Abstract
The insulin-like growth factor-1 receptor (IGF-1R) signaling pathway is a complex and tightly regulated network which is critical for cell proliferation, growth, and survival. IGF-1R is a potential therapeutic target for patients with many different malignancies. This brief review summarizes the results of clinical trials targeting the IGF-1R pathway in patients with breast cancer, sarcoma, and non-small cell lung cancer (NSCLC). Therapeutic agents discussed include both monoclonal antibodies to IGF-1R (dalotuzumab, figitumumab, cixutumumab, ganitumab, R1507, AVE1642) and newer IGF-1R pathway targeting strategies including monoclonal antibodies to IGF-1 and IGF-2 (MEDI-573 and BI 836845) and a small molecule tyrosine kinase inhibitor of IGF-1R (OSI-906). The pullback of trials in patients with breast cancer and NSCLC based on several large negative trials is noted and contrasted with the sustained success of IGF-1R inhibitor monotherapy in a subset of patients with sarcoma. Several different biomarkers have been examined in these trials with varying levels of success, including tumor expression of IGF-1R and its pathway components, serum IGF ligand levels, alternate pathway activation, and specific molecular signatures of IGF-1R pathway dependence. However, there remains a critical need to define predictive biomarkers in order to identify patients who may benefit from IGF-1R directed therapies. Ongoing research focuses on uncovering such biomarkers and elucidating mechanisms of resistance, as this therapeutic target is currently being analyzed from the bedside to bench.
Background
The Insulin-Like Growth Factor (IGF) signaling pathway is a complex and tightly regulated network which is critical for cell proliferation and survival (1). This pathway (Fig. 1) is composed of three receptor tyrosine kinases - insulin-like growth factor-1 receptor (IGF-1R), insulin-like growth factor-2 receptor (IGF-2R), and insulin receptor (INSR); three ligands – insulin, IGF-1, and IGF-2 (2, 3); and six serum Insulin-like Growth Factor Binding Proteins (IGFBP’s), which serve as regulators of the pathway by determining ligand bioavailability (4). The most prevalent of the IGFBP’s is IGFBP3 (5). Both IGF-1 and IGF-2 exert their effects through autocrine, paracrine, and endocrine mechanisms, and both can activate IGF-1R signaling.
Figure 1.
Schematic representation of the IGF-1R signaling network and nodes of therapeutic blockade. The IGF-1R signaling pathway is composed of three receptor tyrosine kinases - insulin-like growth factor-1 receptor (IGF-1R), insulin-like growth factor-2 receptor (IGF-2R), and insulin receptor (INSR); three ligands – Insulin, IGF-1, and IGF-2 (formerly known as somatomedins) (1, 2); and six serum Insulin-like Growth Factor Binding Proteins (IGFBPs). The IGFBPs, of which IGFBP3 is the most common, serve as regulators of the pathway by determining the bioavailability of IGF-1 and IGF-2 ligands (4). Both IGF-1 and IGF-2 exert their effects through autocrine, paracrine, and endocrine mechanisms, and both can activate the IGF-1R pathway. For simplification, IGF-1 ligand only is shown binding to IGF-1R. IGF-1 binding to IGF-1R promotes receptor homodimerization or heterodimerization with INSR. Ligand-activated IGF-1R first binds to intracellular adaptor proteins, such as insulin receptor substrate1 (IRS1) and SHC. These adaptor proteins transmit signals through the phosphatidyl-inositol-3 kinase (PI3K)-AKT1-mammalian target of rapamycin (MTOR) pathway and through the mitogen activated protein kinase (MAPK) pathway. Activated IGF-1R promotes cellular motility through activation of IRS2, which alters integrin expression through poorly understood mechanisms involving the small G protein RHOA, focal adhesion kinase (FAK), Rho-kinase (ROCK), PI3K, and other signaling molecules. Of note, IGF2R is a repository for IGF-2, and it has no intracellular signaling activity. IGF-2R acts as a tumor suppressor gene, as when IGF-2R function is lost, IGF-2 is able to bind IGF-1R and promote tumorigenesis (17). Targets for potential monotherapy and combinatorial therapeutic strategies are noted in the figure. TKI: tyrosine kinase inhibitor. mAb: monoclonal antibody.
IGF-1R is a type 2 tyrosine kinase transmembrane receptor that is normally found as a heterotetramer with two alpha and two beta subunits (6, 7). IGF-1R binding to IGF-1 or IGF-2 can occur with IGF-1R as a homodimer or as a heterodimer with insulin receptor isoforms A or B (INSR-A, INSR-B) (2, 8). While the heterodimer IGF-1R/INSR can bind insulin, it has been shown to preferentially favor IGF-1 mediated signaling (9, 10).
Once activated, IGF-1R activates numerous downstream pathways within the cell. In order to propagate these signals, ligand activated IGF-1R first binds to intracellular adaptor proteins – predominantly insulin receptor substrate1 (IRS1) (11), although other intracellular proteins such as SHC1 (12), GAB (13), and CRK (14) can interact with activated IGF-1R. These adaptor proteins are necessary for IGF-1R to transmit signals downstream in the cell through the phosphatidyl-inositol-3 kinase (PI3K)-AKT1- mammalian target of rapamycin (MTOR) pathway and through the mitogen activated protein kinase (MAPK) pathway. Ligand-activated IGF-1R binds to IRS1, which then binds to the p85 regulatory subunit of PI3K, which then transmits signals to AKT1 and MTOR. Activation of the PI3K-AKT1-MTOR pathway results in pleiotropic effects, including inactivation of the pro-apoptotic protein BAD (15–19). Concurrently, IGF-1R binds to SHC, which interacts with growth factor receptor-bound-2 (GRB2)-son-of-sevenless (SOS) to activate the MAPK pathway (14). Finally, activated IGF-1R is thought to promote cellular motility through activation of IRS2, which acts to alter integrin expression through poorly understood mechanisms involving the small G protein RHOA, focal adhesion kinase (FAK), and Rho-kinase (ROCK) (15, 16). Of note, IGF-2R is a repository for IGF-2, and it has no intracellular signaling activity. In this capacity, IGF-2R acts as a tumor suppressor gene, as when IGF-2R function is lost, IGF-2 is able to bind IGF-1R and promote tumorigenesis (17).
