabolic dysfunction, chronic inammation, oxidative stress, and dysregu-
lated signaling pathways. The coexistence of these diseases in patients further complicates their management, as shared
risk factors and overlapping molecular pathways can exacerbate disease progression and treatment resistance [1].
Cancer represents a signicant global health challenge, contributing signicantly to mortality rates each year and
imposing substantial nancial burdens on patients. Conventional approaches to cancer treatment are either as stan-
dalone treatments or in combination. Radiotherapy is typically used for localized cancers following surgery, while chemo-
therapy is often the rst-line treatment for initial tumors, targeting rapidly dividing cells with cytotoxic drugs. However,
chemotherapy’s eectiveness is hindered by its cytotoxic eects on cells, such as those in the intestinal epithelium and
bone marrow, and the development of drug resistance [2].
* Angeline Julius, angeline.ibt@bharathuniv.ac.in | 1Centre forMaterials Engineering andRegenerative Medicine, Bharath Institute
ofHigher Education andResearch, Chennai, TamilNadu, India. 2AcaDiCell Innovations International Pvt. Ltd., Peerakankaranai,Vandalur,
TamilNadu, India.
Vol:.(1234567890)
Review
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
To overcome these limitations, alternative interventions are being explored. Some studies have investigated the poten-
tial of non-traditional therapeutic agents to address challenges like multidrug resistance and toxicity associated with
conventional anticancer drugs [3]. Epidemiological research has established metabolic links between cancer and type
2 diabetes, characterized by insulin resistance and pancreatic β-cell dysfunction, which increase the risk of cancers [4].
Although the exact mechanisms underlying the relationship between diabetes and cancer are still unknown, hyper-
glycemia, the production of AGEs and oxidative stress, hyperinsulinemia from exogenous insulin use or insulin resistance,
inammation, and obesity are common risk factors for both diseases. Even while diabetes is known to increase the risk
of cancer, several antidiabetic medications have been demonstrated to help prevent cancer [5].
Exogenous insulin therapy is the mainstay of treatment for type 1 diabetes, which is dened as complete insulin insuf-
ciency. Conversely, type 2 diabetes treatment typically involves antidiabetic drugs and lifestyle adjustments. By increas-
ing insulin secretion from pancreatic β-cells, improving insulin sensitivity in peripheral tissues, facilitating glucose uptake
into cells, and reducing glucose reabsorption in the kidneys and intestines, these medications help normalize blood
glucose levels and mitigate associated complications [6]. Remarkably, some of these drugs have demonstrated activity
against cancer cells, potentially inhibiting disease progression. Although some studies suggest that antidiabetic drugs
might pose a cancer risk or accelerate cancer progression, their anti-cancer eects generally outweigh these risks [7].
Traditional approaches to treating cancer and diabetes often focus on targeting single pathways or symptoms. How-
ever, such strategies may fail to address the complex interplay of biological processes underlying these diseases. Multi-
target drugs, designed to simultaneously modulate multiple pathways, represent a paradigm shift in the treatment of
cancer and diabetes. By oering a holistic approach, these drugs hold the potential to improve therapeutic ecacy,
minimize side eects, and address the shared mechanisms that link cancer and diabetes.
Furthermore, the article explores the signicance of multi-target drugs in addressing cancer and diabetes, outlining
challenges and future directions for their development. The objective is to underscore the potential of multi-target
therapies to transform the management of these interlinked diseases, providing insights for researchers and clinicians.
2 Diabetes andcancer
Multi-target drugs represent a transformative approach to managing cancer and diabetes, addressing the shared molecu-
lar and clinical complexities of these diseases. By targeting multiple pathways simultaneously, these therapies oer the
potential to improve ecacy, reduce side eects, and mitigate treatment resistance. Despite challenges in their devel-
opment, advancements in drug design, systems biology, and clinical trial methodologies are paving the way for their
successful implementation. As cancer and diabetes continue to pose signicant global health challenges, multi-target
drugs oer a promising avenue for improving patient outcomes and advancing the eld of medicine [8].
The research landscape exploring the connection between diabetes and cancer has predominantly focused on type
2 diabetes, given its higher prevalence compared to type 1 diabetes. Consequently, there is a signicant gap in under-
standing how type 1 diabetes relates to cancer risk and outcomes. Diabetes type 1 and type 2 have dierent hormonal
and metabolic traits. The autoimmune death of pancreatic beta cells causes type 1 diabetes, which is characterized by
complete insulin insuciency. On the other hand, type 2 diabetes is characterized by relative insulin insuciency and
insulin resistance, frequently combined with hyperglycemia and hyperinsulinemia [9].
Studies investigating diabetes and cancer risk typically include predominantly type 2 diabetes patients, making it chal-
lenging to generalize ndings to type 1 diabetes individuals. This limitation arises because the underlying mechanisms
linking diabetes to cancer can vary signicantly between these two types [10]. For instance, in type 1 diabetes, where
endogenous insulin secretion is decient and exogenous insulin is commonly administered, the dynamics inuencing
cancer development may dier from those in type 2 diabetes, where insulin resistance and higher insulin levels are
prevalent [11].
Several factors inuence the relationship between diabetes and cancer, including obesity, duration of diabetes, and
specic antidiabetic treatments such as insulin therapy. These factors can modify cancer risk through various mechanisms,
aecting cellular processes involved in carcinogenesis. Despite the growing body of research, studies often face limita-
tions such as small sample sizes, ambiguity in dening type 1 diabetes cohorts, short follow-up durations, and inconsist-
ent assessments of diabetes duration [12]. These challenges contribute to varied ndings across dierent cancer types
and hinder precise conclusions about the impact of diabetes, especially type 1 diabetes, on cancer risk and outcomes.
Understanding the mechanistic actions of antidiabetic drugs in the context of cancer management is crucial. Some
medications used in diabetes treatment, such as metformin, have shown potential anticancer eects through pathways
Vol.:(0123456789)
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
Review
independent of their glucose-lowering eects (Table1). However, the specic impacts of these drugs in individuals with
type 1 diabetes and their potential role in modifying cancer risk warrant further investigation.
Metformin activates AMPK (AMP-activated protein kinase), a critical energy sensor that regulates metabolic homeo-
stasis. Metformin inhibits hepatic gluconeogenesis, and directly inhibits the mTOR pathway, limiting tumor growth and
proliferation [19]. In cancer, metformin indirectly reduces insulin and IGF-1 levels, depriving tumors of growth signals.
Retrospective and prospective studies show lower cancer incidence and mortality in diabetic patients on metformin,
particularly for colorectal, breast, and pancreatic cancers [20]. Trials are investigating its ecacy in cancer therapy in
non-diabetic patients. However, ecacy appears to vary by cancer type and patient population. Higher doses required
for direct anti-cancer eects may not be well-tolerated due to gastrointestinal side eects [21, 22].
Pioglitazone activates PPAR-γ, improving insulin sensitivity by modulating gene expression involved in glucose and
lipid metabolism [23]. In cancer, PPAR-γ activation induces tumor dierentiation, inhibits proliferation, and promotes
apoptosis. Pioglitazone reduces systemic inammation and angiogenesis by downregulating pro-inammatory cytokines
[24]. Preclinical studies demonstrate anti-cancer eects in models of bladder and breast cancer. Observational studies
show mixed results, with some linking pioglitazone to reduced cancer risk, while others raise concerns about bladder
cancer. Associated with cardiovascular risks its clinical adoption is still under query and lack of randomized trials speci-
cally targeting its use in cancer prevention or therapy [25].
Aspirin irreversibly inhibits COX-1 and COX-2 (Cyclooxygenase enzymes), reducing prostaglandin production, chronic
inammation, and cancer-promoting signaling pathways such as NF-κB [26]. Reduction of platelet aggregation by aspi-
rin, limit metastatic spread by impairing tumor cell-platelet interactions. Studies like CAPP2 conrm its eectiveness in
Lynch syndrome patients depending on dose and duration, with risks outweighing benets in certain populations [27].
Statins inhibit HMG-CoA reductase, reducing cholesterol synthesis. In cancer, statins disrupt the mevalonate pathway,
which is critical to produce intermediates like isoprenoids that promote tumor cell proliferation. Induces apoptosis and
inhibits angiogenesis and metastasis. Observational studies show a reduction in breast, prostate, and colorectal cancer
incidence in diabetic patients, also in preclinical models [28].
