Insulin-Like Growth Factors and Breast Cancer Therapy

Zeng X, Yee D.

Publication Details

Despite improvements in breast cancer therapy in recent years, additional therapies need to be developed. New therapies may have activity by themselves or may have utility in combination with other agents. Population, preclinical, and basic data suggest the insulin-like growth factor (IGF) system functions to maintain the malignant phenotype in breast cancer. Since the IGFs act via transmembrane tyrosine kinase receptors, targeting of the key receptors could provide a new pathway in breast cancer. In addition, IGF action enhances cell survival, so combination of anti-IGF therapy with conventional cytotoxic drugs could lead to synergistic effects. In this review, we will discuss the rationale for targeting the IGF system, potential methods to disrupt IGF signaling, and identify potential interactions between IGF inhibitors and other anti-tumor strategies. We will also identify important issues to consider when designing clinical trials.

Introduction

Breast cancer is the most common cancer in women responsible for over 40.000 deaths in the United States.1 Since cancer death is almost always caused by growth of breast cancer in metastatic sites, systemic cytotoxic and endocrine therapies are commonly given. In operable breast cancer, systemic adjuvant therapy is employed to reduce the risk of recurrence and prolong overall survival. In women with metastatic disease, systemic therapy is given to control growth of breast cancer in distant organs. Both treatments essentially target metastatic disease; in the adjuvant setting, the goal is to eradicate any microscopic disease that is not clinically detectable while treatment of metastatic disease targets clinically obvious sites.

Though systemic treatment is clearly effective in both adjuvant and advanced settings, therapy is far from completely effective. In the adjuvant setting, administration of chemotherapy reduces the relative risk of recurrence by approximately 30%.2 Identifying methods to enhance the cytotoxicity of chemotherapy are clearly needed. Since normal and cancer cells receive survival and proliferative signals from their extracellular environment, targeting of these signals could enhance the clinical benefit of chemotherapy. Indeed, trastuzumab, an antibody directed against the human epidermal growth factor receptor-2 (HER2), is commonly used in combination with chemotherapy for women with advanced cancer.3 While the exact mechanism for the synergy between trastuzumab and chemotherapy is not completely understood, it is likely that trastuzumab renders cells more sensitive to the apoptotic effects of chemotherapy by attenuating cells survival pathways.

Identifying additional survival pathways could also be used to enhance the benefits of chemotherapy. Insulin-like growth factors (IGFs) and the IGF signaling pathways play a role in development of the normal mammary gland. Numerous studies have now demonstrated that the IGF system regulates all of the key metastatic phenotypes in breast cancer cells: survival, proliferation, and metastasis.

Extensive data are available on the importance of IGF system in growth regulation of breast cancer cell lines.4 The type I insulin-like growth factor receptor (IGF-IR) is significantly overexpressed5 or hyperphosphorylated in tumor cells relative to normal breast epithelium and benign tumors.5,6 In addition, several clinical studies also support a role for IGF-I in breast cancer risk.7,8 In cell line model systems, IGF-IR conferred resistance to trastuzumab-induced growth inhibition9 and a kinase inhibitor to IGF-IR increased radiosensitivity in some breast cancer cells lines.10 Thus, data from population and laboratory studies provide a rationale for targeting IGF-IR in breast cancer. Moreover, disruption of IGF-IR may render cells more sensitive to apoptotic stimuli. Several reviews have already addressed the important role of the IGF system in breast cancer.11-13 Here we will summarize the potential role of IGF action on breast cancer chemotherapy.

The IGF System

The IGF system involves the coordination of growth factors (IGF-I and IGF-II), cell surface receptors (IGF-IR, IGF-IIR, and the insulin receptor IR), six high affinity binding proteins (IGFBP-1 to 6), IGFBP proteases, and the downstream proteins involved in intracellular signaling distal to IGF-IR.

IGF-I and IGF-II are single-chain 7.5 kDa polypeptide growth factors with a high degree of homology to insulin. The main function of IGF-I is to act as an effector molecule of growth hormone (GH), which is fundamental to linear growth and development.14 During puberty, pulsatile GH release from the pituitary stimulates expression of IGF-I in the liver. In addition to its endocrine role, it has been suggested that IGF-I may also have an important role in prenatal growth. Mice with a homozygous deletion of the IGF-I gene have a birth weights less than 60% of their wildtype littermates, these mice have a high post-natal mortality rate.15 Thus, besides its endocrine role, IGF-I plays an important paracrine and autocrine role during normal development and growth of the organism. IGF-II expression is not regulated by GH. However, IGF-II has proliferative and antiapoptotic actions similar to IGF-I. In addition, IGF-II plays a fundamental role in embryonic and fetal growth, this was proven by IGF-II gene knockout mice, which survive but remain smaller than their wildtype littermates.16 Interestingly, size at birth and height at age 14 have been linked to increase breast cancer risk suggesting an etiologic role for the IGFs and breast cancer development.17

The actions of IGFs can be modulated by interaction with a family of six insulin-like growth factor-binding proteins, IGFBP-1 to IGFBP-6, which share 40-60% amino acid identity. IGFBP3 is the largest and most abundant IGFBP, more than 75% IGF is confined to the vascular compartment as a ternary complex with IGFBP3 and the acid labile subunit. By binding IGF-I and IGF-II, IGFBPs regulated the bioavailability of IGFs in the circulation. IGFBP-3 has also been shown to competitively inhibit IGF action at the cellular level in the absence of IGF binding and exert IGF-independent proapoptotic and antiproliferative effects through the activation of caspases involved in a death receptor-mediated pathway.18

Although at least two receptors for IGFs exist, the primary signaling receptor through which both IGF-I and IGF-II exert their biological actions is the IGF-IR. Hybrid IGF-IR/Insulin receptors also exist and could mediate IGF and insulin action in breast cancer cells.19,20 Presence of the hybrid receptor adds an additional layer of complexity to IGF signaling.