Serum IGF-1 and IGFBP3 levels are normally regulated by the pituitary gland (18, 19). Elevated serum levels of IGF-1 and IGF-2 as well as overactivation of the mitogenic, anti-apoptotic, and pro-motility signaling cascades induced by IGF-1R have been implicated in many tumor types, including epithelial malignancies (breast, lung, colorectal, prostate, ovarian), mesenchymal tumors (osteosarcoma, rhabdomyosarcoma), and hematologic malignancies (1, 2, 17, 20, 21). Furthermore, IGF-1R pathway dysregulation acts as an oncogenic signal in the context of both initial tumorigenesis and resistance to cytotoxic and targeted anticancer therapies (2, 3, 22, 23).
Herein, we focus on the role of the IGF-1R pathway in breast cancer, sarcoma, and non-small cell lung cancer (NSCLC), as it is in these three malignancies that IGF-1R pathway blockade has been most extensively studied. In patients with breast cancer, it has been noted that the IGF-1R pathway has extensive crosstalk with the estrogen receptor (ER) and epidermal growth factor receptor 2 (ERBB2) signaling pathways, and IGF-1R has been implicated in resistance to hormonal therapy (24, 25). Furthermore, IGF-1R is directly upstream of the PI3K-AKT1-MTOR pathway, which is aberrantly activated in more than half of human breast cancers (26). Preclinical data in sarcoma tumor models has shown that the IGF-1R pathway is particularly important in tumor growth, metastasis, and angiogenesis in patients with Ewing’s sarcoma and rhabdomyosarcoma, leading to the initial application of IGF-1R inhibitors in patients with these tumor types (27). Finally, IGF-1R protein levels have been shown to be high in NSCLC cell lines and patient samples, both in adenocarcinoma and squamous histologies (28, 29). Also, IGF-1R expression is associated with poor prognosis in patients with NSCLC (28). It is worth mentioning that IGF-1R expression levels have been evaluated in small cell lung cancer (SCLC), however we will only discuss NSCLC.
Numerous therapeutic agents targeting the IGF-1R pathway have been developed. These agents include IGF-1R monoclonal antibodies (mAbs), IGF-1R/INSR tyrosine kinase inhibitors (TKIs), and more recently, IGF-1 and IGF-2 specific mAbs (Fig. 1). Furthermore, several rational combination therapeutic strategies have been employed to attempt to more potently inhibit IGF-1R signaling. To date, the most widely tested combination strategy involves the use of IGF-1R antibodies with MTOR allosteric inhibitors, such as temsirolimus (30) or ridaforolimus (3). There is established pre-clinical rationale for this approach, as numerous studies have now shown that MTOR inhibition paradoxically results in activation of the IGF-1R pathway (31).
In the following sections we describe the current state and future directions of the application of IGF-1R targeting agents in patients with breast cancer, sarcoma, and NSCLC, with a summary of the high-impact trials provided in Table 1.
Table 1.
Published clinical trials involving IGF-1R pathway inhibition in patients with breast cancer, sarcoma, or lung cancer
| Reference | Phase | n | Tumor types | Therapy | Disease control rates |
|---|---|---|---|---|---|
| Atzori et al., 2011 (32) | I | 80 | Colorectal (24%), breast (21%), sarcoma (11%), other (43%) | Dalotuzumab (MK-0646) | SD 8%, PR 4%, CR 0% |
| Di Cosimo et al., 2015 (36) | I | 87 | Breast (26%), colorectal (22%), NSCLC (18%), sarcoma (16%), other (18%) | Dalotuzumab (MK-0646) + ridaforolimus | SD 46%, PR 7%, CR 0% |
| Higano et al., 2015 (33) | I | 40 | Lung (20%), colon (15%), breast (7.5%), other (57.5%) | Cixutumumab (IMC-A12) | SD 25%, PR 0%, CR 0% |
| Ma et al., 2013 (37) | I | 26 | Breast (100%); ER positive (86%) | Cixutumumab (IMC-A12) + temsirolimus | SD 15%, PR 0%, CR 0% |
| Naing et al., 2011 (19) | I | 42 | Adrenocortical (24%), breast (21%), sarcoma (21%), other (41%) | Cixutumumab (IMC-A12) + temsirolimus | SD 43%, PR 0%, CR 0% |
| Tolcher et al., 2009 (34) | I | 53 | Sarcoma (42%), other (58%) | Ganitumab (AMG-479) | SD NA, PR 4%, CR 2% |
| Goto et al., 2012 (47) | I | 19 | NSCLC (100%) | Figitumumab (CP-751,871) + carboplatin and paclitaxel | SD 42%, PR 37%, CR 0% |
| Molife et al., 2010 (45) | I | 46 | Prostate (48%), esophageal (20%), sarcoma (6.5%), NSCLC (4.3%), other (21.2%) | Figitumumab (CP-751,871) + docetaxel | SD 26%, PR 9%, CR 0% |
| Murakami et al., 2012 (5) | I | 19 | Breast (21%), gastric (16%), NSCLC (10%), sarcoma (10%), other (43%) | Ganitumab (AMG-479) | SD 37%, PR 0%, CR 0% |
| Kurzrock et al., 2010 (35) | I | 35 | Sarcoma (51%), lung (5.5%), breast (5.5%), other (38%) | R1507 | SD 35%, PR 5%, CR 0% |
| Macaulay et al., 2013 (18) | I | 58 | Ovarian (21%), sarcoma (9%), breast (7%), NSCLC (5%), other (58%) | AVE1642 + docetaxel OR gemcitabine/erlotinib OR doxorubicin | SD 40–70%, PR 2.5–20%, CR 0% |
| Puzanov et al., 2014 (8) | I | 86 | Colorectal (49%), NSCLC (4%), sarcoma (4%), other (43%) | OSI-906 | SD 36%, PR 1%, CR 0% |
| Haluska et al., 2014 (50) | I | 43 | Urothelial (46.5%), sarcoma (9%), colorectal (5%), breast (2.5%), | MEDI-573 | SD 30%, PR 0%, CR 0% |
| Haluska et al., 2007 (39) | I | 24 | NSCLC (2.5%), other (34.5%) Colorectal (25%), lung (17%), sarcoma (17%), other (41%) | Figitumumab (CP-751,871) | SD 41%, PR 0%, CR 0% |