Sodium-Glucose Co-Transporter 2 (SGLT2) Inhibitors (Empagliozin, Dapagliozin) inhibit SGLT2 in the proximal renal
tubules, promoting glucose excretion and reducing blood glucose levels thereby limiting glucose availability for rapidly
proliferating tumor cells [29]. Preclinical studies suggest additional eects on metabolic reprogramming and reduction
in oxidative stress. Emerging evidence suggests reduced cancer-related mortality in patients using SGLT2 inhibitors,
though direct anti-cancer eects remain under investigation. Ongoing trials are exploring their potential synergistic
use with chemotherapy [30].
Liraglutide, Semaglutidem (Glucagon-Like Peptide-1 receptor agonists) mimic GLP-1, enhancing glucose-dependent
insulin secretion, suppressing glucagon release, reduces hyperinsulinemia and systemic inammation. These agonists
may directly target GLP-1 receptors expressed in certain tumors, inhibiting proliferation and inducing apoptosis. Evi-
dence suggests reduced risk of colorectal, pancreatic, and liver cancers in diabetic patients using GLP-1 agonists. Rare
but severe side eects include pancreatitis and possible associations with thyroid cancer.
While diabetes, particularly type 2 diabetes, has been extensively studied about cancer, the evidence regarding type
1 diabetes remains limited. Future research eorts should prioritize delineating the unique interplay between type 1
diabetes and cancer, considering the distinct metabolic and hormonal proles and the implications of diabetes manage-
ment strategies on cancer outcomes [31, 32].
3 Diabetes‑cancer link‑factors thatcontribute
Diabetes may promote cancer initiation or progression through systemic alterations aecting all tissues and localized
mechanisms specic to certain organs. The key factors involved include hyperglycemia, insulin resistance, chronic inam-
mation, and changes in cellular environments that alter growth signaling, apoptosis, and tissue repair [33]. Chronic
hyperglycemia can lead to the formation of advanced glycation end products (AGEs), which are harmful compounds
that accumulate in tissues. AGEs bind to receptors for advanced glycation end products (RAGE), triggering inamma-
tory pathways, oxidative stress, and cellular dysfunction. This can increase the likelihood of cancerous transformation
and metastasis [34].
Furthermore, insulin can stimulate insulin-like growth factor (IGF)-1, which in turn activates various signaling
pathways such as the PI3K/Akt pathway that promote cell proliferation, survival, and resistance to apoptosis [35].
Vol:.(1234567890)
Review
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
Table 1 Multi-target drugs used for both cancer and diabetes, emphasizing their mechanisms and clinical signicance
Drug Primary use (diabetes) Mechanism in diabetes Mechanism in cancer Evidence
Metformin First-line treatment for Type 2 Diabetes Activates AMPK, reduces hepatic
gluconeogenesis, improves insulin
sensitivity
Activates AMPK to inhibit mTOR,
reduces insulin/IGF-1, induces cell
cycle arrest and apoptosis
Reduced cancer incidence (breast, colo-
rectal, pancreatic); widely supported
by epidemiological studies [13]
Pioglitazone (TZDs) Improves insulin sensitivity via Peroxi-
some Proliferator-Activated Receptor
Gamma (PPAR-γ)activation
Activates PPAR-γ, reduces inamma-
tion, and enhances glucose uptake PPAR-γ activation inhibits tumor pro-
liferation, promotes apoptosis, and
reduces angiogenesis
Some evidence in bladder and breast
cancer; limited use due to side eects
like cardiovascular risks [14]
Aspirin Anti-inammatory and anti-thrombotic
agent Reduces inammation, indirectly
improving insulin sensitivity Inhibits COX-2, reduces prostaglandins,
inammation, and tumor angiogen-
esis
Associated with lower colorectal and
other cancer risks in diabetic popula-
tions [15]
Statins Lowers cholesterol by inhibiting HMG-
CoA reductase Reduces systemic inammation,
improves endothelial function Inhibits the mevalonate pathway, sup-
presses tumor growth, angiogenesis,
and metastasis
Linked to reduced cancer risk (breast,
prostate, colorectal) in diabetic
patients [16]
SGLT2 Inhibitors Promotes glucose excretion via kidney
ltration Lowers blood glucose and insulin levels Limits glucose availability to cancer
cells, may disrupt tumor metabolic
pathways
Emerging evidence suggests reduced
cancer mortality in diabetic patients
[17]
GLP-1 Agonists Enhances glucose-dependent insulin
secretion Activates GLP-1 receptors, reduces
hyperinsulinemia and systemic
inammation
Reduces proliferation in GLP-1
receptor-positive tumors, lowers can-
cer risk indirectly through metabolic
improvement
Shown to reduce risks of colorectal and
pancreatic cancers in clinical studies
[18]
Vol.:(0123456789)
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
Review
Chronic activation of these pathways in cells can lead to uncontrolled growth, a hallmark of cancer. In the colon,
increased insulin and IGF-1 levels contribute to enhanced epithelial cell proliferation and decreased apoptosis that
accelerates the development of colorectal cancer [36]. High levels of estrogen during hyperinsulinemia particularly
in postmenopausal women, has been linked to the initiation and progression of breast cancer. Locally, specific organs
are affected by altered insulin signaling, changes in the tumor microenvironment, and dysregulated metabolic pro-
cesses. These factors increase the risk of developing a variety of cancers, including those of the pancreas, liver, colon,
breast, and endometrium. Understanding these mechanisms is crucial for developing strategies to reduce cancer
risk in people with diabetes [37].
Chronic systemic inflammation in diabetes causes elevated levels of inflammatory cytokines (TNF-α, IL-6, and CRP)
in the blood and have been linked to cancer development and progression by causing DNA damage, mutations,
and genomic instability, all of which are critical for cancer initiation. Inflammation can create a tumor-promoting
microenvironment by modifying the extracellular matrix, increasing the availability of growth factors, and enhancing
angiogenesis. Inflammatory signaling pathways such as the NF-κB and JAK/STAT pathways contribute to both insulin
resistance and tumorigenesis by promoting DNA damage, angiogenesis, and metastasis [38].
Diabetes not only affects cancer incidence but also impacts cancer prognosis and mortality [39, 40]. This increased
risk of mortality applies to various cancers, including colorectal cancer, where diabetes is associated with higher
cancer-specific case fatality [41]. In terms of mortality outcomes, diabetes plays a more significant role downstream,
affecting cancer-specific case fatality and overall mortality rates more than it does in influencing the initial incidence
of cancer. This trend is particularly notable in pancreatic cancer, where diabetes is strongly linked to both incidence
and poorer survival outcomes [42]. Probable population-based investigations and top-notch datasets are crucial
for comprehending these correlations. Potential confounding variables like age, body weight, physical activity lev-
els, food preferences, and other medical issues should all be carefully taken into account and adjusted for in these
investigations [43].
While diabetes clearly associated with increased risks of specific cancers and poorer outcomes in cancer patients,
further research is needed to elucidate the precise mechanisms and to comprehensively assess the impact across
different cancer types and populations [44]. It’s important to note that specific regions or populations may exhibit
unique associations between diabetes and certain cancers. Studies analyzing type 1 diabetes cohorts have indicated
elevated risks for some cancers compared to the general population, although findings have not been consistent
across all studies [45]. Recommendations have been made to focus studies on specific cancer types rather than overall
cancer incidences when exploring the relationship between type 2 diabetes and cancer, considering variations in
site-specific cancer patterns influenced by biological, clinical, or socioeconomic factors [46]. Figure1 illustrates the
linkage between diabetes and cancer.
Fig. 1 Linkage between dia-
betes and cancer
Vol:.(1234567890)
Review
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
3.1 Hyperglycaemia
Patients with cancer frequently exhibit glucose intolerance, which raises concerns regarding a possible connection
between type 2 diabetes and cancer, especially in light of the effects of elevated blood glucose levels [47]. Divergent
theories exist today on how increased blood glucose levels affect the development of cancerous cells. However, it
has been shown that hyperglycemia can enhance the generation of reactive molecules and free radicals, which can
cause oxidative damage to DNA, a crucial stage in the development of cancer. Proto-oncogenes and tumor suppres-
sor genes may experience alterations because of this oxidative stress. Glycated hemoglobin levels, which indicate
non-enzymatic and irreversible hemoglobin glycation and may also contribute to oxidative stress and cancer, can
also be used to evaluate persistent hyperglycemia [48]. This is corroborated by a study conducted in the general
Korean population, which discovered a correlation between a 3.03-fold increase in the risk of all cancer types and a
39.4% increase in glycated hemoglobin, indicating glycated hemoglobin as a causative risk factor. However, evidence
from large randomized controlled trials indicates that improving glycemic control in type 2 diabetes does not reduce
cancer risk, challenging the direct link between hyperglycemia and increased cancer risk. Furthermore, the complex
interaction of hyperglycemia with insulin resistance and obesity complicates the analysis of its independent impact
on cancer risk [49].
