Affiliations of authors: H. Yu, Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport; T. Rohan, Department of Epidemiology and Social Medicine, Albert Einstein College of Medicine, Bronx, NY.
Correspondence to: Herbert Yu, M.D., Ph.D., Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932 (e-mail: hyu{at}lsumc.edu).
![]() |
ABSTRACT |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
![]() |
INTRODUCTION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Functionally, IGF-I not only stimulates cell proliferation but also inhibits apoptosis. It has now been recognized that the combination of these mitogenic and antiapoptotic effects has a profound impact on tumor growth (6). Besides their direct effect on cancer-related cellular activities, members of the IGF family also interact with a variety of molecules that are critically involved in cancer development and progression, including the sex steroid hormones, products of tumor suppressor genes, and other growth factors. Furthermore, the expression and production of IGF-I, a key peptide hormone that is involved in regulating human growth and development, are influenced by nutrition and physical activity. These features of the IGF family underscore its potential importance in the mechanisms that underlie the roles of lifestyle and behavior in influencing cancer risk.
Several extensive reviews (719) have addressed the molecular structure and physiologic function of members of the IGF family. Here, we review briefly the molecular and biochemical aspects of each member of the IGF family and the experimental evidence for the role of IGFs in cancer, discuss the potential impact of lifestyle factors on this group of growth factors, and summarize the findings of clinical and epidemiologic studies of the IGF family in relation to cancer etiology and pathogenesis. Collectively, the evidence reviewed here provides insights into the role of mitogenic growth factors in carcinogenesis. All information used in this review was identified by searching the English-language literature in the MEDLINE® database.
![]() |
THE IGF FAMILY |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
IGFs play an important role in regulating cell proliferation, differentiation, apoptosis, and transformation (11). IGFs exert their actions by interacting with a specific receptor on the cell membrane, namely, the IGF-I receptor (IGF-IR), and the interaction is regulated by a group of specific binding proteins. All of these molecules are considered to be members of the IGF family, which includes the polypeptide ligands IGF-I and IGF-II, two types of cell membrane receptors (i.e., IGF-IR and IGF-IIR), and six binding proteins (i.e., IGFBP-1 through IGFBP-6). In addition, a large group of IGFBP proteases hydrolyzes IGFBPs, resulting in the release of bound IGFs that then resume their ability to interact with IGF-IR. Thus, as far as IGF action is concerned, IGFBP proteases may also be regarded as part of the IGF family because they indirectly regulate the action of IGFs.
Insulin-Like Growth Factors
IGF-I and IGF-II are single-chain polypeptides (8,9). The two molecules have 62% homology in their amino acid sequences. The molecules share additional structural similarities, and their structures resemble the structure of proinsulin. The IGF-I gene is located on chromosome 12 (8,9), and the IGF-II gene is located on chromosome 11, 1.4 kilobases (kb) downstream from the insulin gene (20). Table 1 summarizes some features of IGF molecules and of their encoding genes.
|
In adult tissues, IGF-II transcription is initiated from the P1 promoter. Transcription from promoters P3 and P4 is often seen in fetal tissues (33), and increased transcription from these promoter sites has been observed in certain cancer tissues (3339). Another feature of IGF-II gene transcription is genomic imprinting, in which the expression of a gene is associated with a particular parental allele (40,41). DNA methylation (42), histone acetylation (43), and use of specific promoter sites (44,45) are mechanisms that may control allele-specific transcription. Loss of genomic imprinting in the IGF-II gene is often found in cancer (4650).
Expression of the IGF-I gene is regulated primarily by growth hormone (GH). GH, however, has no regulatory effect on IGF-II expression (11), and the primary regulator of IGF-II gene transcription remains unclear. Nevertheless, expression of IGFs is also influenced by various hormones, including estrogens, adrenocorticotropic hormone, thyrotropin, luteinizing hormone, follicle-stimulating hormone (FSH), and human chorionic gonadotropin, as well as by other growth factors, such as platelet-derived growth factor (PDGF), epidermal growth factor (EGF), and fibroblast growth factor (FGF). Diet and nutrition affect circulating IGF-I levels, suggesting a possible impact on IGF-I production (7,8).
IGF-I has both immediate and long-term effects on various cellular activities, and these effects are mediated mainly through IGF-IR (see next section). IGF-I exerts an acute anabolic action on protein and carbohydrate metabolism by increasing cellular uptake of amino acids and glucose and by stimulating glycogen and protein synthesis (11). IGF-I also has a long-term impact on cell proliferation, differentiation, and apoptosis (11). IGF-I is a potent mitogen for a wide variety of cells and exerts its mitogenic action by increasing DNA synthesis and by stimulating the expression of cyclin D1, which accelerates progression of the cell cycle from G1 to S phase (51,52). The mitogenic effect of IGF-I can be abolished by suppressing the expression of cyclin D1 (53). In addition to stimulating cell cycle progression, IGF-I also inhibits apoptosis. IGF-I is able to stimulate the expression of Bcl proteins and to suppress expression of Bax, which results in an increase in the relative amount of the Bcl/Bax heterodimer, thereby blocking initiation of the apoptotic pathway (5456).
Like IGF-I, IGF-II has mitogenic and antiapoptotic actions and regulates cell proliferation and differentiation. Concentrations of IGF-II in the blood are higher than those of IGF-I in humans of all ages. In adults, IGF-II levels vary between 400 and 600 ng/mL, whereas IGF-I levels range between 100 and 200 ng/mL. Despite its presence at higher concentration in the circulation, IGF-II is believed to play a less important role in postnatal growth than does IGF-I. This conclusion is based on the impact of these IGFs on body growth, their regulation by GH, and their relative binding affinities to IGF-IR and IGFBPs. Animal experiments indicate that the action of IGF-II on body development and growth occurs at a much earlier stage of life than that of IGF-I. Evidence (57,58) suggests that IGF-II may play a key regulatory role during embryonic and fetal growth. After birth, the role of IGF-II is gradually replaced by that of IGF-I (7,11,13). Levels of circulating IGF-I change substantially with agethey increase slowly from birth to puberty, surge at puberty, and decline with age thereafterand these changes are regulated by GH. For IGF-II, however, the circulating levels are relatively stable after puberty, and GH has little influence on them. The actions of both IGF-I and IGF-II are mediated through IGF-IR. IGF-I has a twofold to 15-fold higher binding affinity to IGF-IR than does IGF-II (11). Three of the six IGFBPs have higher affinity to IGF-II than to IGF-I, and the rest have similar binding affinity to both IGFs (14). The combination of high affinity to the receptor and low affinity to the binding proteins results in more IGF-I than IGF-II interacting with IGF-IR.
IGF Receptors
Both IGF-IR and IGF-IIR are glycoproteins and are located on the cell membrane. The two receptors, however, differ completely in structure and function (12,13). IGF-IR is a tetramer of two identical -subunits and two identical ß-subunits (12,13,15). Structurally, IGF-IR resembles the insulin receptor, and there is 60% homology between them. IGFs and insulin are able to cross-bind to each other's receptor, albeit with much weaker binding affinity than that for the preferred ligand (59,60). A hybrid receptor that is composed of one
-subunit and one ß-subunit of IGF-IR and one
-subunit and one ß-subunit of the insulin receptor (11,12) has been identified. The amount of insulin/IGF-I hybrid receptor varies substantially from tissue to tissue. Since its binding affinity for IGF-I is higher than that for insulin, the receptor is thought to function predominantly as an IGF-I receptor, but its biologic importance remains largely unknown.
IGF-IIR is monomeric (13,16,61,62). Three ligand-binding regions are found in the extracellular domain of the receptor, one for IGF-II binding and two for proteins containing mannose-6-phosphate (M6P), including renin, proliferin, thyroglobulin, and the latent form of transforming growth factor (TGF)-ß (11). Binding of IGF-IIR to TGF-ß activates the latter (16,63). Since IGF-IIR can bind both IGF-II and M6P-containing molecules, it is also called the IGF-II/M6P receptor.
The expression of IGF-IR is regulated by steroid hormones and growth factors (13,15). Since high IGF-I levels result in a decline in IGF-IR, IGFs may act as negative feedback signals to repress expression of IGF-IR (64,65). In contrast to the effect of IGFs, other growth factors, including basic FGF, PDGF, and EGF, stimulate IGF-IR expression (15,66,67). The expression of IGF-IR is also stimulated by estrogens, glucocorticoids, GH, FSH, luteinizing hormone, and thyroid hormones (12,15). On the other hand, tumor suppressor gene products, such as wild-type p53 protein and WT1 (Wilms' tumor protein), inhibit expression of IGF-IR (6871). Nutrition also affects IGF-IR levels (7274). Little is known about the regulation of IGF-IIR expression, although some studies (11,13,75,76) have suggested that insulin, IGFs, EGF, and M6P may increase the level of IGF-IIR in the cell membrane.
Binding of IGFs to IGF-IR activates the receptor's tyrosine kinase activity, which triggers a cascade of reactions among a number of molecules involved in the signal transduction pathway. Two distinct signal transduction pathways have been identified for IGF-IR. One pathway activates Ras protein, Raf protein, and mitogen-activated protein kinase, and the other pathway involves phosphoinositol-3-kinase (11,12). Other signal transduction pathways that are initiated by IGF-IR may also exist (77). Activation of IGF-IR by ligand binding is necessary to allow IGF-IR to mediate the actions of IGFs. In addition to mediating the mitogenic and antiapoptotic actions of IGFs, IGF-IR is involved in cell transformation. In vitro experiments (78) have shown that removal of IGF-IR from the cell membrane by eliminating the IGF-IR gene, by suppressing its expression, or by inhibiting its function can abolish cell transformation.
IGF-IIR has no tyrosine kinase activity, and it binds only to IGF-II (16). Since binding of IGF-IIR to IGF-II results in degradation of IGF-II, IGF-IIR acts like an antagonist to IGF-II, reducing its biologic activity (16). Because of this effect, IGF-IIR has been considered to be a potential tumor suppressor molecule. A unique feature of IGF-IIR may contribute to its ability to act as a scavenger for circulating IGF-II. Upon proteolytic cleavage, the extracellular domain of the receptor is disassociated from the cell membrane as a soluble fragment that circulates in the blood with the ability to bind to IGF-II and to facilitate its degradation (7984).
IGF-Binding Proteins
Six IGFBPs with specific binding affinities for IGFs have been identified. These proteins have some structural homology (13,14,8590). Table 1 summarizes the molecular features of these proteins. IGFBPs undergo substantial post-translational modification, which affects their binding affinities for IGFs (17,18,85). IGFBPs are able to bind to specific cell membrane receptors as well as to attach to the cell surface or to the extracellular matrix (17,19,85).
Regulation of IGFBP gene transcription is complex and tissue specific. A number of hormones, including estrogens, glucocorticoids, parathyroid hormone, FSH, GH, thyroid hormone, insulin, vitamin D, and cortisol, have been found to regulate the expression of IGFBPs (17,19,9196). Growth factors, including FGF, EGF, TGF-ß, PDGF, and IGFs themselves, as well as retinoic acid, are also involved in the regulation of expression of these binding proteins (96101). The expression of IGFBP-1 is suppressed by insulin and IGFs (17,19,91). As with its role in the regulation of IGFs, GH plays an important role in stimulating the expression of IGFBP-3 (92). Certain cytokines, such as interleukin 1 and tumor necrosis factor-, are also able to increase IGFBP-3 levels (102).
IGFBPs have multiple and complex functions, which can be either IGF dependent or IGF independent. With respect to IGF-dependent function, IGFBPs are able to inhibit or to enhance the action of IGFs, resulting in either suppression or stimulation of cell proliferation (17,19). These opposing effects of IGFBPs on IGFs are determined by the molecular structures of the binding proteins. When binding to IGFs, IGFBPs play three major roles: 1) transporting IGFs, 2) protecting IGFs from degradation, and 3) regulating the interaction between IGFs and IGF-IR. Normally, IGFBPs have higher binding affinity to IGFs than does IGF-IR; therefore, binding of IGFBPs to IGFs blocks the interaction between IGFs and IGF-IR and suppresses IGF action (17,19). However, binding of IGFBPs to IGFs also protects IGFs from proteolytic degradation, and that protection can enhance the action of IGFs by increasing their bioavailability in local tissue (17,19). The actual impact of IGFBPs on IGFs depends largely on post-translational modification of IGFBPs by phosphorylation and proteolysis (1719,103105). The association of IGFBPs with the cell membrane or extracellular matrix may also affect the binding affinity of IGFBPs to IGFs.
IGFBP-1, IGFBP-2, IGFBP-3, and IGFBP-5 have been found to have dual regulatory effects on IGFs, either suppressing or enhancing the action of IGFs. The effects of IGFBP-2, IGFBP-3, and IGFBP-5 on IGFs are regulated by proteolysis of the binding proteins, whereas the effect of IGFBP-1 on IGFs is affected by phosphorylation (11,1719). In addition to undergoing proteolysis and phosphorylation, IGFBP-1, IGFBP-2, IGFBP-3, and IGFBP-5 bind to specific cell membrane receptors or attach to the cell surface, which reduces their binding affinities for IGFs and results in the release of free IGFs (11,1719). The binding affinity of IGFBP-5 for IGFs also decreases when IGFBP-5 is associated with extracellular matrix (17,18). IGFBP-1, IGFBP-2, and IGFBP-5 are all suspected of having IGF-independent effects on cellular activities, such as cell adhesion and migration (11,17,19).
IGFBP-3 is found not only to regulate the mitogenic action of IGFs but also to inhibit their antiapoptotic effect (106). Besides its IGF-dependent function, IGFBP-3 also has an IGF-independent inhibitory effect on cell growth (17,19). This effect may be mediated through a cell membrane receptor that is specific for IGFBP-3 (107,108).
IGFBP-4 and IGFBP-6 are able to inhibit the mitogenic action of IGFs, but these proteins, unlike the others, do not enhance the actions of IGFs (11,19). IGFBP-4 and IGFBP-6 also undergo proteolysis, but this change seems to have little impact on their functions (11,17,109,110).
IGFBP Proteases
Since the actions of IGFs can be either suppressed or enhanced by IGFBPs, proteolysis of IGFBPs by IGFBP proteases is an important factor in determining the regulatory impact of IGFBPs on IGF action. Prostate-specific antigen (PSA), a serine protease, is able to cleave IGFBP-3 and IGFBP-5 (19,111), and proteolysis of IGFBP-3 by PSA reduces its binding affinity to IGF-I, which restores the mitogenic activity of IGF-I (112). Another serine protease, -nerve growth factor, has 65% homology to PSA and proteolytic activity toward IGFBP-4 and IGFBP-6 (109). Cathepsin D, an acid-activated lysosomal protease, can proteolyze all six IGFBPs (103,110,113). Matrix metalloproteinases, including interstitial collagenase, gelatinase A, stromelysin 1, gelatinase B, and disintegrin metalloproteinase, are involved in the proteolysis of IGFBP-2, IGFBP-3, IGFBP-4, and IGFBP-5 (19,114116). Other proteolytic enzymes that are able to hydrolyze IGFBPs include plasmin, thrombin, and pregnancy-associated plasma protein-A (117119).
