Epidermal growth factor receptor signaling in colorectal cancer: preclinical data and therapeutic perspectives

Epidermal growth factor receptor signaling in colorectal cancer: preclinical data and therapeutic perspectives

Annals of Oncology 16: 189 – 194, 2005 doi:10.1093/annonc/mdi057 Review Epidermal growth factor receptor signaling in colorectal cancer: preclinical...

63KB Sizes 0 Downloads 42 Views

Annals of Oncology 16: 189 – 194, 2005 doi:10.1093/annonc/mdi057

Review

Epidermal growth factor receptor signaling in colorectal cancer: preclinical data and therapeutic perspectives J. P. Spano1*, R. Fagard2, J.-C. Soria3, O. Rixe1, D. Khayat1 & G. Milano4 1

SOMPS, Groupe Hospitalier Pitie´-Salpe´trie`re, Universite´ Pierre et Marie Curie, Paris; 2De´partement de Biochimie et de Biologie Mole´culaire, Hoˆpital Avicenne, Bobigny; 3De´partement de Me´decine, Institut Gustave Roussy, Villejuif; 4Laboratoire d’Oncopharmacologie, CAC Antoine Lacassagne, Nice, France

Received 27 June 2004; revised 11 October 2004; accepted 11 October 2004

Epidermal growth factor receptor (EGFR) belongs to a family of receptors known as the ErbB family (ErbB tyrosine kinase receptors) which comprises four proteins encoded by the c-erbB protooncogene. EGFR is known to activate a cascade of multiple signaling pathways that facilitate tumor growth process. EGFR has been shown to be overexpressed in colorectal cancer patient populations but its prognostic value in colorectal cancer progression remains unclear. The development of a panel of EGFR inhibitors could reduce the proliferation of tumor cells when used alone or in combination with cytotoxic drugs or radiation. This review focuses on the potential role of EGFR signaling in the survival of colorectal tumor cells and the possible modulation of such signaling pathways by EGFR inhibitors so as to increase tumor control or render tumor cells more sensitive to conventional therapy. Key words: colorectal cancer, EGFR, therapy implications

Epidermal growth factor receptor (EGFR) and its signaling pathway The ErbB family of receptor tyrosine kinases comprises ErbB1, 2, 3 and 4. ErbB1, also known as EGFR, is a typical member of the ErbB family having a tyrosine kinase activity, stimulated upon ligand binding [1, 2]. EGFR is a 170-kDa transmembrane glycoprotein composed of an intracellular tyrosine kinase (TK) domain, a transmembrane lipophilic segment and an extracellular ligand-binding domain. Its main autocrine ligands are epidermal growth factor (EGF) and transforming growth factor-alpha (TGF-a), which were described 20 years ago [3, 4] and are known to be activators of DNA synthesis and cell growth in numerous tumor sites [5] including gastrointestinal disease [6]. EGFR catalyzes the transfer of phosphate molecules from ATP to an active site of tyrosine kinase to mediate signals, triggering a cascade of well-identified molecular events that will protect cells from apoptosis, facilitate invasion and promote angiogenesis reaction [7, 8]. After binding its ligand, EGFR is known to homodimerize, then to transphosphorylate several tyrosine kinase domains and thus to initiate intracellular EGFR signaling pathway. This includes the activation of STAT proteins, SRC family kinases, AKT protein and MAP kinases, inducing the transcription of genes involved in cellular processes such as

*Correspondence to: Dr J.-P. Spano, De´partement d’Oncologie Me´dicale, Groupe Hospitalier Pitie´-Salpe´trie`re, 47 Bld de l’Hoˆpital, 75 013 Paris, France. Tel: +33-1-42-16-04-52; Fax: +33-1-42-16-04-65; E-mail: [email protected] q 2005 European Society for Medical Oncology