Furthermore, elevated glucose levels cause the glycation of proteins to increase and the production of AGEs, which
have been linked to the development of cancer and long-term consequences from diabetes. AGEs damage proteins
and modify the extracellular matrix directly, and they bind to the receptor for advanced glycation end products
(RAGE), triggering oxidative stress and inflammation[50]. The AGE-RAGE interaction is implicated in various path-
ways that promote cancer progression, such as inducing platelet-derived growth factor-B in pancreatic cancer and
promoting thrombogenesis, angiogenesis, and inflammation via specific signaling pathways[51].
Furthermore, cancer cells often rely on anaerobic glucose metabolism, leading to increased glycolysis and sub-
sequent AGE formation as a by-product. This dual effect of diabetes and cancer results in heightened AGE-RAGE-
dependent stress responses, fostering oxidative stress and chronic inflammation, which create a conducive environ-
ment for both diseases’ progression [1].
Excessive glucose levels under hyperglycemic circumstances also affect immune function by reducing ascorbic
acid delivery into immune cells in a competitive manner, which hinders immune cells’ capacity to carry out essential
tasks for fighting cancer cells. This phenomenon underscores how hyperglycemia can promote anabolic metabolism
in cancerous cells, fueling their growth and increasing the cancer risk associated with diabetes [52].
Pathways such as Wnt signaling link metabolic diseases like hyperglycemia and obesity with increased cancer risk
[33]. These connections highlight the intricate interplay between diabetes, cancer, and metabolic dysregulation,
emphasizing the need for further research into their shared mechanisms and potential therapeutic targets. Mecha-
nisms involved in linking hyperglycemia and cancer are listed in Table2.
3.2 Hyperinsulinemia
Hyperinsulinemia plays a significant role in cancer development by promoting anabolic and anti-apoptotic processes.
Experimental studies on animals have shown that models lacking insulin or with hyperglycemia exhibit fewer tumors
that progress slowly. Conversely, administering insulin reverses these effects, highlighting its mitogenic properties
[67].
Insulin binds to insulin receptors (IR), hybrid receptors (IR-IGF-IR), or insulin-like growth factor-insulin receptors
(IGF-IR) to produce its effects. IGF-IR is more extensively distributed in all tissue types [68]. A bad prognosis is linked
to the overexpression of IR in breast and prostate cancer cells, indicating that both receptors are involved in promot-
ing tumour growth. Insulin binding to its receptors initiates downstream signaling pathways like RAS/RAF/MEK/ERK
and Akt/protein kinase B, promoting cell proliferation and inhibiting apoptosis. The homology between insulin and
IGF-1 allows them to interact with either IR or IGF-IR, potentially forming hybrid receptors in tissues where both are
co-expressed [69]. These hybrid receptors, particularly in malignant breast and thyroid tissues, contribute to enhanced
cellular proliferation in response to insulin and IGF-1.
Moreover, insulin decreases the liver’s synthesis of IGFBP-1/2, proteins that typically block the effects of IGF-1.
Increased amounts of free circulating IGF-1, a powerful mitogen that stimulates the development of pre-cancerous
Vol.:(0123456789)
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
Review
Table 2 Factors linking hyperglycemia and cancer
Factor Mechanism Examples of associated cancers
Insulin Resistance Increases insulin and IGF-1 levels, promoting cell proliferation and reducing apoptosis Breast [53], Colorectal [54]
Oxidative Stress Elevated glucose leads to excessive ROS production, causing DNA damage and mutations Lung [55], Pancreatic [56]
Chronic Inammation Persistent hyperglycemia activates inammatory pathways (e.g., NF-κB), enhancing tumor growth Liver [57], Prostate [58]
Epigenetic Alterations Hyperglycemia induces histone modications and altered gene expression, fostering malignancy Gastric [59], Colon [60]
Angiogenesis Promotion High glucose increases VEGF levels, supporting new blood vessel formation for tumors Renal Cell Carcinoma [61],
Glioblastoma [62]
Altered Metabolism Provides excess glucose as fuel for cancer cell glycolysis (Warburg eect) Cervical [63], Liver [64]
Immune Evasion Impairs immune surveillance by reducing T-cell activity in a hyperglycemic environment Melanoma [65], Ovarian [66]
Vol:.(1234567890)
Review
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
cells expressing insulin, IGF-1, and hybrid receptors, are the result of this reduction. Proinflammatory cytokines are
released as a result of hyperinsulinemia, which promotes carcinogenesis even more [54]. In summary, hyperinsuline-
mia fosters tumor development through multiple mechanisms involving insulin and IGF-1 signaling, highlighting
its potential role in promoting cancer progression, especially in conditions like obesity and diabetes where insulin
levels are elevated.
3.3 Wnt/β‑catenin signaling pathway
The Wnt/β-catenin and mammalian target of rapamycin (mTOR) signaling pathways are critical in regulating cell processes
and have signicant implications in both cancer and type 2 diabetes [70]. Essential for several physiological processes,
including stem cell maintenance, epithelial-mesenchymal transition, and embryonic development, is the Wnt/β-catenin
pathway. Birth abnormalities and the emergence of cancers such as hepatocellular carcinoma, colon cancer, leukemia, and
metabolic illnesses like type 2 diabetes are linked to modications in this system caused by mutations in its constituent
parts [70]. The two main pathways that drive this process are the non-canonical pathway and the canonical Wnt signal-
ing dependent on β-catenin [71]. Hyperglycemia, characteristic of conditions like obesity and diabetes, enhances Wnt
signaling by promoting nuclear accumulation of β-catenin, thereby stimulating continuous expression of genes crucial
for cell proliferation and survival [72].
High glucose levels promote the formation of the lymphoid enhancer factor/β-catenin complex, leading to increased
acetyltransferase activity of p300 and reduced deacetylase activity of SIRT1. This results in the acetylation of β-catenin
at lysine 354, facilitating its nuclear translocation and activation of Wnt target genes. Dysregulation or mutations in
components such as adenomatous polyposis coli (APC), Axin, and β-catenin can lead to constitutive activation of the
pathway, contributing to malignancies like colorectal and hepatocellular carcinomas (Fig.2) [72].
Dysregulation of mTOR signaling is associated with insulin resistance, tumor formation, angiogenesis, and immune cell
activation. mTOR exists in two complexes: mTORC1, which regulates anabolic processes and cell growth, and mTORC2,
which inuences cell survival and cytoskeletal organization [73].
Activation of mTORC1 promotes cell growth and proliferation by enhancing protein synthesis and inhibiting
autophagy. This pathway is often constitutively activated in cancers due to mutations or alterations in upstream regula-
tors such as PI3K, AKT, and PTEN. Mutations in these components lead to persistent mTORC1 activation, driving tumori-
genesis in various cancers including breast, colorectal, and prostate cancers [74].
Both the Wnt/β-catenin and mTOR signaling pathways play crucial roles in regulating cell growth, proliferation, and
metabolism, and their dysregulation is implicated in the pathogenesis of cancer and type 2 diabetes. Understanding
these pathways’ intricate mechanisms and their interactions with other cellular processes is essential for developing
Fig. 2 Hyperglycemia induced
dysregulation of components
such as adenomatous polypo-
sis coli (APC), Axin, Dishev-
elled (DVL), Casein kinase 1
alpha (CK1α), GSK-3 beta and
formation of β-catenin-LEF
complex, activates Wnt target
genes
Vol.:(0123456789)
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
Review
targeted therapies and improving outcomes for patients aected by these diseases. The mTOR pathway controls basic
physiological functions such as cell survival, growth, proliferation, and metabolism. Dysregulation of mTOR signaling
has profound implications in both cancer and type 2 diabetes. Listed are the mTOR Complexes and Functions and their
implications in diseases [75].
3.4 mTOR complexes andfunctions
3.4.1 mTOR complex 1 (mTORC1)
mTORC1 inhibits catabolic processes like autophagy while controlling anabolic activities including the creation of lipids,
proteins, and organelles [76]. Growth factors such as insulin and insulin-like growth factor (IGF) can activate mTORC1 by
sending signals through the PI3K/AKT signaling pathway [77]. When AKT inhibits the Tuberous Sclerosis Complex (TSC)
1/2, mTORC1 is activated, which promotes cell division and growth [78].
