Abstract
Small molecule kinase inhibitors (SMKIs) are a class of therapeutic drugs that target protein kinases in diseases such as cancer. SMKIs are often designed to inhibit kinases involved in cell proliferation, but these drugs alter cell metabolism and the endocrine control of organismal metabolism. SMKI treatment in diabetic cancer patients reveals that certain SMKIs improve blood glucose levels and can mitigate insulin dependence or diabetic medication requirements in both type 1 diabetes (T1D) and type 2 diabetes (T2D). Certain SMKIs can preserve functional β-cell mass and increase insulin secretion or insulin sensitivity. It is not yet clear why different SMKIs can have opposing effects on insulin and blood glucose. Understanding the therapeutic effects of these drugs in T1D and T2D is complicated by overlapping off-target effects of SMKIs. The potency of inhibition of the intended protein kinase and inhibition of multiple off-target kinases may underpin conflicting reports of how certain SMKIs alter blood glucose and insulin. We summarize the effects of SMKIs on the intended and off-target kinases that can alter blood glucose and insulin, including c-Abl, c-Kit, EGFR, and VEGF. Inhibition of PDGFRβ consistently lowers blood glucose in T1D and T2D. The effects of SMKIs on the kinases that regulate immune pathways, such as BTK and RIPKs, mediate many of the diverse effects of these drugs on metabolism. We highlight that inhibition of RIPK2 by SMKIs is a central node in metabolism that influences key metabolic pathways including lipolysis, blood glucose control, insulin secretion, and insulin resistance.
Protein kinases and small molecule kinase inhibitors
More than 500 protein kinases have been confirmed in the human genome, and these kinases regulate cellular signaling cascades that control fundamental cell function, including proliferation, differentiation, survival, metabolism, and immunity (Adams 2001, Manning et al. 2002). Protein kinases regulate the biological activity of other proteins by transferring a γ-phosphate group from an ATP molecule to a specific serine/threonine residue or a tyrosine residue on the substrate protein (Dhanasekaran & Premkumar Reddy 1998, Manning et al. 2002). Given the critical role of protein kinases in regulating almost all cellular activities and functions, it follows that dysregulation of specific protein kinases is implicated in numerous disease states (Lahiry et al. 2010), and pharmacological inhibition of aberrant kinase activity is a promising drug target for disease treatment. The last 2 decades have seen a rapid and widespread use of more than 60 small molecule kinase inhibitors (SMKIs) approved for clinical use in cancerous malignancies, and many SMKIs are being investigated as potential therapeutic agents in autoimmune and inflammatory conditions, including asthma, psoriasis, inflammatory bowel disease, peritonitis, multiple sclerosis, pulmonary fibrosis, and type 1 and type 2 diabetes (T1D, T2D) (Patterson et al. 2014). Several of these agents have already been approved for use in several non-malignant conditions including dermatitis, ulcerative colitis, rheumatoid arthritis, and graft versus host disease (Roskoski 2020). SMKIs are often used as adjuvants in chemotherapy, but theyare not cytotoxic agents. In general, SMKIs are well-tolerated and widely used. SMKIs in clinical settings have yielded extensive, pre-existing safety and side-effect profiles for many of these drugs (Roskoski 2020).
Kinase inhibitors function by blocking the transfer of the γ-phosphate group from the ATP molecule, either by directly binding to the ATP-binding site (type I inhibitors) or by binding to an allosteric site of the kinase that prevents access to the ATP-binding site (type II–IV inhibitors). Due to the similar 3D structure and highly conserved ATP-binding pocket across the entirety of the mammalian kinome, specificity issues and inhibition of off-target kinases are of major concern in the development and use of SMKIs. Despite their intended selectivity and specificity, clinically approved kinase inhibitors have many off-target effects and inhibit a range of kinases (Davis et al. 2011). The dose is a critical determinant of off-target effects of SMKIs. The effective (i.e. bioavailable) dose is influenced by the metabolism of SMKIs. Most kinase inhibitors are rapidly absorbed and reach peak plasma concentration within 3–7 h after an oral dose. Food consumption only marginally increases the extent of or does not significantly affect the bioavailability of most kinase inhibitors (van Erp et al. 2009). Kinase inhibitors are primarily metabolized by cytochrome P450 (CYP) 3A4 with other CYP enzymes playing a secondary role in the metabolism of some inhibitors. Kinase inhibitors are predominantly excreted in feces, and a minor amount is also excreted in urine (van Erp et al. 2009). The purpose of this review is to highlight how SMKIs that are often designed for cancer treatment can alter blood glucose and insulin. We apologize that not all relevant literature could be included in this review. We selected specific references and focused on literature that highlighted the connection between SMKIs and blood glucose and insulin and also uncovered the mechanisms of action linking metabolism and SMKIs in humans and in pre-clinical studies.
SMKIs have opposing effects on glucose and insulin in type 1 and type 2 diabetes
Both T1D and T2D are diseases defined by elevated blood glucose levels. T1D is caused by autoimmune destruction of pancreatic β-cells, leading to inability to produce and secrete insulin to control blood glucose. In contrast, the early stages of prediabetes that occur before overt T2D are characterized by increased insulin secretion, insulin resistance, and close to normal blood glucose levels. Eventual decline in insulin production and continued insulin resistance equate to hyperglycemia in overt T2D. Despite the differences in the underlying pathophysiology of T1D and T2D, a significant number of clinical reports have demonstrated a decreased need for exogenous insulin or diabetic medications in both T1D and T2D cancer patients receiving SMKI therapy (Table 1). These initial clinical observations spurred interest in re-tasking clinically approved SMKIs for use in diabetes. The underlying mechanisms that mediate the glycemic effects of SMKIs have been studied in vitro and in various in vivo rodent models of T1D and T2D.
Clinical evidence of KIs altering blood glucose or insulin in diabetic cancer patients.
Kinase inhibitor | Summary of effects on blood glucose or insulin | References |
---|---|---|
Brigatinib | ↑ Hyperglycemia in human patients | ARIAD Pharmaceuticals 2017 |
Ceritinib | ↑ Hyperglycemia in human non-diabetic and T2D patients | Novartis Pharmaceuticals Corporation 2014, Khozin et al. 2015, Sakuma et al. 2019, Miyoshi et al. 2021 |
Dasatinib | ↓ Fasting blood glucose and ↓ insulin in human T2D patients | Breccia et al. 2008, Agostino et al. 2011, Ono et al. 2012, Lundholm & Charnogursky 2020, Salaami et al. 2021 |
Erlotinib | ↓ Fasting blood glucose and ↓ insulin in human T2D patients | Costa & Huberman 2006, Brooks 2012 |
Imatinib | ↓ Fasting blood glucose and ↓ insulin in human T1D and T2D diabetic patients | Breccia et al. 2004, Veneri et al. 2005, Agostino et al. 2011, Salaroli et al. 2012 |
Neratinib | ↓ Blood glucose in human T2D patient ↓ Glycemia and ↑ insulin secretion in mouse T1D model |
Ardestani et al. 2019, Angelis et al. 2022 |
Nilotinib | ↑ Blood glucose and ↑ insulin levels in human non-diabetic patients | Saglio et al. 2010, Zdenek et al. 2014 |
↑ Blood glucose and ↓ insulin in human T2D patient | Ito et al. 2013 | |
Pazopanib | ↓ Blood glucose in human T2D patient | Böhm et al. 2010 |
Rociletinib | ↑ Blood glucose levels in human non-diabetic and diabetic patients | Sequist et al. 2015 |
Sorafenib | ↓ Blood glucose levels in human non-diabetic and T2D patients | Agostino et al. 2011, Holstein et al. 2013, Di Costanzo et al. 2017 |
Sunitinib | ↓ Blood glucose and ↓ insulin in human non-diabetic and T1D/T2D patients | Billemont et al. 2008, Templeton et al. 2008, Agostino et al. 2011, Oh et al. 2012, Chen et al. 2013b , Demirci et al. 2014, Huda et al. 2014, Tyrrell & Pwint 2014 |
There is no unifying theory explaining how SMKIs alter blood glucose or insulin, and this is further complicated by conflicting reports of specific SMKIs promoting hyperglycemia or hypoglycemia in different patients or experiment models. For example, there are many reports of the SMKI dasatinib lowering blood glucose in T1D, T2D, and non-diabetic patients (Breccia et al. 2008, Agostino et al. 2011, Ono et al. 2012, Yu et al. 2019, Lundholm & Charnogursky 2020, Salaami et al. 2021). Dasatinib was also reported to promote hyperglycemia in a cohort of non-diabetic patients and in a mouse model of diet-induced obesity (Sylow et al. 2016, Yu et al. 2019). In contrast, hyperglycemia is one of the main dose-limiting side-effects for some SMKIs, including nilotinib, rociletinib, and ceritinib, which has been attributed to impaired pancreatic β-cell insulin secretion, or inhibition of the insulin receptor (IR) and development of insulin resistance in human patients (Novartis Pharmaceuticals Corporation 2014, Zdenek et al. 2014, Sequist et al. 2015, Sakuma et al. 2019). SMKIs have also been reported to have opposing effects on blood insulin levels. Some clinical reports demonstrate increased insulin or c-peptide levels during dasatinib, imatinib, sunitinib, and nilotinib therapy and increased β-cell insulin secretion in vitro, while conflicting reports in the literature suggest nilotinib can also impair insulin secretion in humans (Haap et al. 2007, Ono et al. 2012, Zdenek et al. 2014, Xia et al. 2014, Lutz et al. 2017, Li et al. 2018, Samaha et al. 2019). A schematic summary of the main glycemic and insulinemic effects of some of the most widely used, clinically approved SMKIs is presented in Fig. 1.
