Você está na página 1de 3

Editorial

Epidermal Growth Factor Receptor Inhibitors: A Moving Target?


Susan E. Bates and Tito Fojo

It has been over 20 years since the first agents targeting the Just a few years ago, it was widely expected that EGFR
epidermal growth factor receptor (EGFR) pathway were inhibitors would be effective in a wide range of solid tumors
developed by Mendelsohn and Baselga (1). In vitro studies with high levels of EGFR including breast, ovarian, lung, renal,
demonstrating that antibodies inhibiting the pathway could head and neck, pancreatic, and colorectal malignancies (9).
retard cell growth, and markedly so in some cell lines, led Thus, it was disappointing to find that response rates in phase II
ultimately to clinical trials testing EGFR inhibition as a trials in these diseases were low, as in lung cancer, or nonexis-
therapeutic strategy. As the EGFR pathway was dissected, it tent, as in renal, colon, ovarian, or breast cancer (5, 10 – 15).
was recognized that EGFR was a member of a receptor Why were our expectations so underserved? Our original
tyrosine kinase family comprising four members that dimerize paradigm—that EGFR expression could be equated with
or heterodimerize with activation (2). Diversity in dimeriza- dependence on the pathway and could predict eventual drug
tion and differences in ligand binding likely result in a sensitivity—was incorrect.
spectrum of biological phenotypes. HER2/ErbB2, the second Gefitinib first, and then erlotinib, was licensed for lung
member of the family to be identified, engenders an aggressive cancer based on results in patients with locally advanced or
subtype of breast cancer but is inhibited by the antibody metastatic non – small cell lung cancer after failure of front-line
Herceptin, generating major responses in breast cancer. A chemotherapy. For erlotinib, approval was based on a study of
decade after the introduction of antibodies inhibiting EGFR, 731 patients in which the response rate was 8.9% with a
small-molecule inhibitors were identified that impaired EGFR survival advantage of only 2 months (16). Despite the initial
tyrosine kinase activity (3). Several inhibitors have now been approval of gefitinib based on a 9% to 19% response rate in
brought to clinical trials, leading to the approval and non – small cell lung cancer, a survival advantage could not be
marketing of two of them—gefitinib and erlotinib—in lung shown in later randomized trials and the Food and Drug
cancer. Much has been written recently about these agents and Administration has since limited the scope of its approval (17).
it will not be the task of this editorial to provide a detailed Neither agent showed a survival benefit in combination with
review. chemotherapy in randomized trials. The low response rates
Previous studies, including one by the authors of this underscore the critical question of how to predict which subset
editorial, have noted that the level of expression of EGFR of patients will benefit from the targeted therapy.
cannot predict the sensitivity of cells to inhibitors (4). Bishop How then can differential responsiveness be explained?
et al.’s examination of the National Cancer Institute Drug Herceptin offered the first paradigm for successful inhibition
Screen database revealed that the 60 cell lines could be grouped of an EGFR family member: successful inhibition required
based on sensitivity to EGFR inhibition. Consistent with the significant overexpression. Immunohistochemical analysis of
notion that a threshold level of EGFR is required for sensitivity ErbB2 expression showed that a 2+ or 3+ level of expression,
to agents targeting the EGFR pathway, cells with low levels of predominantly due to amplification of the encoding gene, was
EGFR expression were found to be insensitive to quinazoline required for a meaningful response (18). This observation led
inhibitors. Moreover, cells with high levels of EGFR could be to the development of diagnostic tests for HER2 expression
divided into two groups composed of cells sensitive or and amplification and to careful selection of patients for
insensitive to EGFR inhibition. Yet, inhibition of EGFR and therapy (19).
mitogen-activated protein kinase phosphorylations could be The applicability of the HER2 paradigm to EGFR is
shown in both groups following exposure to AG1478, an uncertain. Until recently, EGFR gene amplification had been
antecedent to gefitinib and erlotinib. These and the observa- principally described in glioma as a cause of overexpression.
tions of others indicated that there were differences in the About 50% of human glioblastomas display amplification of
EGFR pathway among cell lines and an independence from EGFR and about 40% of these have amplified EGFRvIII, a
mitogen-mediated signaling was inferred as an explanation for variant receptor with ligand-independent kinase activity due to
insensitivity to EGFR inhibition. Clinical trials have confirmed deletion of the extracellular ligand-binding domain. This
these in vitro findings, demonstrating that inhibition of both mutant receptor has been shown to be resistant to gefitinib
EGFR and mitogen-activated protein kinase phosphorylations (20, 21). In a recently reported phase I trial of erlotinib in
in tumors can occur in the absence of disease response (5 – 8). glioma, eight responses were noted in 41 patients (22). Ten of
39 tumors were noted to have EGFR amplification; four
responses were noted in the group with amplification and
Authors’ Affiliation: Center for Cancer Research, National Cancer Institute, four in the group without amplification. Two of the amplified,
Bethesda, Maryland nonresponding tumors expressed mutant EGFRvIII. Evaluating
Received 8/22/05; accepted 8/22/05. phospho-Akt as a downstream marker, no tumor positive for
Requests for reprints: Susan E. Bates, Center for Cancer Research, National
Cancer Institute, Building 10, Room 12N226, Bethesda, MD 20892. Phone: 301-
phospho-Akt responded to erlotinib. Thus, gene amplification
402-1357; Fax: 301-402-1608; E-mail: sebates@ helix.nih.gov. alone is not sufficient to provoke a response to EGFR
doi:10.1158/1078-0432.CCR-05-1845 inhibitors.