| Olmos et al., 2010 (27) | I | 29 | Sarcoma (100%); Ewing’s sarcoma (55%) | Figitumumab (CP-751,871) | SD 28.5%, PR 3.5%, CR 3.5% |
| Naing et al., 2012 (30) | I | 20 | Ewing’s sarcoma (85%), desmoplastic small round cell tumor (15%) | Cixutumumab (IMC-A12) + temsirolimus | SD 25%, PR 0%, CR 10% |
| Quek et al., 2011 (43) | I | 21 | Sarcoma (90%), adrenal cortical (5%), colorectal (5%) | Figitumumab (CP-751,871) + everolimus | SD 71%, PR 5%, CR 0% |
| Juergens et al., 2011 (40) | I/II | 31 (I) 107 (II) |
Sarcoma (100%); Ewing’s sarcoma (89%) | Figitumumab (CP-751,871) | SD 24%, PR 14%, CR 0% |
| Schoffski et al., 2013 (42) | II | 111 | Sarcoma (100%); Ewing’s sarcoma (18%) | Cixutumumab (IMC-A12) | SD 40%, PR 2%, CR 0% |
| Schwartz et al., 2013 (44) | II | 174 | Sarcoma (100%); Ewing’s sarcoma (15.5%) | Cixutumumab (IMC-A12) + temsirolimus | SD 38%, PR 5%, CR 0% |
| Pappo et al., 2011 (41) | II | 115 | Ewing’s sarcoma (100%) | R1507 | SD 16%, PR 9%, CR 1% |
| Karp et al., 2009 (48) | II | 98 | NSCLC (100%) | Figitumumab (CP-751,871) + carboplatin and paclitaxel | SD 10–20%, PR + CR 54% -> 37% corrected |
| Robertson et al., 2013 (38) | II | 63 | Breast (100%); ER positive (94%) | Ganitumab (AMG-479) + fulvestrant OR exemestane | SD 27%, PR 8%, CR 0% |
| Ramalingam et al., 2011 (49) | II | 172 | NSCLC (100%) | Erlotinib +/− R1507 | 12-week PFS: 41%/43.5% OS: 8.1 mo/10 mo |
| Langer et al., 2014 (46) | III | 338 | NSCLC (nonadenocarcinoma 100%) | Figitumumab (CP-751,871) + carboplatin and paclitaxel | SD not noted PR + CR 33% |
Abbreviations: CR, complete response; ER, estrogen receptor; mo, months; NA, not available; NSCLC, non-small cell lung cancer; OS, overall survival; PFS, progression free survival; PR, partial response; SD, stable disease.
Clinical-Translational Advances
IGF-1R pathway inhibition in patients with breast cancer
There have been four different anti-IGF1R mAbs tested in early clinical trials involving small numbers of patients with advanced, treatment refractory breast cancer with largely unimpressive results (5, 32–35). Consequently, three phase I clinical trials assessing the combination of IGF-1R mAbs with MTOR inhibitors in patients with advanced, treatment refractory breast cancer have been completed (19, 36, 37). In a phase I clinical trial with dalotuzumab and the MTOR inhibitor ridaforolimus, a subset of patients with ER positive (ER+), highly proliferative disease was shown to have exceptional responses, experiencing a disease control rate (stable disease [SD] plus partial response [PR]) of 55% (6/11 patients). These promising results created momentum for a recently completed phase II clinical trial involving patients with advanced luminal B breast cancer treated with dalotuzumab, ridaforolimus, and hormonal therapy (NCT01234857) (36). In a phase I clinical trial with cixutumumab and the MTOR inhibitor temsirolimus, among 26 patients with breast cancer (86% with ER+ disease), four (15%) had SD, and no PRs or complete responses (CRs) were observed. The results of this trial, in which the median number of prior chemotherapeutic regimens was three, stimulated interest in testing the combination of cixutumumab and temsirolimus in patients with metastatic breast cancer and no more than two prior lines of chemotherapy, but initial trial results have shown no tumor responses (NCT00699491) (37). Finally, unlike combination with MTOR inhibition, the combination of IGF-1R inhibition with exemestane or fulvestrant in patients with advanced breast cancer was unsuccessful in a phase II trial (38), halting the application of combination hormonal therapy and IGF-1R inhibition in patients with breast cancer.
IGF-1R pathway inhibition in patients with sarcoma
Due to successful initial clinical trials in patients with advanced, treatment refractory sarcoma treated with IGF-1R mAbs (5, 32, 34, 35, 39), larger trials with a combined total of 362 patients have been completed (27, 40–42). In summation of these clinical trials, disease stabilization rates have been 16–40%, PRs have ranged from 2–12% of patients, and two of 362 patients have achieved a CR. Overall, the exceptional response of some patients to IGF-1R inhibitor monotherapy has led to speculation that a subset of patients with sarcoma, especially Ewing’s sarcoma, are uniquely dependent on IGF-1R signaling (19).
The combination of MTOR inhibition with IGF-1R inhibition in patients with advanced sarcoma has yielded similar results to IGF-1R monotherapy (19, 30, 36, 43, 44), and the combination of IGF-1R inhibition with cytotoxic chemotherapy has yielded provocative results in patients with leiomyosarcoma (18, 45). Overall, the clinical trials of anti-IGF-1R mAbs in patients with sarcoma have shown occasionally profound responses and disease stabilization rates ranging from 16% to up to 70% when IGF-1R mAbs have been combined with MTOR inhibitors (43). However, larger trials are needed to determine the optimal therapeutic strategy (monotherapy versus combination therapy with MTOR inhibitors) and also to parse out which subset of patients are most likely to benefit.
