Next Article in Journal
Metabolism of Selenium, Selenocysteine, and Selenoproteins in Ferroptosis in Solid Tumor Cancers
Previous Article in Journal
Health and Environmental Effect of Advanced Materials and Fine Particles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tirzepatide—Friend or Foe in Diabetic Cancer Patients?

by
Samson Mathews Samuel
1,*,
Elizabeth Varghese
1,
Peter Kubatka
2 and
Dietrich Büsselberg
1,*
1
Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
2
Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
*
Authors to whom correspondence should be addressed.
Biomolecules 2022, 12(11), 1580; https://doi.org/10.3390/biom12111580
Submission received: 18 September 2022 / Revised: 12 October 2022 / Accepted: 25 October 2022 / Published: 28 October 2022

Abstract

:
It is a well-accepted fact that obesity and diabetes increase the risk of incidence of different cancers and their progression, leading to a decrease in the quality of life among affected cancer patients. In addition to decreasing the risk of cancers, maintaining a healthy body mass index (BMI)/body weight and/or blood glucose levels within the normal range critically impacts the response to anti-cancer therapy among affected individuals. A cancer patient managing their body weight and maintaining blood glucose control responds better to anti-cancer therapy than obese individuals and those whose blood glucose levels remain higher than normal during therapeutic intervention. In some cases, anti-diabetic/glucose-lowering drugs, some of which are also used to promote weight loss, were found to possess anti-cancer potential themselves and/or support anti-cancer therapy when used to treat such patients. On the other hand, certain glucose-lowering drugs promoted the cancer phenotype and risked cancer progression when used for treatment. Tirzepatide (TRZD), the glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide/gastric inhibitory peptide (GIP) agonist, has recently gained interest as a promising injectable drug for the treatment of type 2 diabetes and was approved by the FDA after successful clinical trials (SURPASS 1/2/3/4 and 5, NCT03954834, NCT03987919, NCT03882970, NCT03730662, and NCT04039503). In addition, the reports from the SURMOUNT-1 clinical trial (NCT04184622) support the use of TRZD as an anti-obesity drug. In the current review article, we examine the possibility and molecular mechanisms of how TRZD intervention could benefit cancer therapeutics or increase the risk of cancer progression when used as an anti-diabetic drug in diabetic patients.

1. Incretin Mimetics for the Treatment of Type 2 Diabetes

Obesity and type 2 diabetes (hereafter mentioned as diabetes, unless otherwise specified) are among the most common chronic diseases worldwide, affecting all cross-sections of society, including children, women, youth, and adults. They are the leading causes of morbidity and mortality in the modern world [1,2,3,4]. Although the data from the international diabetes federation (IDF) projects a rise of about 10.9% in global diabetes prevalence by 2045, the obesity prevalence has increased in pandemic proportions over the past five decades [5,6]. Apart from the usual obesity and/or diabetes-related co-morbid complications, such as cardio- and cerebro-vascular diseases, hypertension, liver and renal diseases, and infertility, both obesity and diabetes are closely linked to the incidence, progression, and aggressiveness of many different cancers (breast, ovarian, pancreatic, liver, kidney, and colorectal cancers) [1,6,7,8]. This is partly correlated to the metabolic alterations, pro-inflammatory state, and subdued immune responses characteristic of obesity and diabetes [1,7,8].
The link between diabetes and cancer and the fact that cancer cells are essentially dependent on glucose (glycolysis; Warburg effect; to sustain their rapid rate of growth and proliferation) generated interest in studying the effects of widely prescribed anti-hyperglycemic/anti-diabetic drugs on the risk, incidence, progression, response to therapy, resistance and post-therapeutic relapse in cancers [9]. In addition to the blood glucose-lowering effect of these anti-hyperglycemic drugs (and thus restricting glucose availability to cancer cells), some of the tested anti-hyperglycemic drugs (e.g., metformin) have an anti-cancer potential via the inhibition of several different cancer-promoting pathways and by the activation of pathways that cause cancer cell death [8,9]. Coincidentally, many of these glucose-lowering medications or anti-diabetic drugs (metformin, SGLT2 inhibitors, GLP-1 receptor agonists, DPP4 inhibitors) were also effective for the treatment of obesity and for promoting and maintaining weight loss in both diabetic and non-diabetic individuals, albeit when supported by lifestyle changes, a balanced diet, and exercise [10,11,12,13,14,15,16]. In contrast, anti-hyperglycemic interventions such as insulin, thiazolidinediones, and sulphonylureas promote weight gain [17,18,19].
In May 2022, Tirzepatide (TRZD), marketed as Mounjaro (Eli Lilly and Company, Indianapolis, IN, USA), was approved by the Food and Drug Administration (FDA, Silver Spring, MD, USA) as an anti-hyperglycemic drug that improves glycemic control in adults with diabetes. TRZD, an incretin mimetic class of anti-diabetic drugs, is unique as it is the first and only dual-acting (glucose-dependent insulinotropic polypeptide/gastric inhibitory peptide/GIP and glucagon-like peptide 1 receptor/GLP-1R agonist) injectable anti-hyperglycemic drug showing significant improvements in glycemic control and body weight without the apparent risk of hypoglycemia [20]. Additionally, administration of the anti-hyperglycemic dosage of TRZD (5 mg, 10 mg, and 15 mg TRZD once weekly) was associated with a significant and sustained reduction in body weight [21].
Interestingly, incretin (GLP-1 and GIP) mimetics/agonists are known to cause (1) pancreatic damage (pancreatitis) and possible malignant transformation of the pancreas; and (2) promote the development of medullary thyroid carcinoma (MTC) in mice and possibly in patients with a family history of MTC, and (3) thyroid cancers in patients having multiple endocrine neoplasia syndrome type 2 (MEN2) [22,23,24]. TRZD administration, being a dual-acting incretin mimetic, is thus not recommended (by Eli Lilly and Company in their press release) in patients who have had pancreatitis and should not be used in patients with MTC (or anyone in the family of the patient who has had MTC) or if the patient has multiple MEN2 [25].
Apart from these cautionary suggestions/recommendations, there is little direct scientific evidence to suggest that there is reason to exercise caution regarding TRZD administration, possibly concerning cancer (pancreatic and thyroid) incidence and progression. Additionally, it remains unknown whether TRZD, indirectly via its blood glucose-lowering effect or directly by modulating pro-oncogenic/anti-oncogenic genes or pathways, can prevent cancer incidence and inhibit cancer progression. In this review article, we briefly examine the possibility and the related molecular mechanisms on how TRZD intervention may increase the risk of cancer incidence and progression or whether TRZD has the potential to be used alone or in combination with other commonly used anti-cancer interventions (surgery, chemotherapy, radiation) to enhance the efficacy of cancer treatments.

