Skip to main content

GENERAL COMMENTARY article

Front. Immunol., 07 January 2013
Sec. Vaccines and Molecular Therapeutics

Prediction of response to anti-EGFR antibodies in metastatic colorectal cancer: looking beyond EGFR inhibition


Alessandro Ottaiano* Alessandro Ottaiano1*Maurizio Capuozzo Maurizio Capuozzo2Guglielmo Nasti Guglielmo Nasti1Piera Maiolino Piera Maiolino3Valentina De Angelis Valentina De Angelis4Stefania Scala Stefania Scala5Rosario V. Iaffaioli Rosario V. Iaffaioli1
  • 1Department of Colorectal Oncology at the National Cancer Institute, “G. Pascale” foundation, via M. Semmola, Naples, Italy
  • 2Department of Pharmacy at the ASL-Naples-3, via Marittima 3/B, Ercolano, NA, Italy
  • 3Department of Pharmacy at the National Cancer Institute, “G. Pascale” foundation, via M. Semmola, Naples, Italy
  • 4Institute of Psychological and Systemic Medicine, via F. Giordani 30, Naples, Italy
  • 5Department of Cancer Immunology at the National Cancer Institute, “G. Pascale” foundation, via M. Semmola, Naples, Italy

A commentary on
The evolving role of monoclonal antibodies in colorectal cancer: early presumptions and impact on clinical trial development

by Eng, C. (2010). Oncologist 15, 73–84.

One of the most successfully approach in the treatment of metastatic colorectal cancer (mCRC) is the inhibition of the Epidermal Growth Factor Receptor (EGFR) pathway by antibodies (cetuximab and panitumumab). Notably, randomized trials with anti-EGFR antibodies have shown a significant impact of KRAS [wild type (wt) vs. mutated (mut)] on response and prognosis: the presence of KRAS activating mutations was found to be associated with reduced biological and clinical activity for the treatment (response rate in mut <20% vs. wt >50%). Thus, the mutational status of KRAS is now a widely accepted criteria for selection of patients who would be most likely to respond to these treatments. In the next future, other genes involved in the EGFR pathway could have a role in the prediction of treatment response (BRAF, PIK3CA, PTEN, etc.) (De Roock et al., 2011).

Cetuximab is an IgG1 monoclonal antibody, it binds specifically to the extracellular domain of EGFR inhibiting downstream proliferative, anti-apoptotic and neoangiogenetic signals in kras wt tumors and it has clinical efficacy in mCRC (Eng, 2010). However, one of the accepted anti-tumor mechanism is the antibody-dependent cell-mediated cytotoxicity (ADCC) in which Fc region of the antibody binds to the FcγRs (Fragment c Gamma Receptors) expressed by immune effector cells (Natural Killer cells, macrophages, etc.) (Kohrt et al., 2012). However, the scenario is very complex and the result is not the simple sum of the above phenomena. Very recently, it has been demonstrated that immunologic mechanisms can cooperate (ADCC) but also antagonize with the inhibition of EGFR/kras signal. In fact, CD163+ “tumor-promoting” M2 macrophages which can be abundant in the microenvironment of colorectal carcinomas are activated by cetuximab and in turn they release anti-inflammatory and tumor-promoting mediators, including IL-10 and VEGF (Pander et al., 2011). Furthermore, both ADCC and cetuximab-induced macrophage responses can be more pronounced for FcγRIIIa 158-Val (high-affinity receptor for Fc) carriers (Tsuchiya et al., 2007; Pander et al., 2011). The different abundance and activity of CD163 + M2 macrophages in tumor environment could explain the contrasting results reported in literature on the role of FcγR polymorphisms in mCRC (Zhang et al., 2007; Bibeau et al., 2009).

Very recently, we have demonstrated that homozygous carriers of the 158V allele of the FcγRIIIa show a high response rate and a significantly improved prognosis in kras wt mCRC (Calemma et al., 2012). This was consistent with the hypothesis that variants of human IgG-receptors can influence the extent of ADCC and, thus, response to anti-EGFR therapy. We made, however, the intriguing observation that FcγRIIIa polymorphisms had a prognostic power also in the entire series, including patients with mut kras who did not receive anti-EGFR therapy (data not shown). To confirm this observation, we are extending the analysis of FcγRIIIa polymorphisms to all mCRC patients referring to our center. Our speculation is that ADCC could be triggered by “endogenous” anti-tumor antibodies binding to “high-affinity” FcγR and might be capable of mediating a clinically relevant anti-tumor activity. Such antibodies could be present and work also in mutant kras mCRC patients. The hypothesis that “endogenous” rather than “therapeutic” antibodies might trigger such activity is fascinating but difficult to demonstrate and could be responsible of long-term clinical stabilizations after surgery and/or radio and/or chemotherapy that we see in clinical practice. Indirectly, increased rates of antibody-mediated autoimmune diseases in 158V carriers suggest that the polymorphism also plays a relevant role in the binding of endogenous antibodies (Matsumoto et al., 2005).

