ArticlePDF Available

Function of insulin‑like growth factor 1 receptor in cancer resistance to chemotherapy (Review)

Authors:
ONCOLOGY LETTERS 15: 41-47, 2018
Abstract. Drug resistance is a primary cause of chemo-
therapeutic failure; however, how this resistance develops is
complex. A comprehensive understanding of chemotherapeutic
resistance mechanisms may aid in identifying more effective
drugs and improve the survival rates of patients with cancer.
Insulin-like growth factor 1 receptor (IGF1R), a member of
the insulin receptor family, has been extensively assessed for
biological activity, and its putative contribution to tumor cell
development and progression. Furthermore, researchers have
attended to drugs that target IGF1R since IGF1R functions
as a membrane receptor. However, how IGF1R participates in
chemotherapeutic resistance remains unclear. Therefore, the
present study described the IGF1R gene and its associated
signaling pathways, and offered details of IGF1R-induced tumor
chemoresistance associated with promoting cell proliferation,
inhibition of apoptosis, regulation of ATP-binding cassette
transporter proteins and interactions with the extracellular
matrix. The present study offered additional explanations for
tumor chemotherapy resistance and provided a theoretical
basis of IGF1R and its downstream pathways for future
possible chemotherapy treatment options.
Contents
1. Introduction
2. IGF1R signaling pathway
3. IGF1R and chemotherapy resistance
4. Conclusions
1. Introduction
Insulin-like growth factor 1 receptor (IGF1R) signaling is
a complicated and regulated network essential for cells to
proliferate and survive. The IGF-IGF1R axis consists of three
receptor tyrosine kinases: IGF1R, insulin-like growth factor-2
receptor (IGF2R) and insulin receptor (INSR). The ligands for
these receptors are insulin, insulin-like growth factor-1 (IGF-1),
insulin-like growth factor-2 (IGF-2) and serum insulin-like
growth factor binding proteins (IGFBPs) (1). IGF-1 and IGF-2
possess autocrine, paracrine and endocrine functions, and
activate IGF1R signaling (2). These growth factors and their
receptors are commonly overexpressed in malignant tumors;
this overexpression may be used to assess cancer through
sustained proliferative signals, anti-apoptotic events, invasion,
metastasis and drug resistance in cancer cells (3).
IGF1R expression and activity increases in numerous
tumor types, including ovarian cancer and rhabdomyosar-
coma, and is reported to contribute to cancer cell proliferation
and apoptosis (4,5). Since IGF1R functions as a membrane
receptor, drugs, including IGF1R tyrosine kinase inhibi-
tors, monoclonal antibodies against IGF1R and monoclonal
antibodies against IGF1R ligands targeting this receptor, are
of particular interest (6). Recently, the function of IGF1R in
chemotherapeutic resistance has gained increasing attention,
and relevant mechanisms of inducing resistance in cancer cells
include overexpressing multi-drug-resistant proteins, dysregu-
lating cell survival and death and interacting with the tumor
microenvironment (7).
2. IGF1R signaling pathway
IGF1R structure and function. IGF1R is an insulin receptor
family member, and a disulde‑linked heterotetrameric trans-
membrane glycoprotein (αββα) that contains an extracellular
ligand-binding domain and an intracellular tyrosine kinase
domain (8,9). The ligand‑binding specicity determinant is
reected in the amino‑terminal cysteine‑rich domain of the
extracellular α subunit, primarily recognizing and binding to
IGF-1 and IGF-2. The intracellular signal transduction depends
on the tyrosine kinase activity the ligand in the transmem-
brane β subunit triggers, permitting specic insulin receptor
substrates (IRS-1 to -4) and Src-homology collagen (Shc)
to phosphorylate, activating downstream mitogen-activated
Function of insulin‑like growth factor 1 receptor in
cancer resistance to chemotherapy (Review)
JINGSHENG YUAN, ZHIJIE YIN, KAIXIONG TAO, GUOBING WANG and JINBO GAO
Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
Received May 30, 2017; Accepted September 28, 2017
DOI: 10.3892/ol.2017.7276
Correspondence to: Professor Jinbo Gao, Department of
Gastrointestinal Surgery, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, 1277 Jiefang
Avenue, Wuhan, Hubei 430022, P.R. China
E-mail: jgao@hust.edu.cn
Key wo rds: insulin-like growth factor 1 receptor, cancer,
chemotherapeutic resistance, mechanisms, review
YUA N et al: IGF1R AND CHEMOTHERAPEUTIC RESISTANCE
42
protein kinase (MAPK) and phosphatidylinositol 3-kinase
(PI3K)/protein kinase B (AKT) signaling pathways (6). The
specicity of IGF1R in vivo depends on tissue distribution,
ligand‑binding specicity and receptor differences in intrinsic
signaling (10).
IGF1R is often expressed in normal tissues, serving
multiple physiological functions in growth, development
and feeding (11). The importance of IGF1R in prenatal and
postnatal growth has been demonstrated using knockout
mice (8). In muscle and bone tissues, IGF1R signaling
promotes PI3K/AKT-mediated differentiation and extracel-
lular signal-regulated kinase (ERK) (12). IGF1R also aids in
the maintenance of the myocardium and brain (13).
Cardiac-specific IGF1R signaling promotes protective
physiological hypertrophy, preserving left ventricular function
and inhibiting pathological left ventricular remodeling (14).
Furthermore, IGF1R contributes to glucose metabolism and
neutrophil physiology (15), and is associated with the occur-
rence and development of cardiovascular disease, diabetes and
inammation (16,17).
IGF1R is commonly overexpressed in cancer (18). The
IGF1R signal promotes non-cancerous cells to malignantly
transform (19), and possesses anti-apoptotic and mitogenic
activity (20-22). In addition, IGF1R contributes to invasion,
metastasis and angiogenesis of cancer (23-25). Excessively
activating IGF1R promotes tumors to progress by increasing
glycolysis and biomass production (26), and decreases tumor
sensitivity to hypoxia, low pH and low glucose environ-
ments (27). In addition, expressing IGF1R increases the rate at
which tumor cells proliferate and decreases the rate at which
they are destroyed (28).
IGF1R gene regulation. The 5'‑anking region promoter of
IGF1R is enriched in GC, and lacks the effective transcription
initiation of the majority of eukaryotic genes usually requiring
TATA and CCAAT boxes. This characteristic results in a
partial difference in its gene regulation compared with other
promoter regions (29,30).
IGF1R gene expression is regulated transcriptionally and
post‑transcriptionally. Previous studies have suggested that
numerous transcription factors regulate the IGF1R gene.
Transactivation factors include zinc nger protein specicity
protein 1 (Sp1), forkhead box protein O3 (Foxo3), E2F1 tran-
scription factor, Krüppel-like factor 6, EWS RNA binding
protein 1-Wilms tumor 1 (WT1) fusion protein and high
mobility group A1, all of which bind directly to the IGF1R
promoter (31-34). In contrast, estrogen, BRCA1 DNA repair
associated (BRCA1) and von Hippel-Lindau tumor suppressor
inhibit IGF1R expression by binding to Sp1 (31,35). A previous
study confirmed that WT1 specifically binds to co-WT1
cis‑elements in the IGF1R proximal promoter region, and
decreases IGF1R gene transcription and translation (36).
Overexpressing MYB proto-oncogene transcription factor in
tumor cells increases the expression of IGF-1 and IGF1R by
increasing transcriptional activity (37).
IGF1R‑associated signaling pathways. IGF1R is associated
with multiple signaling pathways via downstream proteins,
including IRS and PI3K (38-40). IGF1R, which mediates
apoptosis-inhibiting signals, and enhances cell metabolism and
protein synthesis via downstream mechanistic ta rget of rapamycin
(MTOR) kinase signaling, activates the PI3K/AKT signaling
pathway (41-43). IGF1R activates the growth factor receptor
bound protein 2 (Grb2)/RAS/RAF/MAPK signaling pathway
to transduce cell growth and proliferation signals (44,45).
IGF1R activation or overexpression is associated with invasion
and metastasis of cancer cells, processes mediated by numerous
signal transduction proteins that affect invasiveness (24,25).
For example, phosphorylating IRS-1 affects the interactions
between epithelial cadherin and β-catenin, and the crosstalk
between the IGF axis and integrins (46). A previous study
demonstrated that protein tyrosine kinase 6 forms a complex
with IGF1R and the adaptor protein IRS-1, which modulates
anchorage-independent growth via the regulation of IGF1R
expression and phosphorylation (23).
Previously, crosstalk between IGF1R and other signaling
pathways has been assessed, with studies focusing on interac-
tions between IGF1R, steroid hormones and other receptor
tyrosine kinases (RTKs) (47). The crosstalk between IGF1R
and focal adhesion kinase (FAK) signaling pathways (38),
IGF1R and the classical Wnt signaling pathways (48,49), and
IGF1R and transforming growth factor β (TGFβ) signaling
pathways have also been further claried (50). In addition,
certain IGF1R signals have been newly identified, namely
RTK heterodimers, including the INSR hybrid receptor, and
IGF1R/INSR that function as dependent receptors intervening
in IGF1R signaling and its regulation (51).