IGF-1R pathway inhibition in patients with non-small cell lung cancer
The combination of IGF-1R inhibition with cytotoxic chemotherapy has been tested in several large clinical trials in patients with NSCLC (46–48). The most well-studied IGF-1R mAb in lung cancer is figitumumab. When the combination of figitumumab, carboplatin, and paclitaxel was used as first line therapy in 98 patients with advanced NSCLC, the objective response rate (ORR) was initially reported as 57%, with an additional 10–20% of patients experiencing SD (48). These encouraging results prompted the completion of a phase III trial comparing figitumumab plus carboplatin/paclitaxel to carboplatin/paclitaxel alone in patients with treatment naïve advanced NSCLC. This clinical trial was closed early due to increased rates of serious adverse events and treatment related deaths in patients treated with figitumumab (46). The phase III trial showed an ORR of 33% for the figitumumab plus carboplatin/paclitaxel arm, and rather than the initially reported 54% ORR in the phase II trial, the actual observed rate was 37%) (46). The serious adverse events that were observed more commonly in patients receiving figitumumab compared to chemotherapy alone included pneumonia (6% vs 4%), hyperglycemia (3% vs <1%), asthenia (3% vs 1%), and dehydration (4% vs 1%). The etiologies of the 17 treatment-related deaths in patients treated with figitumumab included pulmonary hemorrhage, pneumonia, septic shock, hypovolemic shock, sepsis in a neutropenic patient, renal failure, hemorrhage, and etiologies listed as cardiorespiratory arrest, toxicity to various agents, and decrease of performance status (46).
The combination of EGFR plus IGF-1R inhibition has been tested in a cohort of unselected patients with lung adenocarcinoma or squamous cell carcinoma, but there was no improvement in PFS or OS compared to treatment with EGFR inhibition alone. Importantly, in this study less than 5% of patients had an EGFR mutation, as it was proposed that based on preclinical models IGF-1R and EGFR crosstalk was a key mechanism of tumorigenesis and resistance to isolated EGFR inhibition in patients with NSCLC independent of EGFR mutation status (49).
Other therapeutic agents which target the IGF-1R pathway
The growing appreciation of INSR mediated signaling in the IGF pathway has led to two novel strategies to target the IGF-1R pathway in patients with advanced breast cancer, sarcoma, and NSCLC: combined IGF-1R and insulin receptor inhibition (8) and therapeutic antibodies directed against the IGF-1 and IGF-2 ligands (50, 51).
Based on antitumor activity demonstrated in preclinical models in several tumor types, OSI-906 (linsitinib), an oral small-molecule tyrosine kinase inhibitor of IGF-1R and INSR, has been evaluated in 86 patients with advanced, treatment refractory solid tumors. When patients were treated with OSI-906 monotherapy the overall disease stabilization rate was 36% and one patient with melanoma achieved a PR (8). Recently completed phase II trials have evaluated OSI-906 combination therapies with paclitaxel in patients with recurrent ovarian cancer (NCT00889382) and with erlotinib in patients with metastatic EGFR mutant NSCLC (NCT01221077). Results from these trials are pending.
MEDI-573, a mAb to both IGF-1 and IGF-2, has demonstrated the ability to suppress IGF signaling through both IGF-1R and INSR-A without affecting normal INSR-B mediated signaling in cancer cell lines, leading to its use in an early clinical trial in patients with advanced, heavily pretreated solid tumors (50). In this trial, the disease stabilization rate was 30% with no PRs or CRs observed. Based on preclinical studies showing increased INSR-A:INSR-B mRNA ratios in tumor tissue from patients with hormone receptor positive, ERBB2 negative tumors, there is now a phase I/II clinical trial underway assessing the impact of MEDI-573 combined with hormonal therapy in this subset of breast cancer patients (NCT01446159) (50).
A second mAb to both IGF-1 and IGF-2, BI 836845, has been tested in phase I clinical trials involving 81 patients with advanced solid tumors (52, 53). The results have demonstrated tolerability and two patients have experienced a partial response, resulting in additional ongoing clinical trials involving the combination of BI 836845 with afatinib in patients with EGFR-mutant NSCLC in East Asia (NCT02191891) and in combination with everolimus and exemestane in patients with ER+ breast cancer (NCT02123823).
Challenges to clinical applications
The most pressing and as yet undefined challenge to the appropriate clinical application of IGF-1R pathway blockade is the identification of predictive markers that are able to identify patients likely to respond to this therapeutic strategy. As the clinical trials data shows, there are some treatment combinations that have shown disease stabilization rates of one-quarter to one-half of patients, and there has been some intriguing antitumor activity, especially in patients with sarcoma. However, what is now critically needed is development of predictive biomarkers that can guide future clinical trials in applying this therapeutic strategy to the patient populations most likely to benefit.
The identification of predictive biomarkers can be divided into four main categories which have seen varying levels of success: tumor expression of IGF-1R and its pathway components, serum IGF ligand levels, assessment of alternate pathway activation, and attempts at identifying specific molecular signatures of IGF-1R pathway dependence.
Pretreatment IGF-1R expression as assessed by immunohistochemistry has not consistently been correlated with disease control in heterogeneous groups of patients treated with anti-IGF-1R mAbs (32, 35, 44). It is important to note that when tumor expression of a target of a therapeutic agent does not correlate with response, there are many possible etiologies of false negative signals including sampling bias, variability in sample handling, limited assay sensitivity and specificity, and tumor mutations between when a sample is obtained and when treatment is administered (32).