Binding of ligand to the receptor induces autophosphorylation and activation of multiple downstream cell survival and proliferation signaling pathway via recruitment and tyrosine phosphorylation of specific adaptor/effector molecules.21 While individual pathways have been linked to specific cancer phenotypes, it is clear that the intracellular signals initiated after IGF-IR activation constitute a network of interacting molecular events. It is likely oversimplistic to ascribe a specific behavior to a specific pathway. Nonetheless, activation of PI3 kinase downstream of the insulin receptor substrate-1 (IRS-1) adaptor protein has been linked to cell survival and regulation of several proteins involved in apoptosis.22 Induction of cell growth and proliferation has been linked to both the PI3 kinase pathway and the MAP kinase pathway downstream of IGF-IR and its phosphorylated substrates, IRS-1 and Src-collagen homology (Shc) protein. The type II insulin-like growth factor receptor (IGF-IIR) lacks tyrosine kinase activity and appears to exert antiproliferative and proapoptotic activities by sequestration of IGF-II, reducing its availability for interaction with the IGF-IR.23

IGFs and Normal Mammary Tissue

IGFs play a key role in proliferation and survival in the mammary gland, particularly during puberty and pregnancy.24,25 IGF-I is a potent mitogen for mammary epithelial cells and in combination with mammotrophic hormones, such as estrogen receptor (ER), it induces ductal growth in mammary gland explant cultures.26 IGF-IR null mice have deficient mammary development with reductions in the number of terminal end buds, ducts and the per cent of the fat pad occupied by glandular elements. This phenotype is partially restored by administration of des(1-3) IGF-I.27 In addition, IGF-I also plays a role in the maintenance of the adult mammary gland during lactation, lactating transgenic mice overexpressing the Igf1 gene undergo ductal hypertrophy and fail to show normal mammary gland involution following weaning.28 The same group also demonstrated that IGF-I slows the apoptotic loss of mammary epithelial cells during the declining phase of lactation.29

It is known that IGFs is one of the developmental/essential survival factors for the mammary gland, although other factors such as epidermal growth factor (EGF) and its homologues also deliver intracellular signals that suppress apoptosis. Direct evidence for IGFs as survival factors comes from culture studies.24,30,31 IGF-I or IGF-II can suppress the apoptosis of mammary epithelial cells induced by serum withdrawal. It has been recently established that this is achieved through PI3K and MAPK signals that ultimately inhibit activity of a proapoptotic protein, BAD and enhance expression of another antiapoptotic protein Bcl-xL.24,30 Indirect evidence came from the transgenic mice overexpressing IGFBP-5 in the mammary gland, these mice had reduced numbers of alveolar end buds, with decreased ductal branching and increased expression of the pro-apoptotic molecule caspase-3,and decreased expression of pro-survival molecules of the Bcl-2 family.32

IGF and Breast Cancer

The IGFs and IGF-IR function to promote proliferation, inhibit death and stimulate transformation in breast cancer cells.11 High levels of serum IGF-I are associated with an increased risk of breast cancer in premenopausal women.33 There is also substantial evidence that IGF expression occurs locally in breast cancer tissues. Although IGF-I is rarely expressed in primary breast cancer, IGF-II message is more frequently detectable in breast cancer cells compared to normal cells.34 Moreover, the stroma is a rich source of both IGFs.

Studies in transgenic mice have revealed an important role of IGF-I in mammary tumorigenesis. Transgenic mice expressing human des(1-3) IGF-I (under the control of the rat whey acid protein) display an increased incidence of mammary tumors, with 53% of the mice developing mammary adenocarcinomas by 23 months of age.35,36 Furthermore, data in a transgenic mouse system suggest that mice deficient in liver-expressed IGF-I have a reduced ability to develop mammary tumors.37 In human studies, circulating IGF-I levels are higher in breast cancer patients than in controls. In addition, cohort studies have shown that higher levels of circulating IGF-I are associated with an increased risk of breast cancer in premenopausal women.38

The IGF-IR, the primary mediator of the biological actions of IGF-I, has been detected in a majority of primary breast tumor samples with overexpression in 30% to 40% of breast cancers.39 Furthermore, IGF-IR autophosphorylation has been found to be elevated in human breast cancer suggesting that this is an active pathway in primary breast cancer.6,40 Interestingly, a high level of IGF-IR in patients with breast cancer is associated with a greater recurrence risk of recurrence after local radiation therapy.41

Insulin receptor substrate-1 (IRS-1), the primary signaling molecule activated in response to IGF in MCF-7 human breast cancer cells, is reported to be overexpressed in some primary breast tumors and a high IRS-1 are associated with a decreased disease-free survival in a subset of patients with all tumors.42,43 Activation of specific IRS species are associated with distinct biological effects.44 Activation of IRS-1 signaling was associated with cell growth, whereas insulin receptor substrate-2 (IRS-2) signaling was associated with cell motility.44,45 Nagle et al showed that mammary tumor cells obtained from IRS-2 knock-out mice were less invasive and more apoptotic in response to growth factor deprivation than their WT counterparts. In contrast, IRS-1(-/-) tumor cells, which express only IRS-2, were highly invasive and were resistant to apoptotic stimuli.46 These data suggest that signaling pathways downstream of IGF-IR may ultimately be responsible for the malignant phenotype mediated by this growth factor signaling system.

Breast cancer cells also produce several IGFBPs that could modulate IGF action. In addition to the indirect modulation of IGF's mitogenic and antiapoptotic signals by ligand sequestration, IGFBPs also exert IGF-independent effects on cell survival. For example, IGFBP-4 and IGFBP-5 can rescue cells from ceramide or integrin-mediated apoptosis, which may account for the poor prognosis in breast cancers with high IGFBP-4 expression13,47 whereas IGFBP-3 has been found to directly inhibit breast cancer cell growth without interacting with IGFs.13,48

Conventional Chemotherapy for Breast Cancer

It is well established that most chemotherapeutic agents eliminate cells by triggering apoptosis. Most currently approved agents target specific molecules required for a cell to traverse the cell cycle. Targets range from DNA itself, to enzymes (topoisomerases), or structural proteins (tubulin) required for cell division. There are three kinds of cells in a solid tumor: dividing cell that are continuously cycling, resting cell which may potentially enter the cell cycle, and those cells no longer able to divide. In breast cancer, Clarke et al have suggested that only a minority of cells contained within a tumor have the capacity to contribute to all these subpopulations of tumor cells leading to the idea that cancer stem cells exist.49 Essentially only dividing cells are susceptible to the currently available chemotherapy drugs. It is the existence of resting or stem cells that makes it difficult to completely eradicate advanced tumors by chemotherapy; even after a substantial clinical response, a population of cells may still exist that have full capacity to enter the cell cycle.

The primary therapy of localized-early stage I and II- breast cancer is either breast-conserving surgery and radiation therapy or mastectomy with or without reconstruction.50 Systemic adjuvant therapies designed to eradicate clinically undetectable microscopic deposits of cancer cells that may have spread from the primary tumor result in decreased recurrences and improved survival.2,51 Adjuvant therapies include chemotherapy and hormonal therapy. In the adjuvant setting, chemotherapy is usually given in combination for 4-6 months. A wide variety of agents have been effective in breast cancer including DNA alkylators (cyclophosphamide), topoisomerase inhibitors/DNA intercalators (doxorubicin), anti-metabolites (5-fluorouracil, methotrexate), and tubulin interacting agents (paclitaxel).2 Adjuvant chemotherapy effectively reduces the odds of recurrence and death by approximately 20% to 60% of patients. However, since this reduction of risk is not complete, substantial research effort is directed toward improving the benefits of chemotherapy. New target discovery and combination of new agents with “conventional” agents represent an active area of investigation.