Dysregulation or mutations in elements upstream or downstream of mTORC1, such as Ras, Raf, PI3K, AKT, PTEN, and
TSC genes, can result in constitutive activation of mTORC1 that causes cancers, including glioblastoma, melanoma, and
lymphoid malignancies [78].
3.4.2 mTOR complex 2 (mTORC2)
Dysregulation or mutations in elements upstream or downstream of mTORC1, such as Ras, Raf, PI3K, AKT, PTEN, and
TSC genes, can result in constitutive activation of mTORC1 that causes cancers, including glioblastoma, melanoma, and
lymphoid malignancies [79].
3.4.3 Role incancer
Component mutations of the mTOR pathway, particularly in upstream regulators and downstream eectors, lead to
aberrant activation of mTOR signaling, a hallmark of many cancers. Activation of mTORC1 angiogenesis, resulting in
tumour progression both invitro cell lines and invivo models. Activation mutations in oncogenes (Ras, Raf, PI3K, AKT)
and loss-of-function mutations in tumor suppressor genes (PTEN, TSC1/2) contribute to mTOR pathway dysregulation [80].
3.4.4 Role intype 2 diabetes
mTORC1 signaling inuences the function and mass of pancreatic β cells, crucial for glucose homeostasis. Dysregulated
mTORC2 signaling is implicated in insulin resistance, a key feature of type 2 diabetes. Enhanced mTORC1 activity initially
supports β-cell function (increased mass and insulin secretion), but prolonged activation can lead to β-cell dysfunction
and contribute to diabetes progression [81].
3.4.5 Therapeutic implications
mTOR inhibitors, such as rapamycin (sirolimus) and its derivatives, are actively studied for their potential in cancer therapy
due to their ability to inhibit mTORC1 activity and suppress tumor growth [82].
Targeting mTOR signaling remains a promising strategy in both cancer treatment and management of metabolic
disorders like type 2 diabetes. mTOR signaling plays a pivotal role in coordinating cellular responses to environmental
cues, and its dysregulation contributes signicantly to the pathogenesis of cancer and type 2 diabetes. Understanding
the complex interactions within this pathway is essential for developing targeted therapies aimed at mitigating these
diseases [74].
3.5 Role ofsevere inflammation
Severe inammation plays a pivotal role in the complex interplay between diabetes, inammation, and the develop-
ment of liver and pancreatic cancers. Here’s a detailed breakdown of how inammation inuences these processes [83].
Vol:.(1234567890)
Review
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
3.5.1 Inflammation anddiabetes
Poorly controlled diabetes is associated with chronic inammation with increased levels of inammatory cytokines
such as IL-6, TNF-α, and others. These inammatory markers contribute to insulin resistance and systemic inammation,
which are key factors in the pathogenesis of diabetes [84]. Diabetes, especially type 2 diabetes, signicantly increases
the risk of developing liver and pancreatic cancers. Factors contributing to this increased risk include hyperinsulinemia,
hyperglycemia, and chronic inammation [35].
3.5.2 Inflammation inliver cancer (HCC)
Inammatory cytokines such as IL-6, TNF-α, IL-1α, IL-1β, and IL-8 play critical roles in promoting chronic hepatic inam-
mation. They are upregulated in liver diseases like hepatitis and non-alcoholic fatty liver disease (NAFLD), creating an
inammatory microenvironment conducive to carcinogenesis [85].
IL-6 activates the gp130 receptor on hepatocytes, initiating signaling pathways like JAK/STAT3, PI3K/Akt, and Ras/Raf/
MAPK. These pathways not only support liver regeneration but also promote survival and proliferation of hepatocytes,
contributing to the development of HCC [86]. Activation of NF-κB due to inammation further exacerbates the pro-
tumorigenic environment in the liver, enhancing the progression of liver diseases towards cancer.
3.5.3 Inflammation inpancreatic cancer (PDAC)
Similarly, IL-6 is implicated in pancreatic cancer by activating the STAT3 signaling pathway, which stimulates cancer cell
proliferation and survival. This cytokine also modulates other pathways like ERK2, contributing to the aggressive nature
of PDAC. IL-4 and IL-8 also play roles in promoting pancreatic cancer cell growth and metastasis, highlighting the multi-
faceted role of inammation in disease progression [87]. NF-κB Activation: In pancreatic diseases like pancreatitis, NF-κB
activation leads to the production of inammatory cytokines, fostering an inammatory environment that can initiate
and promote tumor formation [88].
3.6 Role ofoxidative stress
Oxidative stress plays a critical role in the interplay between diabetes and cancer, linking these two conditions through
shared metabolic pathways and cellular mechanisms. Here’s a detailed breakdown of how oxidative stress contributes
to both diabetes and cancer and their association.
3.6.1 Oxidative stress indiabetes
Elevated blood glucose levels in diabetes contribute signicantly to oxidative stress. Several pathways amplify oxida-
tive stress in hyperglycemic conditions. Increased oxidative glucose metabolism in mitochondria leads to excessive
production of reactive oxygen species (ROS) such as O2·‾, OH·, and H2O2 [89]. Overactivation of uncoupling proteins
in mitochondria and NADPH oxidase contributes to the generation of superoxides, exacerbating oxidative stress. These
pathways increase ROS production and contribute to cellular damage. Antioxidant systems in the body are overwhelmed,
leading to inadequate protection against oxidative damage. These cells, responsible for insulin production, have limited
antioxidant enzyme expression, making them vulnerable to oxidative stress and contributing to diabetic complications
[90]. Prolonged oxidative stress damages biomolecules such as lipids, proteins, carbohydrates, and DNA, leading to cel-
lular dysfunction and tissue damage characteristic of diabetes complications [91].
3.6.2 Oxidative stress incancer
ROS-induced DNA damage can lead to mutations, replication errors, and genomic instability, all of which contribute to
the initiation and progression of cancer. Persistent oxidative stress induces chronic inammation, which plays a crucial
role in cancer development. Inammatory cells like neutrophils and mast cells release ROS during inammation, further
exacerbating oxidative stress [92]. Soluble mediators such as cytokines (TNF-α, IL-1, IL-6) and chemokines perpetuate
inammation and oxidative stress. Activation of transcription factors (NF-κB, HIF-1α, Nrf2, STAT3, AP-1, NFAT) by these
mediators promotes cell proliferation, survival, and angiogenesis, crucial for tumor growth [93]. Diabetes, characterized
Vol.:(0123456789)
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
Review
by chronic hyperglycemia and oxidative stress, increases the risk of DNA damage and promotes conditions favorable for
carcinogenesis. This association is complex and multifaceted, involving not only direct cellular damage but also systemic
eects on inammation and immune response [89].
4 Implications andResearch Challenges
Given the increasing prevalence of diabetes and its association with cancer, there is a pressing need for preventive strate-
gies targeting oxidative stress. Enhancing antioxidant defenses, reducing hyperglycemia through lifestyle changes or
pharmacotherapy, and targeting inammatory pathways are potential approaches [89].
Antioxidants and drugs that mitigate oxidative stress (like metformin) show promise in both diabetes management
and potentially cancer prevention or treatment. However, careful consideration of their dual eects on metabolism and
cancer risk is crucial. Oxidative stress serves as a common thread linking diabetes and cancer through shared metabolic
pathways and cellular responses [94]. Understanding these interconnected mechanisms oers insights into potential
preventive and therapeutic strategies that could mitigate the risk of cancer in diabetic patients and improve overall health
outcomes. Continued research into oxidative stress management holds promise for addressing the complex relationship
between these two prevalent and challenging diseases [95].
5 Use ofantidiabetic drugs andcancer risk
The relationship between antidiabetic medications and their potential impact on cancer risk is complex and has been
the subject of extensive research. Listed are the cancer risk of Here’s a breakdown of how dierent classes of antidiabetic
drugs, such as biguanides (metformin), sulfonylureas (SUs), and thiazolidinediones (TZDs), are implicated in cancer risk:
5.1 Metformin
Metformin is widely recognized for its potential anticancer eects. Several studies have suggested that metformin may
reduce the risk of cancer development and progression.
It exerts its eects through various mechanisms, including:
(i) AMPK Activation: Metformin activates AMP-activated protein kinase (AMPK), which regulates cellular energy
metabolism and inhibits cancer cell growth [96].
(ii) Insulin Sensitivity: By improving insulin sensitivity and reducing hyperinsulinemia, metformin indirectly mitigates
the growth-promoting eects of insulin and insulin-like growth factor 1 (IGF-1) [96].
(iii) Anti-inammatory Eects: Metformin has anti-inammatory properties that may contribute to its anticancer
eects [97].