In the case of IGF ligand assessments, serum ligand levels have consistently demonstrated predictive value in patients with sarcoma and NSCLC, although it must be noted that the degree of correlation between IGF ligand levels in the serum versus in the tumor microenvironment is unknown. In two clinical trials involving patients with sarcoma treated with IGF-1R inhibitor monotherapy, elevated pretreatment and on-treatment serum IGF-1 levels were associated with improved OS (40, 41). In patients with NSCLC, both a phase I (47) and a phase III clinical trial (46) have demonstrated improved disease control and OS in patients with elevated pre-treatment serum total IGF-1 (46) and greater elevations in serum IGF-1 when treated with figitumumab plus carboplatin/paclitaxel (46, 47). In contrast to serum IGF-1 levels, pretreatment levels and on-treatment changes in serum IGFBP3 have not been associated with disease control (5), combination IGF-1R and MTOR inhibition (19, 37), or IGF-1R inhibition in combination with cytotoxic chemotherapy (18, 47).
The assessment of alternative pathway activation mediating resistance to IGF-1R targeted therapies was the impetus for the combination trials described above. However, the interpretation of the heterogeneous responses to combination therapy necessitate a better understanding of the crosstalk between the IGF-1R pathway and other important signaling molecules such as EGFR, SRC, and ER (54–56) and downstream molecules such as MTOR, PI3K, and AKT1, which have been shown to mediate IGF-1R resistance in preclinical models (57).
Finally, specific gene expression profiles associated with IGF-1R sensitivity or resistance have been identified in models of breast cancer and Ewing’s sarcoma (58–60). This characteristic IGF-1-dependent gene expression profile includes upregulation of transcriptional targets of ER, MAPK3, MAPK1, and components of the PI3K-AKT1-MTOR pathway (58). The assessment of these molecular signatures and alternative pathways mediating IGF-1R resistance within the context of the significant clinical trials data described above is an important next step in improving the patient-specific application of IGF-1R targeting therapies (61).
Conclusions
In conclusion, the IGF-1R pathway is important in the development and maintenance of many different types of malignancies. Drug development targeting this pathway has taken unique routes by different tumor types, from preferential combination therapy in the case of patients with breast cancer to impressive antitumor efficacy in patients with sarcoma treated with anti-IGF-1R monotherapy to a negative phase III trial in combination with cytotoxic chemotherapy in patients with NSCLC. The most pressing needs for the future development of this therapeutic strategy are identifying biomarkers of response by applying a bedside to bench approach with the existing clinical trials data, including an in depth analysis of tumor samples from patients who have responded to IGF-1R directed therapies. These critical analyses will serve as the foundation to guide the most appropriate application of IGF-1R blockade in the clinic.
Acknowledgments
Grant Support
C.M. Lovly is supported in part by the NIH and the NCI under award numbers R01CA121210 and P01CA129243, a Damon Runyon Clinical Investigator Award, and a LUNGevity Career Development Award.
Footnotes
Disclosure of Potential Conflicts of Interest
C.M. Lovly reports receiving commercial research grants from AstraZeneca and Novartis, and is a consultant/advisory board member for ARIAD Pharmaceuticals, Genoptix, Harrison and Star, Novartis, and Sequenom. No potential conflicts of interest were disclosed by the other author.
Disclaimer
The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.
References
- 1.Samani AA, Yakar S, LeRoith D, Brodt P. The role of the IGF system in cancer growth and metastasis: overview and recent insights. Endocr Rev. 2007;28:20–47. doi: 10.1210/er.2006-0001. [DOI] [PubMed] [Google Scholar]
- 2.Casa AJ, Dearth RK, Litzenburger BC, Lee AV, Cui X. The type I insulin-like growth factor receptor pathway: a key player in cancer therapeutic resistance. Front Biosci. 2008;13:3273–87. doi: 10.2741/2925. [DOI] [PubMed] [Google Scholar]
- 3.Brana I, Berger R, Golan T, Haluska P, Edenfield J, Fiorica J, et al. A parallel-arm phase I trial of the humanised anti-IGF-1R antibody dalotuzumab in combination with the AKT inhibitor MK-2206, the mTOR inhibitor ridaforolimus, or the NOTCH inhibitor MK-0752, in patients with advanced solid tumours. Br J Cancer. 2014;111:1932–44. doi: 10.1038/bjc.2014.497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Massague J, Czech MP. The subunit structures of two distinct receptors for insulin-like growth factors I and II and their relationship to the insulin receptor. J Biol Chem. 1982;257:5038–45. [PubMed] [Google Scholar]
- 5.Murakami H, Doi T, Yamamoto N, Watanabe J, Boku N, Fuse N, et al. Phase 1 study of ganitumab (AMG 479), a fully human monoclonal antibody against the insulin-like growth factor receptor type I (IGF1R), in Japanese patients with advanced solid tumors. Cancer Chemother Pharmacol. 2012;70:407–14. doi: 10.1007/s00280-012-1924-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.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;263:11486–92. [PubMed] [Google Scholar]
- 7.Tollefsen SE, Stoszek RM, Thompson K. Interaction of the alpha beta dimers of the insulin-like growth factor I receptor is required for receptor autophosphorylation. Biochemistry. 1991;30:48–54. doi: 10.1021/bi00215a008. [DOI] [PubMed] [Google Scholar]