IGF Signaling Confers Resistance to Chemotherapy

Besides inducing cell cycle progression, IGF-I also protects breast cancer cells from drug-induced apoptosis.52-54 In fibroblasts, protection from apoptosis requires the tyrosine kinase activity of IGF-IR, as kinase defective receptors do not protect fibroblasts from apoptosis.55 In addition to drug-induced apoptosis, IGF-IR activation appears to block other stimuli as well. For example, BNIP3 (Bcl-2/E1B 19 kDa interacting protein) is a proapoptotic member of the Bcl-2 family expressed in hypoxic regions of tumors. Treatment of the breast cancer cell line MCF-7 cells with IGF effectively protected these cells from BNIP3-induced cell death56 likely via activation of PI3K and the Akt/PKB pathways.55,57 Akt/PKB phosphorylates BAD, a member of the Bcl-2 family of proapoptotic proteins, phosphorylated BAD cannot heterodimerize with Bcl-2 or Bcl-Xl, remains in the cytosol and cell death is inhibited.58 It has also been suggested that IGF can inhibit apoptosis by increasing the expression of Bcl-XL at both the mRNA and protein level.59 Thus, signaling from IGF-IR to multiple proteins involved in the intrinsic apoptotic pathways suggest a mechanism for protection from cell death signals.

IGFs provide resistance of breast cancer cells to chemotherapeutic agents.53 IGF-I alters drug sensitivity of HBL100 human breast cancer cell by inhibition of apoptosis induced by diverse anticancer drugs, it increased cell survival of HBL100 cells treated with 5-FU, methotrexate, tamoxifen or camptothecin, but no changes were observed in Bcl-2 protein or Bax mRNA levels.60 IGF-I signaling is also associated with resistance to the growth-inhibitory actions of trastuzumab by upregulation of ubiquitin-related p27kip1 degradation and activation of the PI3K signaling pathway.61 In breast cancer cells, IGF-I can activate JNK which is generally associated with a pro-apoptotic response. However, activation of Akt seems to override pro-apoptotic effects of JNK activation.62 Thus targeting IGF-IR could enhance a pro-apoptotic response initiated by many different agents.

IGF-IR and DNA Repair

Several reports indicate that IGF-IR activation is also important in regulating DNA repair. Fibroblasts can be rescued from DNA damage by IGF-I via activation of the p38 MAP kinase signaling pathway.63,64 Increasing IGF-IR expression increased radioresistance in breast tumor cells,65 and delayed UVB-induced apoptosis by enhancing repair of DNA cyclobutane thymidine dimers in keratinocytes.66,67 IGF-I stimulation supports homologous recombination-directed DNA repair (HRR) via an interaction between IRS-1 and Rad51, a key enzyme of HRR.68 In contrast, IGF-I may actually inhibit the ability of A549 cells to repair potentially lethal DNA damage induced by radiation.69 Though these observations are somewhat conflicting as they suggest IGF-IR may both enhance and inhibit DNA repair, it is possible that these differences relate to the varied experimental model systems and cell types studied. However, these experiments support a link between DNA repair and IGF-I action and the exact IGF effects may be context dependent.

Effects of Breast Cancer Therapy on the IGF System

On the other hand, breast cancer chemotherapy also affects IGFs. It has been reported that serum IGFBP-3 falls significantly following initiation of chemotherapy in breast cancer patients, those individuals with a decrease in IGFBP-3 greater than the median had significantly poorer survival (median survival 5.5 months vs 18 months).70 Another clinical trial showed that plasma IGF-I concentration significantly decreased after the first cycle of cyclophosphamide, methotrexate and 5-fluorouracil adjuvant chemotherapy in breast cancer patients.71 Retinoids such as fenretinide (4-HPR) inhibit breast cell growth while decreasing IGF-I and increase IGFBP-3.72,73

Proline analogues of melphalan can be effectively transported into the MDA-MB 231 cells, evoking higher cytotoxicity, with reduction in IGF-I receptor and MAP kinase expression.74 Tamoxifen also affects the IGF system. IGFBP-3,4,6 levels are lower in breast cancer patients compared to normal controls and levels increased after tamoxifen treatment.75,76 Raloxifene, a selective estrogen receptor modulator being tested in cancer prevention trials, significantly decreased IGF-I and IGF-I/IGFBP-3 ratios when compared to placebo.77 Urokinase plasminogen activator(uPA) inhibitor -17 AAG inhibit MDA-MB-231 cell growth by inhibiting the IGF-IR and ultimately uPA, while expression of the IGF-IR and uPA in breast cancer is associated with poor survival.78

Thus, it is clear that anti-proliferative agents affect IGF system signaling. These associations do not prove a cause and effect relationship, however, given the role of IGF signaling in cell survival, the downregulation of this signaling pathway is consistent with the effects of many anti-cancer drugs.

Anti-IGF Strategies in Breast Cancer

Given the role for IGF signaling in many aspects of the malignant phenotype, it would be valuable to have reagents to disrupt IGF action. Several strategies to interrupt IGF signaling are currently under investigation, including endocrine maneuvers to suppress IGF production; antisense oligonucleotides to reduce functional IGF-IR levels; monoclonal antibodies, dominant negatives, and tyrosine kinase inhibitors to inhibit IGF-IR activation; and neutralization of IGF action using IGFBPs.79,80

Suppression of IGF Production

The majority of circulating IGF-I is produced by the liver in response to growth hormone (GH) released from the pituitary gland, acting through hepatic growth hormone receptors (GHR). GH-releasing hormone antagonists disrupt the pituitary production of GH and reduce circulating levels of GH and have been shown to inhibit the growth of a variety of cancers in animal model systems, including breast cancer.81,82 Somatostatin and its analogues also inhibit the release of GH and thyroid-stimulating hormone from the pituitary gland. Preclinical studies on the anticancer activity of the somatostatin analog octreotide showed 50% reduction in tumor growth using two in vivo breast cancer models, ZR-75-1 breast xenografts and DMBA induced mammary tumors in rats. However, octreotide administered with tamoxifen did not improve response or survival in patients with metastatic breast cancer compared to tamoxifen alone.83 While octreotide was able to reduce serum IGF-I levels, it was possible that this reduction was insufficient to block IGF-IR signaling.

More potent methods to disrupt endocrine IGF-I have been developed. GH-RH antagonists MZ-5-156 or JV-1-36 administered induced the growth-arrest of estrogen-independent MDA-MB-468 human breast cancers xenografted into nude mice.84 Pegvisomant, a competitive antagonist of GHR, is the most potent therapy for reducing serum IGF-I levels in acromegalic patients and may have a role in cancer treatment.84 However, these strategies to disrupt endocrine IGF-I do not address paracrine or autocrine sources of IGF-I. Furthermore, IGF-II is not under GH control, and merely suppressing serum IGF-I levels may be insufficient to block all IGF ligands.