The IGF1R signaling pathway is regulated at multiple
levels; the expression of IGF-2, the presence of IGF2R and
high‑afnity IGFBPs affects ligand‑binding activity (52). In
addition, other extracellular factors, including dendritic cells
and integrins, may contribute to regulating IGF1R activity (53).
Within cells, Notch and apoptosis inducing factor-1 regulates
IGF1R kinase activity (54). Downstream, multiple IGF1R
effectors participate in IRS/PI3K/AKT signal transmission,
including MTOR complex 1, phosphatase and tensin homolog
phosphohydrolase, ribosomal protein S6 kinases, ERK and
c-Jun N-terminal kinase (5,55,56).
3. IGF1R and chemotherapy resistance
Overexpression of IGF1R is associated with poorer chemo-
therapy outcomes for patients with gastric cancer compared
with those with low expression of IGF1R (57). Patients with
co-expression of IGF1R and multi-drug resistance-associated
protein 1 (MRP1) have demonstrated a poorer response with
adjuvant FOLFOX-4 chemotherapy (58). In patients with
human epidermal growth factor receptor 2-negative breast
cancer, the decreased expression of IGF1R was correlated
with an improved response to chemotherapy (59). Blocking
IGF1R signaling facilitates treating bladder cancer cells that
are insensitive to chemotherapy (60). Similar phenomena
have been reported for prostate and ovarian cancer when
IGF1R signaling is blocked (61,62). Although the function
of IGF1R in chemotherapy resistance has been conrmed,
the mechanism remains to be fully elucidated. The present
study assessed IGF1R-associated tolerance mechanisms from
multiple aspects, including promoting proliferation, inhibiting
apoptosis, and inducing changes to ATP-binding cassette
ONCOLOGY LETTERS 15: 41-47, 2018 43
(ABC) transporter proteins and the extracellular matrix
(ECM) (Fig. 1).
Promoting proliferation. A characteristic of tumor cells,
persistent proliferation may be acquired in multiple ways (3).
As chemotherapeutic resistance develops, certain signals
elevate receptor proteins on tumor cell surfaces and permit
cells to avoid growth signal control (57). Changing the recep-
tor's molecular structure, which alters ligand restriction and
promotes the downstream signal to activate, may achieve the
same effect (63).
The Grb2/RAS/RAF/MAPK cascades serve crucial func-
tions in cell proliferation and survival and are aberrantly
activated in drug-tolerant cells. Numerous mechanisms
increase IGF1R expression and activate IGF1R, thereby
promoting signaling cascades and proliferation (64). WT1 is
reportedly silenced in drug-resistant cells, which may degrade
the inhibitory effect of WT1 on IGF1R transcription (65).
Similar effects are reected in the feedback loop between
Foxo3, IGF1R and AKT (31). Micro (mi) RNA inhibits IGF1R
expression by directly targeting the 3' untranslated regions
but CpG methylating the miRNA promoter region results in
the downregulation, and the loss of the inhibitory effects, of
IGF1R expression (66,67). MIR-143, MIR-503 and MIR-1271
regulate cisplatin resistance in human gastric cancer cell
lines by targeting IGF1R (66-69). Normally, insulin-like
growth factor binding protein-7 (IGFBP7) directly binds
to IGF1R and inhibits its function post-transcriptionally;
however, studies indicate that, in chemotherapy-resistant cells,
IGFBP7 expression signicantly decreased (70). Therefore,
IGF1R is overactivated once IGFBP7 inhibitory activity
has decreased (71). Furthermore, inactivating IGF1R inhibits
tumor cell proliferation by blockading G0/G1 and IGF1R binds
to non-IGF ligands from extracellular spaces, cell membranes
and the cytoplasm, which regulates cell proliferation and
survival IGF-independently during chemoresistance (72,73).
In addition to overexpression, IGF1R over-activation is
also important with respect to chemotherapeutic tolerance.
Phosphorylated IGF1R increased in chemotherapeutic
drug-resistant cell lines (74-76) and multiple mechanisms
contribute to the over-activation of IGF1R, including
increased constitutively secreted IGF-1 (63), transgelin
overexpression (77) and the effect of the Src oncogene on
IGF1R (78). By these processes, IGF1R signals promote tumor
cells to proliferate and induce resistance by over-activating
Grb2/RAS/RAF/MAPK cascades (64).
Inhibiting apoptosis. Anti-apoptosis is common to numerous
tumors and chemotherapy-resistant cells evolve diverse
strategies to limit or avoid apoptosis (3). The most common
strategy is to eliminate the tumor suppressor function of
p53 (79). Resistant cells also downregulate pro-apoptotic
Figure 1. IGF1R signaling pathway and its relevant drug resistance mechanisms: Promoting proliferation, inhibiting apoptosis and inducing changes to ABC
transporter proteins and the ECM. Silencing WT1 and mutant p53 causes loss of the inhibitory effects of the IGF1R promoter. Downregulating microRNAs,
including miR-143, miR-503, miR-1271, causes the loss of IGF1R mRNA degradation and IGF1R translation inhibitory activity. Serum insulin-like growth
factor binding proteins decrease the inhibitory effects of IGF1R post-transcriptionally, increasing IGF1R expression and activity. This may promote down-
stream phosphatidylinositol 3-kinase/protein kinase B and Grb2/RAS/RAF/mitogen-activated protein kinase signaling cascades, thereby enhancing cell
proliferation and anti-apoptotic activity. In addition, IGF1R signaling pathways participate in regulating ABC genes and alter cell responses to chemotherapy.
The ECM and IGF1R stabilize and activate the activity of one another. IGF1R, insulin-like growth factor 1 receptor; ABC, ATP-binding cassette; ECM,
extracellular matrix; WT1, Wilms tumor 1; miR, microRNA; Grb2, growth factor receptor bound protein 2.
YUA N et al: IGF1R AND CHEMOTHERAPEUTIC RESISTANCE
44
factors or increase the expression of anti-apoptotic factors to
avoid apoptosis (80). IGF1R participates in apoptosis inhibi-
tion predominantly via the PI3K/AKT signaling pathway in
drug-resistant cell lines but multiple other mechanisms are
associated with IGF1R overexpression and inhibition of apop-
tosis in drug-resistant cells (81,82).
Previous studies have indicated that cancer chemotherapy
is associated with inducing p53-dependent apoptosis
responses (79). p53 is one of the most frequently mutated tumor
suppressors and IGF1R overexpression inhibits wild-type p53
(WT-p53) via phosphorylated (p) AKT (80). This enhances the
ubiquitination-promoting function of murine double minute 2,
which decreases p53 protein production (79). Reciproca l l y,
WT-p53 renders tumor cells more chemosensitive by
inhibiting Sp1-induced transactivation of the IGF1R promoter
and increasing the expression of pro-apoptotic protein
p21 (81). Mutant p53 stimulates IGF1R promoter function in
chemotherapeutic resistant cell lines (82). Furthermore, IGF1R
regulates cisplatin resistance by targeting proto- oncogene Bcl‑2,
which is anti-apoptotic and affects drug resistance by binding
to and inhibiting Bcl 2-associated X protein (BAX) and Bcl 2
homologous antagonist killer protein (83). IGF1R activation
is also associated with decreased expression of IGFBP7,
which is associated with the expression of the anti-apoptotic
gene Bim and chemotherapy tolerance-associated genes,
including annexin A4 and protein kinase C 1 (84). Conversely,
overexpressing IGFBP7 induces apoptosis and reverses tumor
drug resistance (70).
Regulating ABC transporter proteins. The ABC is the largest
protein transporter superfamily present in all organisms (85).
This family of genes codes for different proteins (importers
and exporters) and its increased expression decreases
drug influx and increases efflux, decreasing therapeutic
response (86). IGF1R signals participate in regulating ABC
genes, including multidrug resistance protein 1 (MDR1),
MRP1, multidrug resistance-associated protein 2 (MRP2),
multidrug resistance-associated protein 3 (MRP3) and
breast cancer resistance protein (BCRP) (59,87-89). As such,
IGF1R increases tumor resistance by increasing the expres-
sion of MDR1, a protein implicated in chemotherapeutic
resistance (88). Expression of MRP3 and BCRP decreases
or disappears in the presence of an IGF1R inhibitor (87) and
overexpressing IGF1R results in increased MRP2 promoter
activity via increased pAKT and nuclear factor erythroid
2-related factor 2 in resistant cells (59,88). In addition,
IGF1R silencing increases chemotherapeutic sensitivity via
transcription inhibition of MRP-2 (59). Previous studies have
demonstrated that overexpressing IGF1R and MRP1 was asso-
ciated with chemotherapeutic resistance and poorer prognosis
compared with malignancies with normal or low expression
of IGF1R and MRP1, indicating that the co-expression of
IGF1R/MRP1 in tumors may predict chemotherapeutic
effects (88,89).
Interacting with ECM. The ECM is predominantly composed
of brin (collagen and laminin) and proteoglycans (hyaluronic
acid), which forms the structural framework for the majority
of tissues (90). The ECM transfer signals to the cells via inte-
grin binding and activation, which modulate cell proliferation,
survival and migration and inuence the tumor response to
anti-cancer therapies (91,92).