- 8.Puzanov I, Lindsay CR, Goff LW, Sosman JA, Gilbert J, Berlin J, et al. A phase I study of continuous oral dosing of OSI-906, a dual inhibitor of insulin-like growth factor-1 and insulin receptors in patients with advanced solid tumors. Clin Cancer Res. 2014;21:701–11. doi: 10.1158/1078-0432.CCR-14-0303. [DOI] [PubMed] [Google Scholar]
- 9.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;329:301–7. doi: 10.1038/329301a0. [DOI] [PubMed] [Google Scholar]
- 10.MacDonald RG, Pfeffer SR, Coussens L, Tepper MA, Brocklebank CM, Mole JE, et al. A single receptor binds both insulin-like growth factor II and mannose-6-phosphate. Science. 1988;239:1134–7. doi: 10.1126/science.2964083. [DOI] [PubMed] [Google Scholar]
- 11.Peruzzi F, Prisco M, Dews M, Salomoni P, Grassilli E, Romano G, et al. Multiple signaling pathways of the insulin-like growth factor 1 receptor in protection from apoptosis. Mol Cell Biol. 1999;19:7203–15. doi: 10.1128/mcb.19.10.7203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Koval AP, Blakesley VA, Roberts CT, Jr, Zick Y, Leroith D. Interaction in vitro of the product of the c-Crk-II proto-oncogene with the insulin-like growth factor I receptor. Biochem J. 1998;330:923–32. doi: 10.1042/bj3300923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Dearth RK, Cui X, Kim HJ, Hadsell DL, Lee AV. Oncogenic transformation by the signaling adaptor proteins insulin receptor substrate (IRS)-1 and IRS-2. Cell Cycle. 2007;6:705–13. doi: 10.4161/cc.6.6.4035. [DOI] [PubMed] [Google Scholar]
- 14.Yamauchi K, Pessin JE. Insulin receptor substrate-1 (IRS1) and Shc compete for a limited pool of Grb2 in mediating insulin downstream signaling. J Biol Chem. 1994;269:31107–14. [PubMed] [Google Scholar]
- 15.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;265:21172–8. [PubMed] [Google Scholar]
- 16.Lee AV, Jackson JG, Gooch JL, Hilsenbeck SG, Coronado-Heinsohn E, Osborne CK, 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;13:787–96. doi: 10.1210/mend.13.5.0274. [DOI] [PubMed] [Google Scholar]
- 17.Rowinsky EK, Youssoufian H, Tonra JR, Solomon P, Burtrum D, Ludwig DL. IMC-A12, a human IgG1 monoclonal antibody to the insulin-like growth factor I receptor. Clin Cancer Res. 2007;13:5549s–55s. doi: 10.1158/1078-0432.CCR-07-1109. [DOI] [PubMed] [Google Scholar]
- 18.Macaulay VM, Middleton MR, Protheroe AS, Tolcher A, Dieras V, Sessa C, et al. Phase I study of humanized monoclonal antibody AVE1642 directed against the type 1 insulin-like growth factor receptor (IGF-1R), administered in combination with anticancer therapies to patients with advanced solid tumors. Ann Oncol. 2013;24:784–91. doi: 10.1093/annonc/mds511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Naing A, Kurzrock R, Burger A, Gupta S, Lei X, Busaidy N, et al. Phase I trial of cixutumumab combined with temsirolimus in patients with advanced cancer. Clin Cancer Res. 2011;17:6052–60. doi: 10.1158/1078-0432.CCR-10-2979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Singh P, Alex JM, Bast F. Insulin receptor (IR) and insulin-like growth factor receptor 1 (IGF-1R) signaling systems: novel treatment strategies for cancer. Med Oncol. 2014;31:805. doi: 10.1007/s12032-013-0805-3. [DOI] [PubMed] [Google Scholar]
- 21.Avnet S, Sciacca L, Salerno M, Gancitano G, Cassarino MF, Longhi A, et al. Insulin receptor isoform A and insulin-like growth factor II as additional treatment targets in human osteosarcoma. Cancer Res. 2009;69:2443–52. doi: 10.1158/0008-5472.CAN-08-2645. [DOI] [PubMed] [Google Scholar]
- 22.Pollak M. Insulin and insulin-like growth factor signalling in neoplasia. Nat Rev Cancer. 2008;8:915–28. doi: 10.1038/nrc2536. [DOI] [PubMed] [Google Scholar]
- 23.Gao J, Chang YS, Jallal B, Viner J. Targeting the insulin-like growth factor axis for the development of novel therapeutics in oncology. Cancer Res. 2012;72:3–12. doi: 10.1158/0008-5472.CAN-11-0550. [DOI] [PubMed] [Google Scholar]
- 24.Gee JM, Robertson JF, Gutteridge E, Ellis IO, Pinder SE, Rubini M, et al. Epidermal growth factor receptor/HER2/insulin-like growth factor receptor signalling and oestrogen receptor activity in clinical breast cancer. Endocr Relat Cancer. 2005:S99–S111. doi: 10.1677/erc.1.01005. [DOI] [PubMed] [Google Scholar]
- 25.Fox EM, Miller TW, Balko JM, Kuba MG, Sanchez V, Smith RA, et al. A kinome-wide screen identifies the insulin/IGF-I receptor pathway as a mechanism of escape from hormone dependence in breast cancer. Cancer Res. 2011;71:6773–84. doi: 10.1158/0008-5472.CAN-11-1295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Miller TW, Balko JM, Arteaga CL. Phosphatidylinositol 3-kinase and antiestrogen resistance in breast cancer. J Clin Oncol. 2011;29:4452–61. doi: 10.1200/JCO.2010.34.4879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Olmos D, Postel-Vinay S, Molife LR, Okuno SH, Schuetze SM, Paccagnella ML, et al. Safety, pharmacokinetics, and preliminary activity of the anti-IGF-1R antibody figitumumab (CP-751,871) in patients with sarcoma and Ewing’s sarcoma: a phase 1 expansion cohort study. Lancet Oncol. 2010;11:129–35. doi: 10.1016/S1470-2045(09)70354-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Gong Y, Yao E, Shen R, Goel A, Arcila M, Teruya-Feldstein J, et al. High expression levels of total IGF-1R and sensitivity of NSCLC cells in vitro to an anti-IGF-1R antibody (R1507) PLoS One. 2009;4:e7273. doi: 10.1371/journal.pone.0007273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kim YH, Sumiyoshi S, Hashimoto S, Masago K, Togashi Y, Sakamori Y, et al. Expressions of insulin-like growth factor receptor-1 and insulin-like growth factor binding protein 3 in advanced non-small-cell lung cancer. Clin Lung Cancer. 2012;13:385–90. doi: 10.1016/j.cllc.2011.11.009. [DOI] [PubMed] [Google Scholar]