Ligand Neutralization Using IGFBPs

Since IGF ligands are required to activate IGF-IR, disruption of ligand-receptor interactions is an attractive method to disrupt IGF signaling. Blockade of IGF-mediated cellular effects can be accomplished with overexpression of IGFBPs or by treatment with exogenous IGFBPs. IGFBP-1, either exogenously added or endogenously produced, has been observed to inhibit IGF-IR function resulting in inhibition of IGF-I mediated growth of MCF-7 breast cancer cells.85,86 Silibinin has been shown to have anti-proliferative action against some malignant cell lines by increasing IGFBP3 mRNA and protein levels.87 In vivo, treatment with polyethylene glycol-conjugated recombinant IGFBP-1, inhibited growth of MDA-MB-231 breast tumor xenografts and malignant ascites formation in the MDA-MB-435/LCC6 cells.85 Similar neutralization of IGF ligands has been accomplished using the extracellular domain of IGF-IR88,89 and with neutralizing antibodies.90 Thus, several methods to neutralize IGF ligand interaction with IGF-IR have been tested. Neutralization strategies have the advantage of targeting both IGF-I and IGF-II without the need to identify a specific receptor subtype.

Inhibition of IGF-IR Activation

Abundant evidence implicating IGF-IR is essential for the transformed phenotype and inhibition of apoptosis in breast cancer, targeting this receptor directly may be an effective cancer therapy. Antibody blockade of growth factor receptors is a proven strategy to inhibit receptor-mediated effects, with the effectiveness of trastuzumab against HER2 overexpressing breast cancers being a prime example. Several anti-IGF-IR antibodies have been developed and tested in preclinical model systems. α-IR3, the first monoclonal antibody directed against IGF-IR, inhibited clonal growth and blocked the mitogenic effects of exogenous IGF-I in breast cancer cells in vitro.91 Interestingly, α-IR3 inhibited MDA-MB-231 tumor formation in athymic mice when administered at the time of tumor cell inoculation, but was ineffective against MCF-7 tumor xenografts. Since MCF-7 cells are sensitive to IGF-IR blockade in vitro, it was possible that the pharmacokinetic properties of the antibody are an important determinant of anti-tumor activity. A chimeric humanized single chain antibody (scFv-Fc), a partial agonist of IGF-IR, exhibited dose-dependent growth inhibition of IGF-IR-overexpressing NIH-3T3 cells, and significantly suppresses MCF-7 breast tumor growth in athymic mice.92-94 EM164, a purely antagonistic anti-IGF-IR antibody, displays potent inhibitory activity against IGF-I and IGF-II, and serum-stimulated proliferation and survival of MCF-7 breast cancer cells.95 A high-affinity fully human monoclonal antibody, A12, blocks IGF-I and IGF-II signaling and exhibits strong anti-tumor cell activity against MCF-7 xenograft tumors by enhancing apoptosis.96

An alternative strategy to inhibit IGF action is to target the tyrosine kinase activity of the receptor. Several members of the tyrphostin tyrosine kinase inhibitor family (e.g., AG10124, AG1024, AG538, and I-OMe AG538) were shown to competitively inhibit IGF-IR autophosphorylation and kinase activity in intact IGF-IR-overexpressing NIH-3T3 cells and to inhibit growth of MDA-MB468 and MCF-7 breast cancer cells in monolayer and colony formation in soft agar.97,98 However, cross-reactivity of these compounds with the insulin receptor tyrosine kinase was reported due to the high degree of homology between the two receptors. Newer agents have been developed with apparently more selective IGF-IR activity.99,100 However, it is uncertain if a highly selective IGF-IR tyrosine kinase inhibitor is desirable. Since insulin receptor may mediate some of the biological effects of the IGFs, it is possible that both IGF-IR and insulin receptor will need to be inhibited in tumors. Using anti-sense oligonucleotides, Salatino et al have shown that specific targeting of the IGF-IR in mice inhibits tumor growth,101 supporting the idea that specific inhibition of IGF-IR may block tumor growth. However, since mice have very low serum levels of IGF-II after birth,16 it remains to be seen if targeted disruption of IGF-IR alone is sufficient to inhibit tumor growth in humans.

Combination of Anti-IGF Strategy with Chemotherapy

In theory, inhibition of survival pathways by blocking IGF-IR signaling while enhancing apoptotic stimuli has appeal. Combination of anti-IGFIR antibody αIR3 with doxorubicin resulted in increased cytotoxicity in IGF-I stimulated cells than with chemotherapy alone.102 Similar enhancement of chemotherapy effects have been shown in Ewing's sarcoma cells.103 Tyrphostin AG1024 (a tyrosine kinase inhibitor of IGF-IR) demonstrated a marked enhancement in radiosensitivity and amplification of radiation-induced apoptosis which was associated with increased expression of Bax, p53 and p21, and a decreased expression of Bcl-2.10 Another study demonstrated that cotargeting IGF-IR and c-kit synergistic inhibit proliferation and induction of apoptosis in H209 small cell lung cancer cells.104 There is also evidence that somatostatin analogues may enhance the effect of tamoxifen in animal models by suppressing plasma IGF-I and –II levels.9

In addition to conventional agents, it is also possible that anti-growth factor receptor strategies can be combined. Recent evidence shows that increased levels of IGF-IR signaling appears to interfere with the action of trastuzumab in breast cancer cell models that overexpress HER2/neu.9 Thus, strategies that target IGF-IR signaling may prevent or delay development of resistance to trastuzumab.

Conclusion

The IGF-IR is a promising target in breast cancer therapy because it signals to multiple pathways required for maintenance of the malignant phenotype. Given the role for IGF-IR in cell survival, it is logical to combine anti-IGF therapies with conventional agents. Indeed, the preclinical data suggest that blockade of IGF-IR induces apoptosis and lowering a “survival threshold” with disruption of this signaling system should enhance chemotherapy efficacy.

However, there are several challenges that will need to be addressed before the idea that combination anti-IGF therapy and chemotherapy display synergy. First, there are many ways that IGF signaling could be targeted. As recently noted by Professor Baserga,105 the potential anti-IGF strategies have gone from “rags to riches” in the course of a few short years. Clinical trials to test the most effective strategy will need to be completed before combination trials can begin. Second, the phenotypes regulated by IGF-IR are not restricted to survival alone. Since proliferation is also affected by IGF-IR, it will be important to consider scheduling and choice of chemotherapeutic agent when designing appropriate combinations. For example, it is possible that anti-metabolites would be less efficacious when combined with anti-IGF because of the requirement for cells in S-phase for anti-metabolites to function. Indeed, interference between hormonal therapy and chemotherapy has been noted in breast cancer106 and it is possible that such interference could exist between anti-IGF therapy and certain drugs. On the other hand, agents that have a different mechanism of action, such as DNA alkylators or therapies that induce DNA strand breaks, may be enhanced by blocking IGF-IR due to the receptor's role in DNA repair. Careful preclinical studies will need to be performed before clinical trials should proceed with testing anti-IGF therapy with conventional cytotoxics. Lastly, the idea that multiple molecules are involved in growth factor signaling leads to the potential for “combination targeted therapy” trials. As mentioned, blockade of both HER2 and IGF-IR may have benefit in preclinical systems. It is also highly likely that blockade of IGF-IR and downstream signaling events (MAPK, Akt, etc), could be synergistic. Given the complexity of the cross-talk and feedback between these systems, preclinical studies should also be able to guide us with designing the optimal therapies.