Previous studies have indicated that IGF1R stabilizes
the molecular structure of β1 integrin by protecting it from
proteasomal degradation and promoting tumor cells to grow
and proliferate (93). FAK, a substrate protein of IGF1R, is
activated by integrin, affecting epithelial transformation, inva-
sion and metastasis of tumor cells IGF1R-independently (38).
Extracellular bronectin increases the activity of β1 integrin
to increase the abundance of MAPK-phosphatase-1 and the
receptor of activated C kinase (RACK-1) (62). In addition,
establishing crosstalk between β1 integrin and IGF1R retains
the phosphorylation of IGF1R, which helps stimulate down-
stream signaling of IGF1R, and contributes to cell proliferation
and transformation (94). Previous studies have revealed that, in
the presence of IGF1R, the β1 integrin receptor increased the
recruitment of RACK-1 and mediated tumor cell migration (62).
These changes contribute to chemotherapeutic tolerance.
Other mechanisms. Previous studies have revealed that IGF1R
is sumoylated and translocated to the nucleus, which permits
the receptor to interact with chromatin, and function as a
transcriptional regulator (95-97). Nuclear IGF1R specically
binds to and functions as a transcriptional activator of its
own promoter, and interferes with signaling pathways (98).
Specifically, nuclear IGF1R interferes with Wnt signaling,
which upregulates ABC drug transporters and modulates drug
responses (99). Regarding the tumor microenvironment, acti-
vating IGF1R results in stabilizing hypoxia-inducible factor
(H I F)-1α and HIF-2α, and the upregulation of vascular endo-
thelial growth factor (100). A previous study demonstrated
that overexpressing HIF-1α increased the expression of Bcl-2,
decreased the expression of BAX, and induced the expression
of MDR1 and MRP1 (101). These results offer novel insights
into IGF1R-mediated chemotherapeutic resistance.
4. Conclusions
Chemotherapeutic resistance commonly results in cancer
treatment failing, with previous studies conrming multiple
resistance-associated mechanisms (102,103). Therefore,
understanding how tumors develop resistance may help to
identify improved drugs and increase patient survival rates.
Changes in drug transporter proteins, activating signaling
pathways and ineffectively inducing cell death are primary
mechanisms of chemotherapeutic resistance. IGF1R-mediated
resistance includes promoting cells to proliferate, inhibiting
apoptosis, inducing increased expression of ABC transporter
proteins on cell membranes and inducing changes in the ECM.
Transcription factors and miRNAs also intervene in regulating
IGF1R transcriptionally and cause downstream signaling path-
ways to excessively activate by promoting increased IGF1R
expression or loss of inhibitory effects to the IGF1R promoter.
IGFBPs participate in regulating IGF1R post-transcriptionally,
with the loss of IGF1R inhibition and enhanced expression
of anti-apoptotic and chemotherapy resistance-associated
genes. Following overexpression and over-activation, IGF1R
predominantly triggers the Grb2/RAS/RAF/MAPK and
PI3K/AKT cascades, which induce proliferation and inhibit
apoptosis in chemotherapy-resistant tumor cell lines. IGF1R
ONCOLOGY LETTERS 15: 41-47, 2018 45
signaling regulates the expression of ABC transporter proteins
via multiple mechanisms and renders chemotherapy less effec-
tive. The ECM interacts synergistically with IGF1R activity
as chemotherapy-resistant cells develop; however, how this
occurs remains unclear.
Overall, IGF1R signaling serves a crucial function in tumor
chemotherapeutic tolerance. Recently, drug combinations that
target predicted or identied chemoresistance markers have
been suggested as the future direction of cancer treatment. As
a membrane receptor, IGF1R is of particular interest in cancer
drug targeting; however, IGF1R-mediated resistance mecha-
nisms require further study. Furthermore, RTK heterodimer
and IGF1R nuclear translocation may be associated with drug
resistance, though few reports of this exist in the literature.
Acknowledgements
The present study was supported by the National Natural
Science Foundation of China (grant no. 81572411).
References
1. Cascieri MA and Bayne ML: Analysis of the interaction of IGF-I
analogs with the IGF-I receptor and IGF binding proteins. Adv
Exp Med Biol 343: 33-40, 1993.
2. Hayashi Y, Asuzu DT, Gibbons SJ, Aarsvold KH, Bardsley MR,
Lomberk GA, Mathison AJ, Kendrick ML, Shen KR,
Taguchi T, et al: Membrane-to-nucleus signaling links
insulin-like growth factor-1- and stem cell factor-activated
pathways. PLoS One 8: e76822, 2013.
3. Hanahan D and Weinberg RA: Hallmarks of cancer: The next
generation. Cell 144: 646-674, 2011.
4. Amutha P and Rajkumar T: Role of insulin-like growth factor,
insulin-like growth factor receptors, and insulin-like growth
factor-binding proteins in ovarian cancer. Indian J Med Paediatr
Oncol 38: 198-206, 2017.
5. Wan X, Harkavy B, Shen N, Grohar P and Helman LJ:
Rapamycin induces feedback activation of akt signaling through
an igf-1R-dependent mechanism. Oncogene 26: 1932-1940, 2007.
6. Chen HX and Sharon E: IGF-1R as an anti-cancer target-trials
and tribulations. Chin J Cancer 32: 242-252, 2013.
7. Shi WJ and Gao JB: Molecular mechanisms of chemoresistance
in gastric cancer. World J Gastrointest Oncol 8: 673-681, 2016.
8. Liu JP, Baker J, Perkins AS, Robertson EJ and Efstratiadis A:
Mice carrying null mutations of the genes encoding insulin-like
growth factor I (Igf-1) and type 1 IGF receptor (Igf1r). Cell 75:
59-72, 1993.
9. Ward CW and Garrett TP: Structural relationships between the
insulin receptor and epidermal growth factor receptor families
and other proteins. Curr Opin Drug Discov Devel 7: 630-638,
2004.
10. Siddle K: The insulin receptor and type I IGF receptor:
Comparison of structure and function. Prog Growth Factor
Res 4: 301-32 0, 1992.
11. Wit JM and Walenkamp MJ: Role of insulin-like growth factors
in growth, development and feeding. World Rev Nutr Diet 106:
60-65, 2013.
12. Yakar S, Kim H, Zhao H, Toyoshima Y, Pennisi P, Gavrilova O
and Leroith D: The growth hormone-insulin like growth factor
axis revisited: Lessons from IGF-1 and IGF-1 receptor gene
targeting. Pediatr Nephrol 20: 251-254, 2005.
13. Russo VC, Gluckman PD, Feldman EL and Werther GA: The
insulin-like growth factor system and its pleiotropic functions in
brain. Endocr Rev 26: 916-943, 2005.
14. Laustsen PG, Russell SJ, Cui L, Entingh-Pearsall A,
Holzenberger M, Liao R and Kahn CR: Essential role of insulin
and insulin-like growth factor 1 receptor signaling in cardiac
development and function. Mol Cell Biol 27: 1649-166 4, 2007.
15. Moody G, Beltran PJ, Mitchell P, Cajulis E, Chung YA,
Hwang D, Kendall R, Radinsky R, Cohen P and Calzone FJ:
IGF1R blockade with ganitumab results in systemic effects on
the GH-IGF axis in mice. J Endocrinol 221: 145-155, 2 014.
16. Delafontaine P, Song YH and Li Y: Expression, regulation, and
function of IGF-1, IGF-1R, and IGF-1 binding proteins in blood
vessels. Arterioscler Thromb Vasc Biol 24: 435-444, 2004.
17. Gao S, Wassler M, Zhang L, Li Y, Wang J, Zhang Y, Shelat H,
Williams J and Geng YJ: MicroRNA-133a regulates insulin-like
growth factor-1 receptor expression and vascular smooth muscle
cell proliferation in murine atherosclerosis. Atherosclerosis 232:
171-179, 2014.
18. Werner H: Tumor suppressors govern insulin-like growth factor
signaling pathways: Implications in metabolism and cancer.
Oncogene 31: 2703-2714, 2012.
19. Schayek H, Bentov I, Sun S, Plymate SR and Werner H:
Progression to metastatic stage in a cellular model of prostate
cancer is associated with methylation of the androgen receptor
gene and transcriptional suppression of the insulin-like growth
factor-I receptor gene. Exp Cell Res 316: 1479 -1488, 2010.
20. O'Connor R, Kauffmann-Zeh A, Liu Y, Lehar S, Evan GI,
Baserga R and Blättler WA: Identication of domains of the
insulin-like growth factor I receptor that are required for protec-
tion from apoptosis. Mol Cell Biol 17: 427-435, 1997.
21. Resnicoff M, Abraham D, Yutanawiboonchai W, Rotman HL,
Kajstura J, Rubin R, Zoltick P and Baserga R: The insulin-like
growth factor I receptor protects tumor cells from apoptosis
in vivo. Cancer Res 55: 2463-2469, 1995.
22. Valentinis B and Baserga R: The IGF-I receptor protects tumor
cells from apoptosis induced by high concentrations of serum.
Biochem Biophys Res Commun 224: 362-368, 1996.
23. Baserga R: The IGF-I receptor in cancer research. Exp Cell
Res 253: 1-6, 1999.
24. Samani AA, Yakar S, LeRoith D and Brodt P: The role of the IGF
system in cancer growth and metastasis: Overview and recent
insights. Endocr Rev 28: 20-47, 2007.