2. Possible Anti-Cancer Effects of TRZD

Beyond their pleiotropic effects, the incretin mimetics or GLP-1R/GIPR agonists could be beneficial and protective to multiple organ systems (brain, pancreas, stomach, intestine, kidneys, liver, adipose tissue, heart, and vasculature) in many different aspects (Figure 1). While significantly decreasing blood glucose levels and increasing insulin sensitivity, TRZD treatment also supported a reduction in body weight. It reversed dyslipidemia in the direction of an improved cardiometabolic profile [26,27]. The GLP-1R/GIPR agonists can also regulate the function of the skeletal muscle, immune system, and brown adipose tissue [28,29].
In cancers, owing to the link between high blood glucose levels/diabetes and obesity to the incidence of cancer and its progression, the TRZD treatment-mediated decrease in blood glucose and body weight will most likely inhibit cancer growth and progression. Liragludtide (a GLP-1R agonist) treatment sensitized gemcitabine-resistant pancreatic cell line (PANC-1) to gemcitabine leading to inhibition of cell proliferation and activation of apoptosis [30]. Similar effects were observed in other gemcitabine-insensitive pancreatic cancer cell lines (MiaPaca-1 and MiaPaca-2) [30]. The ability of liraglutide to reverse chemoresistance was mediated by the increased expression of GLP-1R and decreased levels of NF-κB and muti-drug resistance protein ABCG2 in the treated cells [30]. Zhao et al. reported that the GLP-1R activation in response to liraglutide treatment in human pancreatic cancer cell lines and a mouse xenograft model was associated with inhibiting the pancreatic cancer growth and increased apoptosis via the inhibition of Akt and ERK1/2 signaling pathways [31]. Liraglutide inhibits cancer growth and metastasis of human pancreatic cancer cells in vitro and in vivo via the PI3K/Akt signaling [31].
Exenatide similarly exerted its antiproliferative effect on breast cancer cells via the inhibition of NF-κB nuclear translocation and suppression of oncogenic gene expression [32]. Exenatide also promoted apoptosis in breast cancer MCF-7 cells via the upregulation of various apoptotic and death-dependent genes [33]. In MCF-7 breast cancer cells, liraglutide inhibited cancer cell proliferation and promoted apoptosis via the inhibition of microRNA-27a [34]. Ligumsky et al. reported that exenatide attenuated the growth of two breast cancer xenografts (MDA-MB-231 and MDA-MB-468 cells) in athymic mice [35]. Treatment with either GLP-1 or exenatide increased cAMP levels associated with the activation of down-stream target CREB and up-regulation of CRE promoter transcription [35]. The authors concluded that cAMP seems to be a key mediator of exenatide’s anti-cancer activities [35].
Both exenatide and liraglutide inhibited cell proliferation and increased apoptotic cell death in prostate cancer cells in a dose-dependent manner via an increase in the ratio of pro-apoptotic Bax/anti-apoptotic Bcl-2 and activation of the p38 MAPK-dependent cell death pathway [36]. Exenatide treatment reportedly exerted antiproliferative effects in the prostate cancer cell lines (LNCap, PC3, and DU14) and attenuated the growth of LNCap-derived xenografts in athymic mice [37]. Results correlated with the significant downregulation of the tumor expression of P504S, Ki67, and phosphorylated ERK-MAPK [37]. A combination of exenatide and metformin showed an additive effect on the prostate tumor size reduction and inhibition of prostate cancer growth via inhibiting the ERK-MAPK pathway and the AMPK-related inhibition of the mTOR pathway [38].
In colon cancer cells, exenatide reduced the survival of murine colon cancer cell lines via increased levels of cAMP, which inhibits PI3K/Akt, GSK3, and ERK1/2 and activates pro-apoptotic caspase 3/7 [39]. Similar results were reported by Tong et al., using liraglutide in LOVO colon cancer cells [40]. Liraglutide suppressed the cell cycle, reduced cell proliferation, migration, and invasion, and induced apoptosis by inactivating the PI3K/Akt/mTOR signaling [40].
Exenatide treatment also protected against endometrial cancers via AMPK activation and inhibition of the mTOR pathway [41]. In another study, exenatide suppressed hyperglycemia-induced chemoresistance in human endometrial adenocarcinoma Ishikawa and HEC1B cells via ROS-mediated mitochondrial pathway [42]. Pro-apoptotic changes via increased Bax/Bcl-2, cytosolic cytochrome c, PARP, and acetylated-p53 were upregulated by cisplatin but downregulated by high-glucose treatment; however, these changes were reversed by exenatide [42]. Kanda et al., reported that liraglutide dose-dependently suppressed the proliferation of endometrial carcinoma Ishikawa cells [43]. These results were associated with drug-induced autophagy and phosphorylation of AMPK [43]. Moreover, increased GLP-1R expression correlated with positive estrogen receptor and progesterone receptor status and better progression-free survival [43]. Exenatide in ovarian cancer cells promotes apoptotic cell death by the activation of caspase 3/7 and inhibits cell proliferation by suppressing the PI3K/Akt pathway [44,45]. In a translation case-control study, exenatide significantly upregulated co-expressions of PSMA2 protein, and GLP-1 receptor in cervical cancer specimens gained from patients with diabetes were reduced by exenatide [46]. In addition, exenatide suppressed phospho-p65 and -IκB expressions in the NF-κB signaling in vitro and tumor volume and Ki67 expression in vivo [46].
The augmented expression of receptors for advanced glycation end-products (RAGE) and its ligands, advanced glycation end-products (AGEs), high-mobility group box 1 protein (HMGB1), and S100 group of proteins in obese and diabetic individuals was shown to increase the risk, growth, survival, progression, aggressiveness, chemoresistance, metastasis, and poor prognosis among patients suffering from cancers of the pancreas, breast, and colo-rectum [47,48]. The interaction between the AGEs and RAGEs modulates and supports several pro-oncogenic and anti-apoptotic pathways and influences the tumor microenvironment thus controlling the process of inflammation, fibrosis and tumor angiogenesis in growing and metastazing tumors [48,49]. Hence, targeting the AGE-RAGE axis should be beneficial in the effective treatment of several different cancers [47,48]. GLP-1, reportedly reduced the expression of RAGE, in vitro and in vivo, and played a protective role in preventing AGE-related adverse effects in diabetes [50]. In a rat model of non-alcoholic steatohepatitis (NASH), exenatide reportedly displayed hepato-protective effects by downregulating RAGE expression [51]. Similar results of a decrease in hyperglycemia-induced RAGE expression were observed in exenatide-treated rat ventricular myocytes [52]. The anti-inflammatory effect of exenatide treatment was evidenced by the attenuation of AGE-induced production of IL6 and TNFα in rat mesangial cells [53]. However, it remains unclear whether the effect of exenatide or other GLP-1R agonists in the AGE-RAGE axis translates to anti-neoplastic effects for the treatment of cancers.
Whether TRZD could have similar anti-cancer effects through the activation of the GLP-1R/GIPR is yet to be determined.

3. Possible Oncogenic Effects of TRZD

The effects of TRZD are mediated by the GLP-1 receptor (GLP-1R) and the GIP receptor (GIPR). Concerns regarding the pro-oncogenic effects of TRZD are centered on the potential carcinogenic properties and the related molecular pathways. These are mainly triggered by GLP-1R and GIPR agonists and DPP4 (dipeptidyl peptidase 4) inhibitor (inhibits DPP4 mediated GLP-1 and GLP-2 degradation, leading to sustained high levels of both GLP-1 and GLP-2) intervention, mainly on the pancreas and the thyroid.
GLP-1R expression in non-neoplastic tissues has been observed in the pancreatic islets, acini, stomach, duodenal Brunner’s gland, small and large intestinal myenteric plexus, lung and kidney vasculature, breast parenchyma, heart, brainstem, hypothalamus, neurohypophysis, and meninges [54]. Interestingly, the GLP-1R density/expression was significantly higher (overexpressed) in insulinomas (high expression in benign insulinomas and lesser expression in malignant/metastasizing insulinoma) and medullary thyroid carcinomas) and other endocrine, embryonal, and brain tumors [54]. In physiology, the species difference in the expression of GLP-1R is also noteworthy and will play a role in the effect GLP-1R agonists (such as TRZD) on cancers. Although the expression of GLP-1R was higher in the lungs of rats compared to the humans, mice thyroid medullary C-cells showed a significant expression of GLP-1R but were more or less absent in the human thyroid tissue [54,55].
GIPR is expressed in the pancreas, bones, brain, and adipose tissue [56,57]. GIPR was significantly higher in neuroendocrine tumors (NETs), such as functional pancreatic NETs (including insulinomas), gastrinomas, and non-functional pancreatic NETs, compared to non-neoplastic tissue [58]. Colorectal cancers (CRC) express GIPR, which may facilitate GIPR ligand binding and subsequent proliferation of CRC cells via the modulation of pathways that support cancer progression. GIP, as such, is overexpressed in obesity, which could partly explain the higher incidence of CRC in obese individuals [59].