However, it is clear that anti-IGF therapies will soon find their way into clinical trials. Hopefully, the vast experience with preclinical model systems will guide us in the optimal development of these agents.

Acknowledgements

This work was supported by Public Health Service grants CA74285, CA89652, and Cancer Center Support Grant P30 CA77398.

References

1.
Jemal A, Tiwari RC, Murray T. et al. Cancer statistics, 2004. CA Cancer J Clin. 2004;54(1):8–29. [PubMed: 14974761]
2.
Polychemotherapy for early breast cancer: An overview of the randomised trials. Early Breast Cancer Trialists' Collaborative Group. Lancet. 1998;352(9132):930–942. [PubMed: 9752815]
3.
Slamon DJ, Leyland-Jones B, Shak S. et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344(11):783–792. [PubMed: 11248153]
4.
Yee D. The insulin-like growth factors and breast cancer—revisited. Breast Cancer Res Treat. 1998;47(3):197–199. [PubMed: 9516075]
5.
Papa V, Gliozzo B, Clark GM. et al. Insulin-like growth factor-I receptors are overexpressed and predict a low risk in human breast cancer. Cancer Res. 1993;53(16):3736–3740. [PubMed: 8339284]
6.
Resnik JL, Reichart DB, Huey K. et al. Elevated insulin-like growth factor I receptor autophosphorylation and kinase activity in human breast cancer. Cancer Res. 1998;58(6):1159–1164. [PubMed: 9515800]
7.
Li BD, Khosravi MJ, Berkel HJ. et al. Free insulin-like growth factor-I and breast cancer risk. Int J Cancer. 2001;91(5):736–739. [PubMed: 11267989]
8.
Michels KB, Willett WC. Breast cancer—early life matters. N Engl J Med. 2004;351(16):1679–1681. [PubMed: 15483288]
9.
Lu Y, Zi X, Zhao Y. et al. Insulin-like growth factor-I receptor signaling and resistance to trastuzumab (Herceptin). J Natl Cancer Inst. 2001;93(24):1852–1857. [PubMed: 11752009]
10.
Wen B, Deutsch E, Marangoni E. et al. Tyrphostin AG 1024 modulates radiosensitivity in human breast cancer cells. Br J Cancer. 2001;85(12):2017–2021. [PMC free article: PMC2364012] [PubMed: 11747348]
11.
Pollak MN, Schernhammer ES, Hankinson SE. Insulin-like growth factors and neoplasia. Nat Rev Cancer. 2004;4(7):505–518. [PubMed: 15229476]
12.
Sachdev D, Yee D. The IGF system and breast cancer. Endocr Relat Cancer. 2001;8(3):197–209. [PubMed: 11566611]
13.
Moschos SJ, Mantzoros CS. The role of the IGF system in cancer: From basic to clinical studies and clinical applications. Oncology. 2002;63(4):317–332. [PubMed: 12417786]
14.
Laban C, Bustin SA, Jenkins PJ. The GH-IGF-I axis and breast cancer. Trends Endocrinol Metab. 2003;14(1):28–34. [PubMed: 12475609]
15.
Baker J, Liu JP, Robertson EJ. et al. Role of insulin-like growth factors in embryonic and postnatal growth. Cell. 1993;75(1):73–82. [PubMed: 8402902]
16.
DeChiara TM, Efstratiadis A, Robertson EJ. A growth-deficiency phenotype in heterozygous mice carrying an insulin-like growth factor II gene disrupted by targeting. Nature. 1990;345(6270):78–80. [PubMed: 2330056]
17.
Ahlgren M, Melbye M, Wohlfahrt J. et al. Growth patterns and the risk of breast cancer in women. N Engl J Med. 2004;351(16):1619–1626. [PubMed: 15483280]
18.
Kim HS, Ingermann AR, Tsubaki J. et al. Insulin-like growth factor-binding protein 3 induces caspase-dependent apoptosis through a death receptor-mediated pathway in MCF-7 human breast cancer cells. Cancer Res. 2004;64(6):2229–2237. [PubMed: 15026367]
19.
Pandini G, Frasca F, Mineo R. et al. Insulin/insulin-like growth factor I hybrid receptors have different biological characteristics depending on the insulin receptor isoform involved. J Biol Chem. 2002;277(42):39684–39695. [PubMed: 12138094]
20.
Pandini G, Vigneri R, Costantino A. 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(7):1935–1944. [PubMed: 10430101]
21.
Zhang X, Yee D. Tyrosine kinase signalling in breast cancer: Insulin-like growth factors and their receptors in breast cancer. Breast Cancer Res. 2000;2(3):170–175. [PMC free article: PMC138771] [PubMed: 11250706]
22.
Ahmad S, Singh N, Glazer RI. Role of AKT1 in 17beta-estradiol- and insulin-like growth factor I (IGF-I)-dependent proliferation and prevention of apoptosis in MCF-7 breast carcinoma cells. Biochem Pharmacol. 1999;58(3):425–430. [PubMed: 10424760]
23.
Hankins GR, Desouza AT, Bentley RC. et al. M6P/IGF2 receptor: A candidate breast tumor suppressor gene. Oncogene. 1996;12(9):2003–2009. [PubMed: 8649861]
24.
Marshman E, Green KA, Flint DJ. et al. Insulin-like growth factor binding protein 5 and apoptosis in mammary epithelial cells. J Cell Sci. 2003;116(Pt 4):675–682. [PubMed: 12538768]
25.
Deeks S, Richards J, Nandi S. Maintenance of normal rat mammary epithelial cells by insulin and insulin-like growth factor 1. Exp Cell Res. 1988;174(2):448–460. [PubMed: 3276539]
26.
Richert MM, Wood TL. The insulin-like growth factors (IGF) and IGF type I receptor during postnatal growth of the murine mammary gland: Sites of messenger ribonucleic acid expression and potential functions. Endocrinology. 1999;140(1):454–461. [PubMed: 9886857]
27.
Ruan W, Kleinberg DL. Insulin-like growth factor I is essential for terminal end bud formation and ductal morphogenesis during mammary development. Endocrinology. 1999;140(11):5075–5081. [PubMed: 10537134]
28.
Hadsell DL, Greenberg NM, Fligger JM. et al. Targeted expression of des(1-3) human insulin-like growth factor I in transgenic mice influences mammary gland development and IGF-binding protein expression. Endocrinology. 1996;137(1):321–330. [PubMed: 8536631]
29.
Hadsell DL, Bonnette SG, Lee AV. Genetic manipulation of the IGF-I axis to regulate mammary gland development and function. J Dairy Sci. 2002;85(2):365–377. [PubMed: 11913696]