25. Pollak M: The insulin and insulin-like growth factor receptor
family in neoplasia: An update. Nat Rev Cancer 12: 159-169,
2012.
26. Ter Braak B, Siezen CL, Lee JS, Rao P, Voorhoeve C, Ruppin E,
van der Laan JW and van de Water B: Insulin-like growth factor
1 receptor activation promotes mammary gland tumor develop-
ment by increasing glycolysis and promoting biomass production.
Breast Cancer Res 19: 14, 2017.
27. Peretz S, Kim C, Rockwell S, Baserga R and Glazer PM: IGF1
receptor expression protects against microenvironmental stress
found in the solid tumor. Radiat Res 158: 174 -18 0, 2002.
28. Werner H, Sarfstein R, LeRoith D and Bruchim I: Insulin-like
growth factor 1 signaling axis meets p53 genome protection
pathways. Front Oncol 6: 159, 2016.
29. Mamula PW and Goldne ID: Cloning and characterization of
the human insulin‑like growth factor‑I receptor gene 5'‑anking
region. DNA Cell Biol 11: 43-50, 1992.
30. Werner H, Bach MA, Stannard B, Roberts CT Jr and LeRoith D:
Structural and functional analysis of the insulin-like growth
factor I receptor gene promoter. Mol Endocrinol 6: 1545-1558,
1992.
31. Singh RK, Gaikwad SM, Jinager A, Chaudhury S, Maheshwari A
and Ray P: IGF-1R inhibition potentiates cytotoxic effects
of chemotherapeutic agents in early stages of chemoresistant
ovarian cancer cells. Cancer Lett 354: 254-262, 2014.
32. Schayek H, Bentov I, Rotem I, Pasmanik-Chor M, Ginsberg D,
Plymate SR and Werner H: Transcription factor E2F1 is a
potent transactivator of the insulin-like growth factor-I receptor
(IGF-IR) gene. Growth Horm IGF Res 20: 68-72, 2010.
33. Huo X, Liu S, Shao T, Hua H, Kong Q, Wang J, Luo T and
Jiang Y: GSK3 protein positively regulates type I insulin-like
growth factor receptor through forkhead transcription factors
FOXO1/3/4. J Biol Chem 289: 24759-72470, 2014.
34. Fin keltov I, Kuhn S, Glaser T, Idelman G, Wright JJ, Roberts CT Jr
and Werner H: Transcriptional regulation of IGF-I receptor gene
expression by novel isoforms of the EWS-WT1 fusion protein.
Oncogene 21: 1890-1898, 2002.
35. Werner H and Sarfstein R: Transcriptional and epigenetic control
of IGF1R gene expression: Implications in metabolism and
cance. Growth Horm IGF Res 24: 112-118, 2014.
36. Idelman G, Glaser T, Roberts CT Jr and Werner H: WT1-p53
interactions in insulin-like growth factor-I receptor gene regula-
tion. J Biol Chem 278: 3474-3482, 2003.
37. Kim SO, Park JG and Lee YI: Increased expression of the
insulin-like growth factor I (IGF-I) receptor gene in hepatocel-
lular carcinoma cell lines: Implications of IGF-I receptor gene
activation by hepatitis B virus X gene product. Cancer Res 56:
3831-3836, 1996.
YUA N et al: IGF1R AND CHEMOTHERAPEUTIC RESISTANCE
46
38. Taliaferro-Smith L, Oberlick E, Liu T, McGlothen T, Alcaide T,
Tobin R, Donnelly S, Commander R, Kline E, Nagaraju GP, et al:
FAK activation is required for IGF1R-mediated regulation of
EMT, migration, and invasion in mesenchymal triple negative
breast cancer cells. Oncotarget 6: 4757-4772, 2015.
39. Dearth RK, Cui X, Kim HJ, Hadsell DL and Lee AV: Oncogenic
transformation by the signaling adaptor proteins insulin
receptor substrate (IRS)-1 and IRS-2. Cell Cycle 6: 705-713,
2007.
40. Koval AP, Blakesley VA, Roberts CT Jr, Zick Y and Leroith D:
Interaction in vitro of the product of the c-Crk-II proto-oncogene
with the insulin-like growth factor I receptor. Biochem J 330:
923-932, 1998.
41. Kulik G and Weber MJ: Akt- dependent and -independent survival
signaling pathways utilized by insulin-like growth factor I. Mol
Cell Biol 18: 6711-6718, 1998.
42. Peruzzi F, Prisco M, Dews M, Salomoni P, Grassilli E, Romano G,
Calabretta B and Baserga R: Multiple signaling pathways of the
insulin-like growth factor 1 receptor in protection from apop-
tosis. Mol Cell Biol 19: 7203-7215, 1999.
43. Brazil DP, Yang ZZ and Hemmings BA: Advances in protein
kinase B signalling: AKTion on multiple fronts. Trends Biochem
Sci 29: 233-242, 2004.
44. Pollak M: Insulin, insulin-like growth factors and neoplasia. Best
Pract Res Clin Endocrinol Metab 22: 625-638, 2008.
45. Greer EL and Brunet A: FOXO transcription factors at the inter-
face between longevity and tumor suppression. Oncogene 24:
7410-7425, 20 05.
46. Playford MP, Bicknell D, Bodmer WF and Macaulay VM:
Insulin-like growth factor 1 regulates the location, stability,
and transcriptional activity of beta-catenin. Proc Natl Acad Sci
USA 97: 12103-12108, 2000.
47. Liu C, Zhang Z, Tang H, Jiang Z, You L and Liao Y: Crosstalk
between IGF-1R and other tumor promoting pathways. Curr
Pharm Des 20: 2912-2921, 2014.
48. Rota LM, Albanito L, Shin ME, Goyeneche CL, Shushanov S,
Gallagher EJ, LeRoith D, Lazzarino DA and Wood TL: IGF1R
inhibition in mammary epithelia promotes canonical Wnt
signaling and Wnt1-driven tumors. Cancer Res 74: 5668-5679,
2014.
49. Zhang QY, Wang L, Song ZY and Qu XJ: Knockdown of type I
insulin-like growth factor receptor inhibits human colorectal
cancer cell growth and downstream PI3K/Akt, WNT/β-catenin
signal pathways. Biomed Pharmacother 73: 12-18, 2015.
50. Alsina-Sanchis E, Figueras A, Lahiguera Á, Vidal A,
Casanovas O, Graupera M, Villanueva A and Vinals F: The
TGFβ pathway stimulates ovarian cancer cell proliferation by
increasing IGF1R levels. Int J Cancer 139: 1894-4903, 2016.
51. Tognon CE and Sorensen PH: Targeting the insulin-like growth
factor 1 receptor (IGF1R) signaling pathway for cancer therapy.
Expert Opin Ther Targets 16: 33-48, 2012.
52. Firth SM and Baxter RC: Cellular actions of the insulin-like
growth factor binding proteins. Endocr Rev 23: 824-854,
2002.
53. Triplett TA, Cardenas KT, Lancaster JN, Hu Z, Selden HJ,
Jasso GJ, Balasubramanyam S, Chan K, Li L, Chen X, et al:
Endogenous dendritic cells from the tumor microenvironment
support T-ALL growth via IGF1R activation. Proc Natl Acad Sci
USA 113: E1016-E1025, 2016.
54. Medyouf H, Gusscott S, Wang H, Tseng JC, Wai C, Nemirovsky O,
Trumpp A, Pumio F, Carboni J, Gottardis M, et al: High-level
IGF1R expression is required for leukemia-initiating cell activity
in T-ALL and is supported by Notch signaling. J Exp Med 208:
1809-1822, 2011.
55. Manning BD and Cantley LC: AKT/PKB signaling: Navigating
downstream. Cell 129: 1261-1274, 2007.
56. Kim EK, Kim JH, Kim HA, Seol H, Seong MK, Lee JY, Byeon J,
Sohn YJ, Koh JS, Park IC and Noh WC: Phosphorylated S6
kinase-1: A breast cancer marker predicting resistance to
neoadjuvant chemotherapy. Anticancer Res 33: 4073-4079,
2013.
57. Sui P, Cao H, Meng L, Hu P, Ma H and Du J: The synergistic
effect of humanized monoclonal antibodies targeting insulin-like
growth factor 1 receptor (IGF-1R) and chemotherapy. Curr Drug
Targets 15: 674-680, 2014.
58. Ge J, Chen Z, Wu S, Chen J, Li X, Li J, Yin J and Chen Z:
Expression levels of insulin-like growth factor-1 and multidrug
resistance-associated protein-1 indicate poor prognosis in
patients with gastric cancer. Digestion 80: 148-158, 2009.
59. de Groot S, Charehbili A, van Laarhoven HW, Mooyaart AL,
Dekker-Ensink NG, van de Ven S, Janssen LG, Swen JJ, Smit VT,
Heijns JB, et al: Insulin-like growth factor 1 receptor expres-
sion and IGF1R 3129G > T polymorphism are associated with
response to neoadjuvant chemotherapy in breast cancer patients:
Results from the NEOZOTAC trial (BOOG 2010-01). Breast
Cancer Res 18: 3, 2016.