www.aacrjournals.org 7203 Clin Cancer Res 2005;11(20) October 15, 2005


Editorial

Before the clinical trials with gefitinib, overexpression of 1 of the 15 tumors examined to be EGFR positive; in this
EGFR was thought to be the principal mechanism of pathway tumor, both EGFR and mitogen-activated protein kinase
activation in non – small cell lung cancer and solid tumors phosphorylations were reduced after erlotinib therapy. Skin
except for gliomas. This fueled speculation that response to samples from all 15 patients showed inhibition of phospho –
inhibitors would correlate with expression. However, muta- mitogen-activated protein kinase and Ki67 (5). None of the
tions in the EGFR tyrosine kinase domain were then tumors responded to erlotinib. Both studies concluded that
discovered among a subset of lung cancers in patients with the absence of tumor responses was due to a lack of EGFR
tumor responses following gefitinib or erlotinib (23 – 26). dependence in the tumors rather than a failure of receptor
These mutations result in a higher level of receptor phosphor- inhibition.
ylation and activation. In the initial reports, a high concor- Writing in the current issue of Clinical Cancer Research, Van
dance was found between the presence of a mutant EGFR and Schaeybroeck and colleagues offer a surrogate marker for
response to therapy, with mutations reported in 25 of 31 successful inhibition of the EGFR pathway in colon cancer cell
(81%) patients with an objective tumor response (23). By lines. The identification of a surrogate marker for sensitivity
comparison, no mutations were detected in 29 patients whose provides insight into the occasional responses—those tumors
tumors did not respond to therapy. This observation created capable of activating the pathway are those likely to be
an evolving paradigm that EGFR inhibitors might be effective sensitive to its inhibition. Van Schaeybroeck and colleagues
only in cells dependent on EGFR signaling, such as would show that the sensitivity of colon cancer cells to the EGFR
occur in cells with an activating EGFR mutation. Three inhibitor gefitinib can be discriminated by the higher basal
subsequent studies have confirmed and extended these level of EGFR autophosphorylation and by the induction of
findings (26 – 28). Further, in the studies by Taron et al. from phosphorylation that results from exposure to the chemother-
the Spanish Lung Cancer Group and by Mitsudomi et al., in apy agent oxaliplatin. These results, if confirmed clinically,
which a high response rate was documented in patients with provide both a potential surrogate for successful therapy with
tumors harboring mutations, a convincing increase in median an EGFR inhibitor and a potential insight into another
survival following gefitinib was also reported (26, 28). mechanism whereby the EGFR pathway plays a critical role
Whereas the presence of EGFR mutations in non – small cell in oncology. Beyond mutation or constitutive activation, there
lung cancer is well documented, similar EGFR mutations have might be tumors in which cellular stress such as DNA damage
not been systematically observed in other tumor types, results in activation and phosphorylation of the EGFR
potentially limiting this paradigm to non – small cell lung pathway. This activation of the pathway could result in
cancer (29). proliferation or enhanced survival so that disruption of the
The discovery of these mutations led to a paradigm shift for pathway by an EGFR inhibitor could conceivably enhance
lung cancer that did not resolve the question of whether drug sensitivity.