30.
Gilmore AP, Valentijn AJ, Wang P. et al. Activation of BAD by therapeutic inhibition of epidermal growth factor receptor and transactivation by insulin-like growth factor receptor. J Biol Chem. 2002;277(31):27643–27650. [PubMed: 12011069]
31.
Marshman E, Streuli CH. Insulin-like growth factors and insulin-like growth factor binding proteins in mammary gland function. Breast Cancer Res. 2002;4(6):231–239. [PMC free article: PMC137936] [PubMed: 12473169]
32.
Tonner E, Barber MC, Allan GJ. et al. Insulin-like growth factor binding protein-5 (IGFBP-5) induces premature cell death in the mammary glands of transgenic mice. Development. 2002;129(19):4547–4557. [PubMed: 12223411]
33.
Toniolo P, Bruning PF, Akhmedkhanov A. et al. Serum insulin-like growth factor-I and breast cancer. Int J Cancer. 2000;88(5):828–832. [PubMed: 11072255]
34.
Paik S. Expression of IGF-I and IGF-II mRNA in breast tissue. Breast Cancer Res Treat. 1992;22(1):31–38. [PubMed: 1421422]
35.
Ruan W, Newman CB, Kleinberg DL. Intact and amino-terminally shortened forms of insulin-like growth factor I induce mammary gland differentiation and development. Proc Natl Acad Sci USA. 1992;89(22):10872–10876. [PMC free article: PMC50444] [PubMed: 1438291]
36.
Ruan W, Catanese V, Wieczorek R. et al. 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(3):1296–1302. [PubMed: 7867584]
37.
Wu Y, Cui K, Miyoshi K. et al. Reduced circulating insulin-like growth factor I levels delay the onset of chemically and genetically induced mammary tumors. Cancer Res. 2003;63(15):4384–4388. [PubMed: 12907608]
38.
Hankinson SE, Willett WC, Colditz GA. et al. Circulating concentrations of insulin-like growth factor-I and risk of breast cancer. Lancet. 1998;351(9113):1393–1396. [PubMed: 9593409]
39.
Cullen KJ, Yee D, Sly WS. et al. Insulin-like growth factor receptor expression and function in human breast cancer. Cancer Res. 1990;50(1):48–53. [PubMed: 2152773]
40.
Pezzino V, Papa V, Milazzo G. et al. Insulin-like growth factor-I (IGF-I) receptors in breast cancer. Ann NY Acad Sci. 1996;784:189–201. [PubMed: 8651569]
41.
Turner BC, Haffty BG, Narayanan L. 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(15):3079–3083. [PubMed: 9242428]
42.
Rocha RL, Hilsenbeck SG, Jackson JG. 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(1):103–109. [PubMed: 9815544]
43.
Lee AV, Jackson JG, Gooch JL. et al. Enhancement of insulin-like growth factor signaling in human breast cancer: Estrogen regulation of insulin receptor substrate-1 expression in vitro and in vivo. Mol Endocrinol. 1999;13(5):787–796. [PubMed: 10319328]
44.
Hoang CD, Zhang X, Scott PD. et al. Selective activation of insulin receptor substrate-1 and -2 in pleural mesothelioma cells: Association with distinct malignant phenotypes. Cancer Res. 2004;64(20):7479–7485. [PubMed: 15492273]
45.
Zhang X, Kamaraju S, Hakuno F. et al. Motility response to insulin-like growth factor-I (IGF-I) in MCF-7 cells is associated with IRS-2 activation and integrin expression. Breast Cancer Res Treat. 2004;83(2):161–170. [PubMed: 14997047]
46.
Nagle JA, Ma Z, Byrne MA. et al. Involvement of insulin receptor substrate 2 in mammary tumor metastasis. Mol Cell Biol. 2004;24(22):9726–9735. [PMC free article: PMC525494] [PubMed: 15509777]
47.
Perks CM, Bowen S, Gill ZP. et al. Differential IGF-independent effects of insulin-like growth factor binding proteins (1-6) on apoptosis of breast epithelial cells. J Cell Biochem. 1999;75(4):652–664. [PubMed: 10572248]
48.
Oh Y, Muller HL, Lamson G. et al. 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(20):14964–14971. [PubMed: 7686909]
49.
Al-Hajj M, Wicha MS, Benito-Hernandez A. et al. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA. 2003;100(7):3983–3988. [PMC free article: PMC153034] [PubMed: 12629218]
50.
Baselga J, Norton L. Focus on breast cancer. Cancer Cell. 2002;1(4):319–322. [PubMed: 12086846]
51.
Tamoxifen for early breast cancer: An overview of the randomisedtrials. Early Breast Cancer Trialists' Collaborative Group. Lancet. 1998;351(9114):1451–1467. [PubMed: 9605801]
52.
Rubin R, Baserga R. Insulin-like growth factor-I receptor. Its role in cell proliferation, apoptosis, and tumorigenicity. Lab Invest. 1995;73(3):311–331. [PubMed: 7564264]
53.
Gooch JL, Van Den Berg CL, Yee D. Insulin-like growth factor (IGF)-I rescues breast cancer cells from chemotherapy-induced cell death—proliferative and anti-apoptotic effects. Breast Cancer Res Treat. 1999;56(1):1–10. [PubMed: 10517338]
54.
Xie SP, Pirianov G, Colston KW. Vitamin D analogues suppress IGF-I signalling and promote apoptosis in breast cancer cells. Eur J Cancer. 1999;35(12):1717–1723. [PubMed: 10674019]
55.
Kulik G, Klippel A, Weber MJ. Antiapoptotic signalling by the insulin-like growth factor I receptor, phosphatidylinositol 3-kinase, and Akt. Mol Cell Biol. 1997;17(3):1595–1606. [PMC free article: PMC231885] [PubMed: 9032287]
56.
Kothari S, Cizeau J, McMillan-Ward E. et al. BNIP3 plays a role in hypoxic cell death in human epithelial cells that is inhibited by growth factors EGF and IGF. Oncogene. 2003;22(30):4734–4744. [PubMed: 12879018]
57.
Peruzzi F, Prisco M, Dews M. et al. Multiple signaling pathways of the insulin-like growth factor 1 receptor in protection from apoptosis. Mol Cell Biol. 1999;19(10):7203–7215. [PMC free article: PMC84713] [PubMed: 10490655]
58.
Zha J, Harada H, Yang E. et al. Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-X(L). Cell. 1996;87(4):619–628. [PubMed: 8929531]
59.
Singleton JR, Dixit VM, Feldman EL. Type I insulin-like growth factor receptor activation regulates apoptotic proteins. J Biol Chem. 1996;271(50):31791–31794. [PubMed: 8943217]