3.1. In the Pancreas

Therapeutic intervention using GLP-1 receptor agonists, such as exenatide and liraglutide, was correlated to an increased risk of pancreatitis [60,61,62,63,64,65,66]. A study assessing the pancreatic safety of using sitagliptin, a DPP4 inhibitor, reported a small increase in the risk of developing pancreatitis [67]. A significant increase in pancreatitis was described by the US FDA advance event reporting system (FAERS) when using sitagliptin and exenatide in comparison to patients using other anti-diabetic drugs (rosiglitazone, glipizide, nateglinide, reparglinide) to control their blood glucose levels [68].
In the general population, having chronic pancreatitis, possibly due to an inherited (hereditary/familial) gene (PRSS1, SPINK1, CTRC, CASR, and CFTR) mutation, significantly increases the risk of developing pancreatic cancer by about 40% [69,70]. The sustained release of pro-inflammatory cytokines, increase in the levels of reactive oxygen species and related oxidative stress, the dysregulated cellular proliferation, as well as the intraductal pressure due to stenosis of the pancreatic duct(s) characteristic of chronic pancreatitis increases the risk of possible malignant transformation of the pancreas leading to pancreatic cancer (by approximately 26-fold), a diagnosis with a significantly high risk of an unfavorable prognosis [66,70,71,72,73]. Although it is not clear whether an episode of acute pancreatitis (such as those triggered by incretin mimetic intervention) could trigger a similar malignant transformation of the pancreatic tissues, it is worthwhile to educate the at-risk patients and monitor them for any symptoms/signs of pancreatitis.
Retrospective studies have reported that incretin mimetic administration was associated with an increased risk of pancreatic cancer [74]. The occurrence of pancreatic cancer was also more often reported among patients on a GLP-1 agonists-based treatment regimen [68]. A marked expansion in the exocrine and endocrine compartments of the pancreas with an increase in pancreatic mass augmented exocrine cell proliferation, and dysplasia was reported in diabetic patients on incretin mimetic therapy [75]. Evident α-cell hyperplasia and progressive glucagon-producing neuroendocrine tumors were observed in diabetic patients on incretin mimetic therapy [75].
In the pancreatic β-cell, it is possible that TRZD treatment and subsequent GLP-1R or GIPR activation (Figure 2) via the adenylyl cyclase-cAMP-protein kinase A pathway activates the glycolytic and mitochondrial metabolism of glucose, in turn increasing intracellular ATP. The increase in the ATP/ADP leads to the closure of the plasma membrane K+ channels, eventually leading to β-cell depolarization, the subsequent opening of the voltage-gated Ca2+ channels, and the resultant influx of Ca2+ into the cell (the endoplasmic reticulum also releases Ca2+ in response to the initial elevation of cytosolic Ca2+). The increase in cytosolic Ca2+ triggers the release of insulin into the bloodstream. The activation of protein kinase A also, in turn, activates insulin gene transcription leading to the synthesis of insulin, insulin-like growth factors 1 and 2 (IGF 1 and 2), and insulin-like growth factor binding protein 3 (IGF-BP3), which via the insulin receptor or the insulin-like growth factor receptors activates the key pathways such as PI3K/Akt/mTOR, Ras-Raf-ERK, MAPK and JAK-STAT pathway in target cells [58,66]. The aberrant activation of these signaling pathways in an individual with a prior history of pancreatitis could significantly increase the risk of developing pancreatic cancer [76,77,78,79]. Insulin and insulin-like growth factors also activate these oncogenic pathways in other target tissues such as the breast, liver, and colorectum; thereby driving the progression of cancers by supporting cellular proliferation and growth, aberrant cancer metabolism, inhibition of cell death, invasion, and metastasis in these tissues [8,80,81].

3.2. In the Thyroid

Apart from the normal pancreatic cells, the GLP-1R and GIPR are expressed in the parafollicular of C-cells of the thyroid that secrete calcitonin [22,23]. Experimental evidence in rodents suggests a higher incidence of thyroid C-cell tumors in response to GLP-1 agonist intervention [22,23,73].
Anti-hyperglycemic incretin mimetics/GLP-1 analogs/agonists (such as liraglutide, exenatide, taspoglutide, and lixisenatide) caused a potent dose-dependent increase in cAMP levels and protein kinase A activation in the thyroid C-cells, leading to the activation of calcitonin gene expression, and thus triggered the release of calcitonin [22,23]. The sustained activation of these receptors and release of calcitonin is accompanied by C-cell hyperplasia and transformation into C-cell adenomas and MTC [22,73]. Treatment with TRZD (dual GLP-1R and GIPR agonist) could have a similar effect as the other incretin mimetic anti-diabetic drugs (Figure 3).
The effect of TRZD may vary depending on the species-related difference in the expression levels of the GLP-1R and GIPR. In primates and humans, the expression of GLP-1R in the C-cells of the normal thyroid is, however, significantly low or absent compared to that of the GLP-1R expression in thyroids of rodents [82,83]. Cell lines derived from rodent thyroid C-cells, when exposed to GLP-1 receptor agonists, lead to an elevation of cAMP levels and subsequent calcitonin release [23]. This effect of GLP-1 receptor agonists on cAMP levels and calcitonin secretion was not observed in cell lines derived from human thyroid C-cells, possibly due to the lower expression of GLP-1R on the human-derived cells [23]. Hence, the effect of TRZD or other GLP-1 agonists on calcitonin release and MTC may not be as pronounced in other primates and humans as in rodents. However, in rodents and humans, the expression of GLP-1R and GIPR was significantly higher in MTCs compared to normal thyroid tissue across the same species [82,84]. TRZD treatment thus may be a cause of concern if used in patients with MTC (or anyone in the family of the patient has had MTC) or if the patient has multiple MEN2, owing to the possibility of risk of developing and/or progression of MTCs in affected individuals [84].

4. Clinical Data and Trials

Clinical trials (ClincalTrials.gov, accessed on 12 October 2022); search keyword: tirzepatide) are underway to test and continue testing the efficacy of TRZD alone and in comparison with other anti-diabetic drugs for the treatment of type 2 diabetes, obesity, cardiovascular events, heart failure, and obstructive sleep apnea. The drug is also tested for adverse events in participants with impaired liver (hepatic insufficiency and non-alcoholic steatohepatitis) and renal function (renal insufficiency and end-stage renal disease).
Data from ClincalTrials.gov (accessed on 12 October 2022) includes a list of 29 (search performed on 12 October 2022; using keywords diabetes and tirzepatide) tirzepatide administration-based clinical trials concerning/specific to diabetes (Table 1). In total, 16 trials out of the 29 were completed. Moreover, 13 tirzepatide and diabetes-related clinical trials are currently active, out of which 4 are active and recruiting patients, whereas two clinical trials have not started recruiting subjects for the study (Table 1).
On the other hand, a search using the keywords obesity and tirzepatide yielded 14 tirzepatide administration-based clinical trials related to/specific to obesity (Table 1). Though 2 trials out of the 14 are completed, 12 tirzepatide and obesity-related clinical trials are currently active, out of which 4 are active and recruiting patients, and 2 clinical trials have not started recruiting subjects for the study (Table 1).
A search using the keywords cancer and tirzepatide did not yield any results (Table 1).