Targets regulating glycemic effects of SMKIs
It is difficult to ascertain the mechanism of action underlying the ability of a specific SMKIs to regulate blood glucose due to numerous off-target actions on multiple protein kinases. For example, imatinib is designed against, and is considered highly specific for, non-receptor tyrosine kinase abelson (c-Abl) kinase, but imatinib also inhibits numerous other kinases to a comparable degree, including c-Kit and platelet-derived growth factor receptor β (PDGFRβ). Common overlapping targets for many of the SMKIs that have reported effects on blood glucose and insulin dynamics include cAbl, c-Kit, receptor-interacting serine/threonine protein kinase 2 (RIPK2), EGF receptor, ribosomal protein S6 kinase beta-1 (S6K1), PDGFRβ, c-June N-terminal kinase 1 (JNK1), and the insulin receptor (InsR) (Fig. 2). A number of these SMKI targets has been implicated in metabolic homeostasis, but the key target kinases and molecular signals responsible for each specific SMKI’s ability to cause changes in blood glucose or insulin are unknown and dependent on the effective dose. Using publicly available kinase inhibition assay data from the Harvard Medical School LINCS Center database, we have compiled a table describing many clinically approved SMKIs and their targets relevant to metabolism and the relative degree of inhibition (Table 2). The potential metabolic role of each of these target kinases is discussed in detail below.
Clinically approved SMKIs inhibit multiple kinase targets. % values correspond to % inhibition at specified SMKI concentration. For SMKIs where no concentration is listed, nanomolar (nM) values given represent Kd50 values.
SMKI | 1˚ target | RIPK2 | EGFR | c-Kit | cABL | p-cABL | PDGFRβ | S6K1 | JNK1 | InsR |
---|---|---|---|---|---|---|---|---|---|---|
Abemaciclib (1 μM) | CDK4/6 100% | 3% | 0% | 0% | 8% | 2% | 4% | 44% | 96% | 0% |
Afatinib | ErbB1/2/4 0.3 nM | 2700 nM | 0.3 nM | – | 1300 nM | 570 nM | – | – | – | – |
Baricitinib (10 μM) | JAK1/2 100% | 1% | 13% | 44% | 81% | 81% | 83% | 56% | 64% | 33% |
Bosutinib | BCR-Abl 0.12 nM | 3700 nM | 35 nM | 420 nM | 0.12 nM | 0.06 nM | 200 nM | 660 nM | – | – |
Ceritinib (1 μM) | ALK 100% | 7% | 28% | 13% | 10% | 14% | 11% | 0% | 41% | 98% |
Crizotinib (10 μM) | ALK, ROS1 91%, 99% | 89% | 18% | 43% | 100% | 100% | 56% | 98% | 0% | 99% |
Dabrafenib (10 μM) | B-raf 100% | 97% | 24% | 96% n | 99% | 79% | 86% | 91% | 94% | 22% |
Dasatinib | BCR-Abl 0.03 nM | 31 nM | 120 nM | 0.81 nM | 0.03 nM | 0.05 nM | 0.63 nM | – | – | – |
Erlotinib | EGFR 0.7 nM | 680 nM | 0.7 nM | 1700 nM | 330 nM | 76 nM | 1400 nM | – | – | – |
Fedratinib | JAK2 1.1 nM | 6100 nM | 3400 nM | 230 nM | 180 nM | 44 nM | 45 nM | 2000 nM | 260 nM | 850 nM |
Gefitinib | EGFR 1.0 nM | 530 nM | 1.0 nM | – | 480 nM | 2200 nM | – | – | – | – |
Ibrutinib (1 μM) | BTK 100% | 95% | 98% | 56% | 93% | 46% | 67% | 37% | 28% | 63% |
Imatinib | BCR-Abl 1.1 nM | – | – | 13 nM | 1.1 nM | 21 nM | 14 nM | – | 5000 nM | – |
Lapatinib (10 μM) | ErbB1/2 100% | 94% | 100% | 12% | 0% | 0% | 0% | 20% | 0% | 0% |
Neratinib | ErbB2 6 nM | – | 1.1 nM | – | – | – | – | – | 3300 nM | 3000 nM |
Nilotinib | BCR-Abl 10 nM | – | – | 29 nM | 10 nM | 13 nM | 73 nM | – | 450 nM | – |
Nintedanib | FGFR1/2/3, 92–350 nM | – | – | 5.7 nM | 230 nM | 64 nM | 15 nM | 190 nM | 630 nM | 24 nM |
Palbociclib (10 μM) | CDK4/6 98–100% | 0% | 10% | 40% | 47% | 51% | 71% | 44% | 98% | 33% |
Pazopanib | VEGFR1/2/3 14 nM | 580 nM | – | 2.8 nM | 620 nM | 650 nM | 2 nM | – | 2000 nM | – |
Ribociclib (1 μM) | CDK4/6 99–100% | 0% | 0% | 0% | 17% | 15% | 6% | 22% | 0% | 0% |
Ruxolitinib | JAK1/2/3 0.1–3 nM | – | – | – | 3700 nM | – | – | – | 4800 nM | – |
Sorafenib (10 μM) | VEGFR1/2/3 99% | 82% | 15% | 100% | 99% | 75% | 100% | 37% | 0% | 0% |
Sunitinib | VEGFR2 1.5 nM | – | – | 0.4 nM | 270 nM | 150 nM | 0.1 nM | 48 nM | – | 500 nM |
Tofacitinib | JAK3 0.2 nM | – | – | – | – | – | – | – | – | – |
Vandetanib | VEGFR2 820 nM | 4.6 nM | 9.5 nM | 260 nM | 48 nM | 16 nM | 88 nM | 1600 nM | – | – |
Vemurafenib (10 μM) | B-Raf 100% | 96% | 30% | 99% | 97% | 43% | 99% | 14% | 82% | 22% |
c-Abl
Expressed in the majority of cells, c-Abl is a critical regulator of cell growth, motility, cytoskeleton dynamics, receptor endocytosis, DNA repair, cell survival, and autophagy (Hantschel & Superti-Furga 2004). The focus of c-Abl research has largely been in human cancers, including the oncogenic BCR-Abl fusion, which encodes a highly active kinase and contributes to chronic myeloid leukemia (CML) and acute lymphoblastic leukemia (ALL) (Hantschel & Superti-Furga 2004). Imatinib, nilotinib, dasatinib, bosubtinib, and ponatinib represent the main SMKIs developed against c-Abl/BCR-Abl for use in CML and ALL (Lindholm et al. 2016). Interestingly, c-Abl is highly expressed in the s.c. adipose tissue of obese humans and mice fed a high fat diet (HFD), and in vivo data suggest adipose tissue-specific deletion of c-Abl in HFD-fed mice mitigated weight gain, hyperglycemia, and hyperinsulinemia (Wu et al. 2017). Consistent with these reports of reduced HFD-induced weight gain, c-Abl has been implicated as a regulator of adipocyte differentiation via PPARβ2 in in vitro studies (Keshet et al. 2014). In contrast, in vitro work has shown that c-Abl is induced by high glucose levels; however, this can be reversed by c-Abl inhibition via administration of imatinib (Jia et al. 2008, Xia et al. 2014). The results are congruent with in vivo findings from our lab demonstrating that imatinib treatment in obese mice lowers hyperglycemia but induces hyperinsulinemia (Duggan et al. 2020).
Imatinib is a potent inhibitor of c-Abl, and although other kinase targets of imatinib may mediate some of the effects on blood glucose and insulin, clinical reports of T1D and T2D and experimental models consistently show that imatinib lowers blood glucose but restores or enhances insulin production in both T1D and T2D (Breccia et al. 2004, Veneri et al. 2005, Haap et al. 2007, Hägerkvist et al. 2007, Louvet et al. 2008, Han et al. 2009, Agostino et al. 2011, Salaroli et al. 2012, Lau et al. 2014, Samaha et al. 2020, Pichavaram et al. 2021). Additionally, nilotinib is a similarly potent inhibitor of c-Abl, but there are contrasting reports on nilotinib’s effects on blood glucose and insulin. Nilotinib has been reported to increase blood glucose in non-diabetic and T2D patients while increasing insulin levels in non-diabetic patients but decreasing insulin levels in T2D. Furthermore, in a rodent model of T1D, nilotinib preserved serum insulin levels while decreasing blood glucose (Saglio et al. 2010, Ito et al. 2013, Zdenek et al. 2014, Samaha et al. 2019). This is further complicated by the fact both imatinib and nilotinib have a similar inhibition profile regarding kinases implicated in metabolic homeostasis, including c-Kit, PDGFRβ, and JNK to various degrees (Fig. 2 or Table 2). An important study by Louvet et al. attempted to address the difficulty in interpreting SMKI’s effects due to overlapping targets by using a systematic combination of SMKIs and neutralizing antibodies in a genetic rodent model of T1D. Imatinib prevented onset of and led to remission of T1D and modestly decreased leukocyte infiltration in pancreatic islets. Similar results were observed with sunitinb, which is primarily targeted against VEGFR, in the same model. The authors suggest that the efficacy of sunitinib eliminated a role for c-Abl in improving T1D; however, kinase assays demonstrate that sunitinib inhibits c-Abl to a significant degree, as well as c-Kit and PDGFRβ (Table 2). More convincingly, experiments using c-Kit monoclonal antibodies only resulted in a marginal effect on blood glucose, while delivery of a soluble form of PDGFR rapidly reversed T1D in these mice (Louvet et al. 2008). Due to the capability of both imatinib and sunitinib to inhibit PDGFRβ, this strongly supports the concept that inhibiting the kinase activity of PDGFRβ rather than c-Kit promotes lowering of blood glucose and possibly protection from T1D in this model. However, this result does not rule out a role for c-Abl in the regulation of blood glucose and insulin secretion, which should be considered a next step in this line of research.