60. Sun HZ, Wu SF and Tu ZH: Blockage of IGF-1R signaling
sensitizes urinary bladder cancer cells to mitomycin-mediated
cytotoxicity. Cell Res 11: 10 7-115, 2001.
61. Thomas F, Holly JM, Persad R, Bahl A and Perks CM: Fibronectin
confers survival against chemotherapeutic agents but not against
radiotherapy in DU145 prostate cancer cells: involvement of the
insulin like growth factor-1 receptor. Prostate 70: 856-865, 2010.
62. Eckstein N, Servan K, Hildebrandt B, Pölitz A, von Jonquières G,
Wolf-Kümmeth S, Napierski I, Hamacher A, Kassack MU,
Budczies J, et al: Hyperactivation of the insulin-like growth factor
receptor I signaling pathway is an essential event for cisplatin resis-
tance of ovarian cancer cells. Cancer Res 69: 2996-3003, 2009.
63. Montazami N, Aghapour M, Farajnia S and Baradaran B: New
insights into the mechanisms of multidrug resistance in cancers.
Cell Mol Biol (Noisy-le-grand) 61: 70-80, 2015.
64. Worrall C, Nedelcu D, Serly J, Suleymanova N, Oprea I,
Girnita A and Girnita L: Novel mechanisms of regulation of
IGF-1R action: Functional and therapeutic implications. Pediatr
Endocrinol Rev 10: 473-484, 2013.
65. Chen MY, Clark AJ, Chan DC, Ware JL, Holt SE,
Chidambaram A, Fillmore HL and Broaddus WC: Wilms'
tumor 1 silencing decreases the viability and chemoresistance
of glioblastoma cells in vitro: A potential role for IGF-1R
de-repression. J Neurooncol 103: 87-102, 2011.
66. Qian X, Yu J, Yin Y, He J, Wang L, Li Q, Zhang LQ, Li CY,
Shi ZM. Xu Q, et al: MicroRNA-143 inhibits tumor growth and
angiogenesis and sensitizes chemosensitivity to oxaliplatin in
colorectal cancers. Cell Cycle 12: 1385-1394, 2013.
67. Wang T, Ge G, Ding Y, Zhou X, Huang Z, Zhu W, Shu Y and
Liu P: MiR-503 regulates cisplatin resistance of human gastric
cancer cell lines by targeting IGF1R and BCL2. Chin Med J
(Engl) 127: 2357-2362, 2014.
68. Yang M, Shan X, Zhou X, Qiu T, Zhu W, Ding Y, Shu Y and
Liu P: miR-1271 regulates cisplatin resistance of human gastric
cancer cell lines by targeting IGF1R, IRS1, mTOR, and BCL2.
Anticancer Agents Med Chem 14: 884-891,2014.
69. Zhuang M, Shi Q, Zhang X, Ding Y, Shan L, Shan X, Qian J,
Zhou X, Huang Z, Zhu W, et al: Involvement of miR-143 in
cisplatin resistance of gastric cancer cells via targeting IGF1R
and BCL2. Tumour Biol 36: 2737-2745, 2015.
70. Verhagen HJ, de Leeuw DC, Roemer MG, Denkers F, Pouwels W,
Rutten A, Celie PH, Ossenkoppele GJ, Schuurhuis GJ and Smit L:
IGFBP7 induces apoptosis of acute myeloid leukemia cells and
synergizes with chemotherapy in suppression of leukemia cell
survival. Cell Death Dis 5: e1300, 2014.
71. Baxter RC: Insulin-like growth factor (IGF)-binding proteins:
interactions with IGFs and intrinsic bioactivities. Am J Physiol
Endocrinol Metab 278: E967-E976, 2000.
72. Bach LA: Insulin-like growth factor binding proteins-an update.
Pediatr Endocrinol Rev 13: 521-530, 2015.
73. Beattie J, Allan GJ, Lochrie JD and Flint DJ: Insulin-like growth
factor-binding protein-5 (IGFBP-5): A critical member of the
IGF axis. Biochem J 395: 1-19, 2006.
74. Beech DJ, Perer E, Helms J, Gratzer A and Deng N: Insulin-like
growth factor-I receptor activation blocks doxorubicin cytotox-
icity in sarcoma cells. Oncol Rep 10: 181-184, 2003.
75. Dallas NA, Xia L, Fan F, Gray MJ, Gaur P, van Buren G II,
Samuel S, Kim MP, Lim SJ and Ellis LM: Chemoresistant
colorectal cancer cells, the cancer stem cell phenotype, and
increased sensitivity to insulin-like growth factor-I receptor
inhibition. Cancer Res 69: 1951-1957, 2009.
76. Sun Y, Zheng S, Torossian A, Speirs CK, Schleicher S,
Giacalone NJ, Carbone DP, Zhao Z and Lu B: Role of insulin-like
growth factor-1 signaling pathway in cisplatin-resistant lung
cancer cells. Int J Radiat Oncol Biol Phys 82: e563-e572, 2012.
77. Kim TR, Cho EW, Paik SG and Kim IG: Hypoxia-induced SM22α
in A549 cells activates the IGF1R/PI3K/Akt pathway, conferring
cellular resistance against chemo- and radiation therapy. FEBS
Lett 586: 303-309, 2012.
78. Werner H and Le Roith D: New concepts in regulation and function
of the insulin-like growth factors: Implications for understanding
normal growth and neoplasia. Cell Mol Life Sci 57: 932-942, 2000.
ONCOLOGY LETTERS 15: 41-47, 2018 47
79. Tracz AF, Szczylik C, Porta C and Czarnecka AM: Insulin-like
growth factor-1 signaling in renal cell carcinoma. BMC
Cancer 16: 453, 2016.
80. Buck E and Mulvihill M: Small molecule inhibitors of the
IGF-1R/IR axis for the treatment of cancer. Expert Opin Investig
Drugs 20: 605-621, 2011.
81. Ozaki T and Nakagawara A: P53: The attractive tumor suppressor
in the cancer research eld. J Biomed Biotechnol 2011: 603925,
2011.
82. Larsson O, Girnita A and Girnita L: Role of insulin-like growth
factor 1 receptor signalling in cancer. Br J Cancer 96 (Suppl):
R2-R6, 2007.
83. Adams JM and Cory S: Bcl-2-regulated apoptosis: Mechanism
and therapeutic potential. Curr Opin Immunol 19: 488-496, 2007.
84. Kashyap MK: Role of insulin-like growth factor-binding
proteins in the pathophysiology and tumorigenesis of gastro-
esophageal cancers. Tumour Biol 36: 8247-8257, 2015.
85. Jiang ZS, Sun YZ, Wang SM and Ruan JS: Epithelial-mesenchymal
transition: Potential regulator of ABC transporters in tumor
progression. J Cancer 8: 2319-2327, 2017.
86. Zhang YK, Wang YJ, Gupta P and Chen ZS: Multidrug resistance
proteins (MRPs) and cancer therapy. AAPS J 17: 802-812, 2015.
87. Benabbou N, Mirshahi P, Bordu C, Faussat AM, Tang R,
Therwath A, Soria J, Marie JP and Mirshahi M: A subset of
bone marrow stromal cells regulate ATP-binding cassette gene
expression via insulin-like growth factor-I in a leukemia cell
line. Int J Oncol 45: 1372-1380, 2014.
88. Shen K, Cui D, Sun L, Lu Y, Han M and Liu J: Inhibition of
IGF-IR increases chemosensitivity in human colorectal cancer
cells through MRP-2 promoter suppression. J Cell Biochem 113:
2086-2097, 2012.
89. Benabbou N, Mirshahi P, Cadillon M, Soria J, Therwath A
and Mirshahi M: Hospicells promote upregulation of the
ATP-binding cassette genes by insulin-like growth factor-I via
the JAK2/STAT3 signaling pathway in an ovarian cancer cell
line. Int J Oncol 43: 685-694, 2013.
90. Gilkes DM, Semenza GL and Wirtz D: Hypoxia and the
extracellular matrix: Drivers of tumour metastasis. Nat Rev
Cancer 14: 430-439, 2014.
91. Elliott T and Sethi T: Integrins and extracellular matrix: A novel
mechanism of multidrug resistance. Expert Rev Anticancer
Ther 2: 449-459, 2002.
92. L Addison C: Modulation of response to tumor therapies by the
extracellular matrix. Future Oncol 2: 417-429, 2006.
93. Sayeed A, Fedele C, Trerotola M, Ganguly KK and Lang uino LR:
IGF-IR promotes prostate cancer growth by stabilizing α5β1
integrin protein levels. PLoS One 8: e76513, 2013.
94. Prokop I, Konończuk J, Surażyński A and Pałka J: Cross-talk
between integrin receptor and insulin-like growth factor
receptor in regulation of collagen biosynthesis in cultured bro-
blasts. Adv Med Sci 58: 292-297, 2013.
95. Packham S, Warsito D, Lin Y, Sadi S, Karlsson R, Sehat B and
Larsson O: Nuclear translocation of IGF-1R via p150(Glued)
and an importin-β/RanBP2-dependent pathway in cancer cells.
Oncogene 34: 2227-2238, 2015.
96. Aleksic T, Chitnis MM, Perestenko OV, Gao S, Thomas PH,
Turner GD, Protheroe AS, Howarth M and Macaulay VM:
Type 1 insulin-like growth factor receptor translocates
to the nucleus of human tumor cells. Cancer Res 70: 6412- 6 419,
2010.
