Skip to main content
  • AACR Journals
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Radiation Oncology
      • Novel Combinations
      • Reviews
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Journals
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • My Cart

Search

  • Advanced search
Molecular Cancer Therapeutics
Molecular Cancer Therapeutics
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Radiation Oncology
      • Novel Combinations
      • Reviews
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Editorial

Targeting Insulin-Like Growth Factor Signaling: Rational Combination Strategies

David Olmos, Bristi Basu and Johann S. de Bono
David Olmos
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Bristi Basu
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Johann S. de Bono
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/1535-7163.MCT-10-0719 Published September 2010
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Activation of the insulin-like growth factor (IGF) pathway, by binding of the growth factors IGF-I and IGF-II to the receptors IGF-IR and insulin receptor (IR), triggers complex signaling cascades that regulate cell growth, survival, proliferation, and differentiation. There has been intense interest in targeting the IGF axis as an anticancer strategy, following the identification that its dysregulation contributes to malignant transformation (1) and resistance to established anticancer treatments (2). Moreover, knockout of IGF-IR was reported to render nonmalignant cells highly resistant to oncogenic transformation (3). The IGF pathway has been implicated in the pathogenesis of numerous tumors, including Ewing's sarcoma, breast cancer, non–small cell lung cancer, adrenocortical cancer, and prostate cancer. Epidemiologic studies have also related higher circulating IGF ligand levels and polymorphic variants of IGF pathway regulatory genes to cancer risk and prognosis (2). Overall, these data have led to multiple approaches to abrogate IGF-IR signaling, including antisense oligonucleotides, small interference RNA, small-molecule tyrosine kinase inhibitors (TKI), and IGF-IR blocking antibodies (4). Many of these agents reverse transformation in tumor cell lines and increase sensitivity to chemotherapy and irradiation in vitro, inhibiting tumor cell growth and metastasis (2). Clinically, credence for therapeutic IGF-IR blockade has also been strengthened by the reports on tumor responses, for example, with IGF-IR monoclonal antibodies (mAb) in patients with Ewing's sarcoma (5).

Nevertheless, recent clinical trials examining IGF-IR–targeting antibodies have raised concerns about the likely success of this monotherapy strategy. Two recently reported phase II studies have shown that two different human IGF-IR mAbs in Ewing's sarcoma patients as monotherapy, R1507 (Roche) and AMG479 (Amgen), have disappointing progression-free survival at 2.2 and 2.3 months, respectively. Moreover, two other phase III trials investigating the addition of the fully human mAb to IGF-IR, figitumumab (CP-751,871, Pfizer), to either carboplatin and paclitaxel (ADVIGO 1016) or to the epidermal growth factor receptor TKI erlotinib (ADVIGO 1018) in patients with advanced non–small cell lung cancer have been suspended, as planned interim analyses by independent data safety monitoring committees indicated that the addition of figitumumab would be unlikely to meet the primary end point of improving overall survival (6).

Conversely, a phase I clinical trial evaluating a combination of the humanized IGF-IR mAb dalotozumab with the mammalian target of rapamycin (mTOR) inhibitor ridaforolimus (7) has shown antitumor activity in patients with estrogen receptor–positive metastatic breast cancer. Other combination strategies to block this pathway have focused on blocking both IGF-I and IGF-II signaling. This strategy has arisen from preclinical studies indicating that there are different binding epitopes on IGF-IR that have differing biological activities (8, 9). In this issue of Molecular Cancer Therapeutics, Dong and colleagues (10) elegantly describe the antitumor effects of combining two inhibitory IGF-IR antibodies with distinct mechanisms of action, BIIB4 and BIIB5, which block the binding of IGF-I and IGF-II to IGF-IR by competitive and allosteric means, respectively. They show, first biochemically, then in cell-based assays, and finally in xenograft models that combined blockade by these two antibodies results in enhanced inhibition of intracellular signaling through the IGF-IR axis with increased antitumor efficacy in vitro and in vivo when compared with the activity of either single antibody alone. This enhanced reduction in ligand-driven growth was shown to be at least partly due to an accelerated downregulation of IGF-IR following dual targeting by both antibodies. Significantly, however, the enhanced antitumor activity with the two antibodies was most evident at high ligand concentrations, where efficacy of monotherapy was considerably reduced. The combination of BIIB4 and BIIB5 also improved the antitumor efficacy of the epidermal growth factor receptor TKI erlotinib and the mTOR inhibitor rapamycin. These results are timely as combinations of two HER2 targeting therapeutic antibodies against distinct epitopes on HER2, trastuzumab, and pertuzumab have improved response rates in HER2 positive metastatic breast cancer patients in a recent Phase II trial (11), providing a clinically relevant paradigm for dual targeting approaches against different extracellular regions of a cell surface receptor.