Regulation of IGFBP proteolysis is complex and remains poorly understood. Changes in physiologic condition can influence the proteolysis of IGFBPs in the circulation. For example, serum from pregnant women has higher IGFBP proteolytic activity than serum from nonpregnant women (120). The expression or presence of one binding protein affects the proteolysis of the others (121,122). Binding between IGFBPs and IGFs also affects IGFBP proteolysis (17). It has also been shown that IGFs modulate the proteolytic activities of IGFBPs, suggesting that IGFs have an autocrine regulatory loop to control their own action (123). Insulin influences the activities of IGFBP proteases (124,125), but GH has little impact on these proteases (126). Estrogens may regulate the proteolysis of IGFBPs (127).
![]() |
IGFS AND CANCER: EXPERIMENTAL EVIDENCE |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Direct Involvement in Cancer
IGF-I and IGF-II are strong mitogens for a wide variety of cancer cell lines, including sarcoma, leukemia, and cancers of the prostate, breast, lung, colon, stomach, esophagus, liver, pancreas, kidney, thyroid, brain, ovary, and uterus (both cervical and endometrial) (128133). IGFs are also overexpressed in certain cancers (128). Animal experiments (134,135) indicate that overexpression of IGF-I increases the likelihood of tumor development in certain tissues. Overexpression of IGF-II may result from loss of genomic imprinting in IGF-II, loss of function of a transcriptional repressor, or change of transcription promoter sites (130,136138). Cancer cells with a strong tendency to metastasize have higher expression of IGF-II and IGF-IR than those with a low ability to do so (139). The strong impact of IGF-II on cancer growth that is observed consistently in laboratory studies and the paucity of clinical and epidemiologic studies that have found an association between circulating IGF-II and cancer risk suggest that IGF-II may exert its action via paracrine rather than endocrine regulation.
The effects of IGFs on cancer cells are mediated through IGF-IR. Eliminating IGF-IR from the cell membrane, blocking the interaction of IGFs with IGF-IR, or interrupting the signal transduction pathway of IGF-IR can abolish the mitogenic action of IGFs on cancer cells (130,140142). IGF-IR also plays a critical role in cell transformation that is induced by tumor-virus proteins and oncogene products. IGF-IR is involved not only in the induction of cell transformation but also in the maintenance of the transformed phenotype (130). IGF-IR is overexpressed in certain cancers, and its overexpression is associated with aggressive tumors (143,144). The hybrid receptor that binds both IGF-IR and insulin may also mediate the effect of IGFs on cancer (145,146). A recent study (147) indicates that the insulin receptor is involved in mediating the actions of IGF-II on breast cancer.
Since IGF-IIR antagonizes the effect of IGF-II, loss of IGF-IIR function is expected in cancer. One study found cancer-related missense mutations in the IGF-IIR gene with resultant disruption of the binding of IGF-IIR to its ligand. Cancer cells that lack the ability to degrade IGF-II have been shown to have a strong growth potency (148). Suppressing the expression of IGF-IIR yields the same effect as mutation in the IGF-IIR gene (149). Reestablishing the function of IGF-IIR in cancer cells that lack IGF-IIR reduces cancer growth and increases apoptosis (150).
In cancer, IGFBPs regulate the action of IGFs (151156). In most situations, the binding proteins suppress the mitogenic action of IGFs and promote apoptosis (157159). However, because of the presence of IGFBP proteases (1719,160), two in vitro studies (161,162) have found that IGFBPs are able to stimulate the growth of cancer cells. Oh et al. (107) found that IGFBP-3 inhibited breast cancer cell growth without interacting with IGFs. Other studies (163,164) reported that IGFBP-3 could induce apoptosis of breast and prostate cancer cells without the presence of IGFs or IGF-IR.
Interactions With Other Molecules
Many molecules that are known to be involved in cancer have been found to have substantial interactions with members of the IGF family. In general, IGFs interact synergistically with other mitogenic growth factors and steroids and antagonize the effects of antiproliferative molecules in cancer cells.
In breast cancer cells, estrogens enhance the mitogenic effect of IGF-I, induce expression of IGF-I, and stimulate production of IGF-IR (165168). Estrogens also repress synthesis of some IGFBPs in breast tissue (169,170). In breast cancer cells, estrogens decrease the expression of IGF-IIR and increase the level of IGFBP proteases (171). The interaction between estrogens and IGF is reciprocal. IGF-I enhances expression of estrogen receptor (ER) in breast cancer cells, and ER levels in breast tissue are associated with the levels of some IGFBPs (172,173) (see "Cancer Prognosis" section below).
Antiestrogenic agents increase the expression of IGFBPs (174176). Tamoxifen, which is antiestrogenic in breast tissue, abolishes the effects of estrogens on IGF-I, inhibits transcription of IGF-I, and attenuates the response of IGF-IR to IGFs (177,178)
The expression of IGFs in the uterus is regulated by estrogens, and IGFBPs interfere with this process (179181). Since tamoxifen has estrogenic effects in the uterus, synergistic interplay between tamoxifen and IGFs is also observed in endometrial cells. Tamoxifen enhances the IGF-stimulated growth of endometrial cancer cells (182). Furthermore, there is an interaction between the signal transduction pathways of the two systems. IGF-I can enhance ER-induced gene transcription in the absence of estrogen (183). IGFBPs, in contrast, suppress the transcriptional activation that is initiated by ERs (184). IGFs increase the activity of estrone sulfatase, which hydrolyzes estrone sulfate to estrone (185).
Synergistic interaction is also observed between IGFs and EGF, another potent mitogen. In cervical cancer cells, EGF is able to stimulate IGF-II expression and to increase IGF-IR's response to its ligand. Increases in IGF levels, in turn, enhance the mitogenic action of EGF (186,187). In addition, EGF can suppress the expression of IGFBP-3 and increase the availability of free IGFs, further enhancing the mitogenic signal of IGFs (188). In prostate cancer cells, interrupting the signaling pathway for EGF suppresses not only the effect of EGF but also that of IGF-I (189).
Several studies have suggested that IGFs may mediate the effects of tumor viruses. In hepatocellular carcinoma, hepatitis B virus stimulates IGF-IR expression (190) and increases transcription of the IGF-II gene from the P3 and P4 promoters (191). Hepatitis C virus may also be responsible for increased IGF-II transcription from fetal promoters in hepatitis C virus-related liver disease, including hepatocellular carcinoma (192).
Several antiproliferative molecules exert their actions by interfering with IGF signaling. Inhibition of breast cancer cell growth by TGF-ß is mediated through induction of IGFBP-3, which inhibits the mitogenic action of IGFs (98,193). Like TGF-ß, retinoic acid inhibits the growth of breast cancer cells by increasing IGFBP-3 expression (97,101,109). Vitamin D and its synthetic analogues can suppress the stimulatory effect of IGFs on the growth of breast and prostate cancer cells by increasing the expression of IGFBPs and reducing the expression of IGF-IR and IGF-II (94,95,194,195).
Tumor suppressor gene products have a profound impact on the IGF family. Wild-type p53 protein induces the expression of IGFBP-3 (196), represses the transcription of IGF-II from its P3 and P4 promoters (33,197), and suppresses IGF-IR expression (70,198,199). Not only does p53 regulate the action of IGFs but also IGFs influence p53 function. When IGF-I-induced DNA synthesis takes place in breast cancer cells, p53 loses its function by undergoing phosphorylation and relocation from the nucleus to the cytoplasm (200). Other tumor suppressor proteins that interact with IGFs include the Wilms' tumor suppressor gene product WT1 (69,201,202), the mammary-derived growth inhibitor MDGI (203), and the tumor suppressor gene PTEN (204).
Energy Intake
Animal experiments have shown that energy restriction can reduce the risk of cancer and inhibit tumor growth (205208) and that this effect can be attributed in part to IGFs (209,210). Restriction of energy intake in rats transplanted with human prostate cancer cells slows the growth of cancer and accelerates apoptosis. These effects are associated with a decrease in circulating IGF-I (209). Dietary restriction reduces the growth rate of bladder cancer in mice (210). When diet-restricted mice are supplemented with IGF-I, the effect of dietary restriction on cancer growth disappears, and the tumors in these animals grow at the same rate as those in animals that are not on food restriction, suggesting that the effect of energy restriction on cancer is mediated mainly through IGF-I (210).
![]() |
CIRCULATING IGFS AND IGFBPS AND THEIR DETERMINANTS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
More than 90% of IGFs in the circulation are bound to the IGFBPs, mainly IGFBP-3. The complex of IGFBP-3 and IGFs is further bound to another protein, which is called acid-labile subunit and is a glycoprotein with a molecular mass of about 63 kd (211). These three-molecule complexes are the major circulating forms of IGFs in the blood. All IGFs and IGFBPs are detectable in the blood (7,14), but measurement of serum or plasma IGF levels must be preceded by a procedure that separates IGFs from their binding proteins (212). Without the separation step, IGF-I measured in the blood is believed to represent only free IGF-I, which is about 1% of the total IGF-I in the circulation (213).
Although there is considerable interindividual variation in serum and plasma levels of IGFs and IGFBPs, blood levels of IGFs in each individual are relatively constant, and there is no apparent diurnal or circadian variation (7). Besides age, the determinants of this variation remain largely unknown, although the relationship of these growth factors, in particular that of IGF-I, with dietary factors and lifestyle has been investigated in a number of studies (2,3,5,214237).
Similar to levels of IGFs, levels of most of the IGFBPs are also quite stable in the circulation, with the exception of those of IGFBP-1. Because of its close relationship with insulin, serum IGFBP-1 levels decrease in response to increased levels of insulin after food intake (238). Nutritional status also affects the level of IGFBP-2 in the serum. Fasting increases its level substantially; however, this response occurs slowly. A role for insulin in regulating the level of IGFBP-2 in serum has been suggested by animal experiments but has not been seen in humans (11).
Effects of Age and Sex
Levels of IGFs and IGFBPs in the blood show little variation with sex but vary substantially with age. Serum IGF-I level is low at birth and increases gradually until puberty (239244). The rate of increase undergoes a sharp upsurge at puberty, after which the concentration declines slowly with age. Serum IGF-II levels also increase with age from birth to puberty, but after puberty they remain stable (239,240). Concentrations of IGFBP-1 and IGFBP-2 in blood are high at birth and decline with age until puberty (240,241). After that, the levels of these proteins remain relatively constant or increase slightly. The age-specific distributions for IGFBP-3 and acid-labile subunit are similar to the distribution for IGF-I (240,242,243). Limited data suggest that serum IGFBP-5 level declines with age, whereas IGFBP-4 and IGFBP-6 concentrations increase with age (240,244).
Diet, Nutrition, Anthropometric Characteristics, and Physical Activity
Energy intake, body mass index ([weight in kg]/[height in m]2), and physical activity all appear to affect blood levels of IGFs and IGFBPs. However, the intimate relationship among these variables makes disentanglement of their independent effects difficult. Four studies (3,5,220,223) found no association between the level of IGF-I and height. Of studies that investigated the association between IGF-I and body mass index, some showed no association with total IGF-I (3,5,215,220,223,225), whereas others showed inverse associations either with total (217,222) or with free (225) IGF-I.
Nutritional status and dietary energy intake are critical regulators of IGF-I level, and the IGF family links nutrition to growth (72). IGF-I levels are decreased in association with protein-calorie malnutrition, and they increase in response to improvements in energy intake (226). Fasting also results in a decrease in IGF-I level (227), but the effect is smaller in obese subjects who are presumably less dependent on energy intake to maintain IGF-I levels (228). Overnutrition has an effect that is opposite that of fasting, in that it results in an increase in IGF-I level (229). Studies of normal adults have demonstrated a positive correlation between protein intake and serum IGF-I levels (245). A 50% reduction in calorie intake or a 30% reduction in protein intake has been shown to result in a decline in serum IGF-I and IGFBP-3 levels and an increase in IGFBP-2 level (246). These findings are consistent with those of animal experiments (247251), which have also shown that restriction of nutrients has diverse effects on IGF-I gene transcription (252). Although all of these studies demonstrate a substantial impact of energy and protein intake on the IGF family, the exact nature of the doseresponse relationships between food intake and levels of IGFs in circulation remains to be determined.
The association between IGF-I level and physical activity appears to be rather complex, and current evidence does not allow clear conclusions to be drawn. Cross-sectional studies have shown either no association between physical activity and IGF-I levels (5,220,223,230) or positive associations with leisure-time exercise (214), general physical activity (224), or physical fitness (215) and training (232). A nonrandomized study (231) that involved a 16-week training program did not produce a change in IGF-I levels. Some randomized trials have shown that IGF-I levels increase in association with a 2-week training intervention (233) or a strength test (234), but others (235,236,253) have shown that IGF-I decreases after a 5-week period of endurance training in adolescents. One study (234) found no association of IGF-I level with strength training. The inconsistent findings for IGF levels in relation to physical activity may be age related. Exercise increases IGF-I levels in adults (233,234); however, in children and adolescents, the opposite effect is observed (72,235,236).
Since many tissues are able to express the IGF-I gene, the effect of IGF-I is subject to both endocrine and paracrine regulations. Most circulating IGFs and IGFBP-3 are manufactured in the liver. Levels of IGF-I in circulation are closely associated with body growth, especially at puberty. Children with short stature have low circulating IGF-I levels (254,255), and individuals with acromegaly have elevated serum IGF-I levels (256,257).
The assumption that IGF-I exerts its action on body growth mainly through endocrine regulation was challenged recently. Two animal studies (258,259) suggested that hepatic IGF-I was not involved in growth regulation. However, these findings are controversial, since paracrine regulation of IGF-I may have been elevated to compensate for the loss of endocrine-regulated IGF-I, and the action of IGF-II was not evaluated or controlled in the experiment.
