MMP7 and activation of IGF-1R

2 downloads 0 Views 850KB Size Report
Nov 3, 2010 - Di Nicolantonio F, Martini M, Molinari F, Sartore-. Bianchi A, Arena S, Saletti .... Haro H, Crawford HC, Fingleton B, Shinomiya K,. Spengler DM ...
Autophagic Punctum

Cancer Biology & Therapy 11:2, 184-187; January 15, 2011; © 2011 Landes Bioscience

MMP7 and activation of IGF-1R A new insight into anti-EGFR therapeutic resistance in metastatic colorectal cancer Shinya Ohashi, Mitsuteru Natsuizaka and Hiroshi Nakagawa* Gastroenterology Division; University of Pennsylvania; Philadelphia, PA USA

Cetuximab and panitumumab, antiepidermal growth factor receptor (EGFR) specific monoclonal antibodies, have been clinically proven to be efficacious in a small subset (about 10%) of patients with chemotherapy-resistant metastatic colorectal carcinoma (CRC).1,2 In these cases, the tumor cells may be addicted to the EGFR oncogene as EGFR gene amplification was found in patients responding to antiEGFR agents.2 The failure of anti-EGFR therapy has been attributed to activation of the EGFR downstream effector molecules such as KRAS and BRAF, allowing EGFRindependent tumor cell growth and survival. In metastatic CRCs, somatic KRAS mutations are not only the most common (about 40%) but also specifically (> 90%) associated with impaired responsiveness to anti-EGFR agents.3 BRAF mutations (about 10%) also antagonize anti-EGFR antibodies.4 Additionally, PIK3CA mutations (about 10–15%) and loss of PTEN (20–40%) contribute to activation of the prosurvival PI3K pathway and resistance to EGFR-targeted monoclonal antibodies in metastatic CRCs.5-7 Interestingly, PIK3CA has been identified as one of 26 genes that may predict Cetuximab sensitivity and longer progression-free survival in patients with metastatic CRCs harboring wild-type KRAS.8 However, PIK3CA mRNA was found upregulated in this context. A subset of CRC patients with PIK3CA mutations displayed an objective response to Cetuximab.9 Thus, it remains to be clarified as to how the status of PI3K signaling may influence the effect of EGFR inhibition in CRC. In this issue of Cancer Biology & Therapy, a group led by Joan Maurel at

the University of Barcelona reports that concurrent expression of matrix metalloproteinase-7 (MMP7) and insulin-like growth factor 1 receptor (IGF-1R) phosphorylation predicts poor outcome upon anti-EGFR therapy in advanced CRC without KRAS or BRAF mutations.10 The authors have previously demonstrated a reciprocal increase in MMP7 and decrease in circulating insulin-like growth factor binding protein (IGFBP)-3 during disease progression in CRC patients following chemotherapy, implying MMP7-mediated IGFBP3 degradation and enhancement of IGF-dependent cancer cell survival and proliferation.11 Epidermal growth factor (EGF) suppresses IGFBP-3 mRNA expression through activation of MAPK in an EGFR-tyrosine kinase-dependent manner to permit IGF-I-mediated IGF-1R activation.12 IGFBP loss has been implicated in acquired cancer cell resistance to EGFR tyrosine kinase inhibitors.13 In this study, the authors tested a hypothesis that the MMP7-mediated proteolytic activity leads to IGF-1R activation and EGFR transactivation, providing a new model for tumor cells to circumvent the effects of antiEGFR monoclonal antibodies (Fig. 1). The authors carried out immunohistochemistry on CRC tissue microarrays representing 113 informative cases enrolled in a multi-center study for cetuximab or panitumumab treatment to find that concurrent expression of phosphorylated IGF-1R and MMP7 serves as a new marker predicting poor progression-free and overall survival amongst patients carrying neither KRAS nor BRAF mutations, who constituted nearly 60% of the entire cohort. While phosphorylated IGF-1R

©201 1L andesBi os c i enc e. Donotdi s t r i but e. Key words: epidermal growth factor receptor (EGFR), KRAS, insulin-like growth factor 1 receptor (IGF-1R), insulin-like growth factor binding protein 3 (IGFBP3), matrix metalloproteinase-7 (MMP7), colorectal cancer Submitted: 11/03/10 Accepted: 11/08/10 DOI: 10.4161/cbt.11.2.14140 *Correspondence to: Hiroshi Nakagawa; Email: [email protected] Commentary to: Hörndler C, Gallego R, GarcíaAlbeniz X, Alonso-Espinaco V, Alonso V, Escudero P, et al. Co-expression of matrix metalloproteinase-7 (MMP-7) and phosphorylated insulin growth factor receptor I (pIGF-1R) correlates with poor prognosis in patients with wild-type KRAS treated with cetuximab or panitumumab: A GEMCAD study. Cancer Biol Ther 2011; This issue.