Such an IGF-targeting combination approach is supported by other studies that have shown that different IGF-IR antibodies can behave differently in competitive binding assays with IGF-I and IGF-II (12, 13). The strategy suggested by Dong and colleagues—that is, targeting more than one epitope on IGF-IR to improve the blockade of this signaling axis—provides impetus for clinical evaluation of both this and other approaches, including the dual targeting of IGF-I and IGF-II, or using small molecule TKIs that cotarget IR and IGF-1R. Further support for such cotargeting strategies are supported by the reported increases in serum IGF-I levels in cancer patients following treatment with IGF-IR antibodies that may abrogate their therapeutic efficacy through bypass mechanisms (14). Whether IGF-IR blockade clinically also results in raised IGF-II levels requires further investigation. It is noteworthy that preliminary clinical data in adrenocortical patients suggests that figitumumab monotherapy shows no significant activity in this disease, whereas the IGF-IR and IR small-molecule TKI OSI-906 has resulted in tumor responses (15, 16), supporting further investigation of this combined IGF-IR and IR blockade strategy. It is hypothesized that this finding in adrenocortical cancer may be a result of increased expression of IGF-II due to loss of imprinting; such a combination targeting strategy may also be critically important in other tumors with aberrant expression of IGF-II, including Wilms' tumors, hepatocellular cancers, and rhabdomyosarcomas (17).

Overall, it is clear that we now need to consider how we can optimize our therapeutic strategies in light of our understanding of the intrinsic complexities of the IGF axis, which involves three ligands (IGF-I, IGF-II, and insulin) and at least four receptors (IGF-IR, IGF-IIR, IR, and hybrid receptors; ref. 18). Moreover, IR has two isoforms, IR-A and IR-B, as a result of alternative splicing. Whereas IGF-I and insulin are highly selective for binding IGF-IR and IR, respectively, IGF-II can bind IGF-IR as well as the IR-A isoform and IGF-IIR. IGF-IIR may not mediate intracellular signaling and may be a negative regulator of IGF-II. Increasing complexity also arises, however, from the fact that while both IGF-IR and IR function as homodimers, they can also form heteromultimers. These hybrid receptors are composed of both IGF-IR and IR and can be activated by both IGF-I and IGF-II. Signaling downstream of IGF-IR may therefore be activated by both IGF-I and IGF-II ligands through different functional IGF-IR epitopes but encompassing the same ligand-binding site (9, 10). Further studies supporting an important bypass role for IGF-II have arisen from a transgenic mouse model of pancreatic neuroendocrine cancer; this model is dependent on IGF-II signaling, with IR also directly promoting carcinogenesis and compromising the efficacy of strategies targeting IGF-IR alone (19). IR knockout in these tumors resulted in enhanced sensitivity to anti–IGF-IR therapies, implying that blockade of IGF-IR may be bypassed by IGF-II through IR. This may be especially important in malignancies in which IGF-II loss of imprinting is implicated to play a role, and in which direct targeting of IGF-II or use of TKIs with cross-reactivity against IGF-IR and IR may be more efficacious than targeting IGF-IR alone. Nonetheless, molecular events outside the IGF axis may be critical to codriving the growth and survival of tumor cells. Cotargeting strategies will need to also consider both sequential and parallel pathway blockade such as the parallel coinhibition of the erbB axis (20, 21). Sequential pathway inhibition also merits further careful evaluation as indicated by the preliminary results of clinical trials combining IGF-IR/mTOR inhibitors (7), which are consistent with the enhanced antitumor activity observed by such dual inhibition in preclinical models (22).