PDGFRβ
PDGFRβ belongs to a family of membrane-bound receptors activated by platelet-derived growth factors (PDGFs), which control fundamental aspects of cell function, including growth and proliferation. PDGF-PDFRβ signaling regulates hematopoiesis, blood vessel formation, and wound repair (Chen et al. 2013a ). Overactive PDGFR signaling has been implicated in proliferative diseases, including various cancers and atherosclerosis (Chen et al. 2013a ). PDGFRs have been reviewed elsewhere as cancer drug targets (Pietras et al. 2003). Sunitinib and sorafenib are two of the main SMKIs that have been developed against PDGFRβ, but these SMKIs also inhibit cKIT and fms-like tyrosine kinase 3 (FLT3) with equal or even greater potency, according to available kinase assay data (Levitzki 2004, Davis et al. 2011). Nonetheless, sunitinib and sorafenib consistently yield improvements in blood glucose level, lower blood glucose, and/or reductions in insulin requirements in T1D and T2D patients and rodent models (Billemont et al. 2008, Louvet et al. 2008, Templeton et al. 2008, Agostino et al. 2011, Imarisio et al. 2012, Oh et al. 2012, Holstein et al. 2013, Mukai et al. 2014, Di Costanzo et al. 2017, Peng et al. 2018, Karbownik et al. 2020, Mahdi et al. 2022).
PDGFRβ is increased in the white adipose tissue (WAT) of HFD-fed mice. Additionally, adipose-specific knockdown of Pdgfrb resulted in lower accumulation of visceral and s.c. fat and improved blood glucose control and lower blood insulin in HFD-fed mice, coincident with a reduction in new blood vessel formation in adipose tissue (Onogi et al. 2017, Shao et al. 2018). The effects of inhibiting PDGFRβ are not restricted to obesity and T2D, since Louvet et al. showed that targeting PDGFRβ can mitigate aspects of T1D in lean mice (Louvet et al. 2008). There is not much evidence that contradicts the concept of PDGFβ inhibition and lower blood glucose, but some work has implicated PDGFβ as a growth factor that stimulates insulin-independent glucose transport and glucose uptake in adipocytes (Whiteman et al. 2003). In theory, this suggests that inhibition of PDGFRβ could limit blood glucose disposal and increase blood glucose in models of insulin resistance or insufficiency. However, the impact of adipocyte glucose uptake on systemic blood glucose control is limited and any potential effect on adipocyte glucose uptake on blood glucose appears outweighed by the totality of SMKI evidence showing that inhibiting PDGFβ lowers blood glucose. It is not yet clear how targeting PDGFRβ participates in development of, or protection against, T1D and T2D despite the potential for reduced insulin-independent adipocyte glucose uptake.
EGFR and VEGFR
EGF and related vascular endothelial growth factor (VEGF) signaling pathways regulate growth, survival, proliferation, and differentiation in mammalian cells (Press & Lenz 2007). Mutations leading to hyperactive EGFR or VEGFR have been associated with a number of cancers including types of lung and epithelial cancers, renal and thyroid cancers, and glioblastomas (Sullivan & Planchard 2016). Some of the first SMKIs approved for clinical use included the EGFR inhibitors, gefitinib and erlotinib, in 2012 and 2013, respectively, and numerous EGFR and VEGFR inhibitors have been approved in years since (Wu et al. 2015). EGFR inhibitors gefitinib and erlotinib have been demonstrated to reverse hyperglycemia in clinical reports of T2D and improve glucose tolerance and insulin sensitivity in rodent models of T2D, while other EGFR-targeted inhibitors including panitumumab and rociletinib are associated with clinical hyperglycemia in human non-diabetic and T2D cancer patients (Costa & Huberman 2006, Brooks 2012, Zhang et al. 2014, Goldman et al. 2016, Li et al. 2018, Duggan et al. 2020). Although not approved for clinical use, another EGFR SMKI, dubbed PD153035, has been shown to reduce inflammation and improve glucose tolerance and insulin sensitivity in HFD-fed mice (Prada et al. 2009). In this study and others, authors conclude that inhibition of EGFR contributes to the observed therapeutic effects; however, gefitinib, erlotinib, and PD153035 are not specific for EGFR. These SMKIs have been shown to inhibit many other kinases involved in glycemic control, including RIPK2, both in vitro and in vivo (Duggan et al. 2017). Increased expression and activity of EGF have been implicated in diabetes-induced vascular hypertrophy, nephropathy, and vasoconstriction relevant to cardiovascular disease outcomes, but it is unclear if and how EGF signaling may mediate blood glucose control (Gilbert et al. 2000, Chen et al. 2015, Prado et al. 2018). Current evidence is insufficient to determine specific kinase target(s) of EGFR SMKIs that are responsible for these contrasting effects on glycemic control.
Sorafenib, sunitinib, and panzopanib are some of the most widely used SMKIs of VEGFR, and reports in the literature consistently show decreases in blood glucose or hypoglycemia in T1D, T2D, and non-diabetic cancer patients and in rodent models following treatment with these inhibitors (Billemont et al. 2008, Louvet et al. 2008, Templeton et al. 2008, Böhm et al. 2010, Agostino et al. 2011, Oh et al. 2012, Imarisio et al. 2012, Chen et al. 2013b , Holstein et al. 2013, Demirci et al. 2014, Huda et al. 2014, Mukai et al. 2014, Tyrrell & Pwint 2014, Di Costanzo et al. 2017, Lutz et al. 2017, Peng et al. 2018, Karbownik et al. 2020, Mahdi et al. 2022). However, these SMKIs also target c-Kit, c-Abl, and PDGFRβ and there is no clearly established role for VEGFR in blood glucose control. Furthermore, there are conflicting reports on the therapeutic benefits of altering VEGF signaling, and its contribution to angiogenesis during adipose tissue expansion (Tam et al. 2009, Robciuc et al. 2016). It remains unknown if off-target kinases are involved in the VEGF SMKI-mediated changes in blood glucose.
c-KIT
CD117, also known as c-Kit, is a transmembrane stem cell factor receptor expressed predominantly in bone marrow and progenitor cell populations. Gain-of-function mutations in c-Kit have been implicated in human hematopoietic and gastrointestinal cancers (Babaei et al. 2016). Imatinib, sorafenib, and sunitinib are some of the most potent inhibitors of c-Kit with blood glucose-lowering effects (Babaei et al. 2016). c-Kit expression has been found to be increased in the synovium of patients with obesity-associated osteoarthritis (Uchida et al. 2019). Streptozotocin (STZ)-induced hyperglycemia or HFD-induced obesity in rodents is associated with a decrease in the number of c-Kit+ bone marrow progenitor cells (Pierpaoli et al. 2016, Van Den Berg et al. 2016). However, specific inhibition of c-Kit using a monoclonal antibody approach did not affect the development or disease progression in a STZ-induced T1D rodent model, whereas the SMKIs imatinib and sunitinib protected against T1D development (Louvet et al. 2008). This suggests that although imatinib and sunitinib are designed to inhibit c-Kit, the actions of these SMKIs are propagated through other kinases that alter glycemic outcomes, although there is a current lack of convincing evidence identifying the specific kinase(s) and specific mechanism(s) underlying the distinct effects on blood glucose, insulin sensitivity, and β-cell function (Louvet et al. 2008). Multiple studies have identified that c-Kit is required for pancreatic β-cell growth and function, where decreased expression of c-Kit in mutant mouse models results in hyperglycemia and impaired glucose control. These manifestations have been observed as decreased insulin gene expression, β-cell mass, and impaired glucose-stimulated insulin secretion in vivo and in isolated islets. (Krishnamurthy et al. 2007, Feng et al. 2015). Thus, inhibition of c-Kit by SMKIs may be deleterious to glycemic control and limit their therapeutic benefits in T1D and T2D.
SMKIs alter metabolism
SMKIs may alter the connection between the immune system and metabolism. Changes in immune cell populations and immunological messengers (i.e. cytokines) have been implicated in the pathogenesis of metabolic disease and can alter blood glucose, insulin, and insulin resistance during obesity (Hotamisligil 2006, Osborn & Olefsky 2012). SMKIs can alter kinases, such as RIPK2 and Bruton’s tyrosine kinase (BTK) involved in innate immune responses propagating inflammation from pattern recognition receptors (PRRs) and the NLR family pyrin domain containing 3 (NLRP3) inflammasome. Inhibition of specific host kinases by SMKIs can also alter the connection between immunity and metabolism, which has implications for blood glucose and insulin.
RIPK2
RIPK2 is the common adapter for the pattern recognition receptors nucleotide-binding oligomerization domain-containing protein (NOD)1 and NOD2. Our lab and others have defined the innate immune sensors NOD1 and NOD2 as points of convergence between immunity and blood glucose control in mice models. Specifically, activation of NOD1 triggers key features of metabolic syndrome, including lipolysis, inflammation, and dysglycemia. In contrast, activation of NOD2 alleviates metabolic dysfunction and improves blood glucose control during low-dose endotoxin or diet-induced obesity (Schertzer et al. 2011, Chi et al. 2014, Cavallari et al. 2017, 2020, Duggan et al. 2017). Consistent with this, deletion of hematopoietic NOD1 during obesity improves blood glucose control, whereas deletion of NOD2 during obesity worsens blood glucose control (Schertzer et al. 2011, Denou et al. 2015, Chan et al. 2017).