5. Conclusions and Future Perspective

TRZD is considered a novel glucose-lowering drug since it is the first and only dual-acting GLP-1R/GIPR agonist that FDA has approved for the treatment of diabetes. Furthermore, the recommended anti-hyperglycemic dosage of TRZD significantly promoted weight loss and may be used in the treatment of obesity. Reports suggest that other GLP-1R/GIPR agonists, besides their glucose-lowering effect, can be valuable for the prevention and treatment of cardiovascular diseases, metabolic diseases, and neuro- and endocrine disorders owing to their vast array of multi-faceted regulatory effects on the different organ systems. Furthermore, GLP-1R/GIPR agonists were found to have several anti-neoplastic effects in different cancers. Basic and clinical research studies are warranted to ascertain whether TRZD, a dual-acting GLP-1R/GIPR agonist, can bring about similar protective and beneficial effects, as reported in the case of other GLP-1R/GIPR agonists.
Although controversial, TRZD treatment may likely promote/support cancers of the pancreas, thyroid, breast, liver, and colon. However, with the currently available short-term data, it is difficult to ascertain whether the benefits of using tirzepatide for treating diabetes and/or obesity could outweigh its risks. For instance, although it is established that chronic pancreatitis increases the risk of pancreatic cancers, this may not hold in cases of acute pancreatitis such as those caused by the GLP-1/GIP analogs [73]. Based on the data on known mutation rates, it is estimated that it may take over 20 years for normal pancreatic duct cells to transform and grow into a pancreatic carcinoma with metastatic capacity and be diagnosed due to its clinical symptoms [73]. Once diagnosed with diabetes, the individual may be maintained on the anti-diabetic treatment regimen for the rest of his/her life. If the individual is on a GLP-1R/GIPR agonist or TRZD, the long-term exposure to these drugs might accelerate the progression of malignant disease in target tissues. Hence, the study subjects involved in the clinical trials using TRZD need to be followed up and monitored for several years, which will help acquire reliable data on the risks for the malignant disease since the development of cancer can most likely only be influenced over a long period.
Nonetheless, the safety profile of tirzepatide must be precisely monitored, and any serious adverse events (SAEs) should be strictly reported without bias. The completed and currently active clinical trials must educate their participants to identify/recognize and report any adverse event and carry out a long-term follow-up of their participants to identify any SAEs. Basic scientists must come forward to test tirzepatide for its potential cancer-promoting or cancer therapeutic effects. Clinical trials should address the efficacy and safety of using tirzepatide in diabetic non-cancer and cancer patients regarding the possibility of linking it to cancer incidence and progression. There are several outstanding questions (Figure 4) regarding TRZD’s effect on cancer. The pro-oncogenic or anti-cancer effect of TRZD and the possible molecular mechanisms involved are yet to be determined. It is still unknown how TRZD compares with other anti-diabetic drugs with known anti-neoplastic abilities. Whether a combination of TRZD and routinely used chemotherapeutic drugs can efficiently treat cancers is yet to be answered.

Author Contributions

The authors contributed as follows: Conceptualization, S.M.S. and E.V.; literature review and resources, S.M.S.; writing—original draft preparation, S.M.S.; writing—review and editing, S.M.S., E.V., P.K. and D.B.; figure preparation and editing, S.M.S.; visualization, S.M.S. and D.B.; supervision, D.B.; project administration, D.B.; funding acquisition, D.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a National Priorities Research Program grant (NPRP 11S-1214-170101; awarded to Professor Dr. Dietrich Büsselberg, June 2019-Current) from the Qatar National Research Fund (QNRF, a member of Qatar Foundation). The statements made herein are solely the responsibility of the authors.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The publication of this article was funded by the Weill Cornell Medicine—Qatar Health Sciences Library.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the article.

Abbreviations

AGEsAdvanced glycation end-products
AktProtein kinase B
AMPAdenosine monophosphate
ATPAdenosine triphosphate
cAMP3′,5′-cyclic AMP
DPP4Dipeptidyl peptidase 4
ERKExtracellular signal-regulated kinase
FAERSFDA advance event reporting system
FDAFood and drug administration
GIPGIP receptor
GIPGlucose-dependent insulinotropic polypeptide/Gastric inhibitory peptide
GLP-1Glucagon-like peptide 1
GLP-1RGLP-1 receptor
IGF-1RIGF-1 receptor
IGF-BP3Insulin-like growth factor binding protein 3
IGF1/2Insulin-like growth factor 1/2
JAKJanus kinase
MAPKMitogen activated protein kinase
MEN2Multiple endocrine neoplasia syndrome type 2
MTCMedullary thyroid cancer/carcinoma
mTORmammalian target of rapamycin
NF-κBNuclear factor kappa-B
PARPPoly (ADP-ribose) polymerase
PI3KPhosphoinositide 3-kinase
PSMA2Proteasome alpha 2 subunit
RAGEsReceptors of AGEs
SGLT2Sodium glucose co-transporter 2
STATSignal Transducer and Activator of Transcription
TRZDTirzepatide
WHOWorld Health Organization