ADCC could be also responsible of responses to anti-EGFR antibodies seen in KRAS mut tumors. In fact, Ashraf et al. (2012) have demonstrated that higher EGFR expression can predict susceptibility to cetuximab-induced immune killing of CRC cells occurring independently of KRAS/BRAF/PIK3CA mutations (in press on Proc. Natl. Acad. Sci. U.S.A.). Therefore, administration of anti-EGFR antibodies may be considered in CRC tumors with higher target expression and favorable FcγR polymorphisms. However, the context is very complex and other factors can influence the response to anti-tumor antibodies: previous and/or concomitant therapies, HLA expression, cytokines production, immune cell receptors repertoire, etc.

Study of interactions between host and tumors should be urgently improved to optimize the prediction of response to therapeutic antibodies in mCRC.

References

Ashraf, S. Q., Nicholls, A. M., Wilding, J. L., Ntouroupi, T. G., Mortensen, N. J., and Bodmer, W. F. (2012). Direct and immune mediated antibody targeting of ERBB receptors in a colorectal cancer cell-line panel. Proc. Natl. Acad. Sci. U.S.A. 109, 21046–21051.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bibeau, F., Lopez-Crapez, E., Di Fiore, F., Thezenas, S., Ychou, M., Blanchard, F., et al. (2009). Impact of Fc RIIa-Fc RIIIa polymorphisms and KRAS mutations on the clinical outcome of patients with metastatic colorectal cancer treated with cetuximab plus irinotecan. J. Clin. Oncol. 27, 1122–1129.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Calemma, R., Ottaiano, A., Trotta, A. M., Nasti, G., Romano, C., Napolitano, M., et al. (2012). Fc gamma receptor IIIa polymorphisms in advanced colorectal cancer patients correlated with response to anti-EGFR antibodies and clinical outcome. J. Transl. Med. 10, 232–238.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

De Roock, W., De Vriendt, V., Normanno, N., Ciardiello, F., and Tejpar, S. (2011). KRAS, BRAF, PIK3CA, and PTEN mutations: implications for targeted therapies in metastatic colorectal cancer. Lancet Oncol. 12, 594–603.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Eng, C. (2010). The evolving role of monoclonal antibodies in colorectal cancer: early presumptions and impact on clinical trial development. Oncologist 15, 73–84.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kohrt, H. E., Houot, R., Marabelle, A., Cho, H. J., Osman, K., Goldstein, M., et al. (2012). Combination strategies to enhance antitumor ADCC. Immunotherapy 4, 511–527.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Matsumoto, I., Zhang, H., Muraki, Y., Hayashi, T., Yasukochi, T., Kori, Y., et al. (2005). A functional variant of Fcgamma receptor IIIA is associated with rheumatoid arthritis in individuals who are positive for anti-glucose-6-phosphate isomerase antibodies. Arthritis Res. Ther. 7, 1183–1188.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pander, J., Heusinkveld, M., van der Straaten, T., Jordanova, E. S., Baak-Pablo, R., Gelderblom, H., et al. (2011). Activation of tumor-promoting type 2 macrophages by EGFR-targeting antibody cetuximab. Clin. Cancer Res. 17, 5668–5673.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Tsuchiya, N., Kyogoku, C., Miyashita, R., and Kuroki, K. (2007). Diversity of human immune system multigene families and its implication in the genetic background of rheumatic diseases. Curr. Med. Chem. 14, 431–439.

Pubmed Abstract | Pubmed Full Text

Zhang, W., Gordon, M., Schultheis, A. M., Yang, D. Y., Nagashima, F., Azuma, M., et al. (2007). Fc R2A and Fc R3A Polymorphisms associated with clinical outcome of epidermal growth factor receptor–expressing metastatic colorectal cancer patients treated with single-agent cetuximab. J. Clin. Oncol. 25, 3712–3718.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Citation: Ottaiano A, Capuozzo M, Nasti G, Maiolino P, De Angelis V, Scala S and Iaffaioli RV (2013) Prediction of response to anti-EGFR antibodies in metastatic colorectal cancer: looking beyond EGFR inhibition. Front. Immun. 3:409. doi: 10.3389/fimmu.2012.00409

Received: 16 December 2012; Accepted: 17 December 2012;
Published online: 07 January 2013.

Edited by:

Brian J. Czerniecki, University of Pennsylvania, USA

Reviewed by:

Brian J. Czerniecki, University of Pennsylvania, USA

Copyright © 2013 Ottaiano, Capuozzo, Nasti, Maiolino, De Angelis, Scala and Iaffaioli. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in other forums, provided the original authors and source are credited and subject to any copyright notices concerning any third-party graphics etc.

*Correspondence: ale.otto@libero.it

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.