Finally, while the IGF axis clearly remains an important therapeutic target, increasing knowledge of the complexity of this pathway and its altered signaling in human malignancy must guide future studies of IGF pathway inhibitors to accelerate patient benefit and successful drug development. Moreover, there is a pressing need to identify a suite of analytically validated predictive biomarkers to better select the patients who will ultimately derive most benefit from such therapeutic strategies.

Disclosure of Potential Conflicts of Interest

Johann de Bono has served as a paid consultant for Pfizer, Genentech, Novartis, Astellas, Boehringer Ingleheim, Merck, and AstraZeneca.

  • Received August 3, 2010.
  • Accepted August 3, 2010.
  • ©2010 American Association for Cancer Research.

References

  1. ↵
    1. Sell C,
    2. Ptasznik A,
    3. Chang CD,
    4. Swantek J,
    5. Cristofalo VJ,
    6. Baserga R
    . IGF-1 receptor levels and the proliferation of young and senescent human fibroblasts. Biochem Biophys Res Commun 1993;194:259–65.
    OpenUrlCrossRefPubMed
  2. ↵
    1. Pollak M
    . Insulin and insulin-like growth factor signalling in neoplasia. Nat Rev Cancer 2008;8:915–28.
    OpenUrlCrossRefPubMed
  3. ↵
    1. Sell C,
    2. Rubini M,
    3. Rubin R,
    4. Liu JP,
    5. Efstratiadis A,
    6. Baserga R
    . Simian virus 40 large tumor antigen is unable to transform mouse embryonic fibroblasts lacking type 1 insulin-like growth factor receptor. Proc Natl Acad Sci U S A 1993;90:11217–21.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    1. Gualberto A,
    2. Pollak M
    . Emerging role of insulin-like growth factor receptor inhibitors in oncology: early clinical trial results and future directions. Oncogene 2009;28:3009–21.
    OpenUrlCrossRefPubMed
  5. ↵
    1. Olmos D,
    2. Postel-Vinay S,
    3. Molife LR,
    4. 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.
    OpenUrlCrossRefPubMed
  6. ↵
    1. Jassem J,
    2. Langer CJ,
    3. Karp DD,
    4. et al
    . Randomized, open label, phase III trial of figitumumab in combination with paclitaxel and carboplatin versus paclitaxel and carboplatin in patients with non-small cell lung cancer (NSCLC). J Clin Oncol (Meeting Abstracts) 2010;28:7500.
    OpenUrlAbstract
  7. ↵
    1. Di Cosimo S,
    2. Bendell JC,
    3. Cervantes-Ruiperez A,
    4. et al
    . A phase I study of the oral mTOR inhibitor ridaforolimus (RIDA) in combination with the IGF-1R antibody dalotozumab (DALO) in patients (pts) with advanced solid tumors. J Clin Oncol (Meeting Abstracts) 2010;28:3008.
    OpenUrlAbstract
  8. ↵
    1. Doern A,
    2. Cao X,
    3. Sereno A,
    4. et al
    . Characterization of inhibitory anti-insulin-like growth factor receptor antibodies with different epitope specificity and ligand-blocking properties: implications for mechanism of action in vivo. J Biol Chem 2009;284:10254–67.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    1. Sorensen H,
    2. Whittaker L,
    3. Hinrichsen J,
    4. Groth A,
    5. Whittaker J
    . Mapping of the insulin-like growth factor II binding site of the type I insulin-like growth factor receptor by alanine scanning mutagenesis. FEBS Lett 2004;565:19–22.
    OpenUrlCrossRefPubMed
  10. ↵
    1. Dong J,
    2. Demarest SJ,
    3. Sereno A,
    4. et al
    . Combination of two insulin-like growth factor-I receptor inhibitory antibodies targeting distinct epitopes leads to an enhanced anti-tumor response. Mol Cancer Ther 2010;9:2593–604.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    1. Baselga J,
    2. Gelmon KA,
    3. Verma S,
    4. et al
    . Phase II trial of pertuzumab and trastuzumab in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer that progressed during prior trastuzumab therapy. J Clin Oncol 2009;28:1138–44.
    OpenUrlCrossRef
  12. ↵
    1. Burtrum D,
    2. Zhu Z,
    3. Lu D,
    4. 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;63:8912–21.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    1. Beltran PJ,
    2. Mitchell P,
    3. Chung YA,
    4. et al