Propagation of NOD1 and NOD2 signaling requires the kinase activity of the downstream adaptor protein, RIPK2. We have shown that RIPK2 is an obligate component of the contrasting NOD1-driven and NOD2-driven glycemic responses (Duggan et al. 2017, 2020). RIPK2 deletion results in exacerbated obesity and worsened insulin resistance during HFD-feeding in mice (Wang et al. 2013a ), although the relative importance of NOD1-RIPK2 vs NOD2-RIPK2 metabolism during obesity and their individual contributions to glycemic control in metabolic disease states are not well-understood. Since many SMKIs are potent inhibitors of RIPK2, including gefitinib and erlotinib, RIPK2 was positioned as a potential mediator of SMKI effects on blood glucose. Although RIPK2 is not the intended target, many SMKIs with reported inhibitory activity against RIPK2 (Table 2) can attenuate the glycemic consequences of acute NOD1 and NOD2 signaling (Duggan et al. 2017, 2020). Thus, in the context of obesity, inhibition of the NOD1–RIPK2 axis represented a potential mechanism by which SMKIs lower blood glucose. In contrast, inhibition of the NOD2-RIPK2 axis by SMKIs could limit the therapeutic benefits of these drugs on blood glucose. Using a series of genetic knockout mice models, we showed that gefitinib-mediated blood glucose lowering during obesity was entirely independent of NOD1, NOD2, or RIPK2 signaling. Importantly, we found that gefitinib treatment also lowered plasma insulin during a glucose challenge and improved insulin resistance only in Ripk2−/− mice, suggesting that deletion of Ripk2 improves the insulin sensitizing effect of gefitinib during obesity. These findings highlight that inhibition of RIPK2 by SMKIs may be limiting their therapeutic potential to not only lower blood glucose but improve insulin sensitivity in obesity and T2D (Duggan et al. 2020) (Fig. 3).
RIPK1 and RIPK3
RIPK1 and RIPK3 regulate immune, cellular stress, and cell survival responses. These kinases also work to mediate cell apoptosis and necroptosis, which primarily engage RIPK3 (Wegner et al. 2017). Sorafenib, sunitinib, pazopanib, ponatinib, and dabrafenib are all potent inhibitors of RIPK1 and/or RIPK3 and are being investigated for therapeutic repurposing in various inflammatory conditions. Interestingly, emerging evidence connects RIPK1 and RIPK3 to glucose homeostasis and metabolic disease. Overexpressing variants of the Ripk1 gene are associated with obesity in humans, and Ripk1 silencing in mice fed a HFD results in decreased body weight, adiposity, improved insulin sensitivity, and reduced markers of non-alcoholic steatohepatitis (NASH) (Karunakaran et al. 2020, Tao et al. 2021). This evidence positions RIPK1 as a potential therapeutic target for obesity-associated metabolic disease and as a potential mechanism underlying the blood glucose-altering effects of some SMKIs. Similarly, Ripk3 is overexpressed in the adipose tissue of obese mice and humans, and hepatic Ripk3 expression is correlated with the severity of fatty liver disease. In contrast to RIPK1, genetic knockdown of Ripk3 exacerbates inflammation and glucose intolerance during HFD-feeding (Gautheron et al. 2016, Afonso et al. 2021). A recent study established a role for RIPK3 in mediating IL-1β-dependent macrophage infiltration and β-cell dysfunction in response to ER stress or overnutrition, and inhibition of RIPK3 protected against islet inflammation and β-cell dysfunction in mouse pancreatic β-cell line (Yang et al. 2020). While current available evidence suggests that RIPK1 may be a therapeutic target of SMKIs that can contribute to improvements in glycemic control, there is conflicting evidence for the role of RIPK3 in protecting against inflammation and glucose tolerance during obesity versus precipitating β-cell inflammation and potential insulin secretion defects. Therefore, the role of RIPK3 in mediating SMKI’s effects on blood glucose and insulin remains unclear. Given that RIPK1 and RIPK3 have emerged as regulators of metabolism, further studies involving these kinases as potential targets of SMKIs are needed.
Bruton's tyrosine kinase
BTK was initially discovered as the genetic cause of X-linked agammaglobulinemia (XLA), a condition characterized by defective development of B cells resulting in impaired immunoglobulin production (Vetrie et al. 1993). This non-receptor tyrosine kinase is crucial for the activation of B cells and other cells of the hematopoietic system (Liu et al. 2017). BTK influences proliferation, differentiation, and survival of B-cells, which makes this kinase a prime treatment target in many B-cell malignancies (Weber et al. 2017, Pal Singh et al. 2018). BTK has also been implicated in the activation of toll-like receptor (TLR) signaling pathways, demonstrating a role for BTK in mediating both innate and adaptive immunity (Weber et al. 2017). Ibrutinib and acalabrutinib represent the two main FDA-approved SMKIs for BTK, used in the treatment of relapsed/refractory chronic lymphocytic leukemia (CLL), mantle-cell lymphoma (MCL), among several other malignancies (Byrd et al. 2013, Wang et al. 2013b ). Ibrutinib and acalabrutinib are designed to bind Cysteine-481 on BTK; however, off-target binding of several other homologous cysteine-containing kinases, such as the TEC family kinases (ITK, BMX, TEC), EGFR, T-cell X chromosome kinase (TXK), and Janus kinase 3 (JAK3), has been reported, which can lead to adverse side-effects (Honigberg et al. 2010, Byrd et al. 2016, Herman et al. 2017). Although data are not available for acalabrutinib, kinase assay data for ibrutinib indicate that it inhibits RIPK2, EGFR, and c-ABL to a similar degree as BTK and ibrutinib is approximately half as potent for inhibition of c-Kit and PDGFRβ targets (Table 2).
Beyond cancer treatment, studies investigating the deficiency or inhibition of BTK have demonstrated therapeutic potential in numerous inflammatory disease models like autoimmune arthritis, ischemic brain injury, and colitis, among others (Chang et al. 2011, Ito et al. 2015, Mao et al. 2020). The idea of BTK as a point of regulation for NLRP3-associated inflammation has gained traction in recent years (Liu et al. 2017, Purvis et al. 2020, Bittner et al. 2021). The NLRP3 inflammasome mediates the activation and production of several inflammatory cytokines, which can exacerbate insulin resistance, and NLRP3-mediated IL-1β activation contributes significantly to the development of obesity and T2D (Xu et al. 2003, Zhao et al. 2014). Ibrutinib treatment impairs activation of the NLRP3 inflammasome, thereby lowering IL-1β processing and release in human primary macrophages and tumor-associated macrophages (Liu et al. 2017, Benner et al. 2019, Purvis et al. 2020). A recent study demonstrated that BTK is highly expressed in visceral adipose tissue samples collected from obese and diabetic human subjects and a positive correlation has been established between BTK gene expression and pro-inflammatory IL-1 expression in these samples. By targeting BTK via inhibition with ibrutinib, improvement in insulin resistance and increase in glucose uptake were observed in a human liver cancer cell line (Althubiti et al. 2020).
Consistent with this, HFD-feeding in mice has been shown to increase BTK expression and activation in liver, and treatment of HFD-fed mice with ibrutinib led to significantly lower levels of plasma insulin and non-fasting blood glucose and an improvement in insulin signaling in liver (Purvis et al. 2020). As newer generation inhibitors, little to no clinical reports of ibrutinib or acalabrutinib lowering blood glucose in diabetic cancer patients exist, but in vitro and rodent models support a role for targeting BTK to improve glycemic control in the context of metabolic disease, although inhibition of BTK versus other clinically relevant kinase targets (RIPK2, c-ABL, c-Kit, PDGFRβ) must also be considered as an underlying mechanism of action.
Conclusions
Understanding how specific SMKIs can alter blood glucose, insulin, and insulin sensitivity is important for both safe and effective use of SMKIs in their primary therapeutic indications, such as cancer or inflammatory disease. In addition to concerns of SMKI-induced acute hypoglycemia, higher blood glucose can affect the disease course of cancer and inflammatory conditions, and therefore, it is important to understand which SMKIs alter blood glucose and insulin and which kinase targets mediate these effects. The effects of SMKIs on blood glucose and insulin are complex, depend on the effective dose, and are complicated by the multiple kinases inhibited by a given SMKI (Fig. 4). It is important to distinguish between studies that use inhibitors to test the role of a given kinase versus genetic deletion of the target kinase, and ideally, genetic knockout models can be used to validate the proposed target kinase that is mediating the effects of a given SMKI effect. In addition, SMKI drugs in development include inhibitors of phosphoinositide 3-kinases (PI3K) and protein kinase B (PKB)/AKT, which are critical nodes in insulin signaling pathways, where some potential signaling redundancies could be exploited (Liu et al. 2009, Molinaro et al. 2019). How these new SMKIs impact glycemic control and insulin dynamics will need to be carefully considered. An important future goal will be to assess the potential synergy between inhibiting multiple kinases, which occurs with each SMKI, as well as how SMKIs impact the host–microbe relationship, which can have important outcomes in cancer and diabetes progression and therapy.
Declaration of interest
The authors have no conflict of interest related to this research.
Funding
This work was supported by a grant to JDS from the Natural Science and Engineering Research Council (RGPIN-2020-05707). JDS holds a Canada Research Chair in Metabolic Inflammation. DMM was supported by a Canadian Institute for Health Research graduate scholarship. Figures were created with Biorender.com.