Research have shown mixed results regarding metformin’s protective role against cancer. Some research supports
its use as a safe antidiabetic medication with potential benets in reducing cancer risk, particularly in gastrointestinal
cancers and possibly breast cancer.
5.2 Sulfonylureas (SUs)
Unlike metformin, SUs has been associated with an increased risk of certain cancers, particularly when used as mono-
therapy. SUs may stimulate carcinogenesis through mechanisms such as increasing insulin secretion and subsequent
hyperinsulinemia, which can promote cancer cell proliferation. They may also aect IGF-1 levels, which are linked to cell
growth and survival [98].
Population-based studies have shown that long-term use of SUs, especially in higher doses or as monotherapy, may
elevate the risk of pancreatic cancer and potentially other cancers.
Vol:.(1234567890)
Review
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
5.3 Thiazolidinediones (TZDs)
TZDs like pioglitazone have shown conicting results regarding their association with cancer risk. Some studies have
linked pioglitazone to an increased risk of pancreatic cancer. TZDs have also been associated with hepatic and pulmonary
cancers in certain populations, indicating a need for cautious use [99].
TZDs exert their eects through peroxisome proliferator-activated receptor gamma (PPARγ) activation, which regulates
insulin sensitivity and inammation. However, the long-term implications of PPARγ activation in cancer development are
still under investigation [100]. The choice of antidiabetic medication can inuence the risk of cancer in diabetic patients.
Metformin appears to be a favorable option due to its potential anticancer properties and overall safety prole in relation
to cancer risk. In contrast, SUs and TZDs, particularly pioglitazone, may pose a higher risk of certain cancers, especially
with prolonged use or in specic patient populations [33].
Clinicians should carefully consider the balance between glycemic control and potential cancer risks when selecting
antidiabetic therapies for their patients. Further research is needed to elucidate the precise mechanisms linking antidia-
betic medications to cancer risk and to rene treatment strategies accordingly [101].
5.4 Dipeptidyl peptidase‑4 (DPP‑4) inhibitors
DPP-4 inhibitors are a class of medications commonly used to manage type 2 diabetes mellitus. The relationship between
DPP-4 inhibitors and cancer risk has been the subject of various studies [102], yielding mixed results. Concerns have been
raised about a potential association between DPP-4 inhibitors and pancreatic cancer [103]. The association between
DPP-4 inhibitors and colorectal cancer appears to be dose-dependent [102]. A study found that low cumulative doses
of DPP-4 inhibitors were associated with a reduced risk of colorectal cancer, while high cumulative doses were linked
to an increased risk [102]. Research indicates that DPP-4 inhibitors may be associated with a reduced risk of melanoma
but not non-melanoma skin cancer. A study published in 2023 reported a 23% decreased risk of melanoma in patients
using DPP-4 inhibitors compared to those using sulfonylureas [104]. However, these ndings are not consistent across
all studies, and further research is needed to establish denitive conclusions.
5.5 Insulin
While insulin therapy is a cornerstone of diabetes management, concerns have been raised about its potential role in
cancer development mainly based on dose–response relationship and duration of the therapy. Hyperinsulinemia and
insulin therapy have been linked to cancer risk in some studies, particularly with certain cancers including colorectal and
breast cancers [105]. Long-acting insulin analogs, like glargine, have been scrutinized for potential risks, based on struc-
tural similarities to IGF-1, raising concerns about their potential mitogenic eects [106]. Hyperinsulinemia may promote
carcinogenesis through several biological pathways. Insulin acts as a growth factor by binding to insulin receptors (IR)
and insulin-like growth factor-1 receptors (IGF-1R) [54]. Overactivation of these receptors may enhance cell proliferation
and inhibit apoptosis, contributing to tumor development. Additionally high insulin levels suppress the production of
insulin-like growth factor binding proteins (IGFBPs), increasing free IGF-1 levels, which can stimulate tumor growth [53].
6 Conclusion
The repurposing of antidiabetic drugs for cancer management represents a promising avenue for targeted therapy, lev-
eraging shared metabolic pathways such as hyperglycemia, inammation, and oxidative stress. Drugs like sulfonylureas,
biguanides, TZDs, have demonstrated potential anticancer eects, yet challenges remain in fully integrating them into
oncology treatment protocols.
To establish the ecacy and safety of antidiabetic drugs in cancer treatment, large-scale, randomized controlled trials
are essential.
Future research should focus on identifying:
(i) Optimal dosing strategies for cancer patients, which may dier from those used for diabetes management.
Vol.:(0123456789)
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
Review
(ii) Patient selection criteria based on biomarkers or genetic predisposition to maximize therapeutic benets.
(iii) Combination therapies that integrate antidiabetic drugs with conventional or targeted cancer treatments to
enhance ecacy and reduce drug resistance.
Challenges in drug repurposing and development despite their potential, several barriers must be addressed. The
approval of antidiabetic drugs for oncological indications requires extensive preclinical and clinical validation, as their
mechanisms of action in cancer dier from their primary use. Long term safety concerns have to be addressed since
some antidiabetic drugs, particularly sulfonylureas and TZDs, have been linked to an increased risk of certain cancers,
necessitating further investigation into long-term eects and patient-specic risk factors.Pharmacokinetic considera-
tions must be taken into account since drug interactions and metabolic dierences between diabetic and non-diabetic
cancer patients may inuence ecacy and toxicity proles.
While antidiabetic drugs like metformin, sulfonylureas, and TZDs oer exciting possibilities for cancer management,
further clinical trials, mechanistic studies, and regulatory advancements are needed to optimize their use. Overcoming
these challenges could pave the way for more eective, safer, and cost-ecient therapeutic strategies, ultimately improv-
ing outcomes for patients with both diabetes and cancer.
7 Challenges andlimitations ofmulti‑target drug development
Developing multi-target drugs that simultaneously act on multiple pathways involved in diabetes and cancer presents
several challenges, including scientic, nancial, regulatory, and safety-related issues.
7.1 Scientific anddevelopment challenges
(i) Complex Disease Mechanisms
(ii) Both diabetes and cancer are heterogeneous diseases with diverse subtypes and underlying mechanisms.
(iii) Designing a drug that eectively targets multiple pathways without unintended interactions is challenging.
(iv) A multi-target drug must balance ecacy across dierent pathways without over-activating or inhibiting key
biological processes.
7.2 Off‑target effects andtoxicity
Multi-target drugs may lead to unintended interactions with other cellular pathways, increasing the risk of side eects.
For example, PPAR-γ agonists used in diabetes can have potential cardiovascular risks, and targeting metabolic pathways
in cancer can impact normal cell function.
7.3 Pharmacokinetic andpharmacodynamic optimization
A drug acting on multiple targets must have an optimized absorption, distribution, metabolism, and excretion (ADME)
prole to ensure eective bioavailability. Achieving the right balance between dierent targets requires extensive pre-
clinical studies to determine the optimal dosing regimen.
7.4 Drug resistance development
Cancer cells, in particular, are highly adaptable and can develop resistance mechanisms even against multi-target thera-
pies. Metabolic adaptation in diabetes (insulin resistance) can also counteract the eects of a drug over time.
7.5 Financial andcost challenges
Multi-target drug development requires extensive screening and optimization, increasing the cost of discovery and
preclinical research. The complexity of multi-target mechanisms often necessitates advanced computational modeling,
high-throughput screening, and invivo validation, adding to expenses.
Vol:.(1234567890)
Review
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
Because multi-target drugs interact with multiple pathways, the need for more extensive preclinical and clinical test-
ing extends the overall development time. Unexpected side eects or ecacy issues may lead to trial redesigns, further
increasing costs.
7.6 Regulatory andapproval challenges
Regulatory agencies like the FDA and EMA have well-dened guidelines for single-target drugs, but multi-target drugs
require more complex evaluation. The mechanism of action (MOA) for multi-target drugs may not be fully understood,
leading to diculties in dening ecacy endpoints.
7.7 Stringent safety requirements
Regulatory bodies require extensive toxicology studies and clinical trials to ensure that multi-target drugs do not cause
severe adverse eects. Drug interactions and metabolic eects need thorough investigation to avoid long-term risks,
particularly in chronic conditions like diabetes and cancer.
7.8 Complicated clinical trial designs
Clinical trials must evaluate ecacy across multiple targets and disease pathways, requiring larger and more diverse
patient groups. Since multi-target drugs aect multiple biological pathways, they may cause unintended systemic eects
(immune suppression, metabolic imbalances, organ toxicity). Patient-specic variations in genetics, metabolism, and
disease progression may aect drug ecacy and safety. A one-size-ts-all approach is unlikely to work, requiring addi-
tional biomarker-based patient stratication.