97. Sehat B, Togh A, Lin Y, Trocmé E, Liljedahl U, Lagergren J
and Larsson O: SUMOylation mediates the nuclear transloca-
tion and signaling of the IGF-1 receptor. Sci Signal 3: ra10, 2010.
98. Sarfstein R, Pasmanik-Chor M, Yeheskel A, Edry L, Shomron N,
Warman N, Wertheimer E, Maor S, Shochat L and Werner H:
Insulin-like growth factor-I receptor (IGF-IR) translocates to
nucleus and autoregulates IGF-IR gene expression in breast
cancer cells. J Biol Chem 287: 2766-2776, 2 012.
99. Vesel M, Rapp J, Feller D, Kiss E, Jaromi L, Meggyes M,
Miskei G, Duga B, Smuk G, Laszlo T, et al: ABCB1 and ABCG2
drug transporters are differentially expressed in non-small cell
lung cancers (NSCLC) and expression is modied by cisplatin
treatment via altered Wnt signaling. Respir Res 18: 52, 2017.
100. Huang YH, Lin MH, Wang PC, Wu YC, Chiang HL, Wang YL,
Chang JH, Huang YK, Gu SY, Ho HN and Ling TY: Hypoxia
inducible factor 2α/insulin-like growth factor receptor signal
loop supports the proliferation and Oct-4 maintenance of mouse
germline stem cells. Mol Hum Reprod 20: 526-537, 2014.
101. Treins C, Giorgetti-Peraldi S, Murdaca J,
Monthouël-Kartmann MN and Van Obberghen E: Regulation of
hypoxia-inducible factor (HIF)-1 activity and expression of HIF
hydroxylases in response to insulin-like growth factor I. Mol
Endocrinol 19: 1304-1317, 2005.
102. Popeda M, Pluciennik E and Bednarek AK: Proteins in cancer
multidrug resistance. Postepy Hig Med Dosw (Online) 68:
616-632, 2014 (In Polish).
103. Genois MM, Paquet ER, Laftte MC, Maity R, Rodrigue A,
Ouellette M and Masson JY: DNA repair pathways in trypano-
somatids: From DNA repair to drug resistance. Microbiol MoI
Biol Rev 78: 4 0-73, 2 014.
... Instead, it is possible that the catabolism of BSA through macropinocytosis leads to elevated NAD + levels or enhanced NAMPT activity that is important for macropinocytosis-dependent cell growth. In the case of IGF-1R, the underlying mechanism may be related to the observation that IGF-1R activates the RAS/RAF/MAPK signaling pathway [19]. Activation of the RAS pathway is one of the most well-known signaling inputs to enhance macropinocytic uptake [20]. ...
... Among the positive regulators of macropinocytosisdependent growth identified in our study, HDAC and its substrate HSP90 were found to localize to membrane ruffles and enhance macropinosome formation [14]. Similarly, IGF-1R was also identified as a positive regulator of macropinocytosis-dependent growth which is most likely through the activation of the RAS pathway [19]. There are multiple HDAC, HSP90, or IGF-1R inhibitors that are in clinical use or trials and whether these compounds can regulate macropinocytosis-dependent growth in these settings is an important question for future studies [28][29][30]. ...
Article
Full-text available
Cancer cells utilize multiple nutrient scavenging mechanisms to support growth and survival in nutrient-poor, hypoxic tumor microenvironments. Among these mechanisms, macropinocytosis has emerged as an important pathway of extracellular nutrient acquisition in cancer cells, particularly in tumors with activated RAS signaling, such as pancreatic cancer. However, the absence of a clinically available inhibitor, as well as the gap of knowledge in macropinocytosis regulation, remain a hurdle for its use for cancer therapy. Here, we use the Informer set library to identify novel regulators of macropinocytosis-dependent growth in pancreatic cancer cells. Understanding how these regulators function will allow us to provide novel opportunities for therapeutic intervention.
... Insulin-like growth factor 1 (IGF-1) pathway is an evolutionary conserved regulatory module that is implicated in the metabolism of glucose, lipid, and protein [4]. IGF-1 affects almost every tissue in the human body by promoting cell proliferation, growth, and maturation through upregulation of anabolic processes [5,6]. Disruption of the IGF-1 signaling pathway has been associated with the onset of a variety of age-related diseases, including muscle disease, metabolic, cardiovascular, and neurodegenerative diseases, and cancer [7,8]. ...
... The study conducted by Abbasi et al. also confirmed the altered expression of IGF1R in the whole blood after intensive exercise [19]. MiRNAs inhibit IGF1R expression by directly targeting the 3 untranslated region [6,12,20,21]. Due to its proliferation-promoting action, IGF1R has been investigated in many studies as a target in anticancer therapy [22,23]. ...
Article
Full-text available
A deeper insight into the mechanisms responsible for athlete performance that may serve as specific and detailed training indicators is still desired, because conventionally used biomarkers provide limited information about the adaptive processes that occur during exercise. The objective of our study was to assess insulin-like growth factor 1 receptors (IGF1R) gene expression and evaluate plasma concentration of selected microRNAs (miRNAs) during a 10-week training period (sampling times: week 1, 4, 7, and 10) in a group of 12 professional female volleyball players. Circulating miRNAs (miR-223, miR-320a, and miR-486) with established concentration in plasma and documented association with the IGF1 signaling pathway, which is involved in muscle development and recovery, were tested. The levels of analyzed miRNAs, tested by one-way ANOVA, were significantly different between four training periods during a 10-week training cycle (miR-223 p < 0.0001, miR-320a p = 0.00021, miR-486 p = 0.0037, respectively). The levels of IGF1R also appeared to be different (p = 0.00092), and their expression showed a trend to increase between the first and third periods. In the fourth period, the expression decreased, although it was higher compared with the baseline. Correlations between concentration levels of miR-223 and miR-320a (rs = 0.54, p < 0.001), as well as between miR-320a and miR-486 (rs = 0.73, p < 0.001) were also found. In the fourth period, a negative correlation between miR-223 plasma level and leucocyte IGF1R expression was found (rs = −0.63, p = 0.028). Multiple linear regression analysis showed that miR-320a (p = 0.024) and creatine kinase (p = 0.028) had the greatest impact on the expression levels of the IGF1R gene. Future studies are required to define whether these miRNAs, especially miR-320a, as well as IGF1R expression could be useful biomarkers of physiological changes during exercise and to discover their detailed biological roles in mode-specific exercise training adaptations of professional athletes.
... IGF1 impacts local and systemic growth and survival by activating the phosphatidylinositol-3 kinase (PI3K)/Akt pathway through IGF1 receptor (IGF1R) binding. The PI3K/Akt pathway integrates intracellular and environmental cues regarding nutrient availability to regulate cellular proliferation, survival, and protein translation through several downstream mediators, including the mammalian target of rapamycin (mTOR) [11,12]. The IGF1R is commonly expressed in human tumors leading to a mitogenic response to physiological concentrations of IGF1, and both the PI3K/Akt pathway and mTOR are commonly activated in cancers [13]. ...
Article
Full-text available
Simple Summary Breast cancer is the most common cancer in women worldwide. The risk of developing postmenopausal breast cancer is exacerbated by obesity (BMI > 30 kg/m²), and growing evidence suggests that obesity increases the risk of developing triple-negative breast cancer. Calorie restriction (CR), a reduction of calorie intake by 20–40% without causing malnutrition, is associated with decreasing an individual’s risk of developing cancer, enhancing the responses to cancer treatment, and reducing the risk of breast cancer recurrence. The negative impact of CR on breast cancer growth may result from lowering bioavailable levels of IGF1. In this study, we suggest that CR’s antitumor effects are partly mediated by the upregulation of miR-15b, which downregulates IGF1R and other target genes involved in cell cycle control. Our findings suggest that miR-15b could mediate CR’s regulation of IGF1/IGF1R signaling that contributes to the anticancer properties of this dietary intervention. Abstract Calorie restriction (CR) inhibits triple-negative breast cancer (TNBC) progression in several preclinical models in association with decreased insulin-like growth factor 1 (IGF1) signaling. To investigate the impact of CR on microRNAs (miRs) that target the IGF1/IGF1R pathway, we used the spontaneous murine model of TNBC, C3(1)/SV40 T-antigen (C3-TAg). In C3-TAg mice, CR reduced body weight, IGF1 levels, and TNBC progression. We evaluated the tumoral expression of 10 miRs. CR increased the expression of miR-199a-3p, miR-199a-5p, miR-486, and miR-15b. However, only miR-15b expression correlated with tumorigenicity in the M28, M6, and M6C C3-TAg cell lines of TNBC progression. Overexpressing miR-15b reduced the proliferation of mouse (M6) and human (MDA-MB-231) cell lines. Serum restriction alone or in combination with low levels of recombinant IGF1 significantly upregulated miR-15b expression and reduced Igf1r in M6 cells. These effects were reversed by the pharmacological inhibition of IGFR with BMS754807. In silico analysis using miR web tools predicted that miR-15b targets genes associated with IGF1/mTOR pathways and the cell cycle. Our findings suggest that CR in association with reduced IGF1 levels could upregulate miR-15b to downregulate Igf1r and contribute to the anticancer effects of CR. Thus, miR-15b may be a therapeutic target for mimicking the beneficial effects of CR against TNBC.