References
Adams JA 2001 Kinetic and catalytic mechanisms of protein kinases. Chemical Reviews 101 2271–2290. (https://doi.org/10.1021/cr000230w)
Afonso MB, Rodrigues PM, Mateus-Pinheiro M, Simão AL, Gaspar MM, Majdi A, Arretxe E, Alonso C, Santos-Laso A & Jimenez-Agüero R et al.2021 RIPK3 acts as a lipid metabolism regulator contributing to inflammation and carcinogenesis in non-alcoholic fatty liver disease. Gut 70 2359–2372. (https://doi.org/10.1136/GUTJNL-2020-321767)
Agostino NM, Chinchilli VM, Lynch CJ, Koszyk-Szewczyk A, Gingrich R, Sivik J & Drabick JJ 2011 Effect of the tyrosine kinase inhibitors (sunitinib, sorafenib, dasatinib, and imatinib) on blood glucose levels in diabetic and nondiabetic patients in general clinical practice. Journal of Oncology Pharmacy Practice 17 197–202. (https://doi.org/10.1177/1078155210378913)
Althubiti M, Almaimani R, Eid SY, Elzubaier M, Refaat B, Idris S, Alqurashi TA & El-Readi MZ 2020 BTK targeting suppresses inflammatory genes and ameliorates insulin resistance. European Cytokine Network 31 168–179. (https://doi.org/10.1684/ECN.2020.0454)
Angelis V, Johnston SRD, Ardestani A & Maedler K 2022 Case report: neratinib therapy improves glycemic control in a patient with Type 2 diabetes and breast cancer. Frontiers in Endocrinology 13 830097. (https://doi.org/10.3389/FENDO.2022.830097)
Ardestani A, Li S, Annamalai K, Lupse B, Geravandi S, Dobrowolski A, Yu S, Zhu S, Baguley TD & Surakattula M et al.2019 Neratinib protects pancreatic beta cells in diabetes. Nature Communications 10 1–17. (https://doi.org/10.1038/s41467-019-12880-5)
ARIAD Pharmaceuticals 2017 ALUNBRIG® (brigatinib). White Oak, MD, USA: Food and Drug Administration (FDA). (available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/208772s008lbl.pdf)
Babaei MA, Kamalidehghan B, Saleem M, Huri HZ & Ahmadipour F 2016 Receptor tyrosine kinase (c-Kit) inhibitors: a potential therapeutic target in cancer cells. Drug Design, Development and Therapy 10 2443–2459. (https://doi.org/10.2147/DDDT.S89114)
Benner B, Scarberry L, Stiff A, Duggan MC, Good L, Lapurga G, Butchar JP, Tridandapani S & Carson WE 2019 Evidence for interaction of the NLRP3 inflammasome and Bruton’s tyrosine kinase in tumor-associated macrophages: implications for myeloid cell production of interleukin-1beta. Oncoimmunology 8 1659704. (https://doi.org/10.1080/2162402X.2019.1659704)
Billemont B, Medioni J, Taillade L, Helley D, Meric JB, Rixe O & Oudard S 2008 Blood glucose levels in patients with metastatic renal cell carcinoma treated with sunitinib. British Journal of Cancer 99 1380–1382. (https://doi.org/10.1038/sj.bjc.6604709)
Bittner ZA, Liu X, Tortola MM, Tapia-Abellán A, Shankar S, Andreeva L, Mangan M, Spalinger M, Kalbacher H & Düwell P et al.2021 BTK operates a phospho-tyrosine switch to regulate NLRP3 inflammasome activity. Journal of Experimental Medicine 218 19. (https://doi.org/10.1084/jem.20201656)
Böhm S, Hess D, Gillessen S & Brändle M 2010 Improved glycemic control with the multi-receptor tyrosine kinase inhibitor pazopanib. Diabetes Care 33 e82. (https://doi.org/10.2337/DC10-0436)
Breccia M, Muscaritoli M, Aversa Z, Mandelli F & Alimena G 2004 Imatinib mesylate may improve fasting blood glucose in Ph+ chronic myelogenous leukemia patients responsive to treatment. Journal of Clinical Oncology 22 4653–4655. (https://doi.org/10.1200/JCO.2004.04.156)
Breccia M, Muscaritoli M, Cannella L, Stefanizzi C, Frustaci A & Alimena G 2008 Fasting glucose improvement under dasatinib treatment in an accelerated phase chronic myeloid leukemia patient unresponsive to imatinib and nilotinib. Leukemia Research 32 1626–1628. (https://doi.org/10.1016/j.leukres.2008.01.015)
Brooks MB 2012 Erlotinib appears to produce prolonged remission of insulin-requiring type 2 diabetes associated with metabolic syndrome and chronic kidney disease. British Journal of Diabetes and Vascular Disease 12 87–90. (https://doi.org/10.1177/1474651412442694)
Byrd JC, Furman RR, Coutre SE, Flinn IW, Burger JA, Blum KA, Grant B, Sharman JP, Coleman M & Wierda WG et al.2013 Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. New England Journal of Medicine 369 32–42. (https://doi.org/10.1056/NEJMoa1215637)
Byrd JC, Harrington B, O’Brien S, Jones JA, Schuh A, Devereux S, Chaves J, Wierda WG, Awan FT & Brown JR et al.2016 Acalabrutinib (ACP-196) in relapsed chronic lymphocytic leukemia. New England Journal of Medicine 374 323–332. (https://doi.org/10.1056/NEJMoa1509981)
Cavallari JF, Barra NG, Foley KP, Lee A, Duggan BM, Henriksbo BD, Anhê FF, Ashkar AA & Schertzer JD 2020 Postbiotics for NOD2 require nonhematopoietic RIPK2 to improve blood glucose and metabolic inflammation in mice. American Journal of Physiology-Endocrinology and Metabolism 318 E579–E585. (https://doi.org/10.1152/ajpendo.00033.2020)
Cavallari JF, Fullerton MD, Duggan BM, Foley KP, Denou E, Smith BK, Desjardins EM, Henriksbo BD, Kim KJ & Tuinema BR et al.2017 Muramyl dipeptide-based postbiotics mitigate obesity-induced insulin resistance via IRF4. Cell Metabolism 25 1063–1074.e3. (https://doi.org/10.1016/j.cmet.2017.03.021)
Chan KL, Tam TH, Boroumand P, Prescott D, Costford SR, Escalante NK, Fine N, Tu Y, Robertson SJ & Prabaharan D et al.2017 Circulating NOD1 activators and hematopoietic NOD1 contribute to metabolic inflammation and insulin resistance. Cell Reports 18 2415–2426. (https://doi.org/10.1016/j.celrep.2017.02.027)
Chang BY, Huang MM, Francesco M, Chen J, Sokolove J, Magadala P, Robinson WH & Buggy JJ 2011 The Bruton tyrosine kinase inhibitor PCI-32765 ameliorates autoimmune arthritis by inhibition of multiple effector cells. Arthritis Research and Therapy 13 R115. (https://doi.org/10.1186/AR3400)
Chen J, Chen JK & Harris RC 2015 EGF receptor deletion in podocytes attenuates diabetic nephropathy. Journal of the American Society of Nephrology 26 1115–1125. (https://doi.org/10.1681/ASN.2014020192)
Chen PH, Chen X & He X 2013a Platelet-derived growth factors and their receptors: structural and functional perspectives. Biochimica et Biophysica Acta 1834 2176–2186. (https://doi.org/10.1016/J.BBAPAP.2012.10.015)
Chen J, Wang C, Han J, Luan Y, Cui Y, Shen R, Sha D, Cong L, Zhang Z & Wang W 2013b Therapeutic effect of sunitinib malate and its influence on blood glucose concentrations in a patient with metastatic insulinoma. Expert Review of Anticancer Therapy 13 737–743. (https://doi.org/10.1586/ERA.13.45)
Chi W, Dao D, Lau TC, Henriksbo BD, Cavallari JF, Foley KP & Schertzer JD 2014 Bacterial peptidoglycan stimulates adipocyte lipolysis via NOD1. PLoS One 9 e97675. (https://doi.org/10.1371/journal.pone.0097675)
Costa DB & Huberman MS 2006 Improvement of Type 2 Diabetes in a lung cancer patient treated with erlotinib. Diabetes Care 29 1711. (https://doi.org/10.2337/dc06-0558)
Davis MI, Hunt JP, Herrgard S, Ciceri P, Wodicka LM, Pallares G, Hocker M, Treiber DK & Zarrinkar PP 2011 Comprehensive analysis of kinase inhibitor selectivity. Nature Biotechnology 29 1046–1051. (https://doi.org/10.1038/nbt.1990)
Demirci A, Bal O, Durnali A, Ekinci AŞ, Eşbah O, Alkiş N & Öksüzoʇlu B 2014 Sunitinib-induced severe hypoglycemia in a diabetic patient. Journal of Oncology Pharmacy Practice 20 469–472. (https://doi.org/10.1177/1078155213508441)
Denou E, Lolmède K, Garidou L, Pomie C, Chabo C, Lau TC, Fullerton MD, Nigro G, Zakaroff‐Girard A & Luche E et al.2015 Defective NOD2 peptidoglycan sensing promotes diet‐induced inflammation, dysbiosis, and insulin resistance. EMBO Molecular Medicine 7 259–274. (https://doi.org/10.15252/emmm.201404169)
Dhanasekaran N & Premkumar Reddy E 1998 Signaling by dual specificity kinases. Oncogene 17 1447–1455. (https://doi.org/10.1038/sj.onc.1202251)
Di Costanzo GG, Tortora R, Morisco F, Addario L, Guarino M, Cordone G, Falco L & Caporaso N 2017 Impact of diabetes on outcomes of sorafenib therapy for hepatocellular carcinoma. Targeted Oncology 12 61–67. (https://doi.org/10.1007/s11523-016-0454-5)