uncomplicated high-level or even low-level expression of EGFR Thus, we can now hypothesize rules for successful EGFR
can confer sensitivity to EGFR inhibitors. Two studies that inhibitor therapy. The gene can be amplified, mutated, or
conflict with the mutation paradigm suggest that in non – small overexpressed, but it must be critical to the oncogenic
cell lung cancer, survival following a tyrosine kinase inhibitor phenotype. The overriding paradigm seems to be that the
is enhanced in patients whose tumors contain a high copy pathway must be involved in maintenance of the malignant
number of EGFR (27, 30). What mechanisms might regulate phenotype or in cell survival to achieve a successful
EGFR expression and be important in other tumors? One antineoplastic effect. Considering the alacrity with which
example is an intron 1 polymorphism in the EGFR gene—the paradigms have been amended in recent years, this one will
number of CA single sequence repeats (31, 32). This undoubtedly be amended as well and eventually we will get it
polymorphism has been correlated with EGFR expression right. As a scientific community, we must deliver on the
levels and has been suggested as a mediator of responsiveness promise of targeted therapy. Inherent in the often-used (and
to EGFR inhibition. Without gene amplification, head and abused) term ‘‘targeted therapy’’ is the implication that those
neck cancer cells with a lower number of CA dinucleotides had who will benefit from such therapy can be identified
higher levels of EGFR and enhanced responsiveness to erlotinib prospectively. We cannot develop targeted therapy and then
(33). Although these results suggest that EGFR expression alone fail to meet the challenge of finding the subset of patients who
could create an EGFR-dependent environment that would be are likely to benefit. A 2-month survival advantage in a
susceptible to inhibition, the failure of EGFR inhibitors in population derived from a therapy administered to every
colon and renal cell carcinoma argues against such a simple patient with lung cancer cannot be considered to be
explanation. These results do, however, suggest that mutation molecularly targeted therapy. Really, how different is the latter
or gene amplification is not required to activate the EGFR from vincristine, a therapy that specifically targets tubulin and
pathway. has been available to us for nearly 50 years? Let us agree that
Two trials examining the activity of gefitinib in breast cancer by definition, a molecularly targeted therapy will be individ-
showed disappointing results without evidence of objective ualized therapy. Developing a clinically relevant assay to detect
responses (8, 15). In one of the studies, the effect of gefitinib mutations will be a first step. Developing a surrogate assay that
on the EGFR pathway was examined in both skin and tumor will detect tumors with activation of the pathway will be a
biopsies (8). Inhibition of both EGFR and mitogen-activated second step. Then we will see the day when agents such as
protein kinase phosphorylations was observed in normal and erlotinib and gefitinib will be prescribed only for patients
malignant cells. However, Ki67 staining, a marker of proli- who will benefit and the 85% who will not benefit will be
feration, was reduced in skin but not in tumors. Similarly, a spared the cost, toxicity, and disappointment of an ineffective
clinical trial evaluating erlotinib in breast cancer found only therapy.

Clin Cancer Res 2005;11(20) October 15, 2005 7204 www.aacrjournals.org


EGFR Inhibitors: A MovingTarget?