While multi-target drug development oers exciting potential for treating complex diseases like diabetes and cancer,
it faces signicant scientic, nancial, regulatory, and safety challenges. Overcoming these obstacles requires innova-
tive trial designs, personalized medicine approaches, and advanced computational modeling to predict and optimize
drug interactions. Despite the hurdles, advancements in AI-driven drug discovery, biomarker research, and combination
therapies may improve the success rate of multi-target drugs in the future.
8 Future directions
The interconnected molecular pathways between diabetes and cancer have led to the repurposing of several drugs
that target shared mechanisms such as inammation, hyperinsulinemia, and metabolic dysregulation. Among these,
metformin remains the most well-supported by evidence, showing broad potential in reducing cancer incidence and
progression. Drugs like SGLT2 inhibitors and GLP-1 receptor agonists represent newer classes with emerging evidence,
particularly for their dual role in improving metabolic health and potentially limiting cancer progression.
Signicant challenges that remain in translating these drugs into broader oncological practice are:
(i) Long-term safety concerns: Pioglitazone’s association with cardiovascular events and bladder cancer, or statins’
potential myopathies.
(ii) Drug-specic cancer eects: The impact of these drugs can vary based on cancer type, patient comorbidities, and
tumor biology.
(iii) Need for targeted trials: While many ndings stem from observational studies, more randomized controlled trials
are necessary to establish causality and dosing regimens for cancer prevention or adjunctive therapy.
Future directions include conducting well-designed clinical trials to validate the anti-cancer eects of these drugs in
diverse populations and exploring biomarker-based patient stratication to identify individuals most likely to benet.
Additionally, combining these agents with standard cancer therapies may provide synergistic benets while reducing
toxicities.
Emphasizing the clinical potential of these drugs, careful integration into therapeutic guidelines could signicantly
enhance outcomes for patients with diabetes and cancer, though overcoming translational barriers remains key.
Vol.:(0123456789)
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
Review
Author contributions A J conceived the idea, designed the gures and tables, performed formal analysis, supervised, and wrote the rst draft.
SM & RBP performed formal analysis and wrote the rst draft. AJ, SM & RBP made nal revisions, read and approved the nal manuscript.
Data availability No datasets were generated or analysed during the current study.
Declarations
Competing interests The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which
permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to
the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modied the licensed material. You
do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party
material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If
material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds
the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http:// creat iveco
mmons. org/ licen ses/ by- nc- nd/4. 0/.
References
1. Talib WH, Mahmod AI, Abuarab SF, Hasen E, Munaim AA, Haif SK, Ayyash AM, Khater S, Al-Yasari IH, Kury LT Al. Diabetes and cancer:
metabolic association, therapeutic challenges, and the role of natural products. Molecules 2021;26:2179.
2. Pucci C, Martinelli C, Ciofani G. Innovative approaches for cancer treatment: current perspectives and new challenges. Ecancermed Sci.
2019;13.
3. Talib WH, Alsayed AR, Barakat M, Abu-Taha MI, Mahmod AI. Targeting drug chemo-resistance in cancer using natural products. Biomedi-
cines. 2021;9:1353.
4. Li Y, Bian X, Wei S, He M, Yang Y. The relationship between pancreatic cancer and type 2 diabetes: cause and consequence. Cancer Manag
Res 2019;11:8257–68.
5. Zhang AMY, Wellberg EA, Kopp JL, Johnson JD. Hyperinsulinemia in obesity, inammation, and cancer. Diabetes Metab J. 2021;45:285–311.
6. Zhao X, An X, Yang C, Sun W, Ji H, Lian F. The crucial role and mechanism of insulin resistance in metabolic disease. Front Endocrinol
(Lausanne). 2023;14:1149239.
7. Kuo SZ, Honda CO, Li WT, Honda TK, Kim E, Altuna X, Abhold E, Wang-Rodriguez J, Ongkeko WM. Metformin results in diametrically
opposed eects by targeting non-stem cancer cells but protecting cancer stem cells in head and neck squamous cell carcinoma. Int J
Mol Sci. 2019;20:193.
8. Liu B, Zhou H, Tan L, Siu KTH, Guan X-Y. Exploring treatment options in cancer: tumor treatment strategies. Signal Transduct Target Ther.
2024;9:175.
9. Toren E, Burnette KS, Banerjee RR, Hunter CS, Tse HM. Partners in crime: beta-cells and autoimmune responses complicit in type 1 dia-
betes pathogenesis. Front Immunol. 2021;12: 756548.
10. Shahid RK, Ahmed S, Le D, Yadav S. Diabetes and cancer: risk, challenges, management and outcomes. Cancers (Basel). 2021;13:5735.
11. Petersen MC, Shulman GI. Mechanisms of insulin action and insulin resistance. Physiol Rev. 2018;98:2133–223.
12. Thomas NJ, Jones AG. The challenges of identifying and studying type 1 diabetes in adults. Diabetologia. 2023;66:2200–12.
13. Zhu L, Yang K, Ren Z, Yin D, Zhou Y. Metformin as anticancer agent and adjuvant in cancer combination therapy: current progress and
future prospect. Transl Oncol. 2024;44: 101945.
14. Asgharzadeh F, Memarzia A, Alikhani V, Beigoli S, Boskabady MH. Peroxisome proliferator-activated receptors: key regulators of tumor
progression and growth. Transl Oncol. 2024;47: 102039.
15. Wang B, Wu L, Chen J, Dong L, Chen C, Wen Z, Hu J, Fleming I, Wang DW. Metabolism pathways of arachidonic acids: mechanisms and
potential therapeutic targets. Signal Transduct Target Ther. 2021;6:94.
16. Jiang W, Hu J-W, He X-R, Jin W-L, He X-Y. Statins: a repurposed drug to ght cancer. J Exp Clin Cancer Res. 2021;40:1–33.
17. Anastasio C, Donisi I, Del Vecchio V, Colloca A, Mele L, Sardu C, Marfella R, Balestrieri ML, D’Onofrio N. SGLT2 inhibitor promotes mito-
chondrial dysfunction and ER-phagy in colorectal cancer cells. Cell Mol Biol Lett. 2024;29:80.
18. Zheng Z, Zong Y, Ma Y, Tian Y, Pang Y, Zhang C, Gao J. Glucagon-like peptide-1 receptor: mechanisms and advances in therapy. Signal
Transduct Target Ther. 2024;9:234.
19. Goel S, Singh R, Singh V, Singh H, Kumari P, Chopra H, Sharma R, Nepovimova E, Valis M, Kuca K. Metformin: activation of 5′ AMP-activated
protein kinase and its emerging potential beyond anti-hyperglycemic action. Front Genet. 2022;13:1022739.
20. Gandini S, Puntoni M, Heckman-Stoddard BM, Dunn BK, Ford L, DeCensi A, Szabo E. Metformin and cancer risk and mortality: a systematic
review and meta-analysis taking into account biases and confounders. Cancer Prev Res. 2014;7:867–85.
21. Chen K, Li Y, Guo Z, Zeng Y, Zhang W, Wang H. Metformin: current clinical applications in nondiabetic patients with cancer. Aging (Albany
NY). 2020;12:3993.
22. Corleto KA, Strandmo JL, Giles ED. Metformin and breast cancer: current ndings and future perspectives from preclinical and clinical
studies. Pharmaceuticals. 2024;17:396.
Vol:.(1234567890)
Review
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
23. Devchand PR, Liu T, Altman RB, FitzGerald GA, Schadt EE. The pioglitazone trek via human PPAR gamma: from discovery to a medicine
at the FDA and beyond. Front Pharmacol. 2018;9:1093.
24. Tan Y, Wang M, Yang K, Chi T, Liao Z, Wei P. PPAR-α modulators as current and potential cancer treatments. Front Oncol. 2021;11: 599995.
25. Tang H, Shi W, Fu S, Wang T, Zhai S, Song Y, Han J. Pioglitazone and bladder cancer risk: a systematic review and meta-analysis. Cancer
Med. 2018;7:1070–80.
26. Feng Y, Tao L, Wang G, Li Z, Yang M, He W, Zhong X, Zhang Y, Yang J, Cheung S. Aspirin inhibits prostaglandins to prevents colon
tumor formation via down-regulating Wnt production. Eur J Pharmacol. 2021;906: 174173.
27. Tao DL, Tassi Yunga S, Williams CD, McCarty OJT. Aspirin and antiplatelet treatments in cancer. Blood J Am Soc Hematol.