184

Cancer Biology & Therapy

Volume 11 Issue 2

Commentary

Autophagic Punctum

©201 1L andesBi os c i enc e. Donotdi s t r i but e.

Figure 1. IGF-1R-dependent EGFR transactivation involves MMP7-mediated cell surface proteolytic activities and may confer anti-EGFR therapeutic resistance in metastatic CRCs. The study by Hörndler et al.10 indicates concomitant MMP7 and IGF-1R phosphorylation as an independent predictor in clinical outcomes in CRC patients who receive anti-EGFR (α-EGFR) monoclonal antibody therapy. EGFR inhibition can be abrogated by mutations in the EGFR downstream effector molecules indicated by asterisks. IGF-1R activation is negatively regulated by IGFBP3, which is subjected to negative regulation by the RAS-MAPK pathway or epigenetic silencing by promoter methylation. MMP7 can catalyze IGFBP3 proteolysis, liberating IGF to stimulate IGF-1R. The authors hypothesized that IGF-1R can transactivate EGFR to negate the effects of anti-EGFR therapy in patients with wild-type KRAS. Since IGF-1R-mediated EGFR transactivation may involve paracrine EGFR ligands, such as heparin-binding EGF-like growth factor, a future study will be needed to clarify whether IGF-1R can transactivate EGFR in the presence of anti-EGFR antibodies. Substrates for MMP7 include IGFBP3 as well as various cell surface molecules whose degradation may also lead to EGFR transactivation. EGF, epidermal growth factor; FN, Fibronectin; pro-TNFα, pro-tumor necrosis factorα; FasL, Fas ligand.

and MMP7 appeared to be positive in 50% of primary tumors, there was no significant association in expression of these two markers. The IGF-1R phosphorylation status alone influenced neither antiEGFR therapeutic efficacy nor patients’ survival rate. However, CRC patients having tumors double-positive for phosphorylated IGF-1R and MMP7 appeared to be more resistant to first-line treatment despite the KRAS status. Moreover, the double-positive patients with wildtype KRAS (25%) failed to respond to anti-EGFR therapy and progressed more significantly, resulting in poorer overall survival. Multivariate analysis also confirmed that the double positive status was the only factor that can predict survival

www.landesbioscience.com

in the patients with wild-type KRAS. Thus, this study revealed a novel MMP7mediated crosstalk between the EGFR and IGF-1R as a determinant for antiEGFR therapy outcome and prognosis in metastatic CRC patients. The IGF system consists of a family of interacting ligands (IGF-I and IGF-II), receptors and IGFBPs. Both IGF-I and IGF-II exert their biological activities through binding to IGF-1R.14 Amongst six IGFBP secretory glycoproteins, IGFBP3 is the major carrier protein for IGFs in circulation, binding over 90% of IGF detectable in the serum.15,16 In general, IGFBP3 is thought to prevent IGFs from activating IGF-1R. In turn, proteolysis of IGFBP3 increases IGFs available to stimulate

IGF-1R. IGF-1R is frequently overexpressed in CRC.17 IGF-II is amongst most abundantly expressed genes in CRCs.18 Patients with acromegaly characterized by excessive growth hormone and IGF-I in circulation are known to have a substantially increased risk of CRC and other cancers.19 A large scale, prospective, casecontrol study in healthy men implicated circulating IGF-I in the future risk of CRC.20 In addition to these epidemiological studies, many laboratory studies reinforce importance of the IGF system in CRC tumor biology.14 For example, IGF-II has been implicated in the growth of intestinal adenoma in Apc Min/+ mice.21 Amongst many MMPs upregulated in CRCs are MMP1, MMP7 and MMP9,

