In this issue of Clinical Cancer Research, Buzdar et al. (1) report on the success of trastuzumab, a humanized monoclonal antibody against the proto-oncogene HER2 (c-erbB2, HER-2/neu), when administered with 5-fluorouracil, epirubicin, and cyclophosphamide (FEC) and paclitaxel chemotherapy, with high pathologic complete remission rates. These results are consistent with recent data presented by large multicenter trials, confirming the high efficacy of trastuzumab in women with early stage breast cancer.

The epidermal growth factor receptor/HER family of transmembrane type I receptor tyrosine kinases are enzymes that play an important role in fundamental processes like cell proliferation, differentiation, and survival. The ectodomains of HER1, HER3, and HER4 interact with specific sets of ligands, whereas no natural ligand has been identified thus far for HER2. However, HER2 can be activated by heterodimerization with other ligand-activated HER coreceptors. On ligand binding to the active domain of HER1, HER3, or HER4, these receptors preferentially recruit HER2 into a heterodimeric complex in which the HER2 kinase can modulate receptor internalization and prolong signal transduction. On dimerization, conformational changes lead to autophosphorylation and initiation of divergent signal transduction cascades (2). These type I receptors signal through the Ras/Raf/mitogen-activated protein kinase/extracellular signal-regulated kinase pathway, stimulating cell division (3). Cell line evidence also suggests that the type I receptors modulate cell survival through activation of the Akt/phosphoinositol 3-kinase pathway (ref. 4; Fig. 1). Aberrant HER1 and HER2 signaling has been causally associated with cancer cell proliferation and survival.

Fig. 1.

Simplified schema of signaling through Erb receptor on dimerization. Conformational changes lead to autophosphorylation and initiation of divergent signal transduction cascades. The type I receptors signal through the Ras/Raf/mitogen-activated protein kinase (MEK)/extracellular signal–regulated kinase (ERK) pathway, stimulating cell division. Cell line evidence also suggests that the type I receptors modulate cell survival through activation of the Akt/phosphoinositol 3-kinase (PI3-kinase) pathway.

Fig. 1.

Simplified schema of signaling through Erb receptor on dimerization. Conformational changes lead to autophosphorylation and initiation of divergent signal transduction cascades. The type I receptors signal through the Ras/Raf/mitogen-activated protein kinase (MEK)/extracellular signal–regulated kinase (ERK) pathway, stimulating cell division. Cell line evidence also suggests that the type I receptors modulate cell survival through activation of the Akt/phosphoinositol 3-kinase (PI3-kinase) pathway.

Close modal

HER-2 was first identified as an oncogene activated by a point mutation in chemically-induced rat neuroblastomas (5). It encodes a 185-kDa transmembrane protein that is a putative growth factor receptor of the tyrosine kinase family. It was later found to be overexpressed in some human breast carcinomas (6) present on the surface of 20% to 25% of breast cancer cells. The prognosis of those patients whose tumors overexpress HER-2 is poor (714). Based on this association between the members of HER1/HER2 family and worse clinical outcome, antibodies and small molecules that specifically target these receptor tyrosine kinases were developed for their therapeutic efficacy.

In the 1980s, a monoclonal antibody against HER-2, trastuzumab, was developed, and in 1998, it was approved for the treatment of metastatic breast cancer (15). In 2005, the results of five adjuvant trials evaluating trastuzumab, involving >10,000 women, were presented (1618). Despite differences in study design and short follow-ups of only 1 to 2 years, these studies show the same remarkable results—adjuvant trastuzumab therapy halves the recurrence rate and reduces mortality by 30%. This benefit is, on average, higher than that of adjuvant chemotherapy and similar to that seen with adjuvant hormonal therapy. The main setback of trastuzumab is its potential for cardiotoxicity, although benefits seem to outweigh risks and the ensuing congestive heart failure is generally reversible. Today, the evaluation of HER-2 expression should therefore be mandatory in early breast cancer patients who should be offered access to this highly effective therapy.