References

  1. Bellanger, T.M.; Bray, G.A. Obesity related morbidity and mortality. J. La. State Med. Soc. 2005, 157, S42–49; quiz 49. [Google Scholar]
  2. Burki, T. European Commission classifies obesity as a chronic disease. Lancet Diabetes Endocrinol. 2021, 9, 418. [Google Scholar] [CrossRef]
  3. Hu, F.B. Globalization of diabetes: The role of diet, lifestyle, and genes. Diabetes Care 2011, 34, 1249–1257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Rippe, J.M.; Crossley, S.; Ringer, R. Obesity as a chronic disease: Modern medical and lifestyle management. J. Am. Diet. Assoc. 1998, 98, S9–S15. [Google Scholar] [CrossRef]
  5. 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, 9(th) edition. Diabetes Res. Clin. Pract. 2019, 157, 107843. [Google Scholar] [CrossRef] [Green Version]
  6. Blüher, M. Obesity: Global epidemiology and pathogenesis. Nat. Rev. Endocrinol. 2019, 15, 288–298. [Google Scholar] [CrossRef]
  7. Abdelaal, M.; le Roux, C.W.; Docherty, N.G. Morbidity and mortality associated with obesity. Ann. Transl. Med. 2017, 5, 161. [Google Scholar] [CrossRef] [Green Version]
  8. Samuel, S.M.; Varghese, E.; Varghese, S.; Busselberg, D. Challenges and perspectives in the treatment of diabetes associated breast cancer. Cancer. Treat. Rev. 2018, 70, 98–111. [Google Scholar] [CrossRef] [Green Version]
  9. Samuel, S.M.; Varghese, E.; Kubatka, P.; Triggle, C.R.; Büsselberg, D. Metformin: The Answer to Cancer in a Flower? Current Knowledge and Future Prospects of Metformin as an Anti-Cancer Agent in Breast Cancer. Biomolecules 2019, 9, 846. [Google Scholar] [CrossRef] [Green Version]
  10. Seifarth, C.; Schehler, B.; Schneider, H.J. Effectiveness of metformin on weight loss in non-diabetic individuals with obesity. Exp. Clin. Endocrinol. Diabetes 2013, 121, 27–31. [Google Scholar] [CrossRef] [Green Version]
  11. Malin, S.K.; Kashyap, S.R. Effects of metformin on weight loss: Potential mechanisms. Curr. Opin. Endocrinol. Diabetes Obes. 2014, 21, 323–329. [Google Scholar] [CrossRef] [PubMed]
  12. Yerevanian, A.; Soukas, A.A. Metformin: Mechanisms in Human Obesity and Weight Loss. Curr. Obes. Rep. 2019, 8, 156–164. [Google Scholar] [CrossRef] [PubMed]
  13. Hussain, M.; Atif, M.A.; Tunio, A.G.; Ali, B.; Akhtar, L.; Serwar, G. Effect of Sitagliptin On Glycemic Control, Body Weight, Blood Pressure And Serum Lipid Profile In Type 2 Diabetic Hyperlipidemic Patients. J. Ayub. Med. Coll. Abbottabad. 2016, 28, 369–372. [Google Scholar] [PubMed]
  14. Dar, S.; Tahrani, A.A.; Piya, M.K. The role of GLP-1 receptor agonists as weight loss agents in patients with and without type 2 diabetes. Pract. Diabetes 2015, 32, 297–300b. [Google Scholar] [CrossRef]
  15. Curtis, L.; Humayun, M.A.; Walker, J.; Hampton, K.; Partridge, H. Addition of SGLT2 inhibitor to GLP-1 agonist therapy in people with type 2 diabetes and suboptimal glycaemic control. Pract. Diabetes 2016, 33, 129–132. [Google Scholar] [CrossRef]
  16. Michałowska, J.; Miller-Kasprzak, E.; Bogdański, P. Incretin Hormones in Obesity and Related Cardiometabolic Disorders: The Clinical Perspective. Nutrients 2021, 13, 351. [Google Scholar] [CrossRef]
  17. Chaudhury, A.; Duvoor, C.; Reddy Dendi, V.S.; Kraleti, S.; Chada, A.; Ravilla, R.; Marco, A.; Shekhawat, N.S.; Montales, M.T.; Kuriakose, K.; et al. Clinical Review of Antidiabetic Drugs: Implications for Type 2 Diabetes Mellitus Management. Front. Endocrinol. 2017, 8, 6. [Google Scholar] [CrossRef] [Green Version]
  18. Higbea, A.M.; Duval, C.; Chastain, L.M.; Chae, J. Weight effects of antidiabetic agents. Expert Rev. Endocrinol. Metab. 2017, 12, 441–449. [Google Scholar] [CrossRef]
  19. Cheng, V.; Kashyap, S.R. Weight considerations in pharmacotherapy for type 2 diabetes. J. Obes. 2011, 2011, 984245. [Google Scholar] [CrossRef] [Green Version]
  20. Rosenstock, J.; Wysham, C.; Frías, J.P.; Kaneko, S.; Lee, C.J.; Fernández Landó, L.; Mao, H.; Cui, X.; Karanikas, C.A.; Thieu, V.T. Efficacy and safety of a novel dual GIP and GLP-1 receptor agonist tirzepatide in patients with type 2 diabetes (SURPASS-1): A double-blind, randomised, phase 3 trial. Lancet 2021, 398, 143–155. [Google Scholar] [CrossRef]
  21. Jastreboff, A.M.; Aronne, L.J.; Ahmad, N.N.; Wharton, S.; Connery, L.; Alves, B.; Kiyosue, A.; Zhang, S.; Liu, B.; Bunck, M.C.; et al. Tirzepatide Once Weekly for the Treatment of Obesity. N. Engl. J. Med. 2022, 387, 205–216. [Google Scholar] [CrossRef]
  22. Kannan, S.; Nasr, C. Should we be concerned about thyroid cancer in patients taking glucagon-like peptide 1 receptor agonists? Cleve. Clin. J. Med. 2015, 82, 142–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Bjerre Knudsen, L.; Madsen, L.W.; Andersen, S.; Almholt, K.; de Boer, A.S.; Drucker, D.J.; Gotfredsen, C.; Egerod, F.L.; Hegelund, A.C.; Jacobsen, H.; et al. Glucagon-like Peptide-1 receptor agonists activate rodent thyroid C-cells causing calcitonin release and C-cell proliferation. Endocrinology 2010, 151, 1473–1486. [Google Scholar] [CrossRef] [Green Version]
  24. Delfino, M.; Motola, D.; Benini, A.; Franzè, G.P.; Barotto, M.; Campi, A.; Monda, V.M. Incretin-mimetics associated pancreatitis: Evidence from the spontaneous adverse drug reactions reporting in Italy. Expert Opin. Drug Saf. 2014, 13, 151–156. [Google Scholar] [CrossRef]
  25. FDA Approves Lilly’s Mounjaro™ (Tirzepatide) Injection, the First and Only GIP and GLP-1 Receptor Agonist for the Treatment of Adults with Type 2 Diabetes. Eli Lilly and Company. Available online: https://investor.lilly.com/news-releases/news-release-details/fda-approves-lillys-mounjarotm-tirzepatide-injection-first-and (accessed on 13 May 2022).
  26. Pirro, V.; Roth, K.D.; Lin, Y.; Willency, J.A.; Milligan, P.L.; Wilson, J.M.; Ruotolo, G.; Haupt, A.; Newgard, C.B.; Duffin, K.L. Effects of Tirzepatide, a Dual GIP and GLP-1 RA, on Lipid and Metabolite Profiles in Subjects with Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2022, 107, 363–378. [Google Scholar] [CrossRef] [PubMed]
  27. Fisman, E.Z.; Tenenbaum, A. The dual glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) receptor agonist tirzepatide: A novel cardiometabolic therapeutic prospect. Cardiovasc. Diabetol. 2021, 20, 225. [Google Scholar] [CrossRef] [PubMed]
  28. Kalra, S.; Das, A.K.; Sahay, R.K.; Baruah, M.P.; Tiwaskar, M.; Das, S.; Chatterjee, S.; Saboo, B.; Bantwal, G.; Bhattacharya, S.; et al. Consensus Recommendations on GLP-1 RA Use in the Management of Type 2 Diabetes Mellitus: South Asian Task Force. Cardiovasc. Diabetol. 2019, 10, 1645–1717. [Google Scholar] [CrossRef] [Green Version]
  29. Jonik, S.; Marchel, M.; Grabowski, M.; Opolski, G.; Mazurek, T. Gastrointestinal Incretins-Glucose-Dependent Insulinotropic Polypeptide (GIP) and Glucagon-like Peptide-1 (GLP-1) beyond Pleiotropic Physiological Effects Are Involved in Pathophysiology of Atherosclerosis and Coronary Artery Disease-State of the Art. Biology 2022, 11, 288. [Google Scholar] [CrossRef]