Cancer Biology & Therapy 185

which have been associated with poor clinical outcome. Unlike other MMPs detected in the stroma of CRC tissues, MMP7 is expressed specifically by tumor cells and is the only MMP detected in the early stages of colorectal carcinogenesis, namely benign adenomas where KRAS mutation emerges.22 In fact, MMP7 is highly expressed in the intestinal adenomas of Apc Min/+ mice and that loss of MMP7 reduces adenomas significantly.23 Interestingly, K-ras mutation did not affect MMP7 expression in ApcMin/+ tumors,24 suggesting that MMP7 is not downstream of K-ras. How does MMP7 affect IGF signaling or anti-EGFR therapeutic efficiency? IGFBP3 is subjected to proteolysis mediated by various proteases including MMP family members.15 MMP7 may mediate IGFBP3 proteolysis directly or indirectly to facilitate IGF bioavailability.25,26 Other known substrates for MMP7 include E-cadherin,27 NOTCH,28 Fas-ligand,29 pro-tumor necrosis factor,30 Laminin and Fibronectin,31 which are all present on the cell surface. MMP7-mediated Laminin cleavage promotes CRC cell motility.32 Interestingly, Fibronectin has been found to form a molecular complex containing IGF-I and IGFBP3,33 indicating the possibility that MMP7 may influence the IGF-1R activity through Fibronectin cleavage. Therefore, MMP7 may indeed contribute to IGF-1R activation as suggested by the study for discussion. However, other MMP7 substrates seem to have critical roles independent of IGF signaling since IGF-1R phosphorylation alone failed to predict clinical outcomes in this study. In fact, the authors’ group has linked MMP7 to chemotherapeutic resistance of CRC cells through Fas upregulation.34 Given antagonistic crosstalk between E-cadherin and EGFR,35 it is tempting to speculate that these MMP7 substrates may also influence the EGFR activity (Fig. 1). The IGF-1R phosphorylation (50%) observed in this study cannot be attributable to MMP7-mediated IGFBP3 downregulation alone since epigenetic silencing of IGFBP3 is detectable in 30–60% of primary CRCs.36,37 In addition, other proteases including MMP1, MMP2 and cathepsin L are associated with CRC

disease progression38 and known to catalyze IGFBP3 proteolysis.15 Thus, IGFBP3 promoter methylation status should be determined in future studies. Intracellular IGF-1R localization is an independent prognostic factor. CRC tumor tissues exhibiting focal and weak membranous IGF-1R staining pattern predicts a significantly increased risk of recurrence and liver metastasis compared to those with a diffuse and strong membranous expression pattern.39 Since IGF-1R phosphorylation was judged to be positive when it was detected in at least 10% of cancer cells in this study, detailed analysis will be required in the future as to how tumors may respond differentially to anti-EGFR therapeutics, depending upon the phospho-IGF-1R positive cell rate and IGF-1R localization within a single tumor. The authors should also determine EGFR phosphorylation status to test their hypothesis more directly. It will be interesting to explore the effect of EGFRspecific tyrosine kinase inhibitors instead of anti-EGFR monoclonal antibodies, directly targeting EGFR transactivated through IGF-1R in the patients showing concomitant activation of IGF-1R and EGFR without KRAS mutations. This study may also provide the basis for the IGF-1R targeting therapy to treat such patients. Two recent studies corroborate such a premise and warrant further investigation. KRAS wild-type CRC patients with tumors expressing IGF-I appeared to show poorer clinical outcome when treated with Cetuximab and Irinotecan.40 In addition, polymorphisms in IGF-I and IGF-1R were found to be significantly associated with progression-free survival and overall survival in KRAS wild-type metastatic CRC patients.41