cell division and survival [9, 10]. Moreover, it has been shown that EGFR can also mediate the activation of the other members of the ErbB family. This explains, in part, its major role in tumor cell growth [2]. AKT is a well-established antiapoptotic kinase, enabling cell survival via activation of IkB kinase, which plays a fundamental positive regulating role in NF-kB, by leading the transcription of anti-apoptotic genes [10] or by antagonizing P21-mediated cell cycle arrest [11]. In addition, AKT, thanks to its several cell effects, can also activate endothelial nitric oxide synthase, which plays a crucial role in angiogenesis [12]. AKT may also permit the activation of telomerase activity by phosphorylation of the human telomerase reverse transcriptase [13]. In addition to all these major roles in tumor growth and promotion, AKT activation may promote the tumor invasion and metastases process by stimulating the matrix metalloproteinase protein [14]. MAPK pathways with PI3K pathway have also been shown to have anti-apoptotic effects in some cell types [2] and mounting evidence suggests that the anti-apoptotic effect of ErbB receptor signaling may be attributed to the activation of PI3K/AKT pathways [2, 15]. Activation of the MAPK kinase pathway signaling also increases the expression of major proteins involved in the cell cycle regulation, such as Mcl-1, in concert with the activation of Mdm2 protein by AKT [10]. Regarding all these findings, the identification of inhibitors of the different EGFR signaling pathways in a new therapeutic strategies setting could contribute to reduced tumor growth by restoring the pro-apoptotic mechanisms and by reducing cell proliferation.

190

EGFR expression in colorectal cancer and prognosis Enhanced activity or overexpression of EGFR has been found to be associated with tumor progression and poor survival in various malignancies such as head and neck [16], lung [17], breast [18], gastrointestinal tract [19] and bladder [20] cancers. It has been well documented that overexpression of EGFR in colon cancer may be linked to an advanced stage of the disease [21] or may predict a potential metastatic risk [22]. However, the impact of EGFR expression on survival remains controversial and overexpression of EGFR is not uniformly associated with an unfavorable prognosis. In most cases, immunohistochemical methods were used for the detection of EGFR in colorectal cancer. The variability of IHC is well known and thus EGFR overexpression in colorectal cancer, which ranges from 25% to 82% [7, 22 –26], needs to be more clearly defined. It has already been demonstrated, using an immunohistochemical technique, that the basal level of expression of TGF-a, EGF and their common receptor EGFR, as well as EGFR2 and EGFR3 (EGFR4 not already studied), is higher in colorectal cancer tissue than in surrounding mucosa [7, 22, 27, 28]. In primary cultures of human colorectal carcinomas, higher levels of EGFR expression have also been shown in apical, luminal cell membrane during tumor progression suggesting an increased sensitivity of colon cancer cells to a mitogenic stimulation. There might exist an autocrine growth factor in the colon lumen that possibly plays a critical role during tumor progression [29]. However, despite the experimental and clinical data mentioned above, the overexpression of EGFR in colorectal tumors and the relationship between this expression and the other clinical and histological parameters, including the impact on patients’ prognosis and survival, have not been thoroughly investigated. The heterogeneity of EGFR expression in colorectal cancers may partly be attributed to the different detection techniques, although most are based on immunohistochemistry [23]. Despite these known reproducibility and validation difficulties [30–32], immunohistochemical testing remains one of the most common methods used to assess EGFR expression and has also been well validated for the screening of HER2 receptor [33]. Currently, a quantitative immunohistochemical detection scoring system has been approved to evaluate EGFR expression in colon cancer patients [23].

Correlation between EGFR and other clinicopathological parameters in colorectal cancer patients Possible associations between expression (and overexpression of EGFR) and other clinicohistological parameters in colorectal cancer patients remain unclear. Concerning the tumor site, consistent with the main published studies results [23, 34], there are no differences in EGFR expression between the different tumor sites of the colorectal tract. Only one study has