Duggan BM, Cavallari JF, Foley KP, Barra NG & Schertzer JD 2020 RIPK2 dictates insulin responses to tyrosine kinase inhibitors in obese male mice. Endocrinology 161 1–16. (https://doi.org/10.1210/ENDOCR/BQAA086)
Duggan BM, Foley KP, Henriksbo BD, Cavallari JF, Tamrakar AK & Schertzer JD 2017 Tyrosine kinase inhibitors of Ripk2 attenuate bacterial cell wall-mediated lipolysis, inflammation and dysglycemia. Scientific Reports 7 1578. (https://doi.org/10.1038/s41598-017-01822-0)
Elsherbiny NM, El-Sherbiny M & Said E 2015 Amelioration of experimentally induced diabetic nephropathy and renal damage by nilotinib. Journal of Physiology and Biochemistry 71 635–648. (https://doi.org/10.1007/s13105-015-0428-6)
Feng Z, Popell A, Li J, Silverstein J, Oakie A, Yee S & Wang R 2015 c-kit receptor signalling regulates islet vasculature, B-cell survival and function in vivo. Diabetes 64 3852–3866. (https://doi.org/10.2337/db15-0054)
Gautheron J, Vucur M, Schneider AT, Severi I, Roderburg C, Roy S, Bartneck M, Schrammen P, Diaz MB & Ehling J et al.2016 The necroptosis-inducing kinase RIPK3 dampens adipose tissue inflammation and glucose intolerance. Nature Communications 7 11869. (https://doi.org/10.1038/NCOMMS11869)
Gilbert RE, Rumble JR, Cao Z, Cox AJ, Van Eeden P, Allen TJ, Kelly DJ & Cooper ME 2000 Endothelin receptor antagonism ameliorates mast cell infiltration, vascular hypertrophy, and epidermal growth factor expression in experimental diabetes. Circulation Research 86 158–165. (https://doi.org/10.1161/01.RES.86.2.158)
Goldman JW, Mendenhall MA & Rettinger SR 2016 Hyperglycemia associated with targeted oncologic treatment: mechanisms and management. Oncologist 21 1326–1336. (https://doi.org/10.1634/THEONCOLOGIST.2015-0519)
Haap M, Gallwitz B, Thamer C, Müssig K, Häring HU, Kanz L & Hartmann JT 2007 Symptomatic hypoglycemia during imatinib mesylate in a non-diabetic female patient with gastrointestinal stromal tumor. Journal of Endocrinological Investigation 30 688–692. (https://doi.org/10.1007/BF03347451)
Hägerkvist R, Sandler S, Mokhtari D & Welsh N 2007 Amelioration of diabetes by imatinib mesylate (Gleevec®): role of β-cell NF-κB activation and anti-apoptotic preconditioning. FASEB Journal 21 618–628. (https://doi.org/10.1096/fj.06-6910com)
Han MS, Chung KW, Cheon HG, Rhee SD, Yoon CH, Lee MK, Kim KW & Lee MS 2009 Imatinib mesylate reduces ER Stress and Induces Remission of Diabetes in db / db Mice. Diabetes 58 329–336. (https://doi.org/10.2337/db08-0080)
Hantschel O & Superti-Furga G 2004 Regulation of the c-Abl and bcr-abl tyrosine kinases. Nature Reviews. Molecular Cell Biology 5 33–44. (https://doi.org/10.1038/NRM1280)
Herman SEM, Montraveta A, Niemann CU, Mora-Jensen H, Gulrajani M, Krantz F, Mantel R, Smith LL, McClanahan F & Harrington BK et al.2017 The Bruton tyrosine kinase (BTK) inhibitor acalabrutinib demonstrates potent on-target effects and efficacy in two mouse models of chronic lymphocytic leukemia. Clinical Cancer Research 23 2831–2841. (https://doi.org/10.1158/1078-0432.CCR-16-0463)
Holstein A, Kovacs P & Beil W 2013 Severe hypoglycemia due to possible interaction between glibenclamide and sorafenib in a patient with hepatocellular carcinoma. Current Drug Safety 8 148–152. (https://doi.org/10.2174/15748863113089990027)
Honigberg LA, Smith AM, Sirisawad M, Verner E, Loury D, Chang B, Li S, Pan Z, Thamm DH & Miller RA et al.2010 The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy. PNAS 107 13075–13080. (https://doi.org/10.1073/PNAS.1004594107)
Hotamisligil GS 2006 Inflammation and metabolic disorders. Nature 444 860–867. (https://doi.org/10.1038/nature05485)
Huda MSB, Amiel SA, Ross P & Aylwin SJB 2014 Tyrosine kinase inhibitor sunitinib allows insulin independence in long-standing Type 1 diabetes. Diabetes Care 37 e87–e88. (https://doi.org/10.2337/dc13-2132)
Imarisio I, Paglino C, Ganini C, Magnani L, Caccialanza R & Porta C 2012 The effect of sorafenib treatment on the diabetic status of patients with renal cell or hepatocellular carcinoma. Future Oncology 8 1051–1057. (https://doi.org/10.2217/FON.12.74)
Ito M, Shichita T, Okada M, Komine R, Noguchi Y, Yoshimura A & Morita R 2015 Bruton’s tyrosine kinase is essential for NLRP3 inflammasome activation and contributes to ischaemic brain injury. Nature Communications 6 7360. (https://doi.org/10.1038/NCOMMS8360)
Ito Y, Miyamoto T, Chong Y, Maki T, Akashi K & Kamimura T 2013 Nilotinib exacerbates diabetes mellitus by decreasing secretion of endogenous insulin. International Journal of Hematology 97 135–138. (https://doi.org/10.1007/s12185-012-1222-7)
Jia DY, Du ZH, Liu SM, Liu HJ, Wang FW, Ling EA, Liu K & Hao AJ 2008 c-Abl is involved in high glucose-induced apoptosis in embryonic E12.5 cortical neural progenitor cells from the mouse brain. Journal of Neurochemistry 106 1720–1730. (https://doi.org/10.1111/J.1471-4159.2008.05533.X)
Karbownik A, Stachowiak A, Urjasz H, Sobańska K, Szczecińska A, Grabowski T, Stanisławiak-Rudowicz J, Wolc A, Grześkowiak E & Szałek E 2020 The oxidation and hypoglycaemic effect of sorafenib in streptozotocin-induced diabetic rats. Pharmacological Reports 72 254–259. (https://doi.org/10.1007/s43440-019-00021-0)
Karunakaran D, Turner AW, Duchez AC, Soubeyrand S, Rasheed A, Smyth D, Cook DP, Nikpay M, Kandiah JW & Pan C et al.2020 RIPK1 gene variants associate with obesity in humans and can be therapeutically silenced to reduce obesity in mice. Nature Metabolism 2 1113–1125. (https://doi.org/10.1038/S42255-020-00279-2)
Keshet R, Kraitshtein ZB, Shanzer M, Adler J, Reuven N & Shaul Y 2014 C-Abl tyrosine kinase promotes adipocyte differentiation by targeting PPAR-gamma 2. PNAS 111 16365–16370. (https://doi.org/10.1073/pnas.1411086111)
Khozin S, Blumenthal GM, Zhang L, Tang S, Brower M, Fox E, Helms W, Leong R, Song P & Pan Y et al.2015 FDA approval: ceritinib for the treatment of Metastatic anaplastic lymphoma kinase-positive non-small cell lung cancer. Clinical Cancer Research 21 2436–2439. (https://doi.org/10.1158/1078-0432.CCR-14-3157)
Krishnamurthy M, Ayazi F, Li J, Lyttle AW, Woods M, Wu Y, Yee SP & Wang R 2007 c-kit in early onset of diabetes: a morphological and functional analysis of pancreatic beta-cells in c-KitW-v mutant mice. Endocrinology 148 5520–5530. (https://doi.org/10.1210/en.2007-0387)
Lahiry P, Torkamani A, Schork NJ & Hegele RA 2010 Kinase mutations in human disease: interpreting genotype-phenotype relationships. Nature Reviews. Genetics 11 60–74. (https://doi.org/10.1038/nrg2707)
Lau J, Zhou Q, Sutton SE, Herman AE, Schmedt C & Glynne R 2014 Inhibition of c-Kit is not required for reversal of hyperglycemia by imatinib in NOD mice. PLoS One 9 e84900. (https://doi.org/10.1371/journal.pone.0084900)
Levitzki A 2004 PDGF receptor kinase inhibitors for the treatment of PDGF driven diseases. Cytokine and Growth Factor Reviews 15 229–235. (https://doi.org/10.1016/J.CYTOGFR.2004.03.010)
Li Z, Li Y, Overstreet JM, Chung S, Niu A, Fan X, Wang S, Wang Y, Zhang MZ & Harris RC 2018 Inhibition of epidermal growth factor receptor activation is associated with improved diabetic nephropathy and insulin resistance in Type 2 diabetes. Diabetes 67 1847–1857. (https://doi.org/10.2337/DB17-1513)
Lindholm D, Pham DD, Cascone A, Eriksson O, Wennerberg K & Saarma M 2016 C-Abl inhibitors enable insights into the pathophysiology and neuroprotection in Parkinson’s disease. Frontiers in Aging Neuroscience 8 254. (https://doi.org/10.3389/fnagi.2016.00254)
Liu P, Cheng H, Roberts TM & Zhao JJ 2009 Targeting the phosphoinositide 3-kinase pathway in cancer. Nature Reviews. Drug Discovery 8 627–644. (https://doi.org/10.1038/nrd2926)
Liu X, Pichulik T, Wolz OO, Dang TM, Stutz A, Dillen C, Delmiro Garcia M, Kraus H, Dickhöfer S & Daiber E et al.2017 Human NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) inflammasome activity is regulated by and potentially targetable through Bruton tyrosine kinase. Journal of Allergy and Clinical Immunology 140 1054–1067.e10. (https://doi.org/10.1016/J.JACI.2017.01.017)