Effect of Alcohol Consumption and Cigarette Smoking
The relationship is unclear between alcohol consumption and IGF levels. Different levels of alcohol consumption may have opposite effects on IGF levels (5,223,260262). Long-term and heavy consumption of alcohol can cause severe damage to liver function, and loss of liver function may result in a decline in the production of IGFs (260). Indeed, it has been shown that alcoholics have relatively low IGF-I levels (220). But some laboratory experiments have shown that alcohol enhances IGF-I action and expression (261,262). One cross-sectional study (223) found a positive association between moderate alcohol consumption in elderly women and serum IGF-I levels, but the opposite relationship was observed in another study (5).
Different associations between IGF levels and cigarette smoking have been reported. Two studies (223,263) found a positive association between serum levels of IGF-I and cigarette smoking, but one study (220) observed an inverse association and two studies (4,5) showed no association. The level of IGFBP-3 was found to be inversely associated with smoking in one study (263). These associations might have been confounded by the association between alcohol consumption and cigarette smoking.
![]() |
IGFS AND CANCER: CLINICAL AND EPIDEMIOLOGIC EVIDENCE |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
In a number of clinical studies, levels of IGFs, IGF-IR, and IGFBPs were compared in subjects with and without cancer (264274). IGF-I levels have been shown to be higher in the plasma and serum of women with breast cancer than in comparable specimens from women without the disease (264,266,274), although other studies have shown no difference between case and control subjects with respect to levels of IGF-I (267,268) or IGF-II (268) in the serum. One study (268) showed lower serum levels of IGFBP-1, IGFBP-3, and IGFBP-6 in case subjects as compared with control subjects, but another study (267) found no difference. A 10-fold increase in IGF-IR content (measured as nanograms per milligram of protein) has been observed in breast cancer tissue as compared with normal tissue (265).
Two studies of colon cancer (269,270) found similar levels of IGF-I in the sera of case and control subjects, whereas one study (270) reported higher levels of IGF-II, IGFBP-2, and IGFBP-3 in case subjects as compared with control subjects. In endometrial cancer, one study (271) found lower serum IGF-I levels in case subjects as compared with control subjects but no difference in IGFBP-1 levels between the two groups. A study with postmenopausal endometrial cancer patients (272) showed higher serum IGF-I levels, lower IGFBP-1 levels, and no difference in IGFBP-3 levels when compared with healthy control subjects. Another study (275) reported low IGF-I gene transcription in endometrial cancer tissue but no difference in the production of IGF-I and IGF-IR.
Clinical studies have found higher levels of serum IGF-I in prostate cancer patients than in control subjects (276,277) and higher levels of serum IGFBP-2 in prostate cancer patients than in patients with benign prostatic hyperplasia or in healthy control subjects (273,278,279). The transcription and production of IGFBP-2 are also higher in prostate cancer tissue than in normal tissue (280). IGFBP-3 levels have been shown to be low in the serum (279) and tumor tissue (280) of prostate cancer patients. Studies (281,282) also suggest increased expression of IGF-II, IGFBP-4, and IGFBP-5 in prostate cancer tissue.
Differences in levels of some IGFBPs in ovarian cancer patients as compared with those of control subjects are similar to those of prostate cancer patients. Higher levels of IGFBP-2 and lower levels of IGFBP-3 have been observed in sera of patients with epithelial ovarian cancer as compared with normal control subjects (283,284). Elevated IGFBP-2 expression has also been found in ovarian tumor tissue (283,285). Studies (283,286) further suggest that IGF-I levels may be increased in the serum and cancer tissue of patients with ovarian cancer.
In general, the clinical studies reviewed here have been relatively small, have not included adjustment for potential confounding factors, have not always described clearly the source of the study subjects (in particular, the source of the comparison groups), and have not presented estimates of cancer risk in association with IGF levels. Although the inconsistencies in the patterns described above do not allow firm conclusions to be drawn, the clinical data suggest that the IGFs have a role in cancer development.
Epidemiologic Studies
Epidemiologic studies (25,237,287296) have investigated the role of IGFs and IGFBPs in the etiology of cancers of the breast, colon and rectum, prostate, and lung and of childhood leukemia (Table 2). Although some of these studies were relatively small, most were characterized by careful definition of the comparison groups, adjustment for covariates, and estimation of cancer risk by levels of the IGF or IGFBP of interest. To date, most studies have focused on cancer risk in association with serum or plasma levels of IGF-I, IGF-II, and IGFBP-3, or combinations thereof, and they have provided reasonably consistent support for increased risk of solid tumors in association with relatively high levels of IGF-I, decreased risk of solid tumors and of childhood leukemia in association with relatively high levels of IGFBP-3, and increased risk of breast cancer in association with a high ratio of IGF-I to IGFBP-3. For breast cancer, the findings appear to hold largely for premenopausal women. Mutual adjustment of IGF-I and IGFBP-3 levels appears to strengthen the association of each of these factors with cancer risk.
|
Most of the currently available data are from casecontrol studies. Unless they are nested within prospective investigations, casecontrol studies are generally unable to establish the temporal nature of an association due to possible effects of the disease process on blood levels of the molecules of interest and are potentially susceptible to selection and information biases (293). The results of casecontrol studies with respect to IGF levels and the risk of breast, prostate, and colorectal cancers are substantiallybut not completelyin accord with those of cohort studies at the same sites.
Cancer Prognosis
The association between members of the IGF family and various predictors of cancer prognosis has been investigated in a number of cross-sectional studies (265267,297313). Prognostic factors that have been examined in relation to IGF levels include age, menopausal status, ER and progesterone receptor (PR) status, tumor size, lymph node involvement, and histologic grade. Several prospective studies (300302,307309,312,313) have also examined the association of IGF levels with disease-free or overall survival from cancer.
To date, work in this area has focused largely on breast cancer. Many of the investigations have been relatively small, which may have compromised their ability to detect associations. Nevertheless, evidence suggests that IGF-IR is detectable (265,297301) and sometimes amplified (302) in tissues that are ER or PR positive, although two studies (265,274) showed no association. Findings on whether IGF-IR overexpression in breast cancer affects prognosis are contradictory. Two studies (314,315) suggested that IGF-IR expression might lead to poor prognosis, whereas one study (265) found that overexpression of IGF-IR is associated with less aggressive lesions. However, studies (303306,310313) have consistently demonstrated an inverse association between tissue IGFBP-3 levels and ER status. Some (310312) but not all (306,313) of the studies have also suggested a similar association for PR status. Clinical studies (266,267,304307,309,311) have not found consistent evidence for associations between IGF levels and age, menopausal status, tumor size, lymph node involvement, or degree of tumor differentiation.
Studies of the association of IGFs with disease-free or overall survival have also focused largely on breast cancer. One study (300) found no association between IGF-IR level and disease-free survival, one (301) found better disease-free and overall survival in women with IGF-IR-positive tumors after 40 months of follow-up and after adjustment for ER and PR status and other prognostic indicators, and one (302) found worse overall survival in those who had evidence of IGF-IR amplification in their tumors. Detection of IGFBP-3 in breast tissue has been reported to have no association with disease-free survival (312,313) and to have either no association with overall survival (312) or an association with increased risk of death (313); the latter association was independent of other prognostic factors (313).
In relation to the other IGF markers that have been studied, one study (307) showed an association between increased IGFBP-4 levels in breast tissue and decreased overall survival in women with large tumors (>2 cm in diameter), one study (309) showed no association of the level of IGF-II in breast tissue with disease-free or overall survival, and one study (308) showed no association of IGF-I levels in serum with overall 2-year survival.
With the exception of two studies (302,313), the studies described here have been relatively small and have not employed long follow-up periods. Nevertheless, on the basis of the currently available clinical evidence, it would appear that IGF levels have little value in predicting cancer prognosis. This conclusion is supported further by the results of studies of blood levels of IGF-I, one (269) that involved comparing subjects with colonic adenomas with those with carcinoma of the colon and two (3,295) that examined the association between IGF-I levels and stage of prostate cancer. None of these three studies showed any evidence for differences between the compared groups.
![]() |
CONCLUSION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
|
Evidence suggests that lifestyles characterized by a high-energy diet may affect the IGF system, which may, in turn, connect such lifestyles to high rates of cell proliferation and predispose cells to risk of malignant transformation. Currently unknown are the feasibility and validity of implementing dietary interventions to reduce IGF levels with the goal of preventing cancer. Studies that address this issue and the doseresponse relationship between food intake and levels of IGFs in circulation may be of value in developing programs for cancer prevention and control.
![]() |
NOTES |
---|
![]() |
REFERENCES |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
1 Preston-Martin S, Pike MC, Ross RK, Jones PA, Henderson BE. Increased cell division as a cause of human cancer. Cancer Res 1990;50:741521.[Abstract]
2 Hankinson SE, Willett WC, Colditz GA, Hunter DJ, Michaud DS, Deroo B, et al. Circulating concentrations of insulin-like growth factor-1 and risk of breast cancer. Lancet 1998;351:13936.[Medline]
3
Chan JM, Stampfer MJ, Giovannucci E, Gann PH, Ma J, Wilkinson P, et al. Plasma insulin-like growth factor-I and prostate cancer risk: a prospective study. Science 1998;279:5636.
4
Yu H, Spitz MR, Mistry J, Gu J, Hong WK, Wu X. Plasma levels of insulin-like growth factor-I and lung cancer risk: a casecontrol study. J Natl Cancer Inst 1999;91:1516.
5
Ma J, Pollak MN, Giovannucci E, Chan JM, Tao Y, Hennekens CH, et al. Prospective study of colorectal cancer risk in men and plasma levels of insulin-like growth factor (IGF)-I and IGF-binding protein-3. J Natl Cancer Inst 1999;91:6205.
6 Cory S, Vaux DL, Strasser A, Harris AW, Adams JM. Insights from Bcl-2 and Myc: malignancy involves abrogation of apoptosis as well as sustained proliferation. Cancer Res 1999;59(7 suppl):1685s1692s.[Medline]
7 Daughaday WH, Rotwein P. Insulin-like growth factors I and II. Peptide, messenger ribonucleic acid and gene structures, serum, and tissue concentrations. Endocr Rev 1989;10:6891.[Abstract]
8
Sara VR, Hall K. Insulin-like growth factors and their binding proteins. Physiol Rev 1990;70:591614.
9 Rosenfeld RG, Lamson G, Pham H, Oh Y, Conover C, De Leon DD, et al. Insulinlike growth factor-binding proteins. Recent Prog Horm Res 1990;46:99159; discussion 15963.[Medline]
10 Krywicki RF, Yee D. The insulin-like growth factor family of ligands, receptors, and binding proteins. Breast Cancer Res Treat 1992;22:719.[Medline]
11 Jones JI, Clemmons DR. Insulin-like growth factors and their binding proteins: biological actions. Endocr Rev 1995;16:334.[Medline]
12 LeRoith D, Werner H, Beitner-Johnson D, Roberts CT Jr. Molecular and cellular aspects of the insulin-like growth factor I receptor. Endocr Rev 1995;16:14363.[Medline]
13
Stewart CE, Rotwein P. Growth, differentiation, and survival: multiple physiological functions for insulin-like growth factors. Physiol Rev 1996;76:100526.
14
Rajaram S, Baylink DJ, Mohan S. Insulin-like growth factor-binding proteins in serum and other biological fluids: regulation and functions. Endocr Rev 1997;18:80131.
15 Sepp-Lorenzino L. Structure and function of the insulin-like growth factor I receptor. Breast Cancer Res Treat 1998;47:23553.[Medline]
16 Oates AJ, Schumaker LM, Jenkins SB, Pearce AA, DaCosta SA, Arun B, et al. The mannose 6-phosphate/insulin-like growth factor 2 receptor (M6P/IGF2R), a putative breast tumor suppressor gene. Breast Cancer Res Treat 1998;47:26981.[Medline]
17 Kelley KM, Oh Y, Gargosky SE, Gucev Z, Matsumoto T, Hwa V, et al. Insulin-like growth factor-binding proteins (IGFBPs) and their regulatory dynamics. Int J Biochem Cell Biol 1996;28:61937.[Medline]
18 Clemmons DR. Insulin-like growth factor binding proteins and their role in controlling IGF actions. Cytokine Growth Factor Rev 1997;8:4562.[Medline]
19 Collett-Solberg PF, Cohen P. The role of the insulin-like growth factor binding proteins and the IGFBP proteases in modulating IGF action. Endocrinol Metab Clin North Am 1996;25:591614.[Medline]
20 Brissenden JE, Ullrich A, Francke U. Human chromosomal mapping of genes for insulin-like growth factors I and II and epidermal growth factor. Nature 1984;310:7814.[Medline]
21 Jansen M, van Schaik FM, Ricker AT, Bullock B, Woods DE, Gabbay KH, et al. Sequence of cDNA encoding human insulin-like growth factor I precursor. Nature 1983;306:60911.[Medline]
22 Tobin G, Yee D, Brunner N, Rotwein P. A novel human insulin-like growth factor I messenger RNA is expressed in normal and tumor cells. Mol Endocrinol 1990;4:191420.[Abstract]
23 Sussenbach JS, Rodenburg RJ, Scheper W, Holthuizen P. Transcriptional and post-transcriptional regulation of the human IGF-II gene expression. Adv Exp Med Biol 1993;343:6371.[Medline]
24 de Pagter-Holthuizen P, Jansen M, van der Kammen RA, van Schaik FM, Sussenbach JS. Differential expression of the human insulin-like growth factor II gene. Characterization of the IGF-II mRNAs and an mRNA encoding a putative IGF-II-associated protein. Biochim Biophys Acta 1988;950:28295.[Medline]
25 Jansen E, Steenbergh PH, LeRoith D, Roberts CT Jr, Sussenbach JS. Identification of multiple transcription start sites in the human insulin-like growth factor-I gene. Mol Cell Endocrinol 1991;78:11525.[Medline]
26 Shemer J, Adamo ML, Roberts CT Jr, LeRoith D. Tissue-specific transcription start site usage in the leader exons of the rat insulin-like growth factor-I gene: evidence for differential regulation in the developing kidney. Endocrinology 1992;131:27939.[Abstract]
27 West CA, Arnett TR, Farrow SM. Expression of insulin-like growth factor I (IGF-I) mRNA variants in rat bone. Bone 1996;19:416.[Medline]
28 Li X, Cui H, Sandstedt B, Nordlinder H, Larsson E, Ekstrom TJ. Expression levels of the insulin-like growth factor-II gene (IGF2) in the human liver: developmental relationship of the four promoters. J Endocrinol 1996;149:11724.[Abstract]
29 Mittanck DW, Kim SW, Rotwein P. Essential promoter elements are located within the 5` untranslated region of human insulin-like growth factor-I exon I. Mol Cell Endocrinol 1997;126:15363.[Medline]
30 Roy RN, Cecutti A, Gerulath AH, Steinberg WM, Bhavnani BR. Endometrial transcripts of human insulin-like growth factors arise by differential promoter usage. Mol Cell Endocrinol 1997;135:119.[Medline]
31 Pell JM, Saunders JC, Gilmour RS. Differential regulation of transcription initiation from insulin-like growth factor-I (IGF-I) leader exons and of tissue IGF-I expression in response to changed growth hormone and nutritional status in sheep. Endocrinology 1993;132:1797807.[Abstract]
32
Zhang J, Whitehead RE Jr, Underwood LE. Effect of fasting on insulin-like growth factor (IGF)-IA and IGF-IB messenger ribonucleic acids and prehormones in rat liver. Endocrinology 1997;138:31128.