demonstrated higher EGFR expression in cancers of the distal colon than in rectal cancers [35]. In our own study [36], no correlation was found between EGFR expression and tumor site. Some investigators have even suggested that the higher levels of growth factors and EGFR in the left-side large bowel as opposed to the right could be related to different molecular mechanisms between proximal and distal sites [7]. EGFR expression is not uniform throughout the different regions of colorectal tumors, with a higher intensity or reactivity being found in the deepest regions beyond the muscularis propria [23]. In our own experiment [36], a significant association between high EGFR expression and TNM tumor stage at diagnosis was shown, highlighting a relationship between EGFR overexpression and tumor invasion. This is the largest study showing that EGFR expression is marked in TNM tumor stage T3 [36]. These findings are in agreement with an earlier study that examined fewer adenocarcinomas (14 out of 39 colorectal carcinomas examined had an overexpression of EGF and EGFR associated with serosal invasion) [37]. They also confirm the results of Goldstein et al., who demonstrated that EGFR immunoreactivity was significantly higher in the deepest regions of the tumors compared with the superficial or luminal zones [23]. The expression of EGFR protein has also been compared with tumor differentiation grade. The results appear to be controversial with some studies showing a link [24, 38, 39] and others reporting no relationship between tumor differentiation and EGFR levels [7, 23, 35 –37, 40]. No significant correlation was demonstrated between patient gender and patient age in the above-mentioned published studies, or in our study. EGFR expression was reported to be correlated with more aggressive disease [41], increased risk of metastases [22], advanced tumor stage [42] and higher rates of mesenteric lymph-node involvement [37]. Reference was made to the fact that metastatic lesions present genomic differences from the primary tumor [43, 44]. As for EGFR expression, such variations are also possible, probably with higher expression levels in metastatic lesions compared with the primary neoplasm [45, 46]. In the Goldstein study [23], it is noteworthy that EGFR reactivity was higher in both lymph node and metastases than in primary tumors. Also, EGFR reactivity in the deepest region of the primary tumor had the strongest correlation with EGFR reactivity in lymph node and liver metastases. EGFR expression (i.e. positive or negative) was found to be concordant between metastases and primary tumor whereas, in the study by McKay et al. [24], a discrepancy in EGFR protein expression (also determined by immunohistochemical) was described between primary tumors and lymphnode metastases. These controversial findings emphasize the need for additional investigations on this topic.

Expression and prognostic significance of EGFR in colorectal cancer patients Most of the published series, including our findings [7, 23, 24, 34, 36, 40, 47, 48], have shown no association between EGFR

191 overexpression and disease evolution. However, there are relatively few studies in this area and the different patient populations are not comparable. For instance, regarding Goldstein’s findings [23] using a standardized and validated EGFR scoring system, EGFR immunoreactivity (2+ and 3+) was significantly greater in the deepest region than in the mucosa and superficial submucosa. In this study, EGFR expression was associated with decreased survival (P = 0.0252) when considering EGFR reactivity in this deep region, but not EGFR reactivity at all levels of the tumor wall. Our data are in agreement with these observations and show EGFR overexpressed in stage II disease (T3N0M0) in which the tumor extends through the muscularis propria into pericolonic adipose tissue. This highlights the existence of a tumor area overexpressing EGFR, which could carry a more aggressive growth and a higher metastatic potential. These findings could, therefore, offer an opportunity to test the efficacy of EGFR inhibitors at this stage of the disease [49, 50].

How can EGFR inhibitors impair tumor cell growth and improve tumor control? Blocking EGFR activation would obviously represent an innovative and key strategy in patient care because this therapeutic strategy impairs crucial cellular functions linked to proliferation and survival [30]. EGFR signaling can be targeted by either monoclonal antibodies (C225, etc.) or tyrosine kinase inhibitors [ZD 1839 (gefitinib), OSI774 (erlotinib), CI1033, PKI166, GW572016, etc.], or even by antisense approaches (antisense molecules to EGFR or targeting key regulatory regions of the EGFR) [2, 30, 50 –52]. Several mechanisms of action of the different EGFR inhibitors have been reported in preclinical studies and additional data have been gathered in the clinical setting. Antibodies have the propriety of binding to the extracellular portions of ErbB receptors. For instance, C225 seems to interact with the EGFR domain that is associated with ligand binding [50]. For example, C225 can promote growth arrest by blocking the cell cycle in phase G1 [53]. Ye et al. [54] have confirmed these data and have shown an increase in p27 expression and a decrease in CDK2, 4 and 6 expressions, when applying C225 to an ovarian cancer cell model. In addition, C225 may promote apoptosis, as demonstrated in the study by Huang et al. [53], showing an increase in Bax expression and a decrease in Bcl2 expression. Furthermore, Mandal et al. [55] noted a transfer of Bax from the nucleus to the cytoplasm and the activation of caspase 3, 8 and 9 [56] when the DiFi tumor cells were treated with M225 (the murine equivalent of C225). Regarding DNA repair, C225 can trigger a decrease of expression of nuclear DNA-PK, one of the key proteins involved in DNA repair following radiation and cytotoxic effects [57, 58]. Noteworthy, the truncated forms of ErbB1, i.e. EGFR vIII can also be inhibited by C225 [59]. Finally, C225 combined with chemotherapeutic agents or radiotherapy can also help to