- 30.Naing A, LoRusso P, Fu S, Hong DS, Anderson P, Benjamin RS, et al. Insulin growth factor-receptor (IGF-1R) antibody cixutumumab combined with the mTOR inhibitor temsirolimus in patients with refractory Ewing’s sarcoma family tumors. Clin Cancer Res. 2012;18:2625–31. doi: 10.1158/1078-0432.CCR-12-0061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.O’Reilly KE, Rojo F, She QB, Solit D, Mills GB, Smith D, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006;66:1500–8. doi: 10.1158/0008-5472.CAN-05-2925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Atzori F, Tabernero J, Cervantes A, Prudkin L, Andreu J, Rodriguez-Braun E, et al. A phase I pharmacokinetic and pharmacodynamic study of dalotuzumab (MK-0646), an anti-insulin-like growth factor-1 receptor monoclonal antibody, in patients with advanced solid tumors. Clin Cancer Res. 2011;17:6304–12. doi: 10.1158/1078-0432.CCR-10-3336. [DOI] [PubMed] [Google Scholar]
- 33.Higano CS, Berlin J, Gordon M, LoRusso P, Tang S, Dontabhaktuni A, et al. Safety, tolerability, and pharmacokinetics of single and multiple doses of intravenous cixutumumab (IMC-A12), an inhibitor of the insulin-like growth factor-I receptor, administered weekly or every 2 weeks in patients with advanced solid tumors. Invest New Drugs. 2015;33:450–62. doi: 10.1007/s10637-015-0217-7. [DOI] [PubMed] [Google Scholar]
- 34.Tolcher AW, Sarantopoulos J, Patnaik A, Papadopoulos K, Lin CC, Rodon J, et al. Phase I, pharmacokinetic, and pharmacodynamic study of AMG 479, a fully human monoclonal antibody to insulin-like growth factor receptor 1. J Clin Oncol. 2009;27:5800–7. doi: 10.1200/JCO.2009.23.6745. [DOI] [PubMed] [Google Scholar]
- 35.Kurzrock R, Patnaik A, Aisner J, Warren T, Leong S, Benjamin R, et al. A phase I study of weekly R1507, a human monoclonal antibody insulin-like growth factor-I receptor antagonist, in patients with advanced solid tumors. Clin Cancer Res. 2010;16:2458–65. doi: 10.1158/1078-0432.CCR-09-3220. [DOI] [PubMed] [Google Scholar]
- 36.Di Cosimo S, Sathyanarayanan S, Bendell JC, Cervantes A, Stein MN, Brana I, et al. Combination of the mTOR inhibitor ridaforolimus and the anti-IGF1R monoclonal antibody dalotuzumab: preclinical characterization and phase I clinical trial. Clin Cancer Res. 2015;21:49–59. doi: 10.1158/1078-0432.CCR-14-0940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ma CX, Suman VJ, Goetz M, Haluska P, Moynihan T, Nanda R, et al. A phase I trial of the IGF-1R antibody cixutumumab in combination with temsirolimus in patients with metastatic breast cancer. Breast Cancer Res Treat. 2013;139:145–53. doi: 10.1007/s10549-013-2528-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Robertson JF, Ferrero JM, Bourgeois H, Kennecke H, de Boer RH, Jacot W, et al. Ganitumab with either exemestane or fulvestrant for postmenopausal women with advanced, hormone-receptor-positive breast cancer: a randomised, controlled, double-blind, phase 2 trial. Lancet Oncol. 2013;14:228–35. doi: 10.1016/S1470-2045(13)70026-3. [DOI] [PubMed] [Google Scholar]
- 39.Haluska P, Shaw HM, Batzel GN, Yin D, Molina JR, Molife LR, et al. Phase I dose escalation study of the anti insulin-like growth factor-I receptor monoclonal antibody CP-751,871 in patients with refractory solid tumors. Clin Cancer Res. 2007;13:5834–40. doi: 10.1158/1078-0432.CCR-07-1118. [DOI] [PubMed] [Google Scholar]
- 40.Juergens H, Daw NC, Geoerger B, Ferrari S, Villarroel M, Aerts I, et al. Preliminary efficacy of the anti-insulin-like growth factor type 1 receptor antibody figitumumab in patients with refractory Ewing sarcoma. J Clin Oncol. 2011;29:4534–40. doi: 10.1200/JCO.2010.33.0670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Pappo AS, Patel SR, Crowley J, Reinke DK, Kuenkele KP, Chawla SP, et al. R1507, a monoclonal antibody to the insulin-like growth factor 1 receptor, in patients with recurrent or refractory Ewing sarcoma family of tumors: results of a phase II Sarcoma Alliance for Research through Collaboration study. J Clin Oncol. 2011;29:4541–7. doi: 10.1200/JCO.2010.34.0000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Schoffski P, Adkins D, Blay JY, Gil T, Elias AD, Rutkowski P, et al. An open-label, phase 2 study evaluating the efficacy and safety of the anti-IGF-1R antibody cixutumumab in patients with previously treated advanced or metastatic soft-tissue sarcoma or Ewing family of tumours. Eur J Cancer. 2013;49:3219–28. doi: 10.1016/j.ejca.2013.06.010. [DOI] [PubMed] [Google Scholar]
- 43.Quek R, Wang Q, Morgan JA, Shapiro GI, Butrynski JE, Ramaiya N, et al. Combination mTOR and IGF-1R inhibition: phase I trial of everolimus and figitumumab in patients with advanced sarcomas and other solid tumors. Clin Cancer Res. 2011;17:871–9. doi: 10.1158/1078-0432.CCR-10-2621. [DOI] [PubMed] [Google Scholar]