The antitumor effects of HER2 inhibitors require the modulation of key signaling pathways and cell cycle/apoptosis regulatory molecules that mediate the transforming effects of HER2. These pathways may involve heterologous receptor networks and/or heterodimers of the HER (ErbB) family that are not affected by trastuzumab (19). For example, high expression of epidermal growth factor receptor and ligands for the Erb family predicts early escape from trastuzumab therapy (20). Another important reported mechanism of escape, and therefore resistance, is overexpression of the insulin-like growth factor-I receptor (21), a potent inducer of phosphoinositol 3-kinase and Akt. Amplification of the phosphoinositol 3-kinase pathway as a result of loss or low levels of phosphatase and tensin homologue is also associated with resistance to trastuzumab (21). These data are highly consistent with in vivo studies of human primary breast cancers that blocking activation of the phosphoinositol 3-kinase/Akt survival pathway is the main mechanism of action of trastuzumab (22). Similarly, patients with coamplification of cMYC and HER-2 had worse outcome when treated with chemotherapy alone, whereas the addition of trastuzumab reversed this trend with patients achieving high recurrence-free survival. These data suggest that the proapoptotic function of dysregulated cMYC may be counterbalanced by an antiapoptotic signal from another activated oncogene, like HER2 (23). Based on these data, approaches combining trastuzumab with therapeutic agents attacking targets that lead to resistance (e.g., insulin-like growth factor-I receptor inhibitors) are being tested. In addition, somatic mutations in the HER2 gene have recently been reported in ∼4% of non–small-cell-lung cancers (24) and may represent yet another route of escape from trastuzumab therapy. Gene expression and other high throughput studies using biopsies from women undergoing neoadjuvant trastuzumab-containing regimens may shed some light on patients who might not benefit from therapy (25, 26). Early preliminary data presented in the article by Budzar et al. failed to identify patterns to discriminate treatment benefit in this study.

To circumvent trastuzumab resistance, another approach is to block HER2 function with ATP-competitive tyrosine kinase inhibitors. For example, lapatinib is a reversible dual kinase inhibitor against the epidermal growth factor receptor and HER2 (27), which has shown antitumor activity both in vitro and in vivo (28, 29). It has recently shown remarkable activity in patients with HER2-overexpressing breast cancers when given either as first line therapy or after escape from trastuzumab (3032). The addition of lapatinib induces tumor regressions in up to 20% of patients who have progressed previously on trastuzumab alone (33). Recent data presented at the annual meeting of the American Society of Clinical Oncology in 2006 compared capecitabine with or without the addition of lapatinib. The lapatinib arm significantly delayed the progression of breast cancer for nearly twice as long compared with capecitabine alone in patients with advanced breast cancer who had progressed following treatment with trastuzumab, with a median time to disease progression of 8.5 months, compared with 4.5 months for those treated with capecitabine alone. Thus, lapatinib will likely be approved for use in patients with HER2-overexpressing breast cancer.

Trastuzumab has been an unfettered success. Nevertheless, many questions remain, some highlighted by Budzar et al. The optimal duration of trastuzumab in the adjuvant setting, generally administered for at least 52 weeks, remains to be defined. The two studies in early stage breast cancer, Budzar et al.'s and the FinHer (Finland Herceptin) trial (16), show comparable favorable results with trastuzumab given concurrently with chemotherapy for 24 weeks or less. This shorter duration of therapy would be consistent with the body of data suggesting that apoptosis by blocking the phosphoinositol 3-kinase/Akt survival pathway is a major mechanism of action of trastuzumab (22, 23). The current Herceptin Adjuvant (HERA) trial comparing 1 versus 2 years of trastuzumab adjuvant therapy will provide pivotal information on the value of prolonged trastuzumab, and whether shorter durations of trastuzumab therapy should be tested.