Article
Full-text available
The fourth most frequent type of cancer in women and the leading cause of mortality for females worldwide is cervical cancer. Traditionally, medicinal plants have been utilized to treat various illnesses and ailments. The molecular docking method is used in the current study to look into the phytoconstituents of Juglans regia’s possible anticancer effects on cervical cancer target proteins. This work uses the microarray dataset analysis of GSE63678 from the NCBI Gene Expression Omnibus database to find differentially expressed genes. Furthermore, protein-protein interactions of differentially expressed genes were constructed using network biology techniques. The top five hub genes (IGF1, FGF2, ESR1, MYL9, and MYH11) are then determined by computing topological parameters with Cytohubba. In addition, molecular docking research was performed on Juglans regia phytocompounds that were extracted from the IMPPAT database versus hub genes that had been identified. Utilizing molecular dynamics, simulation confirmed that prioritized docked complexes with low binding energies were stable.
Chapter
Cancer Genes is a comprehensive list of the most critical genes known to contribute to cancer imitation and progression. The book delves into their location on each chromosome, providing valuable insights into the mechanisms of cancer gene dysregulation and genetic mutations which provide cancer cells with an advantage during each stage of tumorigenesis. The reference will familiarize readers with the location of cancer genes and equip them with the necessary information to identify relevant gene expression targets for research aimed at preventing the disease. The book is divided into two volumes focusing on cancer-causing genes found in chromosome pairs 1-12 (volume 1), and chromosomes 13-23 (volume 2). A key features of the book is a detailed reference list for advanced readers. The compilation is therefore a quick and handy reference on cancer causing genes for researchers, medical professionals, and anyone interested in understanding the genetic basis of cancer.
Article
Full-text available
Cisplatin resistance is a crucial factor affecting ovarian cancer patient’s survival rate, but the primary mechanism underlying cisplatin resistance in ovarian cancer remains unclear, and this prevents the optimal use of cisplatin therapy. Maggot extract (ME) is used in traditional Chinese medicine for patients with comas and patients with gastric cancer when combined with other drug treatments. In this study, we investigated whether ME enhances the sensitivity of ovarian cancer cells to cisplatin. Two ovarian cancer cells—A2780/CDDP and SKOV3/CDDP—were treated with cisplatin and ME in vitro. SKOV3/CDDP cells that stably expressed luciferase were subcutaneously or intraperitoneally injected into BALB/c nude mice to establish a xenograft model, and this was followed by ME/cisplatin treatment. In the presence of cisplatin, ME treatment effectively suppressed the growth and metastasis of cisplatin-resistant ovarian cancer in vivo and in vitro. RNA-sequencing data showed that HSP90AB1 and IGF1R were markedly increased in A2780/CDDP cells. ME treatment markedly decreased the expression of HSP90AB1 and IGF1R, thereby increasing the expression of the proapoptotic proteins p-p53, BAX, and p-H2AX, while the opposite effects were observed for the antiapoptotic protein BCL2. Inhibition of HSP90 ATPase was more beneficial against ovarian cancer in the presence of ME treatment. In turn, HSP90AB1 overexpression effectively inhibited the effect of ME in promoting the increased expression of apoptotic proteins and DNA damage response proteins in SKOV3/CDDP cells. Inhibition of cisplatin-induced apoptosis and DNA damage by HSP90AB1 overexpression confers chemoresistance in ovarian cancer. ME can enhance the sensitivity of ovarian cancer cells to cisplatin toxicity by inhibiting HSP90AB1/IGF1R interactions, and this might represent a novel target for overcoming cisplatin resistance in ovarian cancer chemotherapy.
Article
Full-text available
Increased insulin-like growth factor (IGF) axis activity is associated with the development and progression of different types of malignancies, including colorectal cancer (CRC). MicroRNAs (miRNAs) belonging to the let-7 family have been reported to target genes involved in this axis and are known as tumor suppressors. In this study, in silico bioinformatic analysis was performed to assess miRNA–mRNA interactions between eight miRNAs belonging to the let-7 family and genes involved in the IGF signaling pathway, coding for receptors and substrates. miRNAs’ expression analysis revealed that hsa-let-7a-5p, hsa-let-7b-5p, hsa-let-7c-5p, hsa-let- 97 7d-5p, hsa-let-7e-5p, hsa-let-7f-5p, and hsa-let-7g-5p were significantly down-regulated in 25 CRC tumoral tissues (T) compared to the corresponding adjacent peritumoral tissues (PT). Moreover, our results showed an upregulation of miR-let-7e-5p in CRC tissues with mutations in KRAS codon 12 or 13, and, for the first time, found a specific dysregulation of let-7a-5p, let-7b-5p, let-7c-5p, let-7d-5p, and let-7i-5p in CRC with perineural invasion. Our results sustain the relationship between the IGF axis, let-7 miRNAs, and CRC and suggest an association between the expression of these miRNAs and perineural invasion.
Article
Full-text available
Triple-negative breast cancer (TNBC) is associated with high recurrence rates, high incidence of distant metastases, and poor overall survival (OS). Taxane and anthracycline-containing chemotherapy (CT) is currently the main systemic treatment option for TNBC, while platinum-based chemotherapy showed promising results in the neoadjuvant and metastatic settings. An early arising of intrinsic or acquired CT resistance is common and represents the main hurdle for successful TNBC treatment. Numerous mechanisms were uncovered that can lead to the development of chemoresistance. These include cancer stem cells (CSCs) induction after neoadjuvant chemotherapy (NACT), ATP-binding cassette (ABC) transporters, hypoxia and avoidance of apoptosis, single factors such as tyrosine kinase receptors (EGFR, IGFR1), a disintegrin and metalloproteinase 10 (ADAM10), and a few pathological molecular pathways. Some biomarkers capable of predicting resistance to specific chemotherapeutic agents were identified and are expected to be validated in future studies for a more accurate selection of drugs to be employed and for a more tailored approach, both in neoadjuvant and advanced settings. Recently, based on specific biomarkers, some therapies were tailored to TNBC subsets and became available in clinical practice: olaparib and talazoparib for BRCA1/2 germline mutation carriers larotrectinib and entrectinib for neurotrophic tropomyosin receptor kinase (NTRK) gene fusion carriers, and anti-trophoblast cell surface antigen 2 (Trop2) antibody drug conjugate therapy for heavily pretreated metastatic TNBC (mTNBC). Further therapies targeting some pathologic molecular pathways, apoptosis, miRNAS, epidermal growth factor receptor (EGFR), insulin growth factor 1 receptor (IGF-1R), and androgen receptor (AR) are under investigation. Among them, phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and EGFR inhibitors as well as antiandrogens showed promising results and are under evaluation in Phase II/III clinical trials. Emerging therapies allow to select specific antiblastics that alone or by integrating the conventional therapeutic approach may overcome/hinder chemoresistance.
Article
Full-text available
Acute myeloid leukemia (AML), the most common form of an acute leukemia, is a malignant disorder of stem cell precursors of the myeloid lineage. Ubiquitination is one of the post-translational modifications (PTMs), and the ubiquitin-like proteins (Ubls; SUMO, NEDD8, and ISG15) play a critical role in various cellular processes, including autophagy, cell-cycle control, DNA repair, signal transduction, and transcription. Also, the importance of Ubls in AML is increasing, with the growing research defining the effect of Ubls in AML. Numerous studies have actively reported that AML-related mutated proteins are linked to Ub and Ubls. The current review discusses the roles of proteins associated with protein ubiquitination, modifications by Ubls in AML, and substrates that can be applied for therapeutic targets in AML.
Article
Full-text available
Epithelial-mesenchymal transition (EMT) can directly contribute to some malignant phenotypes of tumor cells including invasion, metastasis and resistance to chemotherapy. Although EMT is widely demonstrated to play a critical role in chemoresistance and metastasis, the potential signaling network between EMT and drug resistance is still unclear. The distribution of drugs in the internal and external environment of the tumor cells is tightly linked with ATP-binding cassette (ABC) transporters. Recent studies have shown that ABC transporters expression changed continuously during EMT. We believe that EMT is an important regulator of ABC transporters. In this review, we discuss how EMT regulates ABC transporters and their potential linkages. And we hope the knowledge of EMT and ABC transporters will offer more effective targets to experimental research.