improve tumor growth control in experimental models [60, 61]. For instance, in two refractory human colorectal tumor xenografts (Ht-29 and DLD-1), Prewett et al. [41] demonstrated enhanced tumor growth inhibition when C225 was added to CPT-11 therapy. Noteworthy, C225 was able to reverse CPT-11 resistance in both tumor models [41]. Tyrosine kinase inhibitors are small molecules that interact with the intracellular domain of EGFR. Gefitinib is an oral quinazoline derivative that selectively inhibits the EGFR tyrosine kinase and has already demonstrated inhibition in the colon cancer cell lines GEO that express EGFR and TGF-a, in a dose-dependant manner, with anti-angiogenesis effects [62]. Generally, the growth inhibition of gefitinib was cytotoxic, but an apoptosis effect was observed with higher doses. In fact, by binding with the catalytic kinase domain of EGFR, ZD1839, OSI774, PKI166 [2, 30, 63], and for the pan-HER inhibitor CI1033 [64, 65], have a potential and positive impact on apoptosis. This finding has been documented in vitro with the decreased expression of BCl2 concomitant with the increased expression of Bax, with, curiously, no major effect on caspase 3 activity, in CAL33 cultured cells treated by gefitinib [66]. Gefitinib has also been shown to have a dose-dependant anti-proliferative effect when associated with cytotoxic agents such as oxaliplatin [62]. The sequence of administration of cytotoxic drugs with EGFR inhibitors has also been considered. For instance, recently, a schedule-dependant effect was reported between gefitinib and SN-38. The optimal effect was observed when gefitinib was given after SN-38 [67], in two colon cancer cell lines (HT-39 and LoVo). This suggests a new potential active combination in colon cancer, when considering SN-38 as the active CPT-11 metabolite. Gefitinib has also been demonstrated to be a radiosensitizer, especially when given before and/or during radiation therapy [68]. The mechanism of such radiosensitization may be explained by the fact that anti-EGFR agents may attenuate DNA repair [68, 69] and decrease angiogenesis [70]. Previous studies suggest that the greatest growth inhibition obtained by the combination of gefitinib and radiation therapy was observed in those tumor cell lines with the highest EGFR expression [71, 72]. Moreover, EGFR specific ligands such as TGF-a may play a role in radiosensitivity as demonstrated by Sauter et al. [73]. Low plasma TGF-a levels were associated with radioresistance in patients with head and neck cancers [68, 73]. Finally, as mentioned above, the cellular effects of the combination of EGFR-targeting agents and radiation includes, as first event, the induction of DNA damage by the ionizing radiation that activates several mitogenic signaling cascades, including the Raf-MEK-Erk kinase [74]. Finally, the potential antitumor activity of such EGFR inhibitors provides a rationale for relevant clinical implications. Most EGFR inhibitors are currently in phase III clinical trials either as single agents or in combination with chemotherapeutic agents or with ionizing radiation. Some studies [39, 75] have already been published, leading to cetuximab approval in irinotecan-refractory colorectal cancer patients, in several countries or regions (e.g. USA, Europe and Canada).

192 To date, in the clinical setting, only the monoclonal antibodies have really shown activity, and not the small molecules. One issue requiring particular attention involves the predictive response markers that could help select those patients most likely to respond to EGFR-targeting agents. Some investigators have suggested that cutaneous side-effects might be predictors of efficacy. Extensive skin rashes or acne, for instance, seem to be associated with improved survival following anti-EGFR therapy [76, 77]. Of particular note, the presence of specific mutations in the EGFR gene have recently been shown to be associated with dramatic responses to EGFR inhibitors, as recently published in non-small-cell lung cancer (NSCLC) patients treated by gefitinib or erlotinib [77, 78]. Cappuzzo et al. [79] have also suggested that NSCLC with high levels of expression of phosphor-Akt are the ones most likely to benefit from EGFR-inhibitors. In conclusion, although EGFR is not considered to be a prognostic factor in colorectal cancer patients, it plays a major role in tumor cell proliferation. Convincing preclinical and clinical studies have already demonstrated the efficacy of EGFR inhibitors in advanced colorectal carcinomas and their potential synergistic combinations with chemo- and radiation therapy. Additional efforts and trials are needed to evaluate the role of EGFR inhibitors in the adjuvant setting and to help to establish better predictors of the efficacy of such agents. Since EGFR tumor level itself does not seem to be a valuable predictor of response to anti-EGR therapy, other biomarkers need to be evaluated besides EGFR mutated forms, p-Akt expression or EGFR amplification.