Louvet C, Szot GL, Lang J, Lee MR, Martinier N, Bollag G, Zhu S, Weiss A & Bluestone JA 2008 Tyrosine kinase inhibitors reverse type 1 diabetes in nonobese diabetic mice. PNAS 105 18895–18900. (https://doi.org/10.1073/pnas.0810246105)
Lundholm MD & Charnogursky GA 2020 Dasatinib‐induced hypoglycemia in a patient with acute lymphoblastic leukemia. Clinical Case Reports 8 1238–1240. (https://doi.org/10.1002/CCR3.2901)
Lutz SZ, Ullrich A, Häring HU, Ullrich S & Gerst F 2017 Sunitinib specifically augments glucose-induced insulin secretion. Cellular Signalling 36 91–97. (https://doi.org/10.1016/j.cellsig.2017.04.018)
Mahdi A, Jiao T, Tratsiakovich Y, Wernly B, Yang J, Östenson CG, Danser AHJ, Pernow J & Zhou Z 2022 Therapeutic potential of sunitinib in ameliorating endothelial dysfunction in Type 2 diabetic rats. Pharmacology 107 160–166. (https://doi.org/10.1159/000520728)
Manning G, Whyte DB, Martinez R, Hunter T & Sudarsanam S 2002 The protein kinase complement of the human genome. Science 298 1912–1934. (https://doi.org/10.1126/science.1075762)
Mao L, Kitani A, Hiejima E, Montgomery-Recht K, Zhou W, Fuss I, Wiestner A & Strober W 2020 Bruton tyrosine kinase deficiency augments NLRP3 inflammasome activation and causes IL-1β-mediated colitis. Journal of Clinical Investigation 130 1793–1807. (https://doi.org/10.1172/JCI128322)
Miyoshi Y, Ogawa O, Nishida A & Masuzawa M 2021 Recurrent hyperglycemic hyperosmolar state after re-administration of dose-reduced ceritinib, an anaplastic lymphoma kinase inhibitor. Diabetology International 12 126–129. (https://doi.org/10.1007/s13340-020-00442-w)
Molinaro A, Becattini B, Mazzoli A, Bleve A, Radici L, Maxvall I, Sopasakis VR, Molinaro A, Bäckhed F & Solinas G 2019 Insulin-driven PI3K-AKT signaling in the hepatocyte is mediated by redundant PI3Kα and PI3Kβ activities and is promoted by RAS. Cell Metabolism 29 1400–1409.e5. (https://doi.org/10.1016/j.cmet.2019.03.010)
Mukai E, Ohta T, Kawamura H, Lee EY, Morita A, Sasase T, Miyajima K, Inagaki N, Iwanaga T & Miki T 2014 Enhanced vascular endothelial growth factor signaling in islets contributes to β cell injury and consequential diabetes in spontaneously diabetic Torii rats. Diabetes Research and Clinical Practice 106 303–311. (https://doi.org/10.1016/j.diabres.2014.08.023)
Nader MA & Wagih HM 2017 Nilotinib, a tyrosine kinase inhibitor exhibits protection against acute pancreatitis-induced lung and liver damage in rats. Naunyn-Schmiedeberg’s Archives of Pharmacology 390 291–300. (https://doi.org/10.1007/s00210-016-1327-2)
Novartis Pharmaceuticals Corporation 2014 ZYKADIA ® (ceritinib). Cambridge, MA, US: Novartis Pharmaceuticals Corporation. (available at: https://www.novartis.com/us-en/sites/novartis_us/files/zykadia.pdf)
Oh JJ, Hong SK, Joo YM, Lee BK, Min SH, Lee S, Byun SS & Lee SE 2012 Impact of sunitinib treatment on blood glucose levels in patients with metastatic renal cell carcinoma. Japanese Journal of Clinical Oncology 42 314–317. (https://doi.org/10.1093/JJCO/HYS002)
Ono K, Suzushima H, Watanabe Y, Kikukawa Y, Shimomura T, Furukawa N, Kawaguchi T & Araki E 2012 Rapid Amelioration of Hyperglycemia Facilitated by dasatinib in a Chronic myeloid leukemia Patient with type 2 diabetes mellitus. Internal Medicine 51 2763–2766. (https://doi.org/10.2169/internalmedicine.51.8314)
Onogi Y, Wada T, Kamiya C, Inata K, Matsuzawa T, Inaba Y, Kimura K, Inoue H, Yamamoto S & Ishii Y et al.2017 PDGFRβ regulates adipose tissue expansion and glucose metabolism via vascular remodeling in diet-induced obesity. Diabetes 66 1008–1021. (https://doi.org/10.2337/DB16-0881)
Osborn O & Olefsky JM 2012 The cellular and signaling networks linking the immune system and metabolism in disease. Nature Medicine 18 363–374. (https://doi.org/10.1038/nm.2627)
Pal Singh S, Dammeijer F & Hendriks RW 2018 Role of Bruton’s tyrosine kinase in B cells and malignancies. Molecular Cancer 17 1–23. (https://doi.org/10.1186/S12943-018-0779-Z)
Patterson H, Nibbs R, McInnes I & Siebert S 2014 Protein kinase inhibitors in the treatment of inflammatory and autoimmune diseases. Clinical and Experimental Immunology 176 1–10. (https://doi.org/10.1111/cei.12248)
Peng Y, Wen D, Lin F & Mahato RI 2018 Co-delivery of siAlox15 and sunitinib for reversing the new-onset of type 1 diabetes in non-obese diabetic mice. Journal of Controlled Release 292 1–12. (https://doi.org/10.1016/J.JCONREL.2018.10.032)
Pichavaram P, Shawky NM, Hartney TJ, Jun JY & Segar L 2021 Imatinib improves insulin resistance and inhibits injury-induced neointimal hyperplasia in high fat diet-fed mice. European Journal of Pharmacology 890 173666. (https://doi.org/10.1016/J.EJPHAR.2020.173666)
Pierpaoli E, Moresi R, Orlando F, Malavolta M & Provinciali M 2016 Effect of hyperglycemia on the number of CD117 + progenitor cells and their differentiation toward endothelial progenitor cells in young and old ages. Mechanisms of Ageing and Development 159 31–36. (https://doi.org/10.1016/J.MAD.2016.02.006)
Pietras K, Sjöblom T, Rubin K, Heldin CH & Östman A 2003 PDGF receptors as cancer drug targets. Cancer Cell 3 439–443. (https://doi.org/10.1016/S1535-6108(0300089-8)
Prada PO, Ropelle ER, Moura RH, Souza De CT, Pauli JR, Rocco Sa, Rittner R, Franchini KG, Cintra DE & Velloso La et al.2009 EGFR tyrosine kinase inhibitor (PD153035) improves glucose tolerance and insulin action in high-fat diet-fed mice. Diabetes 58 2910–2919. (https://doi.org/10.2337/db08-0506.P.O.P)
Prado AF, Pernomian L, Azevedo A, Costa RAP, Rizzi E, Ramos J, Paes Leme AF, Bendhack LM, Tanus-Santos JE & Gerlach RF 2018 Matrix metalloproteinase-2-induced epidermal growth factor receptor transactivation impairs redox balance in vascular smooth muscle cells and facilitates vascular contraction. Redox Biology 18 181–190. (https://doi.org/10.1016/J.REDOX.2018.07.005)
Press MF & Lenz HJ 2007 EGFR, HER2 and VEGF pathways: validated targets for cancer treatment. Drugs 67 2045–2075. (https://doi.org/10.2165/00003495-200767140-00006)
Purvis GSD, Collino M, Aranda-Tavio H, Chiazza F, O’Riordan CE, Zeboudj L, Mohammad S, Collotta D, Verta R & Guisot NES et al.2020 Inhibition of Bruton’s TK regulates macrophage NF-κB and NLRP3 inflammasome activation in metabolic inflammation. British Journal of Pharmacology 177 4416–4432. (https://doi.org/10.1111/BPH.15182)
Racil Z, Razga F, Drapalova J, Buresova L, Zackova D, Palackova M, Semerad L, Malaskova L, Haluzik M & Mayer J 2013 Mechanism of impaired glucose metabolism during nilotinib therapy in patients with chronic myelogenous leukemia. Haematologica 98 e124–e126. (https://doi.org/10.3324/HAEMATOL.2013.086355)
Robciuc MR, Kivelä R, Williams IM, De Boer JF, Van Dijk TH, Elamaa H, Tigistu-Sahle F, Molotkov D, Leppänen VM & Käkelä R et al.2016 VEGFB/VEGFR1-induced expansion of adipose vasculature counteracts obesity and related metabolic complications. Cell Metabolism 23 712–724. (https://doi.org/10.1016/J.CMET.2016.03.004)
Roskoski R 2020 Properties of FDA-approved small molecule protein kinase inhibitors: a 2020 update. Pharmacological Research 152 104609. (https://doi.org/10.1016/j.phrs.2019.104609)
Saglio G, Kim DW, Issaragrisil S, le Coutre P, Etienne G, Lobo C, Pasquini R, Clark RE, Hochhaus A & Hughes TP et al.2010 Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia. New England Journal of Medicine 362 2251–2259. (https://doi.org/10.1056/NEJMoa0912614)
Sakuma I, Nagano H, Yoshino I, Yokote K & Tanaka T 2019 Ceritinib aggravates glycemic control in insulin-treated patients with diabetes and metastatic ALK-positive lung cancer. Internal Medicine (Tokyo, Japan) 58 817–820. (https://doi.org/10.2169/internalmedicine.1870-18)
Salaami O, Kuo CL, Drake MT, Kuchel GA, Kirkland JL & Pignolo RJ 2021 Antidiabetic effects of the senolytic agent dasatinib. Mayo Clinic Proceedings 96 3021–3029. (https://doi.org/10.1016/J.MAYOCP.2021.06.025)
Salaroli A, Loglisci G, Serrao A, Alimena G & Breccia M 2012 Fasting glucose level reduction induced by imatinib in chronic myeloproliferative disease with TEL-PDGFR? rearrangement and type 1 diabetes. Annals of Hematology 91 1823–1824. (https://doi.org/10.1007/s00277-012-1493-3)