60.
Dunn SE, Hardman RA, Kari FW. et al. Insulin-like growth factor 1 (IGF-1) alters drug sensitivity of HBL100 human breast cancer cells by inhibition of apoptosis induced by diverse anticancer drugs. Cancer Res. 1997;57(13):2687–2693. [PubMed: 9205078]
61.
Lu Y, Zi X, Pollak M. Molecular mechanisms underlying IGF-I-induced attenuation of the growth-inhibitory activity of trastuzumab (Herceptin) on SKBR3 breast cancer cells. Int J Cancer. 2004;108(3):334–341. [PubMed: 14648698]
62.
Mamay CL, Mingo-Sion AM, Wolf DM. et al. An inhibitory function for JNK in the regulation of IGF-I signaling in breast cancer. Oncogene. 2003;22(4):602–614. [PubMed: 12555073]
63.
Heron-Milhavet L, Karas M, Goldsmith CM. et al. Insulin-like growth factor-I (IGF-I) receptor activation rescues UV-damaged cells through a p38 signaling pathway. Potential role of the IGF-I receptor in DNA repair. J Biol Chem. 2001;276(21):18185–18192. [PubMed: 11278917]
64.
Heron-Milhavet L, LeRoith D. Insulin-like growth factor I induces MDM2-dependent degradation of p53 via the p38 MAPK pathway in response to DNA damage. J Biol Chem. 2002;277(18):15600–15606. [PubMed: 11877395]
65.
Shahrabani-Gargir L, Pandita TK, Werner H. Ataxia-telangiectasia mutated gene controls insulin-like growth factor I receptor gene expression in a deoxyribonucleic acid damage response pathway via mechanisms involving zinc-finger transcription factors Sp1 and WT1. Endocrinology. 2004;145(12):5679–5687. [PubMed: 15345673]
66.
Kuhn C, Hurwitz SA, Kumar MG. et al. Activation of the insulin-like growth factor-1 receptor promotes the survival of human keratinocytes following ultraviolet B irradiation. Int J Cancer. 1999;80(3):431–438. [PubMed: 9935186]
67.
Decraene D, Agostinis P, Bouillon R. et al. Insulin-like growth factor-1-mediated AKT activation postpones the onset of ultraviolet B-induced apoptosis, providing more time for cyclobutane thymine dimer removal in primary human keratinocytes. J Biol Chem. 2002;277(36):32587–32595. [PubMed: 12070137]
68.
Trojanek J, Ho T, Del Valle L. et al. Role of the insulin-like growth factor I/insulin receptor substrate 1 axis in Rad51 trafficking and DNA repair by homologous recombination. Mol Cell Biol. 2003;23(21):7510–7524. [PMC free article: PMC207618] [PubMed: 14559999]
69.
Jayanth VR, Belfi CA, Swick AR. et al. Insulin and insulin-like growth factor-1 (IGF-1) inhibit repair of potentially lethal radiation damage and chromosome aberrations and alter DNA repair kinetics in plateau-phase A549 cells. Radiat Res. 1995;143(2):165–174. [PubMed: 7631009]
70.
Holdaway IM, Mason BH, Lethaby AE. et al. Serum insulin-like growth factor-I and insulin-like growth factor binding protein-3 following chemotherapy for advanced breast cancer. ANZ J Surg. 2003;73(11):905–908. [PubMed: 14616567]
71.
Kajdaniuk D, Marek B. Influence of adjuvant chemotherapy with cyclophosphamide, methotrexate and 5-fluorouracil on plasma insulin-like growth factor-I and chosen hormones in breast cancer premenopausal patients. J Clin Pharm Ther. 2000;25(1):67–72. [PubMed: 10771466]
72.
Formelli F. Quality control for HPLC assay and surrogate end point biomarkers from the fenretinide (4-HPR) breast cancer prevention trial. J Cell Biochem Suppl. 2000;34:73–79. [PubMed: 10762018]
73.
Yang LM, Tin UC, Wu K. et al. Role of retinoid receptors in the prevention and treatment of breast cancer. J Mammary Gland Biol Neoplasia. 1999;4(4):377–388. [PubMed: 10705921]
74.
Chrzanowski K, Bielawska A, Palka J. Proline analogue of melphalan as a prodrug susceptible to the action of prolidase in breast cancer MDA-MB 231 cells. Farmaco. 2003;58(11):1113–1119. [PubMed: 14572862]
75.
Gronbaek H, Tanos V, Meirow D. et al. Effects of tamoxifen on insulin-like growth factors, IGF binding proteins and IGFBP-3 proteolysis in breast cancer patients. Anticancer Res. 2003;23(3C):2815–2820. [PubMed: 12926118]
76.
Ferrari L, Bajetta E, Martinetti A. et al. Could exemestane affect insulin-like growth factors, interleukin 6 and bone metabolism in postmenopausal advanced breast cancer patients after failure on aminoglutethimide, anastrozole or letrozole? Int J Oncol. 2003;22(5):1081–1089. [PubMed: 12684675]
77.
Torrisi R, Baglietto L, Johansson H. et al. Effect of raloxifene on IGF-I and IGFBP-3 in postmenopausal women with breast cancer. Br J Cancer. 2001;85(12):1838–1841. [PMC free article: PMC2364027] [PubMed: 11747323]
78.
Nielsen TO, Andrews HN, Cheang M. et al. Expression of the insulin-like growth factor I receptor and urokinase plasminogen activator in breast cancer is associated with poor survival: Potential for intervention with 17-allylamino geldanamycin. Cancer Res. 2004;64(1):286–291. [PubMed: 14729636]
79.
Jerome L, Shiry L, Leyland-Jones B. Anti-insulin-like growth factor strategies in breast cancer. Semin Oncol. 2004;31(1 Suppl 3):54–63. [PubMed: 15052543]
80.
Byron SA, Yee D. Potential therapeutic strategies to interrupt insulin-like growth factor signaling in breast cancer. Semin Oncol. 2003;30(5 Suppl 16):125–132. [PubMed: 14613033]
81.
Szepeshazi K, Schally AV, Armatis P. et al. Antagonists of GHRH decrease production of GH and IGF-I in MXT mouse mammary cancers and inhibit tumor growth. Endocrinology. 2001;142(10):4371–4378. [PubMed: 11564700]
82.
Kahan Z, Varga JL, Schally AV. et al. Antagonists of growth hormone-releasing hormone arrest the growth of MDA-MB-468 estrogen-independent human breast cancers in nude mice. Breast Cancer Res Treat. 2000;60(1):71–79. [PubMed: 10845811]
83.