References 1. Roberts RB, Min LU, Washington MK et al. Importance of epidermal growth factor receptor signaling in establishment of adenomas and maintenance of carcinomas during intestinal tumorigenesis. Proc Natl Acad Sci USA 2002; 99: 1521– 1526. 2. Grant S, Qiao L, Dent P. Roles of erbB family receptor tyrosine kinases, and downstream signalling pathways, in the control of cell growth and survival. Front Biosci 2002; 7: 376– 389. 3. Coffey RJ Jr, Goustin AS, Soderquist AM et al. Transforming growth factor alpha and beta expression in human colon cancer lines: implications for an autocrine model. Cancer Res 1987; 47: 4590–4594. 4. Carpenter G, Cohen S. Epidermal growth factor. J Biol Chem 1990; 265: 7709–7712. 5. Messa C, Russo F, Caruso MG, Di Leo A. EGF, TGF-a, and EGFR in human colorectal adenocarcinoma. Acta Oncol 1998; 37: 285–289. 6. Barnard JA, Beauchamp RD, Russel WE et al. Epidermal growth factor-related peptides and their relevance to gastrointestinal pathophysiology. Gastroenterology 1995; 108: 564–580. 7. Yarden Y, Sliwkoski MX. Untangling the erbB signalling network. Nat Rev Mol Cell Biol 2001; 2: 127–136. 8. Yarden Y, Ullrich A. Growth factor receptor tyrosine kinases. Ann Rev Biochem 1988; 57: 443 –478. 9. Bowman T, Garcia R, Turkson J, Jove R. STATS in oncogenesis. Oncogene 2000; 19: 2474–2488. 10. Testa JR, Bellacosa A. AKT plays a central role in tumorigenesis. Proc Natl Acad Sci USA 2001; 98: 10983–10985.

11. Zhou BP, Liao Y, Xia W et al. Cytoplasmic localization of p21Cip1/WAF1 by Akt-induced phosphorylation in HER-2/neu-overexpressing cells. Nat Cell Biol 2001; 3: 245–252. 12. Fulton D, Gratton JP, McCabe TJ et al. Regulation of endotheliumderived nitric oxide production by the protein kinase Akt. Nature 1999; 399: 597–601. 13. Kang S, Kwon T, Kwon DY, Do SI. Akt protein kinase enhances human telomerase activity through phosphorylation of telomerase reverse transcriptase subunit. J Biol Chem 1999; 274: 13085–13090. 14. Thant AA, Nawa A, Kikkawa F et al. Fibronectin activates matrix metalloproteinase-9 secretion via the MEK1-MAPK and the PI3K-Akt pathways in ovarian cancer cells. Clin Exp Metastasis 2000; 18: 423–428. 15. Kainulainen V, Sundwall M, Maatta SA et al. A natural erbB4 isoform that does not activate phosphoinositide 3-kinase, mediates proliferation but not survival or chemotaxis. J Biol Chem 2000; 275: 8641–8649. 16. Chua DTT, Sham JST, Kwong DLW et al. Prognostic value of paranasopharyngeal extension of nasopharyngeal carcinoma. A significant factor in local control and distant metastasis. Cancer 1996; 78: 202–210. 17. Tateishi M, Ishida T, Mitsudomi T et al. Immunohistochemical evidence of autocrine growth factors in adenocarcinoma of the lung. Cancer Res 1990; 50: 7077–7080. 18. Nicholson S, Richard J, Sainsbury C et al. Epidermal growth factor receptor [EGFR]-results of a 6-year follow-up study in operable breast cancer with emphasis on the node negative group. Br J Cancer 1991; 63: 146–150. 19. Jonjic N, Kovac D, Krasevic M et al. Epidermal growth factor-receptor expression correlates with tumor cell proliferation and prognosis in gastric cancer. Anticancer Res 1997; 17: 3883–3888. 20. Neal DE, Sharples L, Smith K et al. The epidermal growth factor receptor and the prognosis of bladder cancer. Cancer 1990; 65: 1619–1625. 21. Gross ME, Zorbas MA, Daniels YL et al. Cellular growth response to epidermal growth factor in colon carcinoma cells with an amplified epidermal growth factor receptor derived from a familial adenomatous polyposis patient. Cancer Res 1991; 51: 1452–1459. 22. Radinsky R, Risin S, Fan D et al. Level and function of epidermal growth factor receptor predict the metastatic potential of human colon carcinoma cells. Clin Cancer Res 1995; 1: 19– 31. 23. Goldstein NS, Armin M. Epidermal growth factor receptor immunohistochemical reactivity in patients with American Joint Committee on cancer stage IV colon adenocarcinoma. Cancer 2001; 92: 1331–1346. 24. McKay JA, Murray LJ, Curran S et al. Evaluation of the epidermal growth factor receptor (EGFR) in colorectal tumors and lymph node metastases. Eur J Cancer 2002; 38: 2258–2264. 25. Wan CW, McKnight MK, Brattain DE et al. Different epidermal growth factor growth responses and receptor levels in human colon cell lines. Cancer Lett 1988; 43: 139–143. 26. Cunningham D, Humblet Y, Siena S et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med 2004; 351: 337 –345. 27. Kluftinger AM, Robinson BW, Quenville NF et al. Correlation of epidermal growth factor receptor and c-erbB2 oncogene product to known prognostic indicators of colorectal cancers. Surg Oncol 1992; 1: 97–105. 28. Nakae S, Shimada E, Urakawa T. Study of c-erbB-2 receptor expression and DNA ploidy pattern in colorectal carcinoma. J Surg Oncol 1993; 54: 246–251.