33 Zhang L, Kashanchi F, Zhan Q, Zhan S, Brady JN, Fornace AJ, et al. Regulation of insulin-like growth factor II P3 promoter by p53: a potential mechanism for tumorigenesis. Cancer Res 1996;56:136773.[Abstract]
34 Reeve AE, Eccles MR, Wilkins RJ, Bell GI, Millow LJ. Expression of insulin-like growth factor II transcripts in Wilms' tumour. Nature 1985;317:25860.[Medline]
35 Tricoli JV, Rall LB, Karakousis CP, Herrera L, Petrelli NJ, Bell GI, et al. Enhanced levels of insulin-like growth factor messenger RNA in human colon carcinomas and liposarcomas. Cancer Res 1986;46:616973.[Abstract]
36 Gloudemans T, Prinsen I, van Unnik JA, Lips CJ, den Otter W, Sussenbach JS. Insulin-like growth factor gene expression in human smooth muscle tumors. Cancer Res 1990;50:668995.[Abstract]
37 Minniti CP, Tsokos M, Newton WA Jr, Helman LJ. Specific expression of insulin-like growth factor-II in rhabdomyosarcoma tumor cells. Am J Clin Pathol 1994;101:198203.[Medline]
38 Fiorentino M, Grigioni WF, Baccarini P, D'Errico A, De Mitri MS, Pisi E, et al. Different in situ expression of insulin-like growth factor type II in hepatocellular carcinoma. An in situ hybridization and immunohistochemical study. Diagn Mol Pathol 1994;3:5965.[Medline]
39 Lahm H, Amstad P, Wyniger J, Yilmaz A, Fischer JR, Schreyer M, et al. Blockade of the insulin-like growth-factor-I receptor inhibits growth of human colorectal cancer cells: evidence of a functional IGF-II-mediated autocrine loop. Int J Cancer 1994;58:4529.[Medline]
40 Giannoukakis N, Deal C, Paquette J, Goodyer CG, Polychronakos C. Parental genomic imprinting of the human IGF2 gene. Nat Genet 1993;4:98101.[Medline]
41 Feinberg AP. Imprinting of a genomic domain of 11p15 and loss of imprinting in cancer: an introduction. Cancer Res 1999;59(7 Suppl):1743s1746s.[Medline]
42 Wutz A, Barlow DP. Imprinting of the mouse Igf2r gene depends on an intronic CpG island. Mol Cell Endocrinol 1998;140:914.[Medline]
43 Hu JF, Oruganti H, Vu TH, Hoffman AR. The role of histone acetylation in the allelic expression of the imprinted human insulin-like growth factor II gene. Biochem Biophys Res Commun 1998;251:4038.[Medline]
44 Vu TH, Hoffman AR. Promoter-specific imprinting of the human insulin-like growth factor-II gene. Nature 1994;371:7147.[Medline]
45 Yun K, Jinno Y, Sohda T, Niikawa N, Ikeda T. Promoter-specific insulin-like growth factor 2 gene imprinting in human fetal liver and hepatoblastoma. J Pathol 1998;185:918.[Medline]
46
Oda H, Shimizu S, Minami K, Kaneko K, Ishikawa T. Loss of imprinting of the IGF2 gene in a Wilms' tumor in an adult. J Natl Cancer Inst 1997;89:18134.
47 Cui H, Horon IL, Ohlsson R, Hamilton SR, Feinberg AP. Loss of imprinting in normal tissue of colorectal cancer patients with microsatellite instability. Nat Med 1998;4:127680.[Medline]
48 Jarrard DF, Bussemakers MJ, Bova GS, Isaacs WB. Regional loss of imprinting of the insulin-like growth factor II gene occurs in human prostate tissues. Clin Cancer Res 1995;1:14718.[Abstract]
49 Kim HT, Choi BH, Niikawa N, Lee TS, Chang SI. Frequent loss of imprinting of the H19 and IGF-II genes in ovarian tumors. Am J Med Genet 1998;80:3915.[Medline]
50 van Roozendaal CE, Gillis AJ, Klijn JG, van Ooijen B, Claassen CJ, Eggermont AM, et al. Loss of imprinting of IGF2 and not H19 in breast cancer, adjacent normal tissue and derived fibroblast cultures. FEBS Lett 1998;437:10711.[Medline]
51 Furlanetto RW, Harwell SE, Frick KK. Insulin-like growth factor-I induces cyclin-D1 expression in MG63 human osteosarcoma cells in vitro. Mol Endocrinol 1994;8:5107.[Abstract]
52
Dufourny B, Alblas J, van Teeffelen HA, van Schaik FM, van der Burg B, Steenbergh PH, et al. Mitogenic signaling of insulin-like growth factor I in MCF-7 human breast cancer cells requires phosphatidylinositol 3-kinase and is independent of mitogen-activated protein kinase. J Biol Chem 1997;272:3116371.
53
Kornmann M, Arber N, Korc M. Inhibition of basal and mitogen-stimulated pancreatic cancer cell growth by cyclin D1 antisense is associated with loss of tumorigenicity and potentiation of cytotoxicity to cisplatinum. J Clin Invest 1998;101:34452.
54 Minshall C, Arkins S, Straza J, Conners J, Dantzer R, Freund GG, et al. IL-4 and insulin-like growth factor-I inhibit the decline in Bcl-2 and promote the survival of IL-3-deprived myeloid progenitors. J Immunol 1997;159:122532.[Abstract]
55
Parrizas M, LeRoith D. Insulin-like growth factor-1 inhibition of apoptosis is associated with increased expression of the bcl-xL gene product. Endocrinology 1997;138:13558.
56
Wang L, Ma W, Markovich R, Lee WL, Wang PH. Insulin-like growth factor I modulates induction of apoptotic signaling in H9C2 cardiac muscle cells. Endocrinology 1998;139:135460.
57 Schultz GA, Hahnel A, Arcellana-Panlilio M, Wang L, Goubau S, Watson A, et al. Expression of IGF ligand and receptor genes during preimplantation mammalian development. Mol Reprod Dev 1993;35:41420.[Medline]
58 Lighten AD, Hardy K, Winston RM, Moore GE. Expression of mRNA for the insulin-like growth factors and their receptors in human preimplantation embryos. Mol Reprod Dev 1997;47:1349.[Medline]
59
Steele-Perkins G, Turner J, Edman JC, Hari J, Pierce SB, Stover C, et al. Expression and characterization of a functional human insulin-like growth factor I receptor. J Biol Chem 1988;263:1148692.
60
Frattali AL, Pessin JE. Relationship between alpha subunit ligand occupancy and beta subunit autophosphorylation in insulin/insulin-like growth factor-1 hybrid receptors. J Biol Chem 1993;268:7393400.
61 Morgan DO, Edman JC, Standring DN, Fried VA, Smith MC, Roth RA, et al. Insulin-like growth factor II receptor as a multifunctional binding protein. Nature 1987;329:3017.[Medline]
62
Oshima A, Nolan CM, Kyle JW, Grubb JH, Sly WS. The human cation-independent mannose 6-phosphate receptor. Cloning and sequence of the full-length cDNA and expression of functional receptor in COS cells. J Biol Chem 1988;263:255362.
63 Braulke T. Type-2 IGF receptor: a multi-ligand binding protein. Horm Metab Res 1999;31:2426.[Medline]
64 Yang Y, Hoeflich A, Butenandt O, Kiess W. Opposite regulation of IGF-I and IGF-I receptor mRNA and concomitant changes of GH receptor and IGF-II/M6P receptor mRNA in human IM-9 lymphoblasts. Biochim Biophys Acta 1996;1310:31724.[Medline]
65
Hernandez-Sanchez C, Werner H, Roberts CT Jr, Woo EJ, Hum DW, Rosenthal SM, et al. Differential regulation of insulin-like growth factor-I (IGF-I) receptor gene expression by IGF-I and basic fibroblastic growth factor. J Biol Chem 1997;272:466370.
66 Rosenthal SM, Brown EJ, Brunetti A, Goldfine ID. Fibroblast growth factor inhibits insulin-like growth II (IGF-II) gene expression and increases IGF-I receptor abundance in BC3H-1 muscle cells. Mol Endocrinol 1991;5:67884.[Abstract]
67 Rubini M, Werner H, Gandini E, Roberts CT Jr, LeRoith D, Baserga R. Platelet-derived growth factor increases the activity of the promoter of the insulin-like growth factor-1 (IGF-1) receptor gene. Exp Cell Res 1994;211:3749.[Medline]
68 Werner H, Re GG, Drummond IA, Sukhatme VP, Rauscher FJ 3d, Sens DA, et al. Increased expression of the insulin-like growth factor-1 receptor gene, IGF1R, in Wilms tumor is correlated with modulation of IGF1R promoter activity by the WT1 Wilms tumor gene product. Proc Natl Acad Sci U S A 1993;90:582832.[Abstract]
69 Werner H. Dysregulation of the type 1 IGF receptor as a paradigm in tumor progression. Mol Cell Endocrinol 1998;141:15.[Medline]
70
Werner H, Karnieli E, Rauscher FJ, LeRoith D. Wild-type and mutant p53 differentially regulate transcription of the insulin-like growth factor I receptor gene. Proc Natl Acad Sci U S A 1996;93:831823.
71 Prisco M, Hongo A, Rizzo MG, Sacchi A, Baserga R. The insulin-like growth factor I receptor as a physiologically relevant target of p53 in apoptosis caused by interleukin-3 withdrawal. Mol Cell Biol 1997;17:108492.[Abstract]
72 Thissen JP, Ketelslegers JM, Underwood LE. Nutritional regulation of the insulin-like growth factors. Endocr Rev 1994;15:80101.[Abstract]
73 Qu Z, Chow JC, Ling PR, Ziegler TR, Bistrian BR, Smith RJ. Tissue-specific effects of chronic dietary protein restriction and gastrostomy on the insulin-like growth factor-I pathway in the liver and colon of adult rats. Metabolism 1997;46:6917.[Medline]
74
Zhang W, Thornton WH, MacDonald RS. Insulin-like growth factor-I and II receptor expression in rat colon mucosa are affected by dietary lipid intake. J Nutr 1998;128:15865.
75 Hoeflich A, Yang Y, Rascher W, Blum WF, Huber S, Koepf G, et al. Coordinate expression of insulin-like growth factor II (IGF-II) and IGF-II/mannose-6-phosphate receptor mRNA and stable expression of IGF-I receptor mRNA during differentiation of human colon carcinoma cells (Caco-2). Eur J Endocrinol 1996;135:4959.[Medline]
76
Takigawa M, Okawa T, Pan H, Aoki C, Takahashi K, Zue J, et al. Insulin-like growth factors I and II are autocrine factors in stimulating proteoglycan synthesis, a marker of differentiated chondrocytes, acting through their respective receptors on a clonal human chondrosarcoma-derived chondrocyte cell line, HCS-2/8. Endocrinology 1997;138:4390 400.
77 Lopaczynski W. Different regulation of signaling pathways for insulin and insulin-like growth factor I. Acta Biochim Pol 1999;46:5160.[Medline]
78 Baserga R. The insulin-like growth factor I receptor: a key to tumor growth? Cancer Res 1995;55:24952.[Abstract]
79 Kiess W, Greenstein LA, White RM, Lee L, Rechler MM, Nissley SP. Type II insulin-like growth factor receptor is present in rat serum. Proc Natl Acad Sci U S A 1987;84:77204.[Abstract]
80
MacDonald RG, Tepper MA, Clairmont KB, Perregaux SB, Czech MP. Serum form of the rat insulin-like growth factor II/mannose 6-phosphate receptor is truncated in the carboxyl-terminal domain. J Biol Chem 1989;264:325661.
81
Valenzano KJ, Remmler J, Lobel P. Soluble insulin-like growth factor II/mannose 6-phosphate receptor carries multiple high molecular weight forms of insulin-like growth factor II in fetal bovine serum. J Biol Chem 1995;270:164418.
82
Zaina S, Squire S. The soluble type 2 insulin-like growth factor (IGF-II) receptor reduces organ size by IGF-II-mediated and IGF-II-independent mechanism. J Biol Chem 1998;273:286106.
83
Xu Y, Papageorgiou A, Polychronakos C. Developmental regulation of the soluble form of insulin-like growth factor-II/mannose 6-phosphate receptor in human serum and amniotic fluid. J Clin Endocrinol Metab 1998;83:43742.
84
Costello M, Baxter RC, Scott CD. Regulation of soluble insulin-like growth factor II/mannose 6-phosphate receptor in human serum: measurement by enzyme-linked immunosorbent assay. J Clin Endocrinol Metab 1999;84:6117.
85 Oh Y. IGF-independent regulation of breast cancer growth by IGF binding proteins. Breast Cancer Res Treat 1998;47:28393.[Medline]
86 Hwa V, Oh Y, Rosenfeld RG. Insulin-like growth factor binding proteins: a proposed superfamily. Acta Paediatr Suppl 1999;88:3745.
87 Ehrenborg E, Larsson C, Stern I, Janson M, Powell DR, Luthman H. Contiguous localization of the genes encoding human insulin-like growth factor binding proteins 1 (IGBP1) and 3 (IGBP3) on chromosome 7. Genomics 1992;12:497502.[Medline]
88
Allander SV, Larsson C, Ehrenborg E, Suwanichkul A, Weber G, Morris SL, et al. Characterization of the chromosomal gene and promoter for human insulin-like growth factor binding protein-5. J Biol Chem 1994;269:108918.