References
1. Mendelsohn J, Baselga J. The EGF receptor family 12. Dawson NA, Guo C, Zak R, et al. A phase II trial of mutations in the epidermal growth factor receptor un-
as targets for cancer therapy. Oncogene 2000;19: gefitinib (Iressa, ZD1839) in stage IV and recurrent derlying responsiveness of non-small-cell lung cancer
6550 ^ 65. renal cell carcinoma. Clin Cancer Res 2004;10: to gefitinib. N Engl J Med 2004;350:2129 ^ 39.
2. Ritter CA, Arteaga CL. The epidermal growth factor 7812 ^ 9. 24. Paez JG, Janne PA, Lee JC, et al. EGFR mutations
receptor-tyrosine kinase: a promising therapeutic tar- 13. Kindler HL, Friberg G, Skoog L, Wade-Oliver K, in lung cancer: correlation with clinical response to
get in solid tumors. Semin Oncol 2003;30:3 ^ 11. Vokes EE. Phase I/II trial of gefitinib and oxaliplatin in gefitinib therapy. Science 2004;304:1497 ^ 500.
3. Osherov N, Levitzki A. Epidermal-growth-factor- patients with advanced colorectal cancer. Am J Clin 25. Pao W, Miller V, Zakowski M, et al. EGF receptor
dependent activation of the src-family kinases. Eur J Oncol 2005;28:340 ^ 4. gene mutations are common in lung cancers from
Biochem 1994;225:1047 ^ 53. 14. Schilder RJ, Sill MW, Chen X, et al. Phase II study of ‘‘never smokers’’ and are associated with sensitivity of
4. Bishop PC, Myers T, Robey R, et al. Differential sensi- gefitinib in patients with relapsed or persistent ovarian tumors to gefitinib and erlotinib. Proc Natl Acad Sci
tivity of cancer cells to inhibitors of the epidermal or primary peritoneal carcinoma and evaluation of epi- U S A 2004;101:13306 ^ 11.
growth factor receptor family. Oncogene 2002;21: dermal growth factor receptor mutations and immuno- 26. Mitsudomi T, KosakaT, Endoh H, et al. Mutations of
119 ^ 27. histochemical expression: a Gynecologic Oncology the epidermal growth factor receptor gene predict
5. Tan AR,Yang X, Hewitt SM, et al. Evaluation of bio- Group Study. Clin Cancer Res 2005;11:5539 ^ 48. prolonged survival after gefitinib treatment in patients
logic end points and pharmacokinetics in patients with 15. von Minckwitz G, Jonat W, Fasching P, et al. A with non-small-cell lung cancer with postoperative
metastatic breast cancer after treatment with erlotinib, multicentre phase II study on gefitinib in taxane- and recurrence. J Clin Oncol 2005;23:2513 ^ 20.
an epidermal growth factor receptor tyrosine kinase anthracycline-pretreated metastatic breast cancer. 27. Cappuzzo F, Hirsch FR, Rossi E, et al. Epidermal
inhibitor. J Clin Oncol 2004;22:3080 ^ 90. Breast Cancer Res Treat 2005;89:165 ^ 72. growth factor receptor gene and protein and gefitinib
6. Daneshmand M, Parolin DA, Hirte HW, et al. A phar- 16. Cohen MH, Johnson JR, Chen YF, Sridhara R, sensitivity in non-small-cell lung cancer. J Natl Cancer
macodynamic study of the epidermal growth factor Pazdur R. FDA drug approval summary : erlotinib Inst 2005;97:643 ^ 55.
receptor tyrosine kinase inhibitor ZD1839 in meta- (Tarceva) tablets. Oncologist 2005;10:461 ^ 6. 28. Taron M, Ichinose Y, Rosell R, et al. Activating
static colorectal cancer patients. Clin Cancer Res 17. Giaccone G. Epidermal growth factor receptor inhib- mutations in the tyrosine kinase domain of the epider-