- 44.Schwartz GK, Tap WD, Qin LX, Livingston MB, Undevia SD, Chmielowski B, et al. Cixutumumab and temsirolimus for patients with bone and soft-tissue sarcoma: a multicentre, open-label, phase 2 trial. Lancet Oncol. 2013;14:371–82. doi: 10.1016/S1470-2045(13)70049-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Molife LR, Fong PC, Paccagnella L, Reid AH, Shaw HM, Vidal L, et al. The insulin-like growth factor-I receptor inhibitor figitumumab (CP-751,871) in combination with docetaxel in patients with advanced solid tumours: results of a phase Ib dose-escalation, open-label study. Br J Cancer. 2010;103:332–9. doi: 10.1038/sj.bjc.6605767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Langer CJ, Novello S, Park K, Krzakowski M, Karp DD, Mok T, et al. Randomized, phase III trial of first-line figitumumab in combination with paclitaxel and carboplatin versus paclitaxel and carboplatin alone in patients with advanced non-small-cell lung cancer. J Clin Oncol. 2014;32:2059–66. doi: 10.1200/JCO.2013.54.4932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Goto Y, Sekine I, Tanioka M, Shibata T, Tanai C, Asahina H, et al. Figitumumab combined with carboplatin and paclitaxel in treatment-naive Japanese patients with advanced non-small cell lung cancer. Invest New Drugs. 2012;30:1548–56. doi: 10.1007/s10637-011-9715-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Karp DD, Paz-Ares LG, Novello S, Haluska P, Garland L, Cardenal F, et al. Phase II study of the anti-insulin-like growth factor type 1 receptor antibody CP-751,871 in combination with paclitaxel and carboplatin in previously untreated, locally advanced, or metastatic non-small-cell lung cancer. J Clin Oncol. 2009;27:2516–22. doi: 10.1200/JCO.2008.19.9331. [DOI] [PubMed] [Google Scholar]
- 49.Ramalingam SS, Spigel DR, Chen D, Steins MB, Engelman JA, Schneider CP, et al. Randomized phase II study of erlotinib in combination with placebo or R1507, a monoclonal antibody to insulin-like growth factor-1 receptor, for advanced-stage non-small-cell lung cancer. J Clin Oncol. 2011;29:4574–80. doi: 10.1200/JCO.2011.36.6799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Haluska P, Menefee M, Plimack ER, Rosenberg J, Northfelt D, LaVallee T, et al. Phase I dose-escalation study of MEDI-573, a bispecific, antiligand monoclonal antibody against IGFI and IGFII, in patients with advanced solid tumors. Clin Cancer Res. 2014;20:4747–57. doi: 10.1158/1078-0432.CCR-14-0114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Friedbichler K, Hofmann MH, Kroez M, Ostermann E, Lamche HR, Koessl C, et al. Pharmacodynamic and antineoplastic activity of BI 836845, a fully human IGF ligand-neutralizing antibody, and mechanistic rationale for combination with rapamycin. Mol Cancer Ther. 2014;13:399–409. doi: 10.1158/1535-7163.MCT-13-0598. [DOI] [PubMed] [Google Scholar]
- 52.Lin CC, Chang K-Y, Huang DC, Marriott V, Van Beijsterveldt L, Chen L-T, Cheng A-L, et al. A phase I dose escalation study of weekly BI 836845, a fully human, affinity-optimized, insulin-like growth factor (IGF) ligand neutralizing antibody, in patients with advanced solid cancers. J Clin Oncol. 2014;32:5s. (suppl; abstr 2617) [Google Scholar]
- 53.Rihawi K, Ong M, Michalarea V, Bent L, Buschke S, Bogenrieder T, et al. Phase I dose escalation study of 3-weekly BI 836845, a fully human, affinity optimized, insulin-like growth factor (IGF) ligand neutralizing antibody, in patients with advanced solid tumors. J Clin Oncol. 2014;32:5s. (suppl; abstr 2622) [Google Scholar]
- 54.Huang F, Hurlburt W, Greer A, Reeves KA, Hillerman S, Chang H, et al. Differential mechanisms of acquired resistance to insulin-like growth factor-i receptor antibody therapy or to a small-molecule inhibitor, BMS-754807, in a human rhabdomyosarcoma model. Cancer Res. 2010;70:7221–31. doi: 10.1158/0008-5472.CAN-10-0391. [DOI] [PubMed] [Google Scholar]
- 55.Hou X, Huang F, Macedo LF, Harrington SC, Reeves KA, Greer A, et al. Dual IGF-1R/InsR inhibitor BMS-754807 synergizes with hormonal agents in treatment of estrogen-dependent breast cancer. Cancer Res. 2011;71:7597–607. doi: 10.1158/0008-5472.CAN-11-1080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Haluska P, Carboni JM, TenEyck C, Attar RM, Hou X, Yu C, et al. HER receptor signaling confers resistance to the insulin-like growth factor-I receptor inhibitor, BMS-536924. Mol Cancer Ther. 2008;7:2589–98. doi: 10.1158/1535-7163.MCT-08-0493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Cao L, Yu Y, Darko I, Currier D, Mayeenuddin LH, Wan X, et al. Addiction to elevated insulin-like growth factor I receptor and initial modulation of the AKT pathway define the responsiveness of rhabdomyosarcoma to the targeting antibody. Cancer Res. 2008;68:8039–48. doi: 10.1158/0008-5472.CAN-08-1712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Creighton CJ, Casa A, Lazard Z, Huang S, Tsimelzon A, Hilsenbeck SG, et al. Insulin-like growth factor-I activates gene transcription programs strongly associated with poor breast cancer prognosis. J Clin Oncol. 2008;26:4078–85. doi: 10.1200/JCO.2007.13.4429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Litzenburger BC, Creighton CJ, Tsimelzon A, Chan BT, Hilsenbeck SG, Wang T, et al. High IGF-IR activity in triple-negative breast cancer cell lines and tumorgrafts correlates with sensitivity to anti-IGF-IR therapy. Clin Cancer Res. 2011;17:2314–27. doi: 10.1158/1078-0432.CCR-10-1903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Garofalo C, Mancarella C, Grilli A, Manara MC, Astolfi A, Marino MT, et al. Identification of common and distinctive mechanisms of resistance to different anti-IGF-IR agents in Ewing’s sarcoma. Mol Endocrinol. 2012;26:1603–16. doi: 10.1210/me.2012-1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.King H, Aleksic T, Haluska P, Macaulay VM. Can we unlock the potential of IGF-1R inhibition in cancer therapy? Cancer Treat Rev. 2014;40:1096–105. doi: 10.1016/j.ctrv.2014.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]