    . AMG 479, a fully human anti-insulin-like growth factor receptor type I monoclonal antibody, inhibits the growth and survival of pancreatic carcinoma cells. Mol Cancer Ther 2009;8:1095–105.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    1. Lacy MQ,
    2. Alsina M,
    3. Fonseca R,
    4. et al
    . Phase I, pharmacokinetic and pharmacodynamic study of the anti-insulin-like growth factor type 1 receptor monoclonal antibody CP-751,871 in patients with multiple myeloma. J Clin Oncol 2008;26:3196–203.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Carden CP,
    2. Kim ES,
    3. Jones RL,
    4. et al
    . Phase I study of intermittent dosing of OSI-906, a dual tyrosine kinase inhibitor of insulin-like growth factor-1 receptor (IGF-1R) and insulin receptor (IR) in patients with advanced solid tumors. ASCO Meeting Abstracts 2010;28:2530.
    OpenUrlAbstract
  16. ↵
    1. Haluska P,
    2. Worden F,
    3. Olmos D,
    4. et al
    . Safety, tolerability, and pharmacokinetics of the anti-IGF-1R monoclonal antibody figitumumab in patients with refractory adrenocortical carcinoma. Cancer Chemother Pharmacol 2010;65:765–73. Epub 2009 Aug 2.
    OpenUrlCrossRefPubMed
  17. ↵
    1. Feinberg AP,
    2. Tycko B
    . The history of cancer epigenetics. Nat Rev Cancer 2004;4:143–53.
    OpenUrlCrossRefPubMed
  18. ↵
    1. De Meyts P,
    2. Whittaker J
    . Structural biology of insulin and IGF1 receptors: implications for drug design. Nat Rev Drug Discov 2002;1:769–83.
    OpenUrlCrossRefPubMed
  19. ↵
    1. Ulanet DB,
    2. Ludwig DL,
    3. Kahn CR,
    4. Hanahan D
    . Insulin receptor functionally enhances multistage tumor progression and conveys intrinsic resistance to IGF-1R targeted therapy. Proc Natl Acad Sci U S A 2010;107:10791–8.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    1. Chakravarti A,
    2. Loeffler JS,
    3. Dyson NJ
    . Insulin-like growth factor receptor I mediates resistance to anti-epidermal growth factor receptor therapy in primary human glioblastoma cells through continued activation of phosphoinositide 3-kinase signaling. Cancer Res 2002;62:200–7.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    1. Jones HE,
    2. Goddard L,
    3. Gee JM,
    4. et al
    . Insulin-like growth factor-I receptor signalling and acquired resistance to gefitinib (ZD1839; Iressa) in human breast and prostate cancer cells. Endocr Relat Cancer 2004;11:793–814.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    1. O'Reilly KE,
    2. Rojo F,
    3. She QB,
    4. et al
    . mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res 2006;66:1500–8.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top
Molecular Cancer Therapeutics: 9 (9)
September 2010
Volume 9, Issue 9
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Molecular Cancer Therapeutics article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Targeting Insulin-Like Growth Factor Signaling: Rational Combination Strategies
(Your Name) has forwarded a page to you from Molecular Cancer Therapeutics
(Your Name) thought you would be interested in this article in Molecular Cancer Therapeutics.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Targeting Insulin-Like Growth Factor Signaling: Rational Combination Strategies
David Olmos, Bristi Basu and Johann S. de Bono
Mol Cancer Ther September 1 2010 (9) (9) 2447-2449; DOI: 10.1158/1535-7163.MCT-10-0719

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Targeting Insulin-Like Growth Factor Signaling: Rational Combination Strategies
David Olmos, Bristi Basu and Johann S. de Bono
Mol Cancer Ther September 1 2010 (9) (9) 2447-2449; DOI: 10.1158/1535-7163.MCT-10-0719
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Disclosure of Potential Conflicts of Interest
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • A Vision from the New Editor-in-Chief
  • A Multi–FAK-torial Switch
  • A Message from the New Editor-in-Chief
Show more Editorial
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About MCT

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Molecular Cancer Therapeutics
eISSN: 1538-8514
ISSN: 1535-7163

Advertisement