The article by Budzar et al. highlights many unanswered questions on cardiotoxicity. It is accepted that the benefits of trastuzumab therapy far outweigh the generally reversible risk of congestive heart failure. Preclinical data suggest a powerful synergistic interaction between trastuzumab and both platinum and docetaxel. In early trials, platinum-taxane-trastuzumab combinations have exhibited promising clinical activity. The potential for cardiac toxicity when trastuzumab is combined with the anthracyclines suggests a further rationale for the development of non-anthracycline regimens, especially in the adjuvant setting. Molecular markers, like the expression of TOPO2A, may have promise in selecting patients who may be spared anthracycline-containing trastuzumab therapy (34), which remains an important goal. Newer therapeutic agents targeting neovascularization, pan-HER2 inhibition, insulin-like growth factor receptor pathways, and the like, will invariably build on the success of HER2-targeted therapy.

1
Buzdar AU, Valero VV, Ibrahim NK, et al. Neoadjuvant therapy with paclitaxel followed by 5-fluorouracil, epirubicin, and cyclophosphamide chemotherapy and concurrent trastuzumab in human epidermal growth factor receptor 2-positive operable breast cancer: an update of the initial randomized study population and data of additional patients treated with the same regimen.
Clin Cancer Res
2007
;
13
:
228
–33.
2
Riese DJ II, Stern DF. Specificity within the EGF family/ErbB receptor family signaling network.
BioEssays
1998
;
20
:
41
–8.
3
Amundadottir LT, Leder P. Signal transduction pathways activated and required for mammary carcinogenesis in response to specific oncogenes.
Oncogene
1998
;
16
:
737
–46.
4
Okano J, Gaslightwala I, Birnbaum MJ, Rustgi AK, Nakagawa H. Akt/protein kinase B isoforms are differentially regulated by epidermal growth factor stimulation.
J Biol Chem
2000
;
275
:
30934
–42.
5
Schechter AL, Stern DF, Vaidyanathan L, et al. The neu oncogene: an erb-B-related gene encoding a 185,000-Mr tumour antigen.
Nature
1984
;
312
:
513
–6.
6
King CR, Kraus MH, Aaronson SA. Amplification of a novel v-erbB-related gene in a human mammary carcinoma.
Science
1985
;
229
:
974
–6.
7
Walker RA, Gullick WJ, Varley JM. An evaluation of immunoreactivity for c-erbB-2 protein as a marker of poor short-term prognosis in breast cancer.
Br J Cancer
1989
;
60
:
426
.
8
Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene.
Science
1987
;
235
:
177
–82.
9
Wright C, Angus B, Nicholson S, et al. Expression of c-erbB-2 oncoprotein: a prognostic indicator in human breast cancer.
Cancer Res
1989
;
49
:
2087
–90.
10
Lovekin C, Ellis IO, Locker A, et al. c-erbB-2 oncoprotein expression in primary and advanced breast cancer.
Br J Cancer
1991
;
63
:
439
–43.
11
Gullick WJ, Love SB, Wright C, et al. c-erbB-2 protein overexpression in breast cancer is a risk factor in patients with involved and uninvolved lymph nodes.
Br J Cancer
1991
;
63
:
434
–8.
12
Varley JM, Swallow JE, Brammar WJ, Whittaker JL, Walker RA. Alterations to either c-erbB-2(neu) or c-myc proto-oncogenes in breast carcinomas correlate with poor short-term prognosis.
Oncogene
1987
;
1
:
423
–30.
13
May E, Mouriesse H, May-Levin F, Qian JF, May P, Delarue JC. Human breast cancer: identification of populations with a high risk of early relapse in relation to both oestrogen receptor status and c-erbB-2 overexpression.
Br J Cancer
1990
;
62
:
430
–5.
14
Tsuda H, Hirohashi S, Shimosato Y, et al. Correlation between long-term survival in breast cancer patients and amplification of two putative oncogene-coamplification units: hst-1/int-2 and c-erbB-2/ear-1.
Cancer Res
1989
;
49
:
3104
–8.
15
Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2.
N Engl J Med
2001
;
344
:
783
–92.
16
Joensuu H, Kellokumpu-Lehtinen PL, Bono P, et al. Adjuvant docetaxel or vinorelbine with or without trastuzumab for breast cancer.
N Engl J Med
2006
;
354
:
809
–20.
17
Romond EH, Perez EA, Bryant J, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer.
N Engl J Med
2005
;
353
:
1673
–84.
18
Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer.
N Engl J Med
2005
;
353
:
1659
–72.
19
Cho HS, Mason K, Ramyar KX, et al. Structure of the extracellular region of HER2 alone and in complex with the Herceptin Fab.
Nature
2003
;
421
:
756
–60.
20
Smith KL, Robbins PD, Dawkins HJS, et al. Detection of c-erbB-2 amplification in breast cancer by in situ hybridization.
Breast
1993
;
2
:
234
–8.
21
Nahta R, Yuan LX, Zhang B, Kobayashi R, Esteva FJ. Insulin-like growth factor-I receptor/human epidermal growth factor receptor 2 heterodimerization contributes to trastuzumab resistance of breast cancer cells.
Cancer Res
2005
;
65
:
11118
–28.
22
Mohsin SK, Weiss HL, Gutierrez MC, et al. Neoadjuvant trastuzumab induces apoptosis in primary breast cancers.
J Clin Oncol
2005
;
23
:
2460
–8.
23
Kim C, Bryant J, Horne Z, et al. Trastuzumab sensitivity of breast cancer with co-amplification of HER2 and cMYC suggests pro-apoptotic function of dysregulated cMYC in vivo.
Breast Cancer Res Treat
2005
;
94
:
S6
(abstract 46).
24
Shigematsu H, Gazdar AF. Somatic mutations of epidermal growth factor receptor signaling pathway in lung cancers.
Int J Cancer
2006
;
118
:
257
–62.
25
Chang JC, Wooten EC, Tsimelzon A, et al. Gene expression profiling predicts therapeutic response to docetaxel (Taxotere(tm)) in breast cancer patients.
Lancet
2003
;
362
:
280
–7.
26
Ayers M, Symmans WF, Stec J, et al. Gene expression profiles predict complete pathologic response to neoadjuvant paclitaxel and fluorouracil, doxorubicin, and cyclophosphamide chemotherapy in breast cancer.
J Clin Oncol
2004
;
22
:
2284
–93.
27
Rusnak DW, Affleck K, Cockerill SG, et al. The characterization of novel, dual ErbB-2/EGFR, tyrosine kinase inhibitors: potential therapy for cancer.
Cancer Res
2001
;
61
:
7196
–203.
28
Spector N, Xia W, Burris HA, et al. Modulation of tumor growth and survival pathways in cancer patients treated with GW572016.
Proc Am Soc Clin Onco
2004
;
22
:
3003
.
29
Konecny G, Thomssen C, Luck HJ, et al. HER-2/neu gene amplification and response to paclitaxel in patients with metastatic breast cancer.
J Natl Cancer Inst
2004
;
96
:
1141
–51.
30
DeSimone PA, Bence AK, Anderson EB, et al. A phase I study to investigate the safety, tolerability, and pharmacokinetics of single oral escalating doses of GW572016 in healthy volunteers.
38th Annual ASCO Program Proceedings
2002
;
21
:
94a
.
31
Blackwell K, Kaplan E, Franco S, Marcom J. A phase II, open-label, multicenter study of GW572016 in patients with trastuzumab-refractory metastatic breast acncer [abstract 3006].
Proc Am Soc Clin Oncol
2004
;
23
:
196
.
32
Dees EC, Burris H, Hurwitz H, et al. Clinical summary of 67 heavily pre-treated patients with metastatic carcinomas treated with GW572016 in a phase Ib study.
Proc Am Soc Clin Oncol
2004
;
22
:
3188
.
33
Storniolo ABH, Pegram M, Overmoyer B, et al. A phase I, open-label study of lapatinib (GW572016) plus trastuzumab; a clinically active regimen.
Proc Am Soc Clin Oncol
2005
;
23 No. 16S
:
559
.
34
Press MF, Bernstein L, Sauter G, et al. Topoisomerase II-α gene amplification as a predictor of responsiveness to anthracycline-containing chemotherapy in the Cancer International Research Group 006 clinical trial of trastuzumab (Herceptin) in the adjuvant setting.
Breast Cancer Res Treat
2005
;
94
:
S54
(abstract 46).