2021;137:3201–11.
28. Zaky MY, Fan C, Zhang H, Sun X-F. Unraveling the anticancer potential of statins: mechanisms and clinical significance. Cancers
(Basel). 2023;15:4787.
29. Padda IS, Mahtani AU, Parmar M. Sodium-glucose transport protein 2 (SGLT2) inhibitors. 2022.
30. Dabour MS, George MY, Daniel MR, Blaes AH, Zordoky BN. The cardioprotective and anticancer effects of SGLT2 inhibitors: JACC:
cardiooncology state-of-the-art review. Cardio Oncol. 2024;6:159–82.
31. Onitilo AA, Stankowski RV, Berg RL, Engel JM, Williams GM, Doi SA. A novel method for studying the temporal relationship between
type 2 diabetes mellitus and cancer using the electronic medical record. BMC Med Inform Decis Mak. 2014;14:1–9.
32. Hua Y, Zheng Y, Yao Y, Jia R, Ge S, Zhuang A. Metformin and cancer hallmarks: shedding new lights on therapeutic repurposing. J
Transl Med. 2023;21:403.
33. Olatunde A, Nigam M, Singh RK, Panwar AS, Lasisi A, Alhumaydhi FA, Jyoti Kumar V, Mishra AP, Sharifi-Rad J. Cancer and diabetes:
the interlinking metabolic pathways and repurposing actions of antidiabetic drugs. Cancer Cell Int. 2021;21:1–27.
34. Zawada A, Machowiak A, Rychter AM, Ratajczak AE, Szymczak-Tomczak A, Dobrowolska A, Krela-Kaźmierczak I. Accumulation of
advanced glycation end-products in the body and dietary habits. Nutrients. 2022;14:3982.
35. Szablewski L. Insulin resistance: the increased risk of cancers. Curr Oncol. 2024;31:998–1027.
36. Pavlova NN, Zhu J, Thompson CB. The hallmarks of cancer metabolism: Still emerging. Cell Metab. 2022;34:355–77.
37. Lero MW, Shaw LM. Diversity of insulin and IGF signaling in breast cancer: Implications for therapy. Mol Cell Endocrinol. 2021;527:
111213.
38. Stanimirovic J, Radovanovic J, Banjac K, Obradovic M, Essack M, Zafirovic S, Gluvic Z, Gojobori T, Isenovic ER. Role of C-reactive
protein in diabetic inflammation. Mediators Inflamm. 2022;2022:3706508.
39. Vigneri P, Frasca F, Sciacca L, Pandini G, Vigneri R. Diabetes and cancer. Endocr Relat Cancer. 2009;16:1103–23.
40. Renehan AG, Yeh H-C, Johnson JA, Wild SH, Gale EAM, Møller H, Consortium D and CR. Diabetes and cancer (2): evaluating the impact
of diabetes on mortality in patients with cancer. Diabetologia 2012;55:1619–32.
41. Gonzalez N, Prieto I, del Puerto-Nevado L, Portal-Nunez S, Ardura JA, Corton M, Fernandez-Fernandez B, Aguilera O, Gomez-Guerrero
C, Mas S. 2017 update on the relationship between diabetes and colorectal cancer: epidemiology, potential molecular mechanisms
and therapeutic implications. Oncotarget. 2017;8:18456.
42. Menini S, Iacobini C, Vitale M, Pesce C, Pugliese G. Diabetes and pancreatic cancer—a dangerous liaison relying on carbonyl stress.
Cancers (Basel). 2021;13:313.
43. Bammann K, Peplies J, Sjöström M, Lissner L, De Henauw S, Galli C, Iacoviello L, Krogh V, Marild S, Pigeot I. Assessment of diet, physical
activity and biological, social and environmental factors in a multi-centre European project on diet-and lifestyle-related disorders
in children (IDEFICS). J Public Health (Bangkok). 2006;14:279–89.
44. Vincent EE, Yaghootkar H. Using genetics to decipher the link between type 2 diabetes and cancer: shared aetiology or downstream
consequence? Diabetologia. 2020;63:1706–17.
45. Friberg E, Orsini N, Mantzoros CS, Wolk A. Diabetes mellitus and risk of endometrial cancer: a meta-analysis. Diabetologia.
2007;50:1365–74.
46. Johnson JA, Carstensen B, Witte D, Bowker SL, Lipscombe L, Renehan AG, Consortium D and CR. Diabetes and cancer (1): evaluating
the temporal relationship between type 2 diabetes and cancer incidence. Diabetologia. 2012;55:1607–18.
47. Joharatnam-Hogan N, Carter TJ, Reynolds N, Ho JH, Adam S, Board R. Diabetes mellitus in people with cancer. Endotext. 2021.
48. Black HS. Oxidative stress and ROS link diabetes and cancer. J Mol Pathol. 2024;5:96–119.
49. Kim JY, Lee YS, Jo G, Shin M-J. Glycated hemoglobin and cancer risk in korean adults: results from Korean genome and epidemiology
study. Clin Nutr Res. 2018;7:170–7.
50. Lee J, Yun J-S, Ko S-H. Advanced glycation end products and their effect on vascular complications in type 2 diabetes mellitus.
Nutrients. 2022;14:3086.
51. El-Far AH. The role of receptors for advanced glycation end product in pancreatic carcinogenesis. Pancreat Disord Ther.
2016;6:1000166.
52. Xiu F, Stanojcic M, Diao L, Jeschke MG. Stress hyperglycemia, insulin treatment, and innate immune cells. Int J Endocrinol. 2014;2014:
486403.
53. Kotsifaki A, Maroulaki S, Karalexis E, Stathaki M, Armakolas A. Decoding the role of insulin-like growth factor 1 and its isoforms in
breast cancer. Int J Mol Sci. 2024;25:9302.
54. Kasprzak A. Insulin-like growth factor 1 (IGF-1) signaling in glucose metabolism in colorectal cancer. Int J Mol Sci. 2021;22:6434.
55. Zhao Y, Ye X, Xiong Z, Ihsan A, Ares I, Martínez M, Lopez-Torres B, Martínez-Larrañaga M-R, Anadón A, Wang X. Cancer metabolism:
the role of ROS in DNA damage and induction of apoptosis in cancer cells. Metabolites. 2023;13:796.
56. Zhang Z, Huang Q, Zhao D, Lian F, Li X, Qi W. The impact of oxidative stress-induced mitochondrial dysfunction on diabetic micro-
vascular complications. Front Endocrinol (Lausanne). 2023;14:1112363.
57. Yang YM, Kim SY, Seki E. Inflammation and liver cancer: molecular mechanisms and therapeutic targets. Semin Liver Dis.
2019;39:26–42.
58. Staal J, Beyaert R. Inflammation and NF-κB signaling in prostate cancer: mechanisms and clinical implications. Cells. 2018;7:122.
Vol.:(0123456789)
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
Review
59. Babar Q, Saeed A, Tabish TA, Pricl S, Townley H, Thorat N. Novel epigenetic therapeutic strategies and targets in cancer. Biochim
Biophys Acta (BBA)-Mol Basis Dis 2022;1868:166552.
60. Cao Q, Tian Y, Deng Z, Yang F, Chen E. Epigenetic alteration in colorectal cancer: potential diagnostic and prognostic implications.
Int J Mol Sci. 2024;25:3358.
61. Ghalehbandi S, Yuzugulen J, Pranjol MZI, Pourgholami MH. The role of VEGF in cancer-induced angiogenesis and research progress
of drugs targeting VEGF. Eur J Pharmacol. 2023;949: 175586.
62. Liu Z-L, Chen H-H, Zheng L-L, Sun L-P, Shi L. Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct
Target Ther. 2023;8:198.
63. Tian Y, Zhang S, Ni F. Targeting glucose metabolism for HPV-associated cervical cancer: a sweet poison. Biomed Pharmacother.
2024;180: 117519.
64. Tufail M, Jiang C-H, Li N. Altered metabolism in cancer: insights into energy pathways and therapeutic targets. Mol Cancer.
2024;23:203.
65. Zhang S, Zhang X, Yang H, Liang T, Bai X. Hurdle or thruster: glucose metabolism of T cells in anti-tumour immunity. Biochim Biophys
Acta (BBA)-Rev Cancer 2024;1879:189022.
66. Lin X, Kang K, Chen P, Zeng Z, Li G, Xiong W, Yi M, Xiang B. Regulatory mechanisms of PD-1/PD-L1 in cancers. Mol Cancer. 2024;23:108.