3. Linardou H, Dahabreh IJ, Kanaloupiti D, Siannis F, Bafaloukos D, Kosmidis P, et al. Assessment of somatic k-RAS mutations as a mechanism associated with resistance to EGFR-targeted agents: a systematic review and meta-analysis of studies in advanced non-small-cell lung cancer and metastatic colorectal cancer. Lancet Oncol 2008; 9:962-72. 4. Di Nicolantonio F, Martini M, Molinari F, SartoreBianchi A, Arena S, Saletti P, et al. Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. J Clin Oncol 2008; 26:5705-12. 5. Frattini M, Saletti P, Romagnani E, Martin V, Molinari F, Ghisletta M, et al. PTEN loss of expression predicts cetuximab efficacy in metastatic colorectal cancer patients. Br J Cancer 2007; 97:1139-45. 6. Perrone F, Lampis A, Orsenigo M, Di Bartolomeo M, Gevorgyan A, Losa M, et al. PI3KCA/PTEN deregulation contributes to impaired responses to cetuximab in metastatic colorectal cancer patients. Ann Oncol 2009; 20:84-90. 7. Sartore-Bianchi A, Martini M, Molinari F, Veronese S, Nichelatti M, Artale S, et al. PIK3CA mutations in colorectal cancer are associated with clinical resistance to EGFR-targeted monoclonal antibodies. Cancer Res 2009; 69:1851-7. 8. Balko JM, Black EP. A gene expression predictor of response to EGFR-targeted therapy stratifies progression-free survival to cetuximab in KRAS wild-type metastatic colorectal cancer. BMC Cancer 2009; 9:145. 9. Prenen H, De Schutter J, Jacobs B, De Roock W, Biesmans B, Claes B, et al. PIK3CA mutations are not a major determinant of resistance to the epidermal growth factor receptor inhibitor cetuximab in metastatic colorectal cancer. Clin Cancer Res 2009; 15:3184-8. 10. Hörndler C, Gallego R, García-Albeniz X, Alonso-Espinaco V, Alonso V, Escudero P, et al. Co-expression of matrix metalloproteinase-7 (MMP7) and phosphorylated insulin growth factor receptor I (pIGF-1R) correlates with poor prognosis in patients with wild-type KRAS treated with cetuximab or panitumumab. A GEMCAD study. Cancer Biol Ther 2011; 11;177-83. 11. Massoner P, Colleselli D, Matscheski A, Pircher H, Geley S, Jansen Durr P, et al. Novel mechanism of IGF-binding protein-3 action on prostate cancer cells: inhibition of proliferation, adhesion and motility. Endocr Relat Cancer 2009; 16:795-808. 12. Takaoka M, Smith CE, Mashiba MK, Okawa T, Andl CD, El-Deiry WS, et al. EGF-mediated regulation of IGFBP-3 determines esophageal epithelial cellular response to IGF-I. Am J Physiol Gastrointest Liver Physiol 2006; 290:404-16. 13. Guix M, Faber AC, Wang SE, Olivares MG, Song Y, Qu S, et al. Acquired resistance to EGFR tyrosine kinase inhibitors in cancer cells is mediated by loss of IGF-binding proteins. J Clin Invest 2008; 118:2609-19. 14. Donovan EA, Kummar S. Role of insulin-like growth factor-1R system in colorectal carcinogenesis. Crit Rev Oncol Hematol 2008; 66:91-8. 15. Firth SM, Baxter RC. Cellular actions of the insulinlike growth factor binding proteins. Endocr Rev 2002; 23:824-54. 16. Hwa V, Oh Y, Rosenfeld RG. The insulin-like growth factor-binding protein (IGFBP) superfamily. Endocr Rev 1999; 20:761-87. 17. Weber MM, Fottner C, Liu SB, Jung MC, Engelhardt D, Baretton GB. Overexpression of the insulin-like growth factor I receptor in human colon carcinomas. Cancer 2002; 95:2086-95.

©201 1L andesBi os c i enc e. Donotdi s t r i but e.

186

References 1. Cunningham D, Humblet Y, Siena S, Khayat D, Bleiberg H, Santoro A, et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecanrefractory metastatic colorectal cancer. N Engl J Med 2004; 351:337-45. 2. Moroni M, Veronese S, Benvenuti S, Marrapese G, Sartore-Bianchi A, Di Nicolantonio F, et al. Gene copy number for epidermal growth factor receptor (EGFR) and clinical response to antiEGFR treatment in colorectal cancer: a cohort study. Lancet Oncol 2005; 6:279-86.

Cancer Biology & Therapy

Volume 11 Issue 2

18. Zhang L, Zhou W, Velculescu VE, Kern SE, Hruban RH, Hamilton SR, et al. Gene expression profiles in normal and cancer cells. Science 1997; 276:1268-72. 19. Jenkins PJ. Cancers associated with acromegaly. Neuroendocrinology 2006; 83:218-23. 20. Ma J, Pollak MN, Giovannucci E, Chan JM, Tao Y, Hennekens CH, et al. Prospective study of colorectal cancer risk in men and plasma levels of insulin-like growth factor (IGF)-I and IGF-binding protein-3. J Natl Cancer Inst 1999; 91:620-5. 21. Hassan AB, Howell JA. Insulin-like growth factor II supply modifies growth of intestinal adenoma in Apc(Min/+) mice. Cancer Res 2000; 60:1070-6. 22. Wagenaar-Miller RA, Gorden L, Matrisian LM. Matrix metalloproteinases in colorectal cancer: is it worth talking about? Cancer Metastasis Rev 2004; 23:119-35. 23. Wilson CL, Heppner KJ, Labosky PA, Hogan BL, Matrisian LM. Intestinal tumorigenesis is suppressed in mice lacking the metalloproteinase matrilysin. Proc Natl Acad Sci USA 1997; 94:1402-7. 24. Luo F, Brooks DG, Ye H, Hamoudi R, Poulogiannis G, Patek CE, et al. Mutated K-ras(Asp12) promotes tumourigenesis in Apc(Min) mice more in the large than the small intestines, with synergistic effects between K-ras and Wnt pathways. Int J Exp Pathol 2009; 90:558-74. 25. Mochizuki S, Shimoda M, Shiomi T, Fujii Y, Okada Y. ADAM28 is activated by MMP-7 (matrilysin-1) and cleaves insulin-like growth factor binding protein-3. Biochem Biophys Res Commun 2004; 315:79-84. 26. Miyamoto S, Yano K, Sugimoto S, Ishii G, Hasebe T, Endoh Y, et al. Matrix metalloproteinase-7 facilitates insulin-like growth factor bioavailability through its proteinase activity on insulin-like growth factor binding protein 3. Cancer Res 2004; 64:665-71.