  30. Zhao, H.J.; Jiang, X.; Hu, L.J.; Yang, L.; Deng, L.D.; Wang, Y.P.; Ren, Z.P. Activation of GLP-1 receptor enhances the chemosensitivity of pancreatic cancer cells. J. Mol. Endocrinol. 2020, 64, 103–113. [Google Scholar] [CrossRef] [PubMed]
  31. Zhao, H.; Wei, R.; Wang, L.; Tian, Q.; Tao, M.; Ke, J.; Liu, Y.; Hou, W.; Zhang, L.; Yang, J.; et al. Activation of glucagon-like peptide-1 receptor inhibits growth and promotes apoptosis of human pancreatic cancer cells in a cAMP-dependent manner. Am. J. Physiol. Endocrinol. Metab. 2014, 306, E1431–E1441. [Google Scholar] [CrossRef] [Green Version]
  32. Iwaya, C.; Nomiyama, T.; Komatsu, S.; Kawanami, T.; Tsutsumi, Y.; Hamaguchi, Y.; Horikawa, T.; Yoshinaga, Y.; Yamashita, S.; Tanaka, T.; et al. Exendin-4, a Glucagonlike Peptide-1 Receptor Agonist, Attenuates Breast Cancer Growth by Inhibiting NF-κB Activation. Endocrinology 2017, 158, 4218–4232. [Google Scholar] [CrossRef] [PubMed]
  33. Fidan-Yaylalı, G.; Dodurga, Y.; Seçme, M.; Elmas, L. Antidiabetic exendin-4 activates apoptotic pathway and inhibits growth of breast cancer cells. Tumour. Biol. 2016, 37, 2647–2653. [Google Scholar] [CrossRef] [PubMed]
  34. Zhao, W.; Zhang, X.; Zhou, Z.; Sun, B.; Gu, W.; Liu, J.; Zhang, H. Liraglutide inhibits the proliferation and promotes the apoptosis of MCF-7 human breast cancer cells through downregulation of microRNA-27a expression. Mol. Med. Rep. 2018, 17, 5202–5212. [Google Scholar] [CrossRef] [Green Version]
  35. Ligumsky, H.; Wolf, I.; Israeli, S.; Haimsohn, M.; Ferber, S.; Karasik, A.; Kaufman, B.; Rubinek, T. The peptide-hormone glucagon-like peptide-1 activates cAMP and inhibits growth of breast cancer cells. Breast Cancer Res. Treat. 2012, 132, 449–461. [Google Scholar] [CrossRef]
  36. Li, X.N.; Bu, H.M.; Ma, X.H.; Lu, S.; Zhao, S.; Cui, Y.L.; Sun, J. Glucagon-like Peptide-1 Analogues Inhibit Proliferation and Increase Apoptosis of Human Prostate Cancer Cells in vitro. Exp. Clin. Endocrinol. Diabetes 2017, 125, 91–97. [Google Scholar] [CrossRef] [PubMed]
  37. Nomiyama, T.; Kawanami, T.; Irie, S.; Hamaguchi, Y.; Terawaki, Y.; Murase, K.; Tsutsumi, Y.; Nagaishi, R.; Tanabe, M.; Morinaga, H.; et al. Exendin-4, a GLP-1 receptor agonist, attenuates prostate cancer growth. Diabetes 2014, 63, 3891–3905. [Google Scholar] [CrossRef] [Green Version]
  38. Tsutsumi, Y.; Nomiyama, T.; Kawanami, T.; Hamaguchi, Y.; Terawaki, Y.; Tanaka, T.; Murase, K.; Motonaga, R.; Tanabe, M.; Yanase, T. Combined Treatment with Exendin-4 and Metformin Attenuates Prostate Cancer Growth. PLoS ONE 2015, 10, e0139709. [Google Scholar] [CrossRef] [PubMed]
  39. Koehler, J.A.; Kain, T.; Drucker, D.J. Glucagon-like peptide-1 receptor activation inhibits growth and augments apoptosis in murine CT26 colon cancer cells. Endocrinology 2011, 152, 3362–3372. [Google Scholar] [CrossRef] [PubMed]
  40. Tong, G.; Peng, T.; Chen, Y.; Sha, L.; Dai, H.; Xiang, Y.; Zou, Z.; He, H.; Wang, S. Effects of GLP-1 Receptor Agonists on Biological Behavior of Colorectal Cancer Cells by Regulating PI3K/AKT/mTOR Signaling Pathway. Front. Pharmacol. 2022, 13, 901559. [Google Scholar] [CrossRef]
  41. Zhang, Y.; Xu, F.; Liang, H.; Cai, M.; Wen, X.; Li, X.; Weng, J. Exenatide inhibits the growth of endometrial cancer Ishikawa xenografts in nude mice. Oncol. Rep. 2016, 35, 1340–1348. [Google Scholar] [CrossRef] [Green Version]
  42. Zhang, Y.; Cheng, J.; Li, J.; He, J.; Li, X.; Xu, F. The GLP-1R Agonist Exendin-4 Attenuates Hyperglycemia-Induced Chemoresistance in Human Endometrial Cancer Cells Through ROS-Mediated Mitochondrial Pathway. Front. Oncol. 2021, 11, 793530. [Google Scholar] [CrossRef] [PubMed]
  43. Kanda, R.; Hiraike, H.; Wada-Hiraike, O.; Ichinose, T.; Nagasaka, K.; Sasajima, Y.; Ryo, E.; Fujii, T.; Osuga, Y.; Ayabe, T. Expression of the glucagon-like peptide-1 receptor and its role in regulating autophagy in endometrial cancer. BMC Cancer 2018, 18, 657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Kosowska, A.; Gallego-Colon, E.; Garczorz, W.; Kłych-Ratuszny, A.; Aghdam, M.R.F.; Woz Niak, M.; Witek, A.; Wróblewska-Czech, A.; Cygal, A.; Wojnar, J.; et al. Exenatide modulates tumor-endothelial cell interactions in human ovarian cancer cells. Endocr. Connect. 2017, 6, 856–865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. He, W.; Yu, S.; Wang, L.; He, M.; Cao, X.; Li, Y.; Xiao, H. Exendin-4 inhibits growth and augments apoptosis of ovarian cancer cells. Mol. Cell Endocrinol. 2016, 436, 240–249. [Google Scholar] [CrossRef]
  46. Mao, D.; Cao, H.; Shi, M.; Wang, C.C.; Kwong, J.; Li, J.J.X.; Hou, Y.; Ming, X.; Lee, H.M.; Tian, X.Y.; et al. Increased co-expression of PSMA2 and GLP-1 receptor in cervical cancer models in type 2 diabetes attenuated by Exendin-4: A translational case-control study. EBioMedicine 2021, 65, 103242. [Google Scholar] [CrossRef]
  47. Palanissami, G.; Paul, S.F.D. RAGE and Its Ligands: Molecular Interplay Between Glycation, Inflammation, and Hallmarks of Cancer-a Review. Horm. Cancer 2018, 9, 295–325. [Google Scholar] [CrossRef]
  48. Logsdon, C.D.; Fuentes, M.K.; Huang, E.H.; Arumugam, T. RAGE and RAGE ligands in cancer. Curr. Mol. Med. 2007, 7, 777–789. [Google Scholar] [CrossRef]
  49. El-Far, A.H.; Sroga, G.; Jaouni, S.K.A.; Mousa, S.A. Role and Mechanisms of RAGE-Ligand Complexes and RAGE-Inhibitors in Cancer Progression. Int. J. Mol. Sci. 2020, 21, 3613. [Google Scholar] [CrossRef]
  50. Puddu, A.; Mach, F.; Nencioni, A.; Viviani, G.L.; Montecucco, F. An emerging role of glucagon-like peptide-1 in preventing advanced-glycation-end-product-mediated damages in diabetes. Mediat. Inflamm. 2013, 2013, 591056. [Google Scholar] [CrossRef] [Green Version]
  51. Allam, M.M.; El Gazzar, W.B. Exendin-4, a glucagon-like peptide-1 receptor agonist downregulates hepatic receptor for advanced glycation end products in non-alcoholic steatohepatitis rat model. Arch. Physiol. Biochem. 2018, 124, 10–17. [Google Scholar] [CrossRef]
  52. Yi, B.; Hu, X.; Wen, Z.; Zhang, T.; Cai, Y. Exendin-4, a glucagon-like peptide-1 receptor agonist, inhibits hyperglycemia-induced apoptosis in myocytes by suppressing receptor for advanced glycation end products expression. Exp. Ther. Med. 2014, 8, 1185–1190. [Google Scholar] [CrossRef] [PubMed]
  53. Chang, J.T.; Liang, Y.J.; Hsu, C.Y.; Chen, C.Y.; Chen, P.J.; Yang, Y.F.; Chen, Y.L.; Pei, D.; Chang, J.B.; Leu, J.G. Glucagon-like peptide receptor agonists attenuate advanced glycation end products-induced inflammation in rat mesangial cells. BMC Pharmacol. Toxicol. 2017, 18, 67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Körner, M.; Christ, E.; Wild, D.; Reubi, J.C. Glucagon-like peptide-1 receptor overexpression in cancer and its impact on clinical applications. Front. Endocrinol. 2012, 3, 158. [Google Scholar] [CrossRef] [Green Version]