193 29. Tong WM, Ellinger A, Sheinin Y, Cross HS. Epidermal growth factor receptor expression in primary cultured human colorectal carcinoma cells. Br J Cancer 1998; 77: 1792–1798. 30. Baselga J. Targeting the epidermal growth factor receptor with tyrosine kinase inhibitors: small molecules, big hopes. J Clin Oncol 2002; 20: 2217–2219. 31. Waksal HW. Role of an anti-epidermal growth factor receptor in treating cancer. Cancer Metastasis Rev 1999; 18: 427–436. 32. Formento JL, Francoual M, Formento P et al. Epidermal growth factor receptor assay: validation of a single point method and application to breast cancer. Breast Cancer Res Treat 1991; 17: 211–216. 33. Allred DC, Swanson PE. Testing for erbB-2 by immunohistochemistry in breast cancer. Am J Clin Pathol 2000; 113: 171– 175. 34. Koenders PG, Peters WH, Wobbes T et al. Epidermal growth factor receptor levels are lower in carcinomatous than in normal colorectal tissue. Br J Cancer 1992; 65: 189 –192. 35. Koretz K, Schlag P, Moller P. Expression of epidermal growth factor receptor in normal colorectal mucosa, adenoma, and carcinoma. Virchow Arch A Pathol Anat Histopathol 1990; 416: 343–349. 36. Spano JP, Lagorce C, Atlan D et al. Impact of EGFR expression on colorectal cancer patient prognosis and survival. Ann Oncol 2005; 16: 102 –108. 37. Karameris A, Kanavaros P, Aninos D et al. Expression of epidermal growth factor (EGF) and epidermal growth factor receptor (EGFR) in gastric and colorectal carcinomas. An immunohistological study of 63 cases. Pathol Res Pract 1993; 189: 133–137. 38. Steele RJ, Kelly P, Ellul B et al. Epidermal growth factor receptor expression in colorectal cancer. Br J Surg 1990; 77: 1352–1354. 39. Steele RJ, Kelly P, Ellul B et al. Immunohistochemical detection of epidermal growth factor receptors on human colonic carcinomas. Br J Cancer 1990; 61: 325–326. 40. Moorghen M, Ince P, Finney KJ et al. Epidermal growth factor receptors in colorectal carcinoma. Anticancer Res 1990; 10: 605 –611. 41. Prewett MC, Hooper AT, Bassi R et al. Enhanced antitumor activity of anti-epidermal growth factor receptor monoclonal antibody IMC-C225 in combination with irinotecan [CPT-11] against human colorectal tumor xenografts. Clin Cancer Res 2002; 8: 994–1003. 42. Radinsky R. Modulation of tumor cell gene expression and phenotype by the organ-specific metastatic environment. Cancer Metastasis Rev 1995; 14: 323– 338. 43. McKay JA, Douglas JJ, Ross VG et al. Expression of cell cycle control proteins in primary colorectal tumors does not always predict protein expression in lymph node metastases. Clin Cancer Res 2000; 6: 113 –118. 44. McLeod HL, McKay JA, Collie-Duguid ESR, Cassidy J. Therapeutic opportunities from tumor biology in metastatic colon cancer. Eur J Cancer 2000; 36: 1706–1712. 45. Paredes-Zaglul A, Kang JJ, Essig YP et al. Analysis of colorectal cancer by comparative genomic hybridization: evidence for induction of the metastatic phenotype by loss of tumor suppressor genes. Clin Cancer Res 1998; 4: 879–886. 46. Al Mulla F, Keith WN, Pickford IR et al. Comparative genomic hybrodization analysis of primary colorectal carcinomas and their synchronous metastases. Genes Chromosomes Cancer 1999; 24: 306 –314. 47. Saeki T, Salomon DS, Johnson GR et al. Association of epidermal growth factor-related peptides and type I receptor tyrosine kinase with prognosis of human colorectal carcinomas. Jpn J Clin Oncol 1995; 25: 240– 249. 48. Yasui W, Sumiyoshi H, Hata J et al. Expression of epidermal growth factor receptor in human gastric and colonic carcinomas. Cancer Res 1988; 48: 137– 141.