Ingle JN, Suman VJ, Kardinal CG. et al. A randomized trial of tamoxifen alone or combined with octreotide in the treatment of women with metastatic breast carcinoma. Cancer. 1999;85(6):1284–1292. [PubMed: 10189133]
84.
Friend KE. Cancer and the potential place for growth hormone receptor antagonist therapy. Growth Horm IGF Res. 2001;11(Suppl A):S121–123. [PubMed: 11527083]
85.
Van den Berg CL, Cox GN, Stroh CA. et al. Polyethylene glycol conjugated insulin-like growth factor binding protein-1 (IGFBP-1) inhibits growth of breast cancer in athymic mice. Eur J Cancer. 1997;33(7):1108–1113. [PubMed: 9376191]
86.
Yee D, Jackson JG, Kozelsky TW. et al. Insulin-like growth factor binding protein-1 (IGFBP-1) expression inhibits IGF-I action in MCF-7 breast cancer cells. Cell Growth and Differentiation. 1994;5:73–77. [PubMed: 7510125]
87.
Zi X, Zhang J, Agarwal R. et al. Silibinin up-regulates insulin-like growth factor-binding protein 3 expression and inhibits proliferation of androgen-independent prostate cancer cells. Cancer Res. 2000;60(20):5617–5620. [PubMed: 11059749]
88.
D'Ambrosio C, Ferber A, Resnicoff M. et al. A soluble insulin-like growth factor I receptor that induces apoptosis of tumor cells in vivo and inhibits tumorigenesis. Cancer Res. 1996;56(17):4013–4020. [PubMed: 8752172]
89.
Samani AA, Chevet E, Fallavollita L. et al. Loss of tumorigenicity and metastatic potential in carcinoma cells expressing the extracellular domain of the type 1 insulin-like growth factor receptor. Cancer Res. 2004;64(10):3380–3385. [PubMed: 15150088]
90.
Goya M, Miyamoto S, Nagai K. et al. Growth inhibition of human prostate cancer cells in human adult bone implanted into nonobese diabetic/severe combined immunodeficient mice by a ligand-specific antibody to human insulin-like growth factors. Cancer Res. 2004;64(17):6252–6258. [PubMed: 15342412]
91.
Arteaga CL, Kitten LJ, Coronado EB. et al. Blockade of the type I somatomedin receptor inhibits growth of human breast cancer cells in athymic mice. J Clin Invest. 1989;84(5):1418–1423. [PMC free article: PMC304004] [PubMed: 2553774]
92.
Li SL, Liang SJ, Guo N. et al. Single-chain antibodies against human insulin-like growth factor I receptor: Expression, purification, and effect on tumor growth. Cancer Immunol Immunother. 2000;49(4-5):243–252. [PubMed: 10941907]
93.
Sachdev D, Li SL, Hartell JS. et al. A chimeric humanized single-chain antibody against the type I insulin-like growth factor (IGF) receptor renders breast cancer cells refractory to the mitogenic effects of IGF-I. Cancer Res. 2003;63(3):627–635. [PubMed: 12566306]
94.
Ye JJ, Liang SJ, Guo N. et al. Combined effects of tamoxifen and a chimeric humanized single chain antibody against the type I IGF receptor on breast tumor growth in vivo. Horm Metab Res. 2003;35(11-12):836–842. [PubMed: 14710366]
95.
Maloney EK, McLaughlin JL, Dagdigian NE. et al. An anti-insulin-like growth factor I receptor antibody that is a potent inhibitor of cancer cell proliferation. Cancer Res. 2003;63(16):5073–5083. [PubMed: 12941837]
96.
Burtrum D, Zhu Z, Lu D. et al. A fully human monoclonal antibody to the insulin-like growth factor I receptor blocks ligand-dependent signaling and inhibits human tumor growth in vivo. Cancer Res. 2003;63(24):8912–8921. [PubMed: 14695208]
97.
Blum G, Gazit A, Levitzki A. Development of new insulin-like growth factor-1 receptor kinase inhibitors using catechol mimics. J Biol Chem. 2003;278(42):40442–40454. [PubMed: 12869569]
98.
Parrizas M, Gazit A, Levitzki A. et al. Specific inhibition of insulin-like growth factor-1 and insulin receptor tyrosine kinase activity and biological function by tyrphostins. Endocrinology. 1997;138(4):1427–1433. [PubMed: 9075698]
99.
Mitsiades CS, Mitsiades NS, McMullan CJ. et al. Inhibition of the insulin-like growth factor receptor- 1 tyrosine kinase activity as a therapeutic strategy for multiple myeloma, other hematologic malignancies, and solid tumors. Cancer Cell. 2004;5(3):221–230. [PubMed: 15050914]
100.
Garcia-Echeverria C, Pearson MA, Marti A. et al. In vivo antitumor activity of NVP-AEW541-A novel, potent, and selective inhibitor of the IGF-IR kinase. Cancer Cell. 2004;5(3):231–239. [PubMed: 15050915]
101.
Salatino M, Schillaci R, Proietti CJ. et al. Inhibition of in vivo breast cancer growth by antisense oligodeoxynucleotides to type I insulin-like growth factor receptor mRNA involves inactivation of ErbBs, PI-3K/Akt and p42/p44 MAPK signaling pathways but not modulation of progesterone receptor activity. Oncogene. 2004;23(30):5161–5174. [PubMed: 15122317]
102.
Beech DJ, Parekh N, Pang Y. Insulin-like growth factor-I receptor antagonism results in increased cytotoxicity of breast cancer cells to doxorubicin and taxol. Oncol Rep. 2001;8(2):325–329. [PubMed: 11182049]
103.
Benini S, Manara MC, Baldini N. et al. Inhibition of insulin-like growth factor I receptor increases the antitumor activity of doxorubicin and vincristine against Ewing's sarcoma cells. Clin Cancer Res. 2001;7(6):1790–1797. [PubMed: 11410521]
104.
Camirand A, Pollak M. Cotargeting IGF-1R and c-kit: Synergistic inhibition of proliferation and induction of apoptosis in H 209 small cell lung cancer cells. Br J Cancer. 2004;90(9):1825–1829. [PMC free article: PMC2409731] [PubMed: 15150607]
105.
Baserga R. Targeting the IGF-1 receptor: From rags to riches. Eur J Cancer. 2004;40(14):2013–2015. [PubMed: 15341972]
106.
Albain KS, Green SJ, Ravdin PM. et al. Adjuvant chemohormonal therapy for primary breast cancer should be sequential instead of concurrent: Initial results from intergroup trial 0100 (SWOG-8814) Proc ASCO 2002. (Abs#143)