89 Tonin P, Ehrenborg E, Lenoir G, Feunteun J, Lynch H, Morgan K, et al. The human insulin-like growth factor binding protein 4 gene maps to chromosome region 17q12q21.1 and is close to the gene for hereditary breastovarian cancer. Genomics 1993;18:4147.[Medline]
90 Ehrenborg E, Zazzi H, Lagercrantz S, Granqvist M, Hillerbrand U, Allander SV, et al. Characterization and chromosomal localization of the human insulin-like growth factor-binding protein 6 gene. Mamm Genome 1999;10:37680.[Medline]
91 Lee PD, Giudice LC, Conover CA, Powell DR. Insulin-like growth factor binding protein-1: recent findings and new directions. Proc Soc Exp Biol Med 1997;216:31957.[Abstract]
92
Gucev ZS, Oh Y, Kelley KM, Labarta JI, Vorwerk P, Rosenfeld RG. Evidence for insulin-like growth factor (IGF)-independent transcriptional regulation of IGF binding protein-3 by growth hormone in SKHEP-1 human hepatocarcinoma cells. Endocrinology 1997;138:146470.
93 Gabbitas B, Canalis E. Cortisol enhances the transcription of insulin-like growth factor-binding protein-6 in cultured osteoblasts. Endocrinology 1996;137:168792.[Abstract]
94
Rozen F, Yang XF, Huynh H, Pollak M. Antiproliferative action of vitamin D-related compounds and insulin-like growth factor-binding protein 5 accumulation. J Natl Cancer Inst 1997;89:6526.
95
Colston KW, Perks CM, Xie SP, Holly JM. Growth inhibition of both MCF-7 and Hs578T human breast cancer cell lines by vitamin D analogues is associated with increased expression of insulin-like growth factor binding protein-3. J Mol Endocrinol 1998;20:15762.
96 Glantschnig H, Varga F, Klaushofer K. Thyroid hormone and retinoic acid induce the synthesis of insulin-like growth factor-binding protein-4 in mouse osteoblastic cells. Endocrinology 1996;137:2816.[Abstract]
97 Martin JL, Coverley JA, Pattison ST, Baxter RC. Insulin-like growth factor-binding protein-3 production by MCF-7 breast cancer cells: stimulation by retinoic acid and cyclic adenosine monophosphate and differential effects of estradiol. Endocrinology 1995;136:121926.[Abstract]
98
Oh Y, Muller HL, Ng L, Rosenfeld RG. Transforming growth factor-beta-induced cell growth inhibition in human breast cancer cells is mediated through insulin-like growth factor-binding protein-3 action. J Biol Chem 1995;270:1358992.
99 Zhou Y, Mohan S, Linkhart TA, Baylink DJ, Strong DD. Retinoic acid regulates insulin-like growth factor-binding protein expression in human osteoblast cells. Endocrinology 1996;137:97583.[Abstract]
100 Gabbitas B, Canalis E. Growth factor regulation of insulin-like growth factor binding protein-6 expression in osteoblasts. J Cell Biochem 1997;66:7786.[Medline]
101
Chambery D, de Galle B, Babajko S. Retinoic acid stimulates IGF binding protein (IGFBP)-6 and depresses IGFBP-2 and IGFBP-4 in SK-N-SH human neuroblastoma cells. J Endocrinol 1998;159:22732.
102 Olney RC, Wilson DM, Mohtai M, Fielder PJ, Smith RL. Interleukin-1 and tumor necrosis factor-alpha increase insulin-like growth factor-binding protein-3 (IGFBP-3) production and IGFBP-3 protease activity in human articular chondrocytes. J Endocrinol 1995;146:27986.[Abstract]
103
Claussen M, Kubler B, Wendland M, Neifer K, Schmidt B, Zapf J, et al. Proteolysis of insulin-like growth factors (IGF) and IGF binding proteins by cathepsin D. Endocrinology 1997;138:3797803.
104 Jones JI, D'Ercole AJ, Camacho-Hubner C, Clemmons DR. Phosphorylation of insulin-like growth factor (IGF)-binding protein 1 in cell culture and in vivo effect on affinity for IGF-I. Proc Natl Acad Sci U S A 1991;88:74815.[Abstract]
105 Coverley JA, Baxter RC. Phosphorylation of insulin-like growth factor binding proteins. Mol Cell Endocrinol 1997;128:15.[Medline]
106 Nickerson T, Huynh H, Pollak M. Insulin-like growth factor binding protein-3 induces apoptosis in MCF7 breast cancer cells. Biochem Biophys Res Commun 1997;237:6903.[Medline]
107
Oh Y, Muller HL, Lamson G, Rosenfeld RG. Insulin-like growth factor (IGF)-independent action of IGF-binding protein-3 in Hs578T human breast cancer cells. Cell surface binding and growth inhibition. J Biol Chem 1993;268:1496471.
108
Yamanaka Y, Fowlkes JL, Wilson EM, Rosenfeld RG, Oh Y. Characterization of insulin-like growth factor binding protein-3 (IGFBP-3) binding to human breast cancer cells: kinetics of IGFBP-3 binding and identification of receptor binding domain on the IGFBP-3 molecule. Endocrinology 1999;140:131928.
109 Rajah R, Bhala A, Nunn SE, Peehl DM, Cohen P. 7S nerve growth factor is an insulin-like growth factor-binding protein protease. Endocrinology 1996;137:267682.[Abstract]
110
Marinaro JA, Hendrich EC, Leeding KS, Bach LA. HaCaT human keratinocytes express IGF-II, IGFBP-6, and an acid-activated protease with activity against IGFBP-6. Am J Physiol 1999;276:E53642.
111 Cohen P, Graves HC, Peehl DM, Kamarei M, Guidice LC, Rosenfeld RG. Prostate-specific antigen (PSA) is an insulin-like growth factor binding protein-3 protease found in seminal plasma. J Clin Endocrinol Metab 1992;75:104653.[Abstract]
112 Cohen P, Peehl DM, Graves HC, Rosenfeld RG. Biological effects of prostate specific antigen as an insulin-like growth factor binding protein-3 protease. J Endocrinol 1994;142:40715.[Abstract]
113 Braulke T, Claussen M, Saftig P, Wendland M, Neifer K, Schmidt B, et al. Proteolysis of IGFBPs by cathepsin D in vitro and in cathepsin D-deficient mice. Prog Growth Factor Res 1995;6:26571.[Medline]
114
Fowlkes JL, Enghild JJ, Suzuki K, Nagase H. Matrix metalloproteinases degrade insulin-like growth factor-binding protein-3 in dermal fibroblast cultures. J Biol Chem 1994;269:257426.
115
Kubler B, Cowell S, Zapf J, Braulke T. Proteolysis of insulin-like growth factor binding proteins by a novel 50-kilodalton metalloproteinase in human pregnancy serum. Endocrinology 1998;139:155663.
116
Manes S, Llorente M, Lacalle RA, Gomez-Mouton C, Kremer L, Mira E, et al. The matrix metalloproteinase-9 regulates the insulin-like growth factor-triggered autocrine response in DU-145 carcinoma cells. J Biol Chem 1999;274:693545.
117 Lalou C, Silve C, Rosato R, Segovia B, Binoux M. Interactions between insulin-like growth factor-I (IGF-I) and the system of plasminogen activators and their inhibitors in the control of IGF-binding protein-3 production and proteolysis in human osteosarcoma cells. Endocrinology 1994;135:231826.[Abstract]
118
Zheng B, Clarke JB, Busby WH, Duan C, Clemmons DR. Insulin-like growth factor-binding protein-5 is cleaved by physiological concentrations of thrombin. Endocrinology 1998;139:170814.
119
Lawrence JB, Oxvig C, Overgaard MT, Sottrup-Jensen L, Gleich GJ, Hays LG, et al. The insulin-like growth factor (IGF)-dependent IGF binding protein-4 protease secreted by human fibroblasts is pregnancy-associated plasma protein-A. Proc Natl Acad Sci U S A 1999;96:3149 53.
120
Bang P, Fielder PJ. Human pregnancy serum contains at least two distinct proteolytic activities with the ability to degrade insulin-like growth factor binding protein-3. Endocrinology 1997;138:39127.
121 Donnelly MJ, Holly JM. The role of IGFBP-3 in the regulation of IGFBP-4 proteolysis. J Endocrinol 1996;149:R17.[Abstract]
122 Menouny M, Binoux M, Babajko S. IGFBP-2 expression in a human cell line is associated with increased IGFBP-3 proteolysis, decreased IGFBP-1 expression and increased tumorigenicity. Int J Cancer 1998;77:8749.[Medline]
123 Angelloz-Nicoud P, Binoux M. Autocrine regulation of cell proliferation by the insulin-like growth factor (IGF) and IGF binding protein-3 protease system in a human prostate carcinoma cell line (PC-3). Endocrinology 1995;136:548592.[Abstract]
124 Bereket A, Lang CH, Blethen SL, Fan J, Frost RA, Wilson TA. Insulin-like growth factor binding protein-3 proteolysis in children with insulin-dependent diabetes mellitus: a possible role for insulin in the regulation of IGFBP-3 protease activity. J Clin Endocrinol Metab 1995;80:22828.[Abstract]
125
Bang P, Nygren J, Carlsson-Skwirut C, Thorell A, Ljungqvist O. Postoperative induction of insulin-like growth factor binding protein-3 proteolytic activity: relation to insulin and insulin sensitivity. J Clin Endocrinol Metab 1998;83:250915.
126
Skjaerbaek C, Kaal A, Moller J, Vahl N, Weeke J, Orskov H, et al. No effect of growth hormone on serum insulin-like growth factor binding protein-3 proteolysis. J Clin Endocrinol Metab 1998;83:120610.
127 Kudo Y, Iwashita M, Itatsu S, Iguchi T, Takeda Y. Regulation of insulin-like growth factor-binding protein-4 protease activity by estrogen and parathyroid hormone in SaOS-2 cells: implications for the pathogenesis of postmenopausal osteoporosis. J Endocrinol 1996;150:2239.[Abstract]
128 Macaulay VM. Insulin-like growth factors and cancer. Br J Cancer 1992;65:31120.[Medline]
129 Oku K, Tanaka A, Yamanishi H, Nishizawa Y, Matsumoto K, Shiozaki H, et al. Effects of various growth factors on growth of a cloned human esophageal squamous cancer cell line in a protein-free medium. Anticancer Res 1991;11:15915.[Medline]
130
LeRoith D, Baserga R, Helman L, Roberts CT Jr. Insulin-like growth factors and cancer. Ann Intern Med 1995;122:549.
131 Yaginuma Y, Nishiwaki K, Kitamura S, Hayashi H, Sengoku K, Ishikawa M. Relaxation of insulin-like growth factor-II gene imprinting in human gynecologic tumors. Oncology 1997;54:5027.[Medline]
132 Singh P, Dai B, Yallampalli U, Lu X, Schroy PC. Proliferation and differentiation of a human colon cancer cell line (CaCo2) is associated with significant changes in the expression and secretion of insulin-like growth factor (IGF) IGF-II and IGF binding protein-4: role of IGF-II. Endocrinology 1996;137:176474.[Abstract]
133 Frostad S, Bruserud O. In vitro effects of insulin-like growth factor-1 (IGF-1) on proliferation and constitutive cytokine secretion by acute myelogenous leukemia blasts. Eur J Haematol 1999;62:1918.[Medline]
134
Rogler CE, Yang D, Rossetti L, Donohoe J, Alt E, Chang CJ, et al. Altered body composition and increased frequency of diverse malignancies in insulin-like growth factor-II transgenic mice. J Biol Chem 1994;269:1377984.
135 Bates P, Fisher R, Ward A, Richardson L, Hill DJ, Graham CF. Mammary cancer in transgenic mice expressing insulin-like growth factor-II (IGF-II). Br J Cancer 1995;72:118993.[Medline]
136
Toretsky A, Helman LJ. Involvement of IGF-II in human cancer. J Endocrinol 1996;149:36772.
137 Sohda T, Yun K, Iwata K, Soejima H, Okumura M. Increased expression of insulin-like growth factor 2 in hepatocellular carcinoma is primarily regulated at the transcriptional level. Lab Invest 1996;75:30711.[Medline]
138 Li X, Nong Z, Ekstrom C, Larsson E, Nordlinder H, Hofmann WJ, et al. Disrupted IGF2 promoter control by silencing of promoter P1 in human hepatocellular carcinoma. Cancer Res 1997;57:204854.[Abstract]
139 Guerra FK, Eijan AM, Puricelli L, Alonso DF, Bal de Kier Joffe E, Kornblihgtt AR, et al. Varying patterns of expression of insulin-like growth factors I and II and their receptors in murine mammary adenocarcinomas of different metastasizing ability. Int J Cancer 1996;65:812 20.[Medline]
140 Pietrzkowski Z, Mulholland G, Gomella L, Jameson BA, Wernicke D, Baserga R. Inhibition of growth of prostatic cancer cell lines by peptide analogues of insulin-like growth factor 1. Cancer Res 1993;53:11026.[Abstract]
141 Neuenschwander S, Roberts CT Jr, LeRoith D. Growth inhibition of MCF-7 breast cancer cells by stable expression of an insulin-like growth factor I receptor antisense ribonucleic acid. Endocrinology 1995;136:4298303.[Abstract]
142 Jiang Y, Rom WN, Yie TA, Chi CX, Tchou-Wong KM. Induction of tumor suppression and glandular differentiation of A549 lung carcinoma cells by dominant-negative IGF-I receptor. Oncogene 1999;28:60717.
143
Xie Y, Skytting B, Nilsson G, Brodin B, Larsson O. Expression of insulin-like growth factor-1 receptor in synovial sarcoma: association with an aggressive phenotype. Cancer Res 1999;59:358891.
144 Hakam A, Yeatman TJ, Lu L, Mora L, Marcet G, Nicosia SV, et al. Expression of insulin-like growth factor-1 receptor in human colorectal cancer. Hum Pathol 1999;30:112833.[Medline]
145 Belfiore A, Pandini G, Vella V, Squatrito S, Vigneri R. Insulin/IGF-I hybrid receptors play a major role in IGF-I signaling in thyroid cancer. Biochimie 1999;81:4037.[Medline]
146
Pandini G, Vigneri R, Costantino A, Frasca F, Ippolito A, Fujita-Yamaguchi Y, et al. Insulin and insulin-like growth factor-I (IGF-I) receptor overexpression in breast cancers leads to insulin/IGF-I hybrid receptor overexpression: evidence for a second mechanism of IGF-I signaling. Clin Cancer Res 1999;5:193544.
147 Sciacca L, Costantino A, Pandini G, Mineo R, Frasca F, Scalia P, et al. Insulin receptor activation by IGF-II in breast cancers: evidence for a new autocrine/paracrine mechanism. Oncogene 1999;18:24719.[Medline]
148
Byrd JC, Devi GR, de Souza AT, Jirtle RL, MacDonald RG. Disruption of ligand binding to the insulin-like growth factor II/mannose 6-phosphate receptor by cancer-associated missense mutations. J Biol Chem 1999;274:2440816.