49. Neal DE, Marsh C, Bennett MK et al. Epidermal growth factor receptors in human bladder cancer: comparisons of invasive and superficial tumors. Lancet 1985; i: 366– 368. 50. Mendelsohn J. Epidermal growth factor receptor as a target for therapy with antireceptor monoclonal antibodies. J Natl Cancer Inst Monogr 1992; 13: 125–131. 51. Mendelsohn J, Baselga J. Status of epidermal growth factor receptor antagonists in the biology and treatment of cancer. J Clin Oncol 2003; 21: 2787–2799. 52. Niwa H, Wentzel AL, Li M et al. Antitumor effects of epidermal growth factor receptor antisense oligonucleotides in combination with docetaxel in squamous cell carcinoma of the head and neck. Clin Cancer Res 2003; 9: 5028–5035. 53. Huang SM, Bock JM, Harari PM. Epidermal growth factor receptor blockade with C225 modulates proliferation, apoptosis, and radiosensitivity in squamous cell carcinomas of the head and neck. Cancer Res 1999; 59: 1935– 1940. 54. Ye D, Mendelsohn J, Faz Z. Augmentation of a humanized abtiHER2 m Ab 4D5 induced growth inhibition by a human-mouse chimeric anti-EGF receptor mAb C225. Oncogene 1999; 18: 731–738. 55. Mandal M, Adam L, Mendelsohn J, Kumar R. Nuclear targeting of Bax during apoptosis in human colorectal cancer cells. Oncogene 1998; 17: 999–1007. 56. Liu B, Fang M, Schmidt M et al. Induction of apoptosis and activation of the caspase cascade by anti-EGF receptor monoclonal antibodies in DiFi human colon cancer cells do not involve the c-jun Nterminal kinase activity. Br J Cancer 2000; 82: 1991–1999. 57. Wood RD, Mitchell M, Sgouros J, Lindahl T. Human DNA repair genes. Science 2001; 291: 1284– 1289. 58. Bandyopadhay D, Mandal M, Adamn L et al. Physical interaction between epidermal growth factor receptor and DNA-dependent protein kinase in mammalian cells. J Biol Chem 1998; 273: 1568–1573. 59. Luwor RB, Johns TG, Murone C et al. Monoclonal antibody 806 inhibits the growth of tumor xenografts expressing either the de2-7 or amplified epidermal growth factor (EGFR) but not wild-type EGFR. Cancer Res 2001; 61: 5355–5361. 60. Mendelsohn J. Blockade of receptors for growth factors: an anticancer therapy. Clin Cancer Res 2000; 6: 747–753. 61. Huang SM, Harari PM. Modulation of radiation response after epidermal growth factor receptor blockade in squamous cell carcinomas: inhibition of damage repair, cell cycle kinetics and tumor angiogenesis. Clin Cancer Res 2000; 6: 2166–2174. 62. Ciardello F, Caputo R, Bianco R et al. Antitumor effect and potentiation of cytotoxic drugs activity in human cancer cells by ZD-1839 (Iressa), an epidermal growth factor receptor-selective tyrosine kinase inhibitor. Clin Cancer Res 2000; 6: 2053–2063. 63. Hidalgo M, Siu LL, Nemunaitis J et al. Phase I and pharmacologic study of OSI-774, an epidermal growth factor receptor tyrosine kinase inhibitor, in patients with advanced solid malignancies. J Clin Oncol 2001; 19: 3267–3279. 64. Mendelsohn J, Baselga J. The EGFR receptor family as targets for cancer therapy. Oncogene 2000; 19: 6550–6565. 65. Erlichman C, Boerner SA, Hallgren CG et al. The HER tyrosine kinase inhibitor CI1033 enhances cytotoxicity of 7-ethyl-10-hydroxycamptothecin and topotecan by breast cancer resistance proteinmediated drug efflux. Cancer Res 2001; 61: 739 –748. 66. Milano G, Magne N. Pharmacological consequences of EGFR targeting. Bull Cancer 2003; 90: S197–S201. 67. Azzariti A, Xu JM, Porcelli L, Paradiso A. The schedule-dependent enhanced cytotoxic activity of 7-ethyl-10-hydroxy-camptothecin