Samaha MM, Said E & Salem HA 2019 Nilotinib enhances β-islets integrity and secretory functions in a rat model of STZ-induced diabetes mellitus. European Journal of Pharmacology 860 172569. (https://doi.org/10.1016/J.EJPHAR.2019.172569)
Samaha MM, Said E & Salem HA 2020 Modulatory role of imatinib mesylate on pancreatic β-cells’ secretory functions in an STZ rat model of diabetes mellitus. Chemico-Biological Interactions 328 109197. (https://doi.org/10.1016/J.CBI.2020.109197)
Schertzer JD, Tamrakar AK, Magalhães JG, Pereira S, Bilan PJ, Fullerton MD, Liu Z, Steinberg GR, Giacca A & Philpott DJ et al.2011 NOD1 activators link innate immunity to insulin resistance. Diabetes 60 2206–2215. (https://doi.org/10.2337/db11-0004)
Sequist LV, Soria JC, Goldman JW, Wakelee HA, Gadgeel SM, Varga A, Papadimitrakopoulou V, Solomon BJ, Oxnard GR & Dziadziuszko R et al.2015 Rociletinib in EGFR-mutated non–small-cell lung cancer. New England Journal of Medicine 372 1700–1709. (https://doi.org/10.1056/NEJMoa1413654)
Shah S, Akhtar MS, Hassan MQ, Akhtar M, Paudel YN & Najmi AK 2018 EGFR tyrosine kinase inhibition decreases cardiac remodeling and SERCA2a/NCX1 depletion in streptozotocin induced cardiomyopathy in C57/BL6 mice. Life Sciences 210 29–39. (https://doi.org/10.1016/J.LFS.2018.08.018)
Shao M, Vishvanath L, Busbuso NC, Hepler C, Shan B, Sharma AX, Chen S, Yu X, An YA & Zhu Y et al.2018 De novo adipocyte differentiation from Pdgfrβ+ preadipocytes protects against pathologic visceral adipose expansion in obesity. Nature Communications 9 890. (https://doi.org/10.1038/s41467-018-03196-x)
Sierra-Ramirez A, López-Aceituno JL, Costa-Machado LF, Plaza A, Barradas M & Fernandez-Marcos PJ 2020 Transient metabolic improvement in obese mice treated with navitoclax or dasatinib/quercetin. Aging 12 11337–11348. (https://doi.org/10.18632/AGING.103607)
Sullivan I & Planchard D 2016 Next-generation EGFR tyrosine kinase inhibitors for treating EGFR-mutant lung cancer beyond first line. Frontiers in Medicine 3 76. (https://doi.org/10.3389/fmed.2016.00076)
Sylow L, Long JZ, Lokurkar IA, Zeng X, Richter EA & Spiegelman BM 2016 The cancer drug dasatinib increases pgc-1α in adipose tissue but has adverse effects on glucose tolerance in obese mice. Endocrinology 157 4184–4191. (https://doi.org/10.1210/en.2016-1398)
Tam J, Duda DG, Perentes JY, Quadri RS, Fukumura D & Jain RK 2009 Blockade of VEGFR2 and Not VEGFR1 Can Limit Diet-Induced Fat Tissue Expansion: role of Local versus Bone Marrow-Derived Endothelial Cells. PLoS One 4 e4974. (https://doi.org/10.1371/JOURNAL.PONE.0004974)
Tao L, Yi Y, Chen Y, Zhang H, Orning P, Lien E, Jie J, Zhang W, Xu Q & Li Y et al.2021 RIP1 kinase activity promotes steatohepatitis through mediating cell death and inflammation in macrophages. Cell Death and Differentiation 28 1418–1433. (https://doi.org/10.1038/S41418-020-00668-W)
Templeton A, Brandle M, Cerny T & Gillessen S 2008 Remission of diabetes while on sunitinib treatment for renal cell carcinoma. Annals of Oncology 19 824–825. (https://doi.org/10.1093/annonc/mdn047)
Tyrrell HEJ & Pwint T 2014 Sunitinib and improved diabetes control. BMJ Case Reports [epub]. (https://doi.org/10.1136/bcr-2014-207521)
Uchida K, Takano S, Inoue G, Iwase D, Aikawa J, Takata K, Tazawa R, Kawakubo A, Sekiguchi H & Takaso M 2019 Increase in mast cell marker expression in the synovium of obese patients with osteoarthritis of the knee. Diabetes, Metabolic Syndrome and Obesity: Targets and Therapy 12 377–382. (https://doi.org/10.2147/DMSO.S201523)
Van Den Berg SM, PSeijkens TT, HKusters PJ, Beckers L, DenToom M, Smeets E, Levels J, De Winther MPJ & Lutgens E 2016 Diet-induced obesity in mice diminishes hematopoietic stem and progenitor cells in the bone marrow. The FASEB Journal 30 1779–1788. (https://doi.org/10.1096/FJ.201500175)
van Erp NP, Gelderblom H & Guchelaar HJ 2009 Clinical pharmacokinetics of tyrosine kinase inhibitors. Cancer Treatment Reviews 35 692–706. (https://doi.org/10.1016/j.ctrv.2009.08.004)
Veneri D, Franchini M & Bonora E 2005 Imatinib and regression of type 2 diabetes. New England Journal of Medicine 352 1049–1050. (https://doi.org/10.1056/NEJM200503103521023)
Vetrie D, Vořechovský I, Sideras P, Holland J, Davies A, Flinter F, Hammarström L, Kinnon C, Levinsky R & Bobrow M 1993 The gene involved in X-linked agammaglobulinaemia is a member of the src family of protein-tyrosine kinases. Nature 361 226–233. (https://doi.org/10.1038/361226A0)
Wang ML, Rule S, Martin P, Goy A, Auer R, Kahl BS, Jurczak W, Advani RH, Romaguera JE & Williams ME et al.2013b Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. New England Journal of Medicine 369 507–516. (https://doi.org/10.1056/NEJMoa1306220)
Wang XA, Deng S, Jiang D, Zhang R, Zhang S, Zhong J, Yang L, Wang T, Hong S & Guo S et al.2013a CARD3 deficiency exacerbates diet-induced obesity, hepatosteatosis, and insulin resistance in male mice. Endocrinology 154 685–697. (https://doi.org/10.1210/en.2012-1911)
Weber ANR, Bittner Z, Liu X, Dang TM, Radsak MP & Brunner C 2017 Bruton’s tyrosine kinase: an emerging key player in innate immunity. Frontiers in Immunology 8 1454. (https://doi.org/10.3389/FIMMU.2017.01454)
Wegner KW, Saleh D & Degterev A 2017 Complex pathologic roles of RIPK1 and RIPK3: moving beyond necroptosis. Trends in Pharmacological Sciences 38 202–225. (https://doi.org/10.1016/J.TIPS.2016.12.005)
Whiteman EL, Chen JJ & Birnbaum MJ 2003 Platelet-derived growth factor (PDGF) stimulates glucose transport in 3T3-L1 adipocytes overexpressing PDGF receptor by a pathway independent of insulin receptor substrates. Endocrinology 144 3811–3820. (https://doi.org/10.1210/EN.2003-0480)
Wu P, Nielsen TE & Clausen MH 2015 FDA-approved small-molecule kinase inhibitors. Trends in Pharmacological Sciences 36 422–439. (https://doi.org/10.1016/j.tips.2015.04.005)
Wu R, guang SJ, qiu WJ, Li B, Liu Q, Ning G, Jin W & Yuan Z 2017 c-Abl inhibition mitigates diet-induced obesity through improving insulin sensitivity of subcutaneous fat in mice. Diabetologia 60 900–910. (https://doi.org/10.1007/S00125-016-4202-2/FIGURES/7)
Xia CQ, Zhang P, Li S, Yuan L, Xia T, Xie C & Clare-Salzler MJ 2014 C-Abl inhibitor imatinib enhances insulin production by β cells: c-Abl negatively regulates insulin production via interfering with the expression of NKx2.2 and GLUT-2. PLoS One 9 e97694. (https://doi.org/10.1371/journal.pone.0097694)
Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS & Tartaglia LA et al.2003 Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. Journal of Clinical Investigation 112 1821–1830. (https://doi.org/10.1172/JCI19451)
Yang B, Maddison LA, Zaborska KE, Dai C, Yin L, Tang Z, Zang L, Jacobson DA, Powers AC & Chen W 2020 RIPK3-mediated inflammation is a conserved β cell response to ER stress. Science Advances 6 eabd7272. (https://doi.org/10.1126/SCIADV.ABD7272)
Yu L, Liu J, Huang X & Jiang Q 2019 Adverse effects of dasatinib on glucose-lipid metabolism in patients with chronic myeloid leukaemia in the chronic phase. Scientific Reports 9 17601. (https://doi.org/10.1038/s41598-019-54033-0)
Zdenek R, Belohlavkova P, Cetkovsky P, Faber E, Klamova H, Ludmila M, Sacha T, Jaroslava V, Zackova D & Zak P et al.2014 Comparison of glucose and lipid metabolism abnormality during nilotinib, imatinib and dasatinib therapy – results of Enigma 2 study. Blood 124 1813–1813. (https://doi.org/10.1182/blood.V124.21.1813.1813)
Zhang MZ, Wang Y, Paueksakon P & Harris RC 2014 Epidermal growth factor receptor inhibition slows progression of diabetic nephropathy in association with a decrease in endoplasmic reticulum stress and an increase in autophagy. Diabetes 63 2063–2072. (https://doi.org/10.2337/DB13-1279)
Zhao G, Dharmadhikari G, Maedler K & Meyer-Hermann M 2014 Possible role of interleukin-1β in Type 2 diabetes onset and implications for anti-inflammatory therapy strategies. PLOS Computational Biology 10 e1003798. (https://doi.org/10.1371/JOURNAL.PCBI.1003798)