149
O'Gorman DB, Costello M, Weiss J, Firth SM, Scott CD. Decreased insulin-like growth factor-II/mannose 6-phosphate receptor expression enhances tumorigenicity in JEG-3 cell. Cancer Res 1999;59:56924.
150 Souza RF, Wang S, Thakar M, Smolinski KN, Yin J, Zou TT, et al. Expression of the wild-type insulin-like growth factor II receptor gene suppresses growth and causes death in colorectal carcinoma cells. Oncogene 1999;18:40638.[Medline]
151 Yee D, Favoni RE, Lippman ME, Powell DR. Identification of insulin-like growth factor binding proteins in breast cancer cells. Breast Cancer Res Treat 1991;18:310.[Medline]
152 Pekonen F, Nyman T, Ilvesmaki V, Partanen S. Insulin-like growth factor binding proteins in human breast cancer tissue. Cancer Res 1992;52:52047.[Abstract]
153 Sheikh MS, Shao ZM, Clemmons DR, LeRoith D, Roberts CT Jr, Fontana JA. Identification of the insulin-like growth factor binding proteins 5 and 6 (IGFBP-5 and 6) in human breast cancer cells. Biochem Biophys Res Commun 1992;183:100310.[Medline]
154 Bachrach LK, Nanto-Salonen K, Tapanainen P, Rosenfeld RG, Gargosky SE. Insulin-like growth factor binding protein production in human follicular thyroid carcinoma cells. Growth Regul 1995;5:10918.[Medline]
155 Kimura G, Kasuya J, Giannini S, Honda Y, Mohan S, Kawachi M, et al. Insulin-like growth factor (IGF) system components in human prostatic cancer cell-lines: LNCaP, DU145, and PC-3 cells. Int J Urol 1996;3:3946.[Medline]
156 Tennant MK, Thrasher JB, Twomey PA, Birnbaum RS, Plymate SR. Insulin-like growth factor-binding proteins (IGFBP)-4, -5, and -6 in the benign and malignant human prostate: IGFBP-5 messenger ribonucleic acid localization differs from IGFBP-5 protein localization. J Clin Endocrinol Metab 1996;81:378392.[Abstract]
157 Reeve JG, Morgan J, Schwander J, Bleehen NM. Role for membrane and secreted insulin-like growth factor-binding protein-2 in the regulation of insulin-like growth factor action in lung tumors. Cancer Res 1993;53:46805.[Abstract]
158 Yee D, Jackson JG, Kozelsky TW, Figueroa JA. Insulin-like growth factor binding protein-1 expression inhibits insulin-like growth factor I action in MCF-7 breast cancer cells. Cell Growth Differ 1994;5:737.[Abstract]
159 Perks CM, Bowen S, Gill ZP, Newcomb PV, Holly JM. Differential IGF-independent effects of insulin-like growth factor binding proteins (16) on apoptosis of breast epithelial cells. J Cell Biochem 1999;75:65264.[Medline]
160 Schmid C, Rutishauser J, Schlapfer I, Froesch ER, Zapf J. Intact but not truncated insulin-like growth factor binding protein-3 (IGFBP-3) blocks IGF-I-induced stimulation of osteoblasts: control of IGF signalling to bone cells by IGFBP-3-specific proteolysis? Biochem Biophys Res Commun 1991;179:57985.[Medline]
161 Chen JC, Shao ZM, Sheikh MS, Hussain A, LeRoith D, Roberts CT Jr, et al. Insulin-like growth factor-binding protein enhancement of insulin-like growth factor-I (IGF-I)-mediated DNA synthesis and IGF-I binding in a human breast cancer carcinoma cell line. J Cell Physiol 1994;158:6978.[Medline]
162 Camacho-Hubner C, McCusker RH, Clemmons DR. Secretion and biological actions of insulin-like growth factor binding proteins in two human tumor-derived cell lines in vitro. J Cell Physiol 1991;148:2819.[Medline]
163
Gill ZP, Perks CM, Newcomb PV, Holly JM. Insulin-like growth factor-binding protein (IGFBP-3) predisposes breast cancer cells to programmed cell death in a non-IGF-dependent manner. J Biol Chem 1997;272:256027.
164
Rajah R, Valentinis B, Cohen P. Insulin-like growth factor (IGF)-binding protein-3 induces apoptosis and mediates the effects of transforming growth factor-beta1 on programmed cell death through a p53- and IGF-independent mechanism. J Biol Chem 1997;272:121818.
165 Ruan W, Catanese V, Wieczorek R, Feldman M, Kleinberg DL. Estradiol enhances the stimulatory effect of insulin-like growth factor-I (IGF-I) on mammary development and growth hormone-induced IGF-I messenger ribonucleic acid. Endocrinology 1995;136:1296302.[Abstract]
166
Stewart AJ, Johnson MD, May FE, Westley BR. Role of insulin-like growth factors and the type I insulin-like growth factor receptor in the estrogen-stimulated proliferation of human breast cancer cells. J Biol Chem 1990;265:211728.
167 Thorsen T, Lahooti H, Rasmussen M, Aakvaag A. Oestradiol treatment increases the sensitivity of MCF-7 cells for the growth stimulatory effect of IGF-I. J Steroid Biochem Mol Biol 1992;41:53740.[Medline]
168 Clarke RB, Howell A, Anderson E. Type I insulin-like growth factor receptor gene expression in normal human breast tissue treated with oestrogen and progesterone. Br J Cancer 1997;75:2517.[Medline]
169 Owens PC, Gill PG, De Young NJ, Weger MA, Knowles SE, Moyse KJ. Estrogen and progesterone regulate secretion of insulin-like growth factor binding proteins by human breast cancer cells. Biochem Biophys Res Commun 1993;193:46773.[Medline]
170 Manni A, Badger B, Wei L, Zaenglein A, Grove R, Khin S, et al. Hormonal regulation of insulin-like growth factor II and insulin-like growth factor binding protein expression by breast cancer cells in vivo: evidence for stromal epithelial interactions. Cancer Res 1994;54:293442.[Abstract]
171 Mathieu M, Vignon F, Capony F, Rochefort H. Estradiol down-regulates the mannose-6-phosphate/insulin-like growth factor-II receptor gene and induces cathepsin-D in breast cancer cells: a receptor saturation mechanism to increase the secretion of lysosomal proenzymes. Mol Endocrinol 1991;5:81522.[Abstract]
172 Clemmons DR, Camacho-Hubner C, Coronado E, Osborne CK. Insulin-like growth factor binding protein secretion by breast carcinoma cell lines: correlation with estrogen receptor status. Endocrinology 1990;127:267986.[Abstract]
173 Figueroa JA, Jackson JG, McGuire WL, Krywicki RF, Yee D. Expression of insulin-like growth factor binding proteins in human breast cancer correlates with estrogen receptor status. J Cell Biochem 1993;52:196205.[Medline]
174 Pratt SE, Pollak MN. Estrogen and antiestrogen modulation of MCF7 human breast cancer cell proliferation is associated with specific alterations in accumulation of insulin-like growth factor-binding proteins in conditional media. Cancer Res 1993;53:51938.[Abstract]
175
Huynh H, Yang X, Pollak M. Estradiol and antiestrogens regulate a growth inhibitory insulin-like growth factor binding protein 3 autocrine loop in human breast cancer cells. J Biol Chem 1996;271:101621.
176 Huynh H, Yang XF, Pollak M. A role of insulin-like growth factor binding protein 5 in the antiproliferative action of the antiestrogen ICI 182780. Cell Growth Differ 1996;7:15016.[Abstract]
177 Huynh HT, Tetenes E, Wallace L, Pollak M. In vivo inhibition of insulin-like growth factor I gene expression by tamoxifen. Cancer Res 1993;53:172730.[Abstract]
178 Guvakova MA, Surmacz E. Tamoxifen interferes with the insulin-like growth factor I receptor (IGF-IR) signaling pathway in breast cancer cells. Cancer Res 1997;57:260610.[Abstract]
179 Murphy LJ, Murphy LC, Friesen HG. Estrogen induces insulin-like growth factor-I expression in the rat uterus. Mol Endocrinol 1987;1:44550.[Abstract]
180 Huynh HT, Pollak M. Insulin-like growth factor I gene expression in the uterus is stimulated by tamoxifen and inhibited by the pure antiestrogen ICI 182780. Cancer Res 1993;53:55858.[Abstract]
181 Rajkumar K, Dheen T, Krsek M, Murphy LJ. Impaired estrogen action in the uterus of insulin-like growth factor binding protein-1 transgenic mice. Endocrinology 1996;137:125864.[Abstract]
182 Kleinman D, Karas M, Danilenko M, Arbell A, Roberts CT, LeRoith D, et al. Stimulation of endometrial cancer cell growth by tamoxifen is associated with increased insulin-like growth factor (IGF)-I induced tyrosine phosphorylation and reduction in IGF binding proteins. Endocrinology 1996;137:108995.[Abstract]
183 Ignar-Trowbridge DM, Pimentel M, Parker MG, McLachlan JA, Korach KS. Peptide growth factor cross-talk with the estrogen receptor requires the A/B domain and occurs independently of protein kinase C or estradiol. Endocrinology 1996;137:173544.[Abstract]
184 Lee AV, Weng CN, Jackson JG, Yee D. Activation of estrogen receptor-mediated gene transcription by IGF-I in human breast cancer cells. J Endocrinol 1997;152:3947.[Abstract]
185 Hwa IA, Reimann K, Lim PK, Lai LC. Effects of insulin-like growth factors I and II on oestrone sulphatase activity in human breast cancer cell lines. Int J Mol Med 1999;4:1758.[Medline]
186 Steller MA, Delgado CH, Zou Z. Insulin-like growth factor II mediates epidermal growth factor-induced mitogenesis in cervical cancer cells. Proc Natl Acad Sci U S A 1995;92:119704.[Abstract]
187 Steller MA, Delgado CH, Bartels CJ, Woodworth CD, Zou Z. Overexpression of the insulin-like growth factor-1 receptor and autocrine stimulation in human cervical cancer cells. Cancer Res 1996;56:17615.[Abstract]
188 Hembree JR, Agarwal C, Eckert RL. Epidermal growth factor suppresses insulin-like growth factor binding protein 3 levels in human papillomavirus type 16-immortalized cervical epithelial cells and thereby potentiates the effects of insulin-like growth factor 1. Cancer Res 1994;54:31606.[Abstract]
189
Putz T, Culig Z, Eder IE, Nessler-Menardi C, Bartsch G, Grunicke H, et al. Epidermal growth factor (EGF) receptor blockade inhibits the action of EGF, insulin-like growth factor I, and a protein kinase A activator on the mitogen-activated protein kinase pathway in prostate cancer cell lines. Cancer Res 1999;59:22733.
190 Kim SO, Park JG, Lee YI. Increased expression of the insulin-like growth factor I (IGF-I) receptor gene in hepatocellular carcinoma cell lines: implications of IGF-I receptor gene activation by hepatitis B virus X gene product. Cancer Res 1996;56:38316.[Abstract]
191 Lee YI, Lee S, Lee Y, Bong YS, Hyun SW, Yoo YD, et al. The human hepatitis B virus transactivator X gene product regulates Sp1 mediated transcription of an insulin-like growth factor II promoter 4. Oncogene 1998;16:236780.[Medline]
192 Nardone G, Romano M, Calabro A, Pedone PV, de Sio I, Persico M, et al. Activation of fetal promoters of insulinlike growth factors II gene in hepatitis C virus-related chronic hepatitis, cirrhosis, and hepatocellular carcinoma. Hepatology 1996;23:130412.[Medline]
193 Gucev ZS, Oh Y, Kelley KM, Rosenfeld RG. Insulin-like growth factor binding protein 3 mediates retinoic acid- and transforming growth factor beta2-induced growth inhibition in human breast cancer cells. Cancer Res 1996;56:154550.[Abstract]
194
Xie SP, James SY, Colston KW. Vitamin D derivatives inhibit the mitogenic effects of IGF-I on MCF-7 human breast cancer cells. J Endocrinol 1997;154:495504.
195 Huynh H, Pollak M, Zhang JC. Regulation of insulin-like growth factor (IGF) II and IGF binding protein 3 autocrine loop in human PC-3 prostate cancer cells by vitamin D metabolite 1,25 (OH) 2D3 and its analog EB1089. Int J Oncol 1998;13:13743.[Medline]
196 Buckbinder L, Talbott R, Velasco-Miguel S, Takenaka I, Faha B, Seizinger BR, et al. Induction of the growth inhibitor IGF-binding protein 3 by p53. Nature 1995;377:6469.[Medline]
197 Zhang L, Zhan Q, Zhan S, Kashanchi F, Fornace AJ Jr, Seth P, et al. p53 regulates human insulin-like growth factor II gene expression through active P4 promoter in rhabdomyosarcoma cells. DNA Cell Biol 1998;17:12531.[Medline]
198 Webster NJ, Resnik JL, Reichart DB, Strauss B, Haas M, Seely BL. Repression of the insulin receptor promoter by the tumor suppressor gene product p53: a possible mechanism for receptor overexpression in breast cancer. Cancer Res 1996;56:27818.[Abstract]
199
Ohlsson C, Kley N, Werner H, LeRoith D. p53 regulates insulin-like growth factor-I (IGF-I) receptor expression and IGF-I-induced tyrosine phosphorylation in an osteosarcoma cell line: interaction between p53 and Sp1. Endocrinology 1998;139:11017.
200 Takahashi K, Suzuki K. Association of insulin-like growth-factor-I-induced DNA synthesis with phosphorylation and nuclear exclusion of p53 in human breast cancer MCF-7 cells. Int J Cancer 1993;55:4538.[Medline]
201
Werner H, Rauscher FJ 3rd, Sukhatme VP, Drummond IA, Roberts CT Jr, LeRoith D. Transcriptional repression of the insulin-like growth factor I receptor (IGF-I-R) gene by the tumor suppressor WT1 involves binding to sequences both upstream and downstream of the IGF-I-R gene transcription start site. J Biol Chem 1994;269:1257782.
202
Tajinda K, Carroll J, Roberts CT Jr. Regulation of insulin-like growth factor I receptor promoter activity by wild-type and mutant versions of the WT1 tumor suppressor. Endocrinology 1999;140:471324.