194

68.

69.

70.

71.

72.

73.

(SN-38) in combination with gefitinib (Iressa, ZD1839). Biochem Pharmacol 2004; 68: 135–144. Magne N, Fischel JL, Dubreuil A et al. Sequence-dependent effects of ZD1839 (IRESSA) in combination with cytotoxic treatment in human head and neck cancer. Br J Cancer 2002; 86: 819–827. Pietras M, Salomon D. Biological rationale for HER2/neu (c-rebB2) as a target for monoclonal antibody therapy. Semin Oncol 2000; 27: 13 –19. Hirata A, Ogawa S, Kometani T et al. ZD1839 (Iressa) induces antiangiogenic effects though inhibition of epidermal growth factor receptor tyrosine kinase. Cancer Res 2002; 62: 2554– 2560. Bonner JA, Maihle NJ, Folven BR et al. The interaction of epidermal growth factor and radiation in human head and neck squamous cell carcinoma cell lines with vastly different radiosensitivity. Int J Radiat Oncol Biol Phys 1994; 29: 243–247. Bonner JA, Raish KP, Trummell HQ et al. Enhanced apoptosis with combination C225/radiation treatment serves as the impetus for clinical investigation in head and neck cancers. J Clin Oncol 2000; 18: 47–53. Sauter ER, Coia LR, Eisenberg BL, Ridge JA. Radiation treatment decreases transforming growth factor alpha expression in squamous carcinoma of the tongue. Cancer Lett 1994; 78: 159 –162.

74. Todd DG, Mikkelsen RB, Rorrer WK et al. Ionizing radiation stimulates existing signal transduction pathways involving the activation of epidermal growth factor receptor and erb-B-3, and changes of intracellular calcium in A431 human squamous carcinoma cells. J Recept Signal Transduct Res 1999; 19: 885–908. 75. Saltz LB, Meropol NJ, Loehrer PJ Sr et al. Phase II trial of cetuximab in patients with refractory colorectal cancer that expresses the epidermal growth factor receptor. J Clin Oncol 2004; 22: 1201–1208. 76. Ellis LM, Hoff PM. Targeting the epidermal growth factor receptor: an important incremental step in the battle against colorectal cancer. J Clin Oncol 2004; 22: 1177–1179. 77. Lynch TJ, Bell DW, Sordella R et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of nonsmall-cell lung cancer to gefitinib. N Engl J Med 2004; 350: 2129–2139. 78. Pao W, Miller V, Zakowski M et al. EGF receptor gene mutations are common in lung cancers from ‘never smokers’ and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci USA 2004; 101: 13306–13311. 79. Cappuzzo F, Magrini E, Ceresoli GL et al. Akt phosphorylation and gefitinib efficacy in patients with advanced non-small-cell lung cancer. J Natl Cancer Inst 2004; 96: 1133–1141.