67. Janssen JA. Hyperinsulinemia and its pivotal role in aging, obesity, type 2 diabetes, cardiovascular disease and cancer. Int J Mol Sci.
2021;22:7797.
68. Werner H, Bruchim I. The insulin-like growth factor-I receptor as an oncogene. Arch Physiol Biochem. 2009;115:58–71.
69. Kooijman R. Regulation of apoptosis by insulin-like growth factor (IGF)-I. Cytokine Growth Factor Rev. 2006;17:305–23.
70. Benchoula K, Parhar IS, Wong EH. The crosstalk of hedgehog, PI3K and Wnt pathways in diabetes. Arch Biochem Biophys. 2021;698:
108743.
71. Barker N. The canonical Wnt/β-catenin signalling pathway. Wnt Signal Pathw Methods Mamm Model 2008;5–15.
72. García-Jiménez C, García-Martínez JM, Chocarro-Calvo A, De la Vieja A. A new link between diabetes and cancer: enhanced WNT/β-
catenin signaling by high glucose. J Mol Endocrinol. 2014;52:R51–66.
73. Papadopoli D, Boulay K, Kazak L, Pollak M, Mallette FA, Topisirovic I, Hulea L. mTOR as a central regulator of lifespan and aging.
F1000Research 2019;8.
74. Bouyahya A, El Allam A, Aboulaghras S, Bakrim S, El Menyiy N, Alshahrani MM, Al Awadh AA, Benali T, Lee L-H, El Omari N. Targeting
mTOR as a cancer therapy: recent advances in natural bioactive compounds and immunotherapy. Cancers (Basel). 2022;14:5520.
75. Tian T, Li X, Zhang J. mTOR signaling in cancer and mTOR inhibitors in solid tumor targeting therapy. Int J Mol Sci. 2019;20:755.
76. Rabanal-Ruiz Y, Otten EG, Korolchuk VI. mTORC1 as the main gateway to autophagy. Essays Biochem. 2017;61:565–84.
77. Khan KH, Yap TA, Yan L, Cunningham D. Targeting the PI3K-AKT-mTOR signaling network in cancer. Chin J Cancer. 2013;32:253.
78. Polchi A, Magini A, Di MD, Tancini B, Emiliani C. mTOR signaling and neural stem cells: the tuberous sclerosis complex model. Int J
Mol Sci. 2018;19:1474.
79. Zhou H, Huang S. The complexes of mammalian target of rapamycin. Curr Protein Pept Sci. 2010;11:409–24.
80. Pópulo H, Lopes JM, Soares P. The mTOR signalling pathway in human cancer. Int J Mol Sci. 2012;13:1886–918.
81. Xie J, Herbert TP. The role of mammalian target of rapamycin (mTOR) in the regulation of pancreatic β-cell mass: implications in the
development of type-2 diabetes. Cell Mol Life Sci. 2012;69:1289–304.
82. Xie J, Wang X, Proud CG. mTOR inhibitors in cancer therapy. F1000Research 2016;5.
83. Tsalamandris S, Antonopoulos AS, Oikonomou E, Papamikroulis G-A, Vogiatzi G, Papaioannou S, Deftereos S, Tousoulis D. The role
of inflammation in diabetes: current concepts and future perspectives. Eur Cardiol Rev. 2019;14:50.
84. Spranger J, Kroke A, Mohlig M, Hoffmann K, Bergmann MM, Ristow M, Boeing H, Pfeiffer AFH. Inflammatory cytokines and the risk to
develop type 2 diabetes: results of the prospective population-based European Prospective Investigation into Cancer and Nutrition
(EPIC)-Potsdam Study. Diabetes. 2003;52:812–7.
85. Duan Y, Pan X, Luo J, Xiao X, Li J, Bestman PL, Luo M. Association of inflammatory cytokines with non-alcoholic fatty liver disease.
Front Immunol. 2022;13: 880298.
86. Nenu I, Toadere TM, Topor I, Țichindeleanu A, Bondor DA, Trella Șerban E, Sparchez Z, Filip GA. Interleukin-6 in hepatocellular car-
cinoma: a dualistic point of view. Biomedicines. 2023;11:2623.
87. Bhatia R, Bhyravbhatla N, Kisling A, Li X, Batra SK, Kumar S. Cytokines chattering in pancreatic ductal adenocarcinoma tumor micro-
environment. Semin Cancer Biol. 2022;86:499–510.
88. Silke J, O’Reilly LA. NF-κB and pancreatic cancer; chapter and verse. Cancers (Basel). 2021;13:4510.
89. Caturano A, D’Angelo M, Mormone A, Russo V, Mollica MP, Salvatore T, Galiero R, Rinaldi L, Vetrano E, Marfella R. Oxidative stress in
type 2 diabetes: impacts from pathogenesis to lifestyle modifications. Curr Issues Mol Biol. 2023;45:6651–66.
90. Hass DT, Barnstable CJ. Uncoupling proteins in the mitochondrial defense against oxidative stress. Prog Retin Eye Res. 2021;83:
100941.
91. Tangvarasittichai S. Oxidative stress, insulin resistance, dyslipidemia and type 2 diabetes mellitus. World J Diabetes. 2015;6:456.
92. Waris G, Ahsan H. Reactive oxygen species: role in the development of cancer and various chronic conditions. J Carcinog. 2006;5:14.
93. Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol
Med. 2010;49:1603–16.
94. Bosso M, Haddad D, Al Madhoun A, Al-Mulla F. Targeting the metabolic paradigms in cancer and diabetes. Biomedicines. 2024;12:211.
95. Forman HJ, Zhang H. Targeting oxidative stress in disease: promise and limitations of antioxidant therapy. Nat Rev Drug Discov.
2021;20:689–709.
96. Cao W, Li J, Hao Q, Vadgama JV, Wu Y. AMP-activated protein kinase: a potential therapeutic target for triple-negative breast cancer.
Breast Cancer Res. 2019;21:1–10.
97. Sakata N. The anti-inflammatory effect of metformin: the molecular targets. Genes Cells. 2024;29:183–91.
98. Kim HJ, Lee S, Chun KH, Jeon JY, Han SJ, Kim DJ, Kim YS, Woo J-T, Nam M-S, Baik SH. Metformin reduces the risk of cancer in patients
with type 2 diabetes: an analysis based on the Korean National Diabetes Program Cohort. Medicine (Baltimore). 2018;97: e0036.
Vol:.(1234567890)
Review
Discover Medicine (2025) 2:87 | https://doi.org/10.1007/s44337-025-00286-w
99. Bosetti C, Rosato V, Buniato D, Zambon A, La Vecchia C, Corrao G. Cancer risk for patients using thiazolidinediones for type 2 diabetes:
a meta-analysis. Oncologist. 2013;18:148–56.
100. Choi S-S, Park J, Choi JH. Revisiting PPARγ as a target for the treatment of metabolic disorders. BMB Rep. 2014;47:599.
101. Harris SB, Cheng AYY, Davies MJ, Gerstein HC, Green JB, Skolnik N. Person-centered, outcomes-driven treatment: a new paradigm
for type 2 diabetes in primary care. 2020.
102. Chou C-L, Juan S-H, Li C-H, Chen H-H, Kao C-C, Chen L-Y, Chien L-N, Fang T-C. Association between DPP-4 inhibitors and events of
colorectal and liver cancers in patients with diabetes receiving second-line agents: a nested case-control study. Front Oncol. 2022;12:
840142.
103. Pinto LC, Rados DV, Barkan SS, Leitao CB, Gross JL. Dipeptidyl peptidase-4 inhibitors, pancreatic cancer and acute pancreatitis: a
meta-analysis with trial sequential analysis. Sci Rep. 2018;8:782.
104. Pradhan R, Oriana HY, Platt RW, Azoulay L. Dipeptidyl peptidase-4 inhibitors and the risk of skin cancer among patients with type 2
diabetes: a UK population-based cohort study. BMJ Open Diabetes Res Care. 2023;11: e003550.
105. Zhu B, Qu S. The relationship between diabetes mellitus and cancers and its underlying mechanisms. Front Endocrinol (Lausanne).
2022;13:75.
106. Juárez-Vázquez CI, Gurrola-Díaz CM, Vargas-Guerrero B, Domínguez-Rosales JA, Rodriguez-Ortiz JF, Barros-Núñez P, Flores-Martínez
SE, Sánchez-Corona J, Rosales-Reynoso MA. Insulin glargine aects the expression of Igf-1r, Insr, and Igf-1 genes in colon and liver of
diabetic rats. Iran J Basic Med Sci. 2018;21:489.
Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional aliations.