203 Huynh H, Beamer W. Suppression of mammary-derived growth inhibitor gene expression by growth hormone and insulin-like growth factor I. Int J Oncol 1998;13:57782.[Medline]
204 Wick W, Furnari FB, Naumann U, Cavenee WK, Weller M. PTEN gene transfer in human malignant glioma: sensitization to irradiation and CD95L-induced apoptosis. Oncogene 1999;18:393643.[Medline]
205 Cohen LA, Choi KW, Wang CX. Influence of dietary fat, caloric restriction, and voluntary exercise on N-nitrosomethylurea-induced mammary tumorigenesis in rats. Cancer Res 1988;48:427683.[Abstract]
206 Weindruch R. Effect of caloric restriction on age-associated cancers. Exp Gerontol 1992;27:57581.[Medline]
207 Kritchevsky D. Caloric restriction and experimental carcinogenesis. Adv Exp Med Biol 1992;322:13441.
208 Hursting SD, Perkins SN, Phang JM. Caloric restriction delays spontaneous tumorigenesis in p53-knockout transgenic mice. Proc Natl Acad Sci U S A 1994;91:703640.[Abstract]
209
Mukherjee P, Sotnikov AV, Mangian HJ, Zhou JR, Visek WJ, Clinton SK. Energy intake and prostate tumor growth, angiogenesis, and vascular endothelial growth factor expression. J Natl Cancer Inst 1999;91:51223.
210 Dunn SE, Kari FW, French J, Leininger JR, Travlos G, Wilson R, et al. Dietary restriction reduces insulin-like growth factor I levels, which modulates apoptosis, cell proliferation, and tumor progression in p53-deficient mice. Cancer Res 1997;57:466772.[Abstract]
211
Baxter RC, Martin JL, Beniac VA. High molecular weight insulin-like growth factor binding protein complex. Purification and properties of the acid-labile subunit from human serum. J Biol Chem 1989;264:118438.
212
Khosravi MJ, Diamandi A, Mistry J, Krishna RG, Khare A. Acid-labile subunit of human insulin-like growth factor-binding protein complex: measurement, molecular, and clinical evaluation. J Clin Endocrinol Metab 1997;82:394451.
213
Juul A, Holm K, Kastrup KW, Pedersen SA, Michaelsen KF, Scheike T, et al. Free insulin-like growth factor I serum levels in 1430 healthy children and adults, and its diagnostic value in patients suspected of growth hormone deficiency. J Clin Endocrinol Metab 1997;82:2497502.
214 Poehlman ET, Copeland KC. Influence of physical activity on insulin-like growth factor-I in healthy younger and older men. J Clin Endocrinol Metab 1990;71:146873.[Abstract]
215 Kelly PJ, Eisman JA, Stuart MC, Pocock NA, Sambrook PN, Gwinn TH. Somatomedin-C, physical fitness, and bone density. J Clin Endocrinol Metab 1990;70:71823.[Abstract]
216 Donahue LR, Hunter SJ, Sherblom AP, Rosen C. Age-related changes in serum insulin-like growth factor-binding proteins in women. J Clin Endocrinol Metab 1990;71:5759.[Abstract]
217 Colletti RB, Copeland KC, Devlin JT, Roberts JD, McAuliffe TL. Effect of obesity on plasma insulin-like growth factor-I in cancer patients. Int J Obes 1991;15:5237.[Medline]
218 Yamamoto H, Sohmiya M, Oka N, Kato Y. Effects of aging and sex on plasma insulin-like growth factor I (IGF-I) levels in normal adults. Acta Endocrinol (Copenh) 1991;124:497500.[Medline]
219 Marin P, Kvist H, Lindstedt G, Sjostrom, Bjorntorp P. Low concentrations of insulin-like growth factor-I in abdominal obesity. Int J Obes Relat Metab Disord 1993;17:839.[Medline]
220 Landin-Wilhelmsen K, Wilhelmsen L, Lappas G, Rosen T, Lindstedt G, Lundberg PA, et al. Serum insulin-like growth factor I in a random population sample of men and women: relation to age, sex, smoking habits, coffee consumption and physical activity, blood pressure and concentrations of plasma lipids, fibrinogen, parathyroid hormone and osteocalcin. Clin Endocrinol (Oxf) 1994;41:3517.[Medline]
221 Rudman D, Drinka PJ, Wilson CR, Mattson DE, Scherman F, Cuisinier MC, et al. Relations of endogenous anabolic hormones and physical activity to bone mineral density and lean body mass in elderly men. Clin Endocrinol (Oxf) 1994;40:65361.[Medline]
222 Veldhuis JD, Liem AY, South S, Weltman A, Weltman J, Clemmons DA, et al. Differential impact of age, sex steroid hormones, and obesity on basal versus pulsatile growth hormone secretion in men as assessed in ultrasensitive chemiluminescence assay. J Clin Endocrinol Metab 1995;80:320922.[Abstract]
223 Goodman-Gruen D, Barrett-Connor E. Epidemiology of insulin-like growth factor-1 in elderly men and women. The Rancho Bernardo Study. Am J Epidemiol 1997;145:9706.[Abstract]
224 Porch JV, Jain K, Reilly A, Valdez C, Mazariegos M, Ziegler TR, et al. Aging, physical activity, insulin-like growth factor I, and body composition in Guatemalan women. Am J Clin Nutr 1997;66:8749.[Abstract]
225 Janssen JA, Stolk RP, Pols HA, Grobbee DE, de Jong FH, Lamberts SW. Serum free IGF-I, total IGF-I, IGFBP-1 and IGFBP-3 levels in an elderly population: relation to age and sex steroid levels. Clin Endocrinol (Oxf) 1998;48:4718.[Medline]
226 Soliman AT, Hassan AE, Aref MK, Hintz RL, Rosenfeld RG, Rogol AD. Serum insulin-like growth factors I and II concentrations and growth hormone and insulin responses to arginine infusion in children with protein-energy malnutrition before and after nutritional rehabilitation. Pediatr Res 1986;20:112230.[Abstract]
227 Clemmons DR, Klibanski A, Underwood LE, McArthur JW, Ridgway EC, Beitins IZ, et al. Reduction of plasma immunoreactive somatomedin C during fasting in humans. J Clin Endocrinol Metab 1981;53:124750.[Abstract]
228 Snyder DK, Clemmons DR, Underwood LE. Treatment of obese, diet-restricted subjects with growth hormone for 11 weeks: effects of anabolism, lipolysis, and body composition. J Clin Endocrinol Metab 1988;67:5461.[Abstract]
229 Forbes GB, Brown MR, Welle SL, Underwood LE. Hormonal response to overfeeding. Am J Clin Nutr 1989;49:60811.[Abstract]
230 Rudman D, Mattson DE. Serum insulin-like growth factor I in healthy older men in relation to physical activity. J Am Geriatr Soc 1994;42:716.[Medline]
231 Nicklas BJ, Ryan AJ, Treuth MM, Harman SM, Blackman MR, Hurley BF, et al. Testosterone, growth hormone and IGF-I responses to acute and chronic resistive exercise in men aged 5570 years. Int J Sports Med 1995;16:44550.[Medline]
232 Horber F, Kohler SA, Lippuner K, Jaeger P. Effect of regular physical training on age-associated alteration of body composition in men. Eur J Clin Invest 1996;26:27985.[Medline]
233 Roelen CA, de Vries WR, Koppeschaar HP, Vervoorn C, Thijssen JH, Blankenstein MA. Plasma insulin-like growth factor-I and high affinity growth hormone-binding protein levels increase after two weeks of strenuous physical training. Int J Sports Med 1997;18:23841.[Medline]
234 Bermon S, Ferrari P, Bernard P, Altare S, Dolisi C. Responses of total and free insulin-like growth factor-I and insulin-like growth factor binding protein-3 after resistance exercise and training in elderly subjects. Acta Physiol Scand 1999;165:516.[Medline]
235 Eliakim A, Brasel JA, Mohan S, Barstow TJ, Berman N, Cooper DM. Physical fitness, endurance training, and the growth hormone-insulin-like growth factor I system in adolescent females. J Clin Endocrinol Metab 1996;81:398692.[Abstract]
236
Eliakim A, Brasel JA, Mohan S, Wong WL, Cooper DM. Increased physical activity and the growth hormone-IGF-I axis in adolescent males. Am J Physiol 1998;275:R30814.
237 Bruning PF, Van Doorn J, Bonfrer JM, Van Noord PA, Korse CM, Linders TC, et al. Insulin-like growth-factor-binding protein 3 is decreased in early-stage operable pre-menopausal breast cancer. Int J Cancer 1995;62:26670.[Medline]
238 Underwood LE. Nutritional regulation of IGF-I and IGFBPs. J Pediatr Endocrinol Metab 1996;9 Suppl 3:30312.[Medline]
239 Juul A, Bang P, Hertel NT, Main K, Dalgaard P, Jorgensen K, et al. Serum insulin-like growth factor-I in 1030 healthy children, adolescents, and adults: relation to age, sex, stage of puberty, testicular size, and body mass index. J Clin Endocrinol Metab 1994;78:74452.[Abstract]
240 Yu H, Mistry J, Nicar MJ, Khosravi MJ, Diamandis A, van Doorn J, et al. Insulin-like growth factors (IGF-I, free IGF-I and IGF-II) and insulin-like growth factor binding proteins (IGFBP-2, IGFBP-3, IGFBP-6, and ALS) in blood circulation. J Clin Lab Anal 1999;13:16672.[Medline]
241 Nystrom FH, Ohman PK, Ekman BA, Osterlund MK, Karlberg BE, Arnqvist HJ. Population-based reference values for IGF-I and IGF-binding protein-1: relations with metabolic and anthropometric variables. Eur J Endocrinol 1997;136:16572.[Medline]
242 Juul A, Dalgaard P, Blum WF, Bang P, Hall K, Michaelsen KF, et al. Serum levels of insulin-like growth factor (IGF)-binding protein-3 (IGFBP-3) in healthy infants, children, and adolescents: the relation to IGF-I, IGF-II, IGFBP-1, IGFBP-2, age, sex, body mass index, and pubertal maturation. J Clin Endocrinol Metab 1995;80:253442.[Abstract]
243
Juul A, Moller S, Mosfeldt-Laursen E, Rasmussen MH, Scheike T, Pedersen SA, et al. The acid-labile subunit of human ternary insulin-like growth factor binding protein complex in serum: hepatosplanchnic release, diurnal variation, circulating concentrations in healthy subjects, and diagnostic use in patients with growth hormone deficiency. J Clin Endocrinol Metab 1998;83:440815.
244 Mohan S, Farley JR, Baylink DJ. Age-related changes in IGFBP-4 and IGFBP-5 levels in human serum and bone: implications for bone loss with aging. Prog Grow Factor Res 1995;6:46573.
245 Devine A, Rosen C, Mohan S, Baylink D, Prince RL. Effects of zinc and other nutritional factors on insulin-like growth factor I and insulin-like growth factor binding proteins in postmenopausal women. Am J Clin Nutr 1998;68:2006.[Abstract]
246 Smith WJ, Underwood LE, Clemmons DR. Effects of caloric or protein restriction on insulin-like growth factor-I (IGF-I) and IGF-binding proteins in children and adults. J Clin Endocriol Metab 1995;80:4439.[Abstract]
247 Lowe WL Jr, Adamo M, Werner H, Roberts CT Jr, LeRoith D. Regulation by fasting of rat insulin-like growth factor I and its receptor. Effects on gene expression and binding. J Clin Invest 1989;84:61926.[Medline]
248 Straus DS, Takemoto CD. Effect of fasting on insulin-like growth factor-I (IGF-I) and growth hormone receptor mRNA levels and IGF-I gene transcription in rat liver. Mol Endocrinol 1990;4:91100.[Abstract]
249 Straus DS, Takemoto CD. Effect of dietary protein deprivation on insulin-like growth factor (IGF)-I and -II, IGF binding protein-2, and serum albumin gene expression in rat. Endocrinology 1990;127:184960.[Abstract]
250 VandeHaar MJ, Moats-Staats BM, Davenport ML, Walker JL, Ketelslegers JM, Sharma BK, et al. Reduced serum concentrations of insulin-like growth factor-I (IGF-I) in protein-restricted growing rats are accompanied by reduced IGF-I mRNA levels in liver and skeletal muscle. J Endocrinol 1991;130:30512.[Abstract]
251 Miura Y, Kato H, Noguchi T. Effect of dietary proteins on insulin-like growth factor-1 (IGF-1) messenger ribonucleic acid content in rat liver. Br J Nutr 1992;67:25765.[Medline]
252
Straus DS. Nutritional regulation of hormones and growth factors that control mammalian growth. FASEB J 1994;8:612.
253 Scheett TP, Mills PJ, Ziegler MG, Stoppani J, Cooper DM. Effect of exercise on cytokines and growth mediators in prepubertal children. Pediatr Res 1999;46:42934.[Abstract]
254 Rosenfeld RG, Wilson DM, Lee PD, Hintz RL. Insulin-like growth factors I and II in evaluation of growth retardation. J Pediatr 1986;109:42833.[Medline]
255 Cohen P, Rosenfeld RG. Physiologic and clinical relevance of the insulin-like growth factor binding proteins. Curr Opin Pediatr 1994;6:4627.[Medline]
256 Clemmons DR, Van Wyk JJ, Ridgway EC, Kliman B, Kjellberg RN, Underwood LE. Evaluation of acromegaly by radio-immunoassay of somatomedin-C. N Engl J Med 1979;301:113842.[Abstract]
257 Jorgensen JO, Moller N, Moller J, Weeke J, Blum WF. Insulin-like growth factors (IGF)-I and -II and IGF binding protein-1, -2, and -3 in patients with acromegaly before and after adenomectomy. Metabolism 1994;43:57983.[Medline]
258
Sjogren K, Liu JL, Blad K, Skrtic S, Vidal O, Wallenius V, et al. Liver-derived insulin-like growth factor I (IGF-I) is the principal source of IGF-I in blood but is not required for postnatal body growth in mice. Proc Natl Acad Sci U S A 1999;96:708892.
259
Yakar S, Liu JL, Stannard B, Butler A, Accili D, Sauer B, et al. Normal growth and development in the absence of hepatic insulin-like growth factor I. Proc Natl Acad Sci U S A 1999;96:73249.
260 Santolaria F, Gonzalez-Gonzalez G, Gonzalez-Reimers E, Martinez-Riera A, Milena A, Rodgiguez-Moreno F, et al. Effects of alcohol and liver cirrhosis on the GH-IGF-I axis. Alcohol Alcohol 1995;30:7038.[Abstract]
261 Tomono M, Kiss Z. Ethanol enhances the stimulatory effects of insulin and insulin-like growth factor-1 on DNA synthesis in NIH 3T3 fibroblasts. Biochem Biophys Res Commun 1995;208:637.[Medline]
262 Srivastava VK, Hiney JK, Dees WL. Effects of ethanol on the intraovarian insulin-like growth factor-1 system in the prepubertal rat. Alcohol Clin Exp Res 1999;23:293300.[Medline]
263
Kaklamani VG, Linos A, Kaklamani E, Markaki I, Mantzoros C. Age, sex, and smoking are predictors of circulating insulin-like growth factor 1 and insulin-like growth factor-binding protein 3. J Clin Oncol 1999;17:8137.