Article
Full-text available
Background Lung cancer (LC) is still the most common cause of cancer related deaths worldwide. Non-small cell lung cancer (NSCLC) accounts for 85% of all LC cases but is not a single entity. It is now accepted that, apart from the characteristic driver mutations, the unique molecular signatures of adeno- (AC) and squamous cell carcinomas (SCC), the two most common NSCLC subtypes should be taken into consideration for their management. Therapeutic interventions, however, frequently lead to chemotherapy resistance highlighting the need for in-depth analysis of regulatory mechanisms of multidrug resistance to increase therapeutic efficiency. Methods Non-canonical Wnt5a and canonical Wnt7b and ABC transporter expressions were tested in primary human LC (n = 90) resections of AC and SCC. To investigate drug transporter activity, a three dimensional (3D) human lung aggregate tissue model was set up using differentiated primary human lung cell types. Following modification of the canonical, beta-catenin dependent Wnt pathway or treatment with cisplatin, drug transporter analysis was performed at mRNA, protein and functional level using qRT-PCR, immunohistochemistry, immune-fluorescent staining and transport function analysis. ResultsNon-canonical Wnt5a is significantly up-regulated in SCC samples making the microenvironment different from AC, where the beta-catenin dependent Wnt7b is more prominent. In primary cancer tissues ABCB1 and ABCG2 expression levels were different in the two NSCLC subtypes. Non-canonical rhWnt5a induced down-regulation of both ABCB1 and ABCG2 transporters in the primary human lung aggregate tissue model recreating the SCC-like transporter pattern. Inhibition of the beta-catenin or canonical Wnt pathway resulted in similar down-regulation of both ABC transporter expression and function. In contrast, cisplatin, the frequently used adjuvant chemotherapeutic agent, activated beta-catenin dependent signaling that lead to up-regulation of both ABCB1 and ABCG2 transporter expression and activity. Conclusions The difference in the Wnt microenvironment in AC and SCC leads to variations in ABC transporter expression. Cisplatin via induction of canonical Wnt signaling up-regulates ABCB1 and ABCG2 drug transporters that are not transporters for cisplatin itself but are transporters for drugs that are frequently used in combination therapy with cisplatin modulating drug response.
Article
Full-text available
Background The insulin-like growth factor 1 (IGF1) signaling axis plays a major role in tumorigenesis. In a previous experiment, we chronically treated mice with several agonists of the IGF1 receptor (IGF1R). We found that chronic treatment with insulin analogues with high affinity towards the IGF1R (IGF1 and X10) decreased the mammary gland tumor latency time in a p53R270H/+WAPCre mouse model. Frequent injections with insulin analogues that only mildly activated the IGF1R in vivo (glargine and insulin) did not significantly decrease the tumor latency time in this mouse model. Methods Here, we performed next-generation RNA sequencing (40 million, 100 bp reads) on 50 mammary gland tumors to unravel the underlying mechanisms of IGF1R-promoted tumorigenesis. Mutational profiling of the individual tumors was performed to screen for treatment-specific mutations. The transcriptomic data were used to construct a support vector machine (SVM) classifier so that the phenotypic characteristics of tumors exposed to the different insulin analogue treatments could be predicted. For translational purposes, we ran the same classifiers on transcriptomic (micro-array) data of insulin analogue-exposed human breast cancer cell lines. Genome-scale metabolic modeling was performed with iMAT. ResultsWe found that chronic X10 and IGF1 treatment resulted in tumors with an increased and sustained proliferative and invasive transcriptomic profile. Furthermore, a Warburg-like effect with increased glycolysis was observed in tumors of the X10/IGF1 groups and, to a lesser extent, also in glargine-induced tumors. A metabolic flux analysis revealed that this enhanced glycolysis programming in X10/IGF1 tumors was associated with increased biomass production programs. Although none of the treatments induced genetic instability or enhanced mutagenesis, mutations in Ezh2 and Hras were enriched in X10/IGF1 treatment tumors. Conclusions Overall, these data suggest that the decreased mammary gland tumor latency time caused by chronic IGF1R activation is related to modulation of tumor progression rather than increased tumor initiation.
Article
Full-text available
Gastric cancer is the fourth most common cancer and the second leading cause of cancer deaths worldwide. Chemotherapy is one of the major treatments for gastric cancer, but drug resistance limits the effectiveness of chemotherapy, which results in treatment failure. Resistance to chemotherapy can be present intrinsically before the administration of chemotherapy or it can develop during chemotherapy. The mechanisms of chemotherapy resistance in gastric cancer are complex and multifactorial. A variety of factors have been demonstrated to be involved in chemoresistance, including the reduced intracellular concentrations of drugs, alterations in drug targets, the dysregulation of cell survival and death signaling pathways, and interactions between cancer cells and the tumor microenvironment. This review focuses on the molecular mechanisms of chemoresistance in gastric cancer and on recent studies that have sought to overcome the underlying mechanisms of chemoresistance.
Article
Full-text available
Renal cell carcinoma (RCC) incidence is highest in highly developed countries and it is the seventh most common neoplasm diagnosed. RCC management include nephrectomy and targeted therapies. Type 1 insulin-like growth factor (IGF-1) pathway plays an important role in cell proliferation and apoptosis resistance. IGF-1 and insulin share overlapping downstream signaling pathways in normal and cancer cells. IGF-1 receptor (IGF1R) stimulation may promote malignant transformation promoting cell proliferation, dedifferentiation and inhibiting apoptosis. Clear cell renal cell carcinoma (ccRCC) patients with IGF1R overexpression have 70 % increased risk of death compared to patients who had tumors without IGF1R expression. IGF1R signaling deregulation may results in p53, WT, BRCA1, VHL loss of function. RCC cells with high expression of IGF1R are more resistant to chemotherapy than cells with low expression. Silencing of IGF1R increase the chemosensitivity of ccRCC cells and the effect is greater in VHL mutated cells. Understanding the role of IGF-1 signaling pathway in RCC may result in development of new targeted therapeutic interventions. First preclinical attempts with anti-IGF-1R monoclonal antibodies or fragment antigen-binding (Fab) fragments alone or in combination with an mTOR inhibitor were shown to inhibit in vitro growth and reduced the number of colonies formed by of RCC cells.
Article
Full-text available
Clinical, epidemiological and experimental evidence indicate that the insulin-like growth factors (IGFs) are important mediators in the biochemical chain of events that lead from a phenotypically normal to a neoplastic cell. The IGF1 receptor (IGF1R), which mediates the biological actions of IGF1 and IGF2, exhibits potent pro-survival and anti-apoptotic activities. The IGF1R is highly expressed in most types of cancer and is regarded as a promising therapeutic target in oncology. P53 is a transcription factor with tumor suppressor activity that is usually activated in response to DNA damage and other forms of cellular stress. On the basis of its protective activities, p53 is commonly regarded as the guardian of the genome. We provide evidence that the IGF signaling axis and p53 genome protection pathways are tightly interconnected. Wild type, but not mutant, p53 suppresses IGF1R gene transcription, leading to abrogation of the IGF signaling network, with ensuing cell cycle arrest. Gain-of-function, or loss-of-function, mutations of p53 in tumor cells may disrupt its inhibitory activity, thus generating oncogenic molecules capable of transactivating the IGF1R gene. The interplay between the IGF1 and p53 pathways is also of major relevance in terms of metabolic regulation, including glucose transport and glycolysis. A better understanding of the complex physical and functional interactions between these important signaling pathways will have major basic and translational relevance.
Article
Insulin and IGFs play an important role in cancer initiation and progression, including ovarian cancer (OC). Epithelial ovarian cancer (EOC) is the most frequent type of OC in women and it is the most lethal gynecological malignancy worldwide. Generally, insulin is associated with metabolism, whereas Insulin like growth factors (IGFs) are involved in cell proliferation. Hence, Insulin-like growth factor binding proteins (IGFBPs) determines the bioavailability of IGFs in circulation. The interplay between these molecules such as insulin, IGFs, IGFBPs and insulin-like growth factor receptor 1 (IGF1R) may be crucial for ovarian cancer cell biology and cancer progression. However, the IGF1R inhibitors exhibiting potent activity on IGF/IGF1R also demonstrated activity against OC cells. The combination therapy of drugs may prove to be beneficial in clinical management of OC. This review describes both molecular and clinical associations between insulin and IGF1 signaling pathways in ovarian cancer. The data was collected using PubMed search engine with the following key words such as ovarian cancer, IGFs, IGFBP, IGF1Rs and ovarian cancer. © 2017 Indian Journal of Medical and Paediatric Oncology | Published by Wolters Kluwer - Medknow.
Article
The insulin-like growth factor (IGF) system is essential for normal growth and development, and its perturbation is implicated in a number of diseases. IGF activity is finely regulated by a family of six high-affinity IGF binding proteins (IGFBPs). 1GFBPs usually inhibit IGF actions but may enhance them under certain conditions. Additionally, IGFBPs bind non-IGF ligands in the extracellular space, cell membrane, cytoplasm and nucleus, thereby modulating cell proliferation, survival and migration in an IGF-independent manner. IGFBP activity is regulated by transcriptional mechanisms as well as by post-translational modifications and proteolysis. Understanding the balance between the various actions of IGFBPs in vivo may lead to novel insights into disease processes and possible IGFBP-based therapeutics.
Article
Significance T-cell acute lymphoblastic leukemia (T-ALL) is a malignancy of developing T cells. Cancer cell growth is often driven by cell-intrinsic alterations in signaling pathways as well as extrinsic signals from the tumor microenvironment. Here we identify tumor-associated dendritic cells as a key endogenous cell type in the tumor microenvironment that promotes murine T-ALL growth and survival at both primary and metastatic tumor sites. We also find that tumor-associated dendritic cells activate the insulin-like growth factor I receptor in T-ALL cells, which is critical for their survival. Analysis of primary patient T-ALL samples reveals phenotypically analogous tumor microenvironments. Our findings suggest that targeting signals from the tumor microenvironment could expand therapeutic options for T-ALL.