2003;9:2457 ^ 64. itors in the treatment of non-small-cell lung cancer. mal growth factor receptor are assocated with im-
7. Baselga J, Rischin D, Ranson M, et al. Phase I safety, J Clin Oncol 2005;23:3235 ^ 42. proved survival in gefitinib-treated chemorefractory
pharmacokinetic, and pharmacodynamic trial of 18. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of lung adenocarcinomas. Clin Cancer Res 11:5878 ^ 85.
ZD1839, a selective oral epidermal growth factor re- chemotherapy plus a monoclonal antibody against 29. Lee JW, SoungYH, Kim SY, et al. Absence of EGFR
ceptor tyrosine kinase inhibitor, in patients with five HER2 for metastatic breast cancer that overexpresses mutation in the kinase domain in common human can-
selected solid tumor types. J Clin Oncol 2002;20: HER2. N Engl J Med 2001;344:783 ^ 92. cers besides non-small cell lung cancer. Int J Cancer
4292 ^ 302. 19. Ross JS, Fletcher JA, Bloom KJ, et al.Targeted ther- 2005;113:510 ^ 1.
8. Baselga J, Albanell J, Ruiz A, et al. Phase II and apy in breast cancer: the HER-2/neu gene and protein. 30. Tsao MS, Sakurada A, Cutz JC, et al. Erlotinib in lung
tumor pharmacodynamic study of gefitinib in patients Mol Cell Proteomics 2004;3:379 ^ 98. cancerHmolecular and clinical predictors of outcome.
with advanced breast cancer. J Clin Oncol 2005;23: 20. Learn CA, Hartzell TL, Wikstrand CJ, et al. Resis- N Engl J Med 2005;353:133 ^ 44.
5323 ^ 33. tance to tyrosine kinase inhibition by mutant epidermal 31. Buerger H, Gebhardt F, Schmidt H, et al. Length and
9. Salomon DS, Brandt R, Ciardiello F, Normanno N. growth factor receptor variant III contributes to the loss of heterozygosity of an intron 1 polymorphic se-
Epidermal growth factor-related peptides and their neoplastic phenotype of glioblastoma multiforme. Clin quence of egfr is related to cytogenetic alterations
receptors in human malignancies. Crit Rev Oncol Cancer Res 2004;10:3216 ^ 24. and epithelial growth factor receptor expression.
Hematol 1995;19:183 ^ 232. 21. Li B, Chang CM, Yuan M, McKenna WG, Shu HK. Cancer Res 2000;60:854 ^ 7.
10. Perez-Soler R, Chachoua A, Hammond LA, et al. Resistance to small molecule inhibitors of epidermal 32. Gebhardt F, Zanker KS, Brandt B. Modulation of epi-
Determinants of tumor response and survival with growth factor receptor in malignant gliomas. Cancer dermal growth factor receptor gene transcription by a
erlotinib in patients with non-small-cell lung cancer. Res 2003;63:7443 ^ 50. polymorphic dinucleotide repeat in intron 1. J Biol
J Clin Oncol 2004;22:3238 ^ 47. 22. Haas-Kogan DA, Prados MD, Tihan T, et al. Epider- Chem 1999;274:13176 ^ 80.
11. Jermann M, Stahel RA, Salzberg M, et al. A phase II, mal growth factor receptor, protein kinase B/Akt, and 33. Amador ML, Oppenheimer D, Perea S, et al. An epi-
open-label study of gefitinib (IRESSA) in patients with glioma response to erlotinib. J Natl Cancer Inst 2005; dermal growth factor receptor intron 1 polymorphism
locally advanced, metastatic, or relapsed renal-cell 97:880 ^ 7. mediates response to epidermal growth factor recep-
carcinoma. Cancer Chemother Pharmacol 2005;1 ^ 7. 23. Lynch TJ, Bell DW, Sordella R, et al. Activating tor inhibitors. Cancer Res 2004;64:9139 ^ 43.

www.aacrjournals.org 7205 Clin Cancer Res 2005;11(20) October 15, 2005

Você também pode gostar