264 Peyrat JP, Bonneterre J, Hecquet B, Vennin P, Louchez MM, Fournier C, et al. Plasma insulin-like growth factor-1 (IGF-1) concentrations in human breast cancer. Eur J Cancer 1993;29A:4927.
265 Papa V, Gliozzo B, Clark GM, McGuire WL, Moore D, Fujita-Yamaguchi Y, et al. Insulin-like growth factor-I receptors are overexpressed and predict a low risk in human cancer. Cancer Res 1993;53:373640.[Abstract]
266 Barni S, Lissoni P, Brivio F, Fumagalli L, Merlini D, Cataldo M, et al. Serum levels of insulin-like growth factor-I in operable breast cancer in relation to the main prognostic variables and their perioperative changes in relation to those of prolactin. Tumori 1994;80:2125.[Medline]
267 Favoni R, de Cupis A, Perrotta A, Sforzini S, Amoroso D, Pensa F, et al. Insulin-like growth factor-I (IGF-I) and IGF-binding proteins blood serum levels in women with early- and late-stage breast cancer: mutual relationship and possible correlations with patients' hormonal status. J Cancer Res Clin Oncol 1995;121:67482.[Medline]
268 Kaulsay KK, Ng EH, Ji CY, Ho GH, Aw TC, Lee KO. Serum IGF-binding protein-6 and prostate specific antigen in breast cancer. Eur J Endocrinol 1999;140:1648.[Medline]
269 Glass AR, Kikendall JW, Sobin LH, Bowen PE. Serum concentrations of insulin-like growth factor 1 in colonic neoplasia. Acta Oncol 1994;33:701.[Medline]
270 el Atiq F, Garrouste F, Remacle-Bonnet M, Sastre B, Pommier G. Alterations in serum levels of insulin-like growth factors and insulin-like growth-factor-binding proteins in patients with colorectal cancer. Int J Cancer 1994;57:4917.[Medline]
271 Rutanen EM, Stenman S, Blum W, Karkkainen T, Lehtovirta P, Stenman UH. Relationship between carbohydrate metabolism and serum insulin-like growth factor system in postmenopausal women: comparison of endometrial cancer patients with healthy controls. J Clin Endocrinol Metab 1993;77:199204.[Abstract]
272 Ayabe T, Tsutsumi O, Sakai H, Yoshikawa H, Yano T, Kurimoto F, et al. Increased circulating levels of insulin-like growth factor-I and decreased circulating levels of insulin-like growth factor binding protein-1 in postmenopausal women with endometrial cancer. Endocr J 1997;44:41924.[Medline]
273 Ho PJ, Baxter RC. Insulin-like growth factor-binding protein-2 in patients with prostate carcinoma and benign prostatic hyperplasia. Clin Endocrinol (Oxf) 1997;46:33342.[Medline]
274 Vadgama JV, Wu Y, Datta G, Khan H, Chillar R. Plasma insulin-like growth factor-I and serum IGF-binding protein 3 can be associated with the progression of breast cancer, and predict the risk of recurrence and the probability of survival in African-Amercian and Hispanic women. Oncology 1999;57:33040.[Medline]
275 Maiorano E, Loverro G, Viale G, Giannini T, Napoli A, Perlino E. Insulin-like growth factor-I expression in normal and diseased endometrium. Int J Cancer 1999;80:18893.[Medline]
276 Mantzoros CS, Tzonou A, Signorello LB, Stampfer M, Trichopoulos D, Adami HO. Insulin-like growth factor 1 in relation to prostate cancer and benign prostatic hyperplasia. Br J Cancer 1997;76:11158.[Medline]
277 Djavan B, Bursa B, Seitz C, Soeregi G, Remzi M, Basharkhah A, et al. Insulin-like growth factor 1 (IGF-1), IGF-1 density, and IGF-1/PSA ratio for prostate cancer detection. Urology 1999;54:6036.[Medline]
278 Cohen P, Peehl DM, Stamey TA, Wilson KF, Clemmons DR, Rosenfeld RG. Elevated levels of insulin-like growth factor-binding protein-2 in the serum of prostate cancer patients. J Clin Endocrinol Metab 1993;76:10315.[Abstract]
279 Kanety H, Madjar Y, Dagan Y, Levi J, Papa MZ, Pariente C, et al. Serum insulin-like growth factor-binding protein-2 (IGFBP-2) is increased and IGFBP-3 is decreased in patients with prostate cancer: correlation with serum prostate-specific antigen. J Clin Endocrinol Metab 1993;77:22933.[Abstract]
280 Tennant MK, Thrasher JB, Twomey PA, Birnbaum RS, Plymate SR. Insulin-like growth factor-binding protein-2 and -3 expression in benign human prostate epithelium, prostate intraepithelial neoplasia, and adenocarcinoma of the prostate. J Clin Endocrinol Metab 1996;81:41120.[Abstract]
281 Tennant MK, Thrasher JB, Twomey PA, Drivdahl RH, Birnbaum RS, Plymate SR. Protein and messenger ribonucleic acid (mRNA) for the type 1 insulin-like growth factor (IGF) receptor is decreased and IGF-II mRNA is increased in human prostate carcinoma compared to benign prostate epithelium. J Clin Endocrinol Metab 1996;81:377482.[Abstract]
282 Tennant MK, Thrasher JB, Twomey PA, Birnbaum RS, Plymate SR. Insulin-like growth factor-binding proteins (IGFBP)-4, -5, and -6 in the benign and malignant human prostate: IGFBP-5 messenger ribonucleic acid localization differs from IGFBP-5 protein localization. J Clin Endocrinol Metab 1996;81:378392.[Abstract]
283 Karasik A, Menczer J, Pariente C, Kanety H. Insulin-like growth factor-I (IGF-I) and IGF-binding protein-2 are increased in cyst fluids of epithelial ovarian cancer. J Clin Endocrinol Metab 1994;78:2716.[Abstract]
284
Flyvbjerg A, Mogensen O, Mogensen B, Nielsen OS. Elevated serum insulin-like growth factor-binding protein 2 (IGFBP-2) and decreased IGFBP-3 in epithelial ovarian cancer: correlation with cancer antigen 125 and tumor-associated trypsin inhibitor. J Clin Endocrinol Metab 1997;82:230813.
285 Kanety H, Kattan M, Goldberg I, Kopolovic J, Ravia J, Menczer J, et al. Increased insulin-like growth factor binding protein-2 (IGFBP-2) gene expression and protein production lead to high IGFBP-2 content in malignant ovarian cyst fluid. Br J Cancer 1996;73:106973.[Medline]
286 Shah NG, Bhatavdekar JM, Doctor SS, Suthar TP, Balar DB, Dave RS. Circulating epidermal growth factor (EGF) and insulin-like growth factor-I (IGF-I) in patients with epithelial ovarian carcinoma. Neoplasm 1994;41:2413.
287 Bohlke K, Cramer DW, Trichopoulos D, Mantzoros CS. Insulin-like growth factor-I in relation to premenopausal ductal carcinoma in situ of the breast. Epidemiology 1998;9:5703.[Medline]
288 Del Giudice ME, Fantus IG, Ezzat S, McKeown-Eyssen G, Page D, Goodwin PJ. Insulin and related factors in premenopausal breast cancer risk. Breast Cancer Res Treat 1998;47:11120.[Medline]
289 Agurs-Collins T, Adams-Campbell L, Sook K, Cullen KJ. Insulin-like growth factor-1 and breast cancer risk in postmenopausal African-American women [abstract]. Proc Am Assoc Cancer Res 1999;40:152.
290 Pollak M, Jazmaji V, Jenkins D, Marcon N, Saibil F, Stern H, et al. Relationship of colorectal cancer risk to serum insulin-like growth factor I and insulin-like growth factor binding protein 3 levels [abstract]. Late breaking session. Philadelphia (PA): 90th annual meeting of the American Association for Cancer Research; April 1014, 1999.
291 Giovannucci E, Pollak MN, Platz EA, Willett WC, Stampfer MJ, Majeed N, et al. Plasma insulin-like growth factor-1 and binding protein-3 and risk of colorectal cancer and adenoma in women [abstract]. Proc Am Assoc Cancer Res 1999;40:211.
292 Manousos O, Souglakos J, Bosetti C, Tzonou A, Chatzidakis V, Trichopoulos D, et al. IGF-I and IGF-II in relation to colorectal cancer. Int J Cancer 1999;83:157.[Medline]
293 Petridou E, Dessypris N, Spanos E, Mantzoros C, Skalkidou A, Kalmanti M, et al. Insulin-like growth factor-I and binding protein-3 in relation to childhood leukaemia. Int J Cancer 1999;80:4946.[Medline]
294 Mantzoros CS, Tzonou A, Signorello LB, Stampfer M, Trichopoulos D, Adami HO. Insulin-like growth factor 1 in relation to prostate cancer and benign prostatic hyperplasia. Br J Cancer 1997;76:11158.[Medline]
295
Wolk A, Mantzoros CS, Andersson SO, Bergstrom R, Signorello LB, Lagiou P, et al. Insulin-like growth factor 1 and prostate cancer risk: a population-based, casecontrol study. J Natl Cancer Inst 1998;90:9115.
296
Signorello LB, Brismar K, Bergstrom R, Andersson SO, Wolk A, Trichopoulos D, et al. Insulin-like growth factor-binding protein-1 and prostate cancer. J Natl Cancer Inst 1999;91:19657.
297 Peyrat JP, Bonneterre J, Beuscart R, Djiane J, Demaille A. Insulin-like growth factor 1 receptors in human breast cancer and their relation to estradiol and progesterone receptors. Cancer Res 1988;48:642933.[Abstract]
298 Pekonen F, Partanen S, Makinen T, Rutanen EM. Receptors for epidermal growth factor and insulin-like growth factor I and their relation to steroid receptors in human breast cancer. Cancer Res 1988;48:13437.[Abstract]
299 Foekens JA, Portengen H, Janssen M, Klijn JG. Insulin-like growth factor-1 receptors and insulin-like growth factor-1-like activity in human primary breast cancer. Cancer 1989;63:213947.[Medline]
300 Foekens JA, Portengen H, van Putten WL, Trapman AM, Reubi JC, Alexieva-Figusch J, et al. Prognostic value of receptors for insulin-like growth factor 1, somatostatin, and epidermal growth factor in human breast cancer. Cancer Res 1989;49:70029.[Abstract]
301 Bonneterre J, Peyrat JP, Beuscart R, Demaille A. Prognostic significance of insulin-like growth factor 1 receptors in human breast cancer. Cancer Res 1990;50:69315.[Abstract]
302 Berns EM, Klijn JG, van Staveren IL, Portengen H, Foekens JA. Sporadic amplification of the insulin-like growth factor 1 receptor gene in human breast tumors. Cancer Res 1992;52:10369.[Abstract]
303 Shao ZM, Sheikh MS, Ordonez JV, Feng P, Kute T, Chen JC, et al. IGFBP-3 gene expression and estrogen receptor status in human breast carcinoma. Cancer Res 1992;52:51003.[Abstract]
304 Pekonen F, Nyman T, Ilvesmaki V, Partanen S. Insulin-like growth factor binding proteins in human breast cancer tissue. Cancer Res 1992;52:52047.[Abstract]
305 Figueroa JA, Jackson JG, McGuire WL, Krywicki RF, Yee D. Expression of insulin-like growth factor binding proteins in human breast cancer correlates with estrogen receptor status. J Cell Biochem 1993;52:196205.[Medline]
306 McGuire SE, Hilsenbeck SG, Figueroa JA, Jackson JG, Yee D. Detection of insulin-like growth factor binding proteins (IGFBPs) by ligand blotting in breast cancer tissues. Cancer Lett 1994;77:2532.[Medline]
307 Yee D, Sharma J, Hilsenbeck SG. Prognostic significance of insulin-like growth factor-binding protein expression in axillary lymph node-negative breast cancer. J Natl Cancer Inst 1994;86:17859.[Abstract]
308 Bhatavdekar JM, Patel DD, Karelia NH, Vora HH, Ghosh N, Shah NG, et al. Tumor markers in patients with advanced breast cancer as prognosticators: a preliminary study. Breast Cancer Res Treat 1994;30:2937.[Medline]
309 Toropainen EM, Lipponen PK, Syrjanen KJ. Expression of insulin-like growth factor II in female breast cancer as related to established prognostic factors and long-term prognosis. Anticancer Res 1995;15:266974.[Medline]
310
Rocha RL, Hilsenbeck SG, Jackson JG, Lee AV, Figueroa JA, Yee D. Correlation of insulin-like growth factor-binding protein-3 messenger RNA with protein expression in primary breast cancer tissues: detection of higher levels in tumors with poor prognostic features. J Natl Cancer Inst 1996;88:6016.
311 Yu H, Levesque MA, Khosravi MJ, Papanastasiou-Diamandi A, Clark GM, Diamandis EP. Associations between insulin-like growth factors and their binding proteins and other prognostic indicators in breast cancer. Br J Cancer 1996;74:12427.[Medline]
312 Rocha RL, Hilsenbeck SG, Jackson JG, VanDenBerg CL, Weng CN, Lee AV, et al. Insulin-like growth factor binding protein-3 and insulin receptor substrate-1 in breast cancer: correlation with clinical parameters and disease-free survival. Clin Cancer Res 1997;3:1039.[Abstract]
313 Yu H, Levesque MA, Khosravi MJ, Papanastasiou-Diamandi A, Clark GM, Diamandis EP. Insulin-like growth factor-binding protein-3 and breast cancer survival. Int J Cancer 1998;79:6248.[Medline]
314 Turner BC, Haffty BG, Narayanan L, Yuan J, Havre PA, Gumbs AA, et al. Insulin-like growth factor-I receptor overexpression mediates cellular radioresistance and local breast cancer recurrence after lumpectomy and radiation. Cancer Res 1997;57:307983.[Abstract]
315 Resnik JL, Reichart DB, Huey K, Webster NJ, Seely BL. Elevated insulin-like growth factor I receptor autophosphorylation and kinase activity in human breast cancer. Cancer 1998;58:115964.
Manuscript received January 13, 2000; revised June 28, 2000; accepted July 16, 2000.
This article has been cited by other articles in HighWire Press-hosted journals:
![]() |
||||
|
Oxford University Press Privacy Policy and Legal Statement |