27. Noe V, Fingleton B, Jacobs K, Crawford HC, Vermeulen S, Steelant W, et al. Release of an invasion promoter E-cadherin fragment by matrilysin and stromelysin-1. J Cell Sci 2001; 114:111-8. 28. Sawey ET, Johnson JA, Crawford HC. Matrix metalloproteinase 7 controls pancreatic acinar cell transdifferentiation by activating the Notch signaling pathway. Proc Natl Acad Sci USA 2007; 104:19327-32. 29. Mitsiades N, Yu WH, Poulaki V, Tsokos M, Stamenkovic I. Matrix metalloproteinase-7-mediated cleavage of Fas ligand protects tumor cells from chemotherapeutic drug cytotoxicity. Cancer Res 2001; 61:577-81. 30. Haro H, Crawford HC, Fingleton B, Shinomiya K, Spengler DM, Matrisian LM. Matrix metalloproteinase-7-dependent release of tumor necrosis factoralpha in a model of herniated disc resorption. J Clin Invest 2000; 105:143-50. 31. Yamamoto K, Miyazaki K, Higashi S. Cholesterol sulfate alters substrate preference of matrix metalloproteinase-7 and promotes degradations of pericellular laminin-332 and fibronectin. J Biol Chem 2010; 285:28862-73. 32. Remy L, Trespeuch C, Bachy S, Scoazec JY, Rousselle P. Matrilysin 1 influences colon carcinoma cell migration by cleavage of the laminin-5 beta3 chain. Cancer Res 2006; 66:11228-37. 33. Gui Y, Murphy LJ. Insulin-like growth factor (IGF)binding protein-3 (IGFBP-3) binds to fibronectin (FN): demonstration of IGF-I/IGFBP-3/fn ternary complexes in human plasma. J Clin Endocrinol Metab 2001; 86:2104-10. 34. Almendro V, Ametller E, Garcia-Recio S, Collazo O, Casas I, Auge JM, et al. The role of MMP7 and its cross-talk with the FAS/FASL system during the acquisition of chemoresistance to oxaliplatin. PLoS One 2009; 4:4728.

35. Andl CD, Rustgi AK. No one-way street: cross-talk between e-cadherin and receptor tyrosine kinase (RTK) signaling: a mechanism to regulate RTK activity. Cancer Biol Ther 2005; 4:28-31. 36. Tomii K, Tsukuda K, Toyooka S, Dote H, Hanafusa T, Asano H, et al. Aberrant promoter methylation of insulin-like growth factor binding protein-3 gene in human cancers. Int J Cancer 2007; 120:566-73. 37. Kawasaki T, Nosho K, Ohnishi M, Suemoto Y, Kirkner GJ, Fuchs CS, et al. IGFBP3 promoter methylation in colorectal cancer: relationship with microsatellite instability, CpG island methylator phenotype and p53. Neoplasia 2007; 9:1091-8. 38. Troy AM, Sheahan K, Mulcahy HE, Duffy MJ, Hyland JM, O’Donoghue DP. Expression of Cathepsin B and L antigen and activity is associated with early colorectal cancer progression. Eur J Cancer 2004; 40:1610-6. 39. Nakamura M, Miyamoto S, Maeda H, Zhang SC, Sangai T, Ishii G, et al. Low levels of insulin-like growth factor type 1 receptor expression at cancer cell membrane predict liver metastasis in Dukes’ C human colorectal cancers. Clin Cancer Res 2004; 10:8434-41. 40. Scartozzi M, Mandolesi A, Giampieri R, Pierantoni C, Loupakis F, Zaniboni A, et al. Insulin-like growth factor 1 expression correlates with clinical outcome in K-RAS wild type colorectal cancer patients treated with cetuximab and irinotecan. Int J Cancer 2010; 127:1941-7. 41. Winder T, Zhang W, Yang D, Ning Y, Bohanes P, Gerger A, et al. Germline polymorphisms in genes involved in the IGF1-pathway predict efficacy of cetuximab in wild-type KRAS mCRC patients. Clin Cancer Res 2010; 16:5591-602.

©201 1L andesBi os c i enc e. Donotdi s t r i but e.

www.landesbioscience.com

Cancer Biology & Therapy 187

Suggest Documents