  55. Körner, M.; Stöckli, M.; Waser, B.; Reubi, J.C. GLP-1 receptor expression in human tumors and human normal tissues: Potential for in vivo targeting. J. Nucl. Med. 2007, 48, 736–743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Gasbjerg, L.S.; Gabe, M.B.N.; Hartmann, B.; Christensen, M.B.; Knop, F.K.; Holst, J.J.; Rosenkilde, M.M. Glucose-dependent insulinotropic polypeptide (GIP) receptor antagonists as anti-diabetic agents. Peptides 2018, 100, 173–181. [Google Scholar] [CrossRef] [PubMed]
  57. Chandarana, R.; D’Souza, J.S.; Coutinho, E.C. Glucose-Dependent Insulinotropic Polypeptide Receptor (GIPR). In Encyclopedia of Signaling Molecules; Choi, S., Ed.; Springer: New York, NY, USA, 2012; pp. 773–779. [Google Scholar] [CrossRef]
  58. Sen, S.; He, Y.; Koya, D.; Kanasaki, K. Cancer biology in diabetes. J. Diabetes Investig. 2014, 5, 251–264. [Google Scholar] [CrossRef] [PubMed]
  59. Prabakaran, D.; Wang, B.; Feuerstein, J.D.; Sinclair, J.A.; Bijpuria, P.; Jepeal, L.I.; Wolfe, M.M. Glucose-dependent insulinotropic polypeptide stimulates the proliferation of colorectal cancer cells. Regul. Pept. 2010, 163, 74–80. [Google Scholar] [CrossRef] [Green Version]
  60. Denker, P.S.; Dimarco, P.E. Exenatide (Exendin-4)–Induced Pancreatitis: A case report. Diabetes Care 2006, 29, 471. [Google Scholar] [CrossRef] [Green Version]
  61. Cure, P.; Pileggi, A.; Alejandro, R. Exenatide and Rare Adverse Events. N. Engl. J. Med. 2008, 358, 1969–1972. [Google Scholar] [CrossRef]
  62. Tripathy, N.R.; Basha, S.; Jain, R.; Shetty, S.; Ramachandran, A. Exenatide and acute pancreatitis. J. Assoc. Physicians India 2008, 56, 987–988. [Google Scholar]
  63. Dolan, R.D.; Bazarbashi, A.N.; Lo, A.; Smith, B.N. Liraglutide-Induced Hemorrhagic Pancreatitis in a Nondiabetic Patient. ACG Case Rep. J. 2020, 7, e00380. [Google Scholar] [CrossRef] [PubMed]
  64. Fatakhova, K.; Rahman, A.; Al-Naqeeb, G.; Al Yassin, S.; Alqaisi, S.; Ramdass, A. 1311 Acute Pancreatitis in a Patient Using Liraglutide for Weight Loss. Off. J. Am. Coll. Gastroenterol. 2019, 114, S728–S729. [Google Scholar] [CrossRef]
  65. Gameil, M.A.; Elsebaie, A.H. Mildly symptomatic liraglutide-induced acute pancreatitis in a patient with type 2 diabetes mellitus: A case report. Egypt. J. Intern. Med. 2020, 32, 26. [Google Scholar] [CrossRef]
  66. Vangoitsenhoven, R.; Mathieu, C.; Van der Schueren, B. GLP1 and cancer: Friend or foe? Endocr. Relat. Cancer 2012, 19, F77–F88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Buse, J.B.; Bethel, M.A.; Green, J.B.; Stevens, S.R.; Lokhnygina, Y.; Aschner, P.; Grado, C.R.; Tankova, T.; Wainstein, J.; Josse, R.; et al. Pancreatic Safety of Sitagliptin in the TECOS Study. Diabetes Care 2017, 40, 164–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Elashoff, M.; Matveyenko, A.V.; Gier, B.; Elashoff, R.; Butler, P.C. Pancreatitis, Pancreatic, and Thyroid Cancer with Glucagon-Like Peptide-1–Based Therapies. Gastroenterology 2011, 141, 150–156. [Google Scholar] [CrossRef] [Green Version]
  69. Vujasinovic, M.; Dugic, A.; Maisonneuve, P.; Aljic, A.; Berggren, R.; Panic, N.; Valente, R.; Pozzi Mucelli, R.; Waldthaler, A.; Ghorbani, P.; et al. Risk of Developing Pancreatic Cancer in Patients with Chronic Pancreatitis. J. Clin. Med. 2020, 9, 3720. [Google Scholar] [CrossRef]
  70. Lowenfels, A.B.; Maisonneuve, P.; Cavallini, G.; Ammann, R.W.; Lankisch, P.G.; Andersen, J.R.; Dimagno, E.P.; Andren-Sandberg, A.; Domellof, L. Pancreatitis and the Risk of Pancreatic Cancer. N. Engl. J. Med. 1993, 328, 1433–1437. [Google Scholar] [CrossRef]
  71. Jura, N.; Archer, H.; Bar-Sagi, D. Chronic pancreatitis, pancreatic adenocarcinoma and the black box in-between. Cell Res. 2005, 15, 72–77. [Google Scholar] [CrossRef] [Green Version]
  72. Rebours, V.; Boutron-Ruault, M.C.; Schnee, M.; Férec, C.; Le Maréchal, C.; Hentic, O.; Maire, F.; Hammel, P.; Ruszniewski, P.; Lévy, P. The natural history of hereditary pancreatitis: A national series. Gut 2009, 58, 97. [Google Scholar] [CrossRef] [Green Version]
  73. Nauck, M.A.; Friedrich, N. Do GLP-1-based therapies increase cancer risk? Diabetes Care 2013, 36, S245–S252. [Google Scholar] [CrossRef] [PubMed]
  74. Boniol, M.; Franchi, M.; Bota, M.; Leclercq, A.; Guillaume, J.; van Damme, N.; Corrao, G.; Autier, P.; Boyle, P. Incretin-Based Therapies and the Short-term Risk of Pancreatic Cancer: Results from Two Retrospective Cohort Studies. Diabetes Care 2018, 41, 286–292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Butler, A.E.; Campbell-Thompson, M.; Gurlo, T.; Dawson, D.W.; Atkinson, M.; Butler, P.C. Marked expansion of exocrine and endocrine pancreas with incretin therapy in humans with increased exocrine pancreas dysplasia and the potential for glucagon-producing neuroendocrine tumors. Diabetes 2013, 62, 2595–2604. [Google Scholar] [CrossRef] [Green Version]
  76. Mortazavi, M.; Moosavi, F.; Martini, M.; Giovannetti, E.; Firuzi, O. Prospects of targeting PI3K/AKT/mTOR pathway in pancreatic cancer. Crit. Rev. Oncol. Hematol. 2022, 176, 103749. [Google Scholar] [CrossRef] [PubMed]
  77. Furukawa, T. Impacts of activation of the mitogen-activated protein kinase pathway in pancreatic cancer. Front. Oncol. 2015, 5, 23. [Google Scholar] [CrossRef] [PubMed]
  78. Yu, J.H.; Kim, H. Role of janus kinase/signal transducers and activators of transcription in the pathogenesis of pancreatitis and pancreatic cancer. Gut Liver 2012, 6, 417–422. [Google Scholar] [CrossRef] [Green Version]
  79. Collisson, E.A.; Trejo, C.L.; Silva, J.M.; Gu, S.; Korkola, J.E.; Heiser, L.M.; Charles, R.P.; Rabinovich, B.A.; Hann, B.; Dankort, D.; et al. A central role for RAF→MEK→ERK signaling in the genesis of pancreatic ductal adenocarcinoma. Cancer. Discov. 2012, 2, 685–693. [Google Scholar] [CrossRef] [Green Version]
  80. Vigneri, P.G.; Tirrò, E.; Pennisi, M.S.; Massimino, M.; Stella, S.; Romano, C.; Manzella, L. The Insulin/IGF System in Colorectal Cancer Development and Resistance to Therapy. Front. Oncol. 2015, 5, 230. [Google Scholar] [CrossRef] [Green Version]
  81. Chettouh, H.; Lequoy, M.; Fartoux, L.; Vigouroux, C.; Desbois-Mouthon, C. Hyperinsulinaemia and insulin signalling in the pathogenesis and the clinical course of hepatocellular carcinoma. Liver Int. 2015, 35, 2203–2217. [Google Scholar] [CrossRef] [Green Version]
  82. Madsen, L.W.; Knauf, J.A.; Gotfredsen, C.; Pilling, A.; Sjögren, I.; Andersen, S.; Andersen, L.; de Boer, A.S.; Manova, K.; Barlas, A.; et al. GLP-1 receptor agonists and the thyroid: C-cell effects in mice are mediated via the GLP-1 receptor and not associated with RET activation. Endocrinology 2012, 153, 1538–1547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Rosol, T.J. On-target effects of GLP-1 receptor agonists on thyroid C-cells in rats and mice. Toxicol. Pathol. 2013, 41, 303–309. [Google Scholar] [CrossRef] [PubMed]
  84. Regazzo, D.; Bertazza, L.; Galletta, E.; Barollo, S.; Mondin, A.; Zovato, S.; Iacobone, M.; Zilio, E.; Scaroni, C.; Radu, C.M.; et al. The GIP/GIPR axis in medullary thyroid cancer: Clinical and molecular findings. Endocr. Relat. Cancer 2022, 29, 273–284. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Possible target sites of GLP-1R/GIPR agonists and possibly TRZD. The incretin hormones GLP-1 and GIP secreted by the gastrointestinal tract have a wide array of target tissue that modulate various organ system functions. GLP-1R/GIPR agonists and possibly TRZD play several beneficial and protective functions in the various organ systems. The green upward arrow indicates in-crease/activation, while the red downward arrow indicates decrease/inhibition. EC = endothelial cell, NASH = non-alcoholic steatohepatitis and VLDL = very low-density lipoprotein. Created with Biorender.com (accessed on 14 September 2022).
Figure 1. Possible target sites of GLP-1R/GIPR agonists and possibly TRZD. The incretin hormones GLP-1 and GIP secreted by the gastrointestinal tract have a wide array of target tissue that modulate various organ system functions. GLP-1R/GIPR agonists and possibly TRZD play several beneficial and protective functions in the various organ systems. The green upward arrow indicates in-crease/activation, while the red downward arrow indicates decrease/inhibition. EC = endothelial cell, NASH = non-alcoholic steatohepatitis and VLDL = very low-density lipoprotein. Created with Biorender.com (accessed on 14 September 2022).
Biomolecules 12 01580 g001
Figure 2. Possible pro-oncogenic effect of tirzepatide (TRZD) on the pancreas, breast, colorectal, and liver cancers. TRZD, possibly via GLP-1R or GIPR activation, leads to intracellular Ca2+-dependent release of insulin from the pancreatic β-cells. The GLP-1R activation can also lead to protein kinase A-dependent gene transcription leading to the synthesis and release of insulin-like growth factor 1 or 2 (IGF 1/2) and insulin-like growth factor binding protein 3 (IGF-BP3). Insulin and IGF 1/2 via the insulin receptor (IR) and the IGF receptor 1 (IGF-1R) in turn activate the PI3K/Akt/mTOR, Ras-Raf-ERK, MAPK, and JAK-STAT pathway, which supports several oncogenic and cancer progression traits such as resisting cell death, induction of angiogenesis, sustained proliferative signals, tumor inflammation, and activation of invasion and metastasis in target tissues (pancreas, breast, colorectum and liver). The black arrows indicate the pathway and flow of components involved in the pathway. The green upward arrow indicates an increase, while the red downward arrow indicates a decrease. Created with Biorender.com (accessed on 15 September 2022).
Figure 2. Possible pro-oncogenic effect of tirzepatide (TRZD) on the pancreas, breast, colorectal, and liver cancers. TRZD, possibly via GLP-1R or GIPR activation, leads to intracellular Ca2+-dependent release of insulin from the pancreatic β-cells. The GLP-1R activation can also lead to protein kinase A-dependent gene transcription leading to the synthesis and release of insulin-like growth factor 1 or 2 (IGF 1/2) and insulin-like growth factor binding protein 3 (IGF-BP3). Insulin and IGF 1/2 via the insulin receptor (IR) and the IGF receptor 1 (IGF-1R) in turn activate the PI3K/Akt/mTOR, Ras-Raf-ERK, MAPK, and JAK-STAT pathway, which supports several oncogenic and cancer progression traits such as resisting cell death, induction of angiogenesis, sustained proliferative signals, tumor inflammation, and activation of invasion and metastasis in target tissues (pancreas, breast, colorectum and liver). The black arrows indicate the pathway and flow of components involved in the pathway. The green upward arrow indicates an increase, while the red downward arrow indicates a decrease. Created with Biorender.com (accessed on 15 September 2022).
Biomolecules 12 01580 g002
Figure 3. Possible pro-oncogenic effect of tirzepatide (TRZD) on the thyroid gland. TRZD via GLP-1R or GIPR activation leads to an increase in the intracellular levels of cAMP and protein kinase A activation, which drives the expression and secretion of calcitonin. The sustained activation of these receptors and release of calcitonin is accompanied by C-cell hyperplasia and transformation into C-cell adenomas and MTC. The black arrows indicate the pathway and flow of components involved in the pathway. The green upward arrow indicates an increase, while the red downward arrow indicates a decrease. Created with Biorender.com (accessed on 13 September 2022).
Figure 3. Possible pro-oncogenic effect of tirzepatide (TRZD) on the thyroid gland. TRZD via GLP-1R or GIPR activation leads to an increase in the intracellular levels of cAMP and protein kinase A activation, which drives the expression and secretion of calcitonin. The sustained activation of these receptors and release of calcitonin is accompanied by C-cell hyperplasia and transformation into C-cell adenomas and MTC. The black arrows indicate the pathway and flow of components involved in the pathway. The green upward arrow indicates an increase, while the red downward arrow indicates a decrease. Created with Biorender.com (accessed on 13 September 2022).
Biomolecules 12 01580 g003
Figure 4. Outstanding questions regarding the use of tirzepatide (TRZD) in diabetic/obese or diabetic-/obese-cancer patients. Created with Biorender.com (accessed on 15 September 2022).
Figure 4. Outstanding questions regarding the use of tirzepatide (TRZD) in diabetic/obese or diabetic-/obese-cancer patients. Created with Biorender.com (accessed on 15 September 2022).
Biomolecules 12 01580 g004
Table 1. Status of tirzepatide administration-related clinical trials. (Search keywords: Diabetes/Obesity/Cancer + Tirzepatide) (https://clinicaltrials.gov/ (accessed on 12 October 2022).
Table 1. Status of tirzepatide administration-related clinical trials. (Search keywords: Diabetes/Obesity/Cancer + Tirzepatide) (https://clinicaltrials.gov/ (accessed on 12 October 2022).
Serial No.ConditionTotal Registered TrialsCompletedActive, Not RecruitingActive, Not Yet RecruitingActive, Recruiting
1Diabetes2916724
2Obesity142624
3Cancers---- -
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Samuel, S.M.; Varghese, E.; Kubatka, P.; Büsselberg, D. Tirzepatide—Friend or Foe in Diabetic Cancer Patients? Biomolecules 2022, 12, 1580. https://doi.org/10.3390/biom12111580

AMA Style

Samuel SM, Varghese E, Kubatka P, Büsselberg D. Tirzepatide—Friend or Foe in Diabetic Cancer Patients? Biomolecules. 2022; 12(11):1580. https://doi.org/10.3390/biom12111580

Chicago/Turabian Style

Samuel, Samson Mathews, Elizabeth Varghese, Peter Kubatka, and Dietrich Büsselberg. 2022. "Tirzepatide—Friend or Foe in Diabetic Cancer Patients?" Biomolecules 12, no. 11: 1580. https://doi.org/10.3390/biom12111580

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop