Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
A THERAPEUTIC EMERGENCY FOR HER-2 POSITIVE PATIENTS WHO HAVE BEEN GIVEN EXTENDED TAMOXIFEN THERAPY prepared by Michael J. Halliwell, Ph.D. California State University Long Beach CA 90840 Home address: 2930 Colorado Ave. # D-18 Santa Monica, CA 90404-3647 Home phone: 310-829-2821 Home fax: 310-828-9174 TABLE OF CONTENTS INTRODUCTION. . . . . . . . . . . . . . . . . . . . . . . . . . 1 A. Impact of HER-2 Amplification among Untreated Patients. . . 2 B. Interaction between HER-2 Status and Tamoxifen Therapy. . . 7 C. Interplay between Host Defense and Body Tumor Burden. . . . 23 D. Elevated Proliferation Rate of HER-2 Positive Tumors. . . . 26 E. Immune Surveillance Effects on Early-Stage HER-2+ Tumors. . 28 F. Adjuvant Chemotherapy Avoids HER-2 Stimulation by Tamoxifen 33 REFERENCES. . . . . . . . . . . . . . . . . . . . . . . . . . . 36 INTRODUCTION De Placido et al (1998) review the adjuvant effects of HER-2: Borg et al (1994) reported the survival in 737 patients with primary breast cancer: 445 had received tamoxifen after radical surgery. Among patients treated with adjuvant tamoxifen, survival was significantly worse for patients with cerbB2 amplified tumors (46% vs 67%, p<0.0001), while no significant difference was observed in the 292 patients treated with surgery alone (59% for c-erbB2-positive vs 66% for c-erbB2-negative; p = 0.34). Similarly, Tetu and Brisson (1994) found that c-erbB2 overexpression (determined by immunohistochemistry) played a prognostic role in a consecutive series of 888 cases of primary breast cancer, 656 of which were given some form of adjuvant therapy after surgery. . . . This suggests that the unfavorable prognostic role of c-erbB2 overexpression is relevant especially when endocrine therapy is given. A similar result has been very recently reported by Sjogren et al (1998). Id. at 148-149. After reviewing GUN-1 findings, De Placido et al continue: In summary, although these results have been obtained from either non randomized or small trials, all available data suggest that c-erbB2 overexpression is predictive of decreased tamoxifen efficacy. Id. at 150. If a drug has been found to cause bad results for a group of patients, many ethicists object to risking further use to refine such a finding. As a result data relating to such a misuse of a drug is usually too sparse to allow much clarification through use of subgroup analysis. Allred et al (1992) discuss this problem: We recognize that the process of subset analysis is fraught with difficulties. The small number of patients in many subsets reduces the power to detect significant differences, but the multiple statistical tests greatly increase the likelihood of finding a significant result by chance alone. Therefore, to interpret more confidently findings based on subset analyses, a biologic basis for the subset and the result should be proposed and the hypothesis should be confirmed in other data sets. Id. at 603. The various strands of evidence analyzed below clearly show the peril from adjuvant tamoxifen for HER-2+ patients. 1 A. Impact of HER-2 Amplification among Untreated Patients Slamon et al (1989) summarize their pioneering findings: In the current study, we collected a total of 668 human breast cancer specimens. Of these, 526 had sufficient clinical follow-up to allow for evaluation of an association between gene amplification and disease outcome. Id. at 709. We evaluated 345 patients with node-positive disease in a blinded fashion (Table 1). Of these, 101 (27%) had evidence of HER-2/neu amplification. Univariate survival analysis showed amplification of the HER-2/neu gene to be a significant predictor of both disease-free survival and overall survival for these patients (Table 1). Id. We also evaluated DNA from tumors of 181 node-negative patients with a median follow-up of 59 months (62 months for those still alive). Of these, 45 (25%) had amplification of the HER-2/neu gene. Univariate and multivariate analysis did not show an association between gene amplification and disease outcome in this group of patients. Id. Patterson et al (1989) analyze data from 158 breast cancers: The presence of HER-2/neu oncogene amplification was as good, if not better than nuclear grade and tumor size in predicting relapse in this group of retrospectively selected nodenegative patients.... However, the amplification of HER-2/neu oncogene occurs quite infrequently (about 15% of cases) in this group of retrospectively analyzed node-negative patients using microtomed sections from paraffin blocks. Costantino et al (1994) provide 5-year disease-free survival (DFS) data from NSABP Trial B-14 for tamoxifen-treated patients (78% in aneuploid tumors, 88% in diploid tumors) which suggests an overall 5-year DFS of 82%. Since 470 tamoxifen patients were evaluated for c-erbB-2, if 15% were positive, the standard error for comparing them with the whole would be 4.6% (.82 x .18/70)1/2. The reported p = 0.3 in the c-erbB-2+ test of significance is consistent with a DFS one standard error below the population value, or 77.4%. These trends are merged with NSABP B-14 data (Figure 2 2 and Table 3) from Fisher et al (1989b) in the following table: A.01 NSABP B-14 Five-Year Distant Recurrence Rates (Estrogen-Receptor Positive, Node-Negative Breast Cancer) HER-2 Negative Fail Rate TOT Rand #1 HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 5-Yr Tam 31 7.8% 399 8 11.3% 71 39 8.3% 470 Placebo 48 12.0% 399 9 12.7% 71 57 12.1% 470 Tam/Plac 0.65 0.89 0.68 Borg et al (1994) report that among 292 South Swedish breast cancer patients treated with surgery alone five-year survival was 59% for c-erbB2-positive vs 66% for c-erbB2-negative tumors with p = 0.34. Data in Figure 3 shows that the strongest trend in the control group was for PgR-positive, node-positive patients where the survival disadvantage for erbB-2+ patients is 54% vs 66% with p = 0.26. Because this report gives the group sizes and trend probabilities for the components of the control group in Figure 2, one can deduce the values for PgR-positive, node-negative patients from the overall control-group margin: A.02 South Swedish Five-Year Death Rates (Progesterone-Receptor Positive Breast Cancer) No Therapy Node-Negative Dead Rate TOT Node-Positive Dead Rate TOT Overall Dead Rate TOT HER-2 Pos. 3 21% 14 5 45% 11 8 32% 25 HER-2 Neg. 15 19% 78 17 33% 51 32 25% 129 HER+/HER- 1.11 1.36 1.28 Sjogren et al (1998) report overall survival (OAS) data for 3 Uppsala patients, mostly (84%) denied systemic adjuvant therapy: In the 202 lymph node-negative patients, statistically nonsignificant trends of shorter OAS were seen for c-erbB-2positives compared with c-erbB-2-negatives: 76% versus 88% at 5 years, respectively (P = .07; data not shown. Similar differences in OAS with regard to c-erbB-2 status were seen in the two larger treatment groups; the 94 treated with radiotherapy alone (P = .01; data not shown) and the 74 who did not receive adjuvant treatment (P = .1; data not shown). No difference was seen in relapse-free survival. Id. at 468. A.03 Uppsala Five-Year Death Rates (Mixed ER-Status, Node-Negative Breast Cancer) No Adjuvant Dead Rate TOT Radiotherapy Dead Rate TOT Overall Dead Rate TOT HER-2 Pos. 3 20% 15 4 25% 16 7 23% 31 HER-2 Neg. 7 12% 59 9 12% 78 16 12% 137 HER+/HER- 1.7 2.1 1.9 McCann et al (1991) contrast significant and insignificant trends in data from seven years of breast cancer patients treated (mostly without adjuvant therapy) at an Irish hospital: Of those patients with pathologically confirmed lymph-node (LN) status, 115 had metastases to the regional lymph nodes and 113 were LN negative. For those patients with LN+ disease, c-erbB-2+ tumors identified a poorer prognostic group (P = 0.003; Fig. 5). Overexpression of c-erbB-2 oncoprotein did not predict a worse disease outcome in LNdisease (P = 0.27). Id. at 3298. A.04 Irish Nine-Year Death Rates (Progesterone-Receptor Positive Breast Cancer) Node-Negative Dead Rate TOT Node-Positive Dead Rate TOT Overall Dead Rate TOT HER-2 Pos. 6 50% 12 19 100% 19 25 81% 31 HER-2 Neg. 35 35% 101 58 60% 96 93 47% 197 HER+/HER- 1.43 1.67 4 1.72 The node-negative results are almost entirely from patients who received no adjuvant chemotherapy or hormonal therapy. The shift from the South Swedish 1.21 adverse impact observed in fiveyear deaths for HER-2 positive tumors, to a 1.43 fold increase in deaths at nine years in the same subgroup of Irish breast cancer patients, may indicate a gradual erosion of the body's anti-tumor defenses. Liu et al (1992) note HER-2/neu findings from prior studies: In large studies, 15-40% of node-positive (stage II) cases exhibit HER2 gene amplification, which is associated with a poorer prognosis. Analysis of node-negative (stage I) breast lesions reveals a similar 10-30% incidence of HER2 amplification [6 citations].Id. at 1029-1030. Ravdin and Chamness (1995) review HER-2 status and prognosis: Examination of 11 studies of node-negative patients (Tables V and VI) suggests little clinical utility in this group. In only one of these 11 studies is there a positive finding in multivariate analysis. Even in the univariate analyses the correlations appear weak. Thus there seems to be little support for the use of ErbB-2 in node-negative patients, which is the group for whom prognostic factors are most important for making adjuvant treatment decisions. Id. at 23. The proportion of breast cancers with HER2 gene amplification is set at the midpoint of reported ranges for node-positive (27.5%) and node-negative (20%) tumors from Liu et al (1992). These tables also incorporate a small increase with time in the adverse impact of this gene and a much larger impact among nodepositive tumors (consistent with Slamon et al, 1989; Costantino et al, 1994; Borg et al, 1994; Sjogren et al, 1998; and McCann et al, 1991). 5 A.05 Composite Distant Recurrence Rates (Estrogen-Receptor Positive, Node-Negative Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 5 Years 28 13.0% 216 6 13.0% 46 34 13.0% 262 10 Years 49 22.7% 216 12 26.1% 46 61 23.3% 262 15 Years 68 31.5% 216 16 34.8% 46 84 32.1% 262 A.06 Composite Distant Recurrence Rates (ER-Negative and/or Node-Positive Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 5 Years 79 27.2% 290 37 35.6% 104 116 29.4% 394 10 Years 132 45.5% 290 63 60.6% 104 195 49.5% 394 15 Years 166 57.2% 290 79 76.0% 104 245 62.2% 394 Borg et al (1991) comment on the prognostic impact of ERBB2: The reason for failure to predict disease outcome in nodenegative breast cancer is unclear. Possibly ERBB2 is involved in cell proliferation and thus confers a growth advantage in the early stages and in local cancer. Though, to express its full potential in systemic disease, the gene may have to act in concert with other events which render the cell also capable of metastasizing. Id. at 137. It seems likely that the eventual failure of natural killer cells to harvest metastases is the key "other event" which must occur to allow distant metastases to become established. There is no doubt that breast cancer cells shed into the blood by nodenegative patients reach the lymph nodes. If failure to establish tumor colonies there were an unchanging inability to complete the metastatic process, the increasingly early detection and removal of the primary tumor on account of widespread mammographic screening, would not produce the large increase in the proportion 6 of node-negative tumors which has been observed. B. Interaction between HER-2 Status and Tamoxifen Therapy Benz et al (1992) report mouse innoculation experiments which confirm the growth sustaining effects of tamoxifen on a line of breast cancer cells (MCF/HER2-18) with a 45-fold overexpression of the HER2 receptor. When athymic nude mice were injected with five million cells of this and several other lines, daily intravenous tamoxifen caused an immediate and sustained cessation of tumor growth for the other lines, while the MCF/HER2-18 tumors showed an accelerated growth rate lasting weeks. The tripled tumor volumes for the MCF/HER2-18 line after 80 days were said by the authors to support the possibility that HER2 overexpression in human breast cancers may be linked to similar tamoxifen effects. Horwitz (1993) reports results of an experiment comparing the eight-week growth of a breast cancer cell line with and without exposure to 1 micro-mole of tamoxifen. While tamoxifen suppressed cell growth by 40%, reflecting a complete loss or decrease in the level of progesterone receptor (PgR), Horwitz notes: Our data suggest that subsets of cells may actually be stimulated by tamoxifen. . . . This subpopulation represents 5.2% of the cells in this experiment and contains an average PgR [level] greater than one million PgR molecules per cell - levels that none of the untreated cells attains. Thus tamoxifen, while decreasing PgR levels in a majority of cells, appears paradoxically to increase PR levels in a selected subset of cells. The ominous consequence of tumor cell populations that may be stimulated by tamoxifen requires little comment. Id. at 70. Perren (1991) notes the prognostic impact of HER-2 status: 7 Some papers find the prognostic effect of c-erbB-2 expression to be restricted to the node positive subset (Slamon et al, 1989; Tandon et al, 1989; Borg et al, 1990) and similar results are described by O'Reilly et al (1991) who, however, show that the independent prognostic effect of c-erbB-2 expression was restricted to recurrence free survival analysis. Other groups have made the potentially very important observation that there is a significant prognostic effect of c-erbB-2 expression in groups that would generally be considered to have a good prognosis. This includes the node negative subset where an association between c-erbB-2 expression and poor survival is described in papers by Wright et al (1989a) and Ro et al (1989). Paik et al (1990) demonstrated that the maximum prognostic effect of c-erbB-2 expression, in terms of survival, was seen in those patients who had well differentiated tumors, particularly in those who were also node negative. A similar effect was noted in a study of 79 node negative patients (Richner et al, 1990) where although there was no overall association between cerbB-2 expression and overall survival, such an association was found in the ER positive subset (P = 0.001). Id. at 330. It is not clear how c-erbB-2 expression exerts its prognostic effect and it may be simply that tumor cells positive for cerbB-2 have a growth advantage over cells negative for this oncogene. There may, however, also be some relationship between c-erbB-2 and responsiveness to treatment. Id. Ravdin and Chamness (1995) note the impact of HER-2 status: The suggestion that ErbB-2 may be more predictive of overall survival (OS) than of disease-free survival is provocative, because in general it would be expected that a prognostic factor indicating aggressive tumor behavior would at least be as significant a predictor of recurrence as of survival -indeed, because relapse occurs before death from breast cancer, the event rate for relapse would be higher especially in shorter studies, so that significance might show up earlier. In addition death (OS) is confounded in many studies by natural mortality. That the predictive value of c-erbB-2 is nevertheless greater in predicting OS, and is also greater in node-positive populations (most of whom receive adjuvant therapy), is suggestive that ErbB-2 level may be a predictor of treatment resistance. Id. at 24. Pegram et al (1998) note HER-2/neu resistance to tamoxifen: The first report of an association between HER-2/neu overexpression and failure to respond to tamoxifen therapy [for relapsing patients] was by Nicholson et al (1990). Id. at 70. 8 The first randomized clinical study with long term follow-up to demonstrate a significant interaction between HER-2/neu overexpression by immunohistochemistry and treatment response to adjuvant tamoxifen was reported by Carlomagno et al (1996).... The [GUN study] authors caution that the apparent detrimental effect of tamoxifen in the HER-2/neu positive group was based on a very small subgroup of patients, so that it cannot be definitively concluded that tamoxifen treatment worsens prognosis in HER-2/neu-positive patients. Recently, Sjogren el al (1998), reporting on data from 312 consecutive primary breast cancers, found that HER-2/neu overexpression . . . . showed a stronger correlation with survival that did ER status in node positive women treated with adjuvant tamoxifen. . . . Although these findings are considered preliminary in light of the comparatively small number of patients in the subset analysis, the results nevertheless are consistent across all of the clinical studies analyzed to date. Id. at 71. Hormonal therapy (primarily tamoxifen) given after relapse does so little to extend survival it is termed palliative. While few clinical trials have an untreated control group in such dire circumstances, the modest benefit of treatment suggests that a sharp reduction in progression-free survival is evidence of tumor stimulation by the therapy. data on the odds ratio De Laurentiis et al (2000) compile (OR) for treatment failure in HER-2 positive patients (compared to HER-2 negatives) who were given endocrine therapy to treat metastatic breast cancer: An overall estimate of the OR was computed by the MantellHaenszel method. Seven studies were included in the metanalysis (1110 patients). The overall OR was 2.46 (95% CI = 1.81-3.34). Nicholson et al (1990) discuss HER-2/neu's impact on therapy: There was a policy of first-line treatment with tamoxifen for relapsing patients without immediate life-threatening visceral disease. In 61 such patients, immunochemical assessment of neu expression was carried out. Id. at 811. 9 The correlation of neu expression with response to tamoxifen in patients with recurrent disease was assessed immunochemically. Response rate [see Table 5 infra] was reduced in the presence of neu from 50% to 17% for ER+ cases and from 26% to 0% for ER- cases. Id. Nicholson Table 5. Neu and Tamoxifen Response Neu-/ER+ Neu-/ER- Neu+/ER+ Neu+/ER- Responders 12 6 1 0 Non-Responders 12 17 5 8 Berns et al (1995) discuss their post-recurrence therapy: Noticeably in our study is the low response rate and short progression free survival (PFS) of patients with HER-2/neuamplified tumors to endocrine treatment with tamoxifen. Only two (12%) of the 16 patients with an exclusive [this is the only mutation] HER-2/neu amplification, but 17% of all 23 patients with HER-2/neu amplification (either combined or exclusively) in their tumors showed a response to endocrine therapy. Moreover, amplification of the HER-2/neu gene was related to a shorter PFS (p=0.004; Table II and Fig. 2A). Progressive disease from start of endocrine therapy at three months occurred in 79% of the patients with HER-2/neuamplified tumors and only in 47% of the patients without this amplification (Fig. 2A). This difference continued over 12 months (Table II). Id. at 15-16. Soble et al (1997) analyze treatment of 43 Chicago patients: the HER-2 gene encodes a transmembrane growth factor receptor and is overexpressed in 30% of human breast cancers. Early reports suggest that overexpression correlates with reduced effectiveness of adjuvant tamoxifen and diminished response to palliative tamoxifen. . . . Disease-free interval (DFI) for adjuvant patients and progression-free interval (PFI) for palliative patients were calculated by the Kaplan-Meier method and log-rank analyses were used to compare DFI and PFI for HER-2 positive vs negative patients. The median DFI was 67 weeks for HER-2 positive vs 204 weeks for HER-2 negative patients (p <.001). The median PFI was 13 weeks for HER-2 positive vs 62 weeks for HER-2 negative patients (p = .045). Tetu and Brisson (1994) analyze the treatment of 888 patients with node-positive breast cancer treated over a seven year period in Quebec, Canada. Cox-adjusted distant metastasis free survival 10 (DMFS) and overall survival (OS) were calculated to compensate for prognostic imbalances among those choosing the various treatments: The curves reveal that the difference between negative and positive (membrane) staining was most significant in the subgroups of patients treated with adjuvant chemotherapy and hormone therapy, whereas, the difference did not reach significance in the group of patients who received no adjuvant therapy (Fig. 4). For patients submitted to adjuvant chemotherapy or hormone therapy (Fig. 5) the 5-year DMFS for negative tumors was 57% as opposed to 35% for positive cases (P < 0.0001). Similarly the 5-year OS was 69% for negative tumors and 41% for those expressing HER-2/neu (P = 0.0001). After adjustment by multivariate analyses for other prognostic factors, the hazard ratio for occurrence of distant metastasis or death was 2.110 for those submitted to adjuvant therapy (Table 1). This means that HER-2/neu oncoprotein expression was associated with a 2.11-fold increased risk of dying or developing distant metastases during the follow-up period, compared with the absence of expression of the marker. However, surprisingly, no difference in DMFS and OS was found in the subgroup of patients treated with both chemotherapy and hormone therapy. Id. at 2362-2363. B.01 Quebec Cox-Adjusted Five-Year Distant Recurrence Rates (Node-Positive Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT CMF x 6 84 39.8% 211 25 61.0% 41 109 43.3% 252 Tamoxifen 71 42.8% 166 26 81.2% 32 97 49.0% 198 CMF + TAM 67 39.0% 172 16 47.1% 34 83 40.3% 206 No Adjuv. 101 51.5% 196 21 58.3% 36 122 52.6% 232 Total 323 43.4% 745 88 61.5% 143 411 46.3% 888 Borg et al (1991) note an important trend in their data: Furthermore, erbB-2 amplification was correlated to poor prognosis mainly in the patient group receiving adjuvant tamoxifen (data not shown). Id. at 141. Borg et al (1994) amplify findings of their earlier study: 11 We have previously reported erbB-2 amplification to be associated with poor disease-free and overall survival mainly in steroid receptor-positive breast cancer (Borg et al, 1991). In an extended study, we now demonstrate that this effect may be due to an unresponsiveness of the erbB-2 expressing receptor-positive tumors to adjuvant tamoxifen therapy, and that tamoxifen in fact may enhance the aggressiveness of those tumors. Id. at 138. Borg et al (1994) discuss the effects of adjuvant tamoxifen therapy (mostly 20 mg per day for 1 or 2 years): Analyzing the material as a whole with regard to clinical course, DFS and OS were significantly reduced in the group of patients with erbB-2+ tumors compared to the erbB-2- group (5-year OS, 50% vs 65%; P < 0.0001). If adjuvant therapy was also taken into consideration, this effect was found to be entirely confined to the subgroup of 445 patients receiving adjuvant tamoxifen (5-year OS, 46% vs 67%; P < 0.001; Fig. 1a). Id. at 139. Borg et al (1994) point out their strongest tendency: Thus when analyzing N+PgR+ patients, erbB-2 was a highly significant predictor of early recurrence and death in the group receiving adjuvant tamoxifen (5-year OS, 34% vs 73%; P < 0.0001; Fig. 3a). Id. at 141. Sjogren et al (1998) present (in Figure 3) five-year overall survival for 47 node-positive Uppsala breast cancer patients who were treated with adjuvant tamoxifen therapy. Of the ten who were HER-2 positive nine died (90%), of the 37 who were HER-2 negative ten died (27%); thus amplification of the HER-2 gene raised the death rate by a factor of 3.33. McCann et al (1991) show (in Figure 5) that among 105 Irish node-positive breast cancer patients (who were mainly treated with adjuvant tamoxifen therapy) the last of the 19 HER-2 positive patients died at the 5 1/2 year mark, at this time deaths were only 43% of the 96 HER-2 negative 12 patients (a risk ratio of 2.32). If five-year death rates for untreated patients would have been about the same (50% for HER-2 positives and 33% for HER-2 negatives) as in the South Swedish data, adjuvant tamoxifen therapy almost doubled mortality (for both studies combined) among the HER-2 positive patients and had little net impact in the HER-2 negative group. Bianco et al (2000) report fifteen years of follow-up for the node-positive premenopausal patients in GUN-2 (all of whom received 9 cycles of adjuvant CMF) where no interaction between tamoxifen therapy and HER-2 status was found. Since Carlomagno et al (1996) separate out node negative patients in the GUN-1 trial, the overall relationships reported by Bianco et al (1998) can be extrapolated from 14 to 15 years and subdivided as follows: B.02 GUN-2 Trial Fifteen-Year Distant Recurrence Rates (Premenopausal, Node-Positive Breast Cancer) HER-2 Negative Fail Rate TOT CMF x 9 HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 2-Yr Tam 26 60% 43 10 62% 16 36 61% 59 Controls 34 74% 46 13 76% 17 47 75% 63 Tam/Cont B.03 0.81 0.82 0.81 GUN-1 Trial Fifteen-Year Distant Recurrence Rates (Postmenopausal, Node-Positive Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 2-Yr Tam 25 50% 50 16 94% 17 41 61% 67 Controls 36 84% 43 12 86% 14 48 84% 57 Tam/Cont 0.60 1.10 13 0.73 B.04 GUN-1 Trial Fifteen-Year Distant Recurrence Rates (Node-Negative Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 2-Yr Tam 12 24% 50 15 75% 20 27 39% 70 Controls 32 50% 64 13 46% 28 45 49% 92 Tam/Cont 0.48 1.63 0.80 Dividing this table to fit data (shown on slides at the 1998 ASCO convention) from Bianco et al (1998) produces these trends: B.05 GUN-1 Trial Fifteen-Year Distant Recurrence Rates (Estrogen-Receptor Positive, Node-Negative Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 2-Yr Tam 5 16% 32 8 67% 12 13 30% 44 Controls 20 48% 42 8 44% 18 28 47% 60 Tam/Cont B.06 0.33 1.50 0.63 GUN-1 Trial Fifteen-Year Distant Recurrence Rates (Estrogen-Receptor Negative, Node-Negative Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 2-Yr Tam 7 39% 18 7 87% 8 14 54% 26 Controls 11 50% 22 5 50% 10 17 53% 32 Tam/Cont The 0.78 above relationships 1.75 are somewhat 1.01 conservative with respect to the impact of HER-2 status, because only 57% (245 of 433) had this status determined. Bianco et al (2000) found mortality risk ratios (tam/cont) for the GUN-1 patients of 0.54 (CI = 0.35-0.84) among HER-2 negative cases and 2.23 (CI = 0.95- 14 5.23) for the HER-2 positive group. patients (where no adjuvant The same GUN-1 categories of chemotherapy was given) are consolidated below: B.07 GUN-1 Trial Fifteen-Year Distant Recurrence Rates (Mixed ER- and Nodal-Status Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 2-Yr Tam 37 34% 100 31 84% 37 68 50% 137 Controls 68 64% 107 25 60% 42 93 62% 149 Tam/Cont 0.58 1.41 0.80 The 0.58 ratio for HER-2 negative tumors is close to the 0.54 reported by Bianco et al (2000) for these cases, not surprising for this large, statistically stable category. There is simply no room for a 2.23 ratio in the GUN-1 HER-2 positive tumors, since a 100% failure rate in the tamoxifen arm would only yield a ratio of 1.67. Carlomagno et al (1996) reports on most of the GUN-1 tumors whose HER-2 status is known, and confirms an unusually large amount of "room for failure" in the HER-2 positive arm; Figures 1 and 2 show that for untreated patients the fifteen-year mortality rate was 38% for 102 HER-2 negative patients, but only 16% for 43 HER-2 positive patients. Nordenskjold et al (1999) report a similar tendency in their analysis of data from the Swedish Breast Cancer Cooperative Group (1996) for the impact of extending tamoxifen therapy from two years to five years: "Whereas HER2-neu-negative patients showed benefit from prolonged treatment (relative risk = 0.65, 95% CI 15 0.42-1.02), no benefit was evident for HER2-neu-positive patients (relative risk = 1.9, 95% CI 0.54-6.6)." Applying these ratios to the 1996 trends for the fraction (1/8) of patients (and the experience base) with reported HER-2 data produces these tables for the 87% of such patients with a negative HER-2 rating and the 13% with tumors found to be HER-2 positive: B.08 Swedish Third-to-Tenth Year Distant Recurrence Rates (Postmenopausal, Any Nodal- or ER-Status Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 5-Yr Tam 24 19.2% 125 16 84.2% 19 40 27.8% 144 2-Yr Tam 37 30.6% 121 8 44.4% 18 45 32.4% 139 5yr/2yr 0.63 1.90 0.86 Nordenskjold et al (1999) note that if Estrogen-Receptor negative cases in the 1996 data (21%) are excluded from the analysis, the tendency reaches borderline statistical significance (p = 0.044). These tables for ER-positive patients reflect this situation (where chi-square = 4.05): B.09 Swedish Third-to-Tenth Year Recurrence Rates (Postmenopausal, ER-Positive, Mixed Nodes Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 5-Yr Tam 16 16.0% 100 11 78.6% 14 27 23.7% 114 2-Yr Tam 28 28.9% 97 6 46.2% 13 34 30.9% 110 5yr/2yr 0.55 1.70 0.77 One can use data from one of the centers (Stockholm) which contributed to the Swedish Breast Cancer Cooperative Group (1996) 16 trials, which are reported by Rutqvist et al (1987) at 261, to fill in the shared early years for the two arms of the above tables and a control group of untreated patients: B.10 Stockholm Five-Year Distant Recurrence Rates (Postmenopausal, Node-Negative, < 31 mm Breast Cancer) ER-Negative Fail Rate TOT ER-Positive Fail Rate TOT Overall Fail Rate TOT 2-Yr Tam 14 17.9% 78 32 10.3% 311 46 11.8% 389 Controls 13 17.3% 75 54 17.9% 302 67 17.8% 377 Tam/Cont B.11 1.03 0.58 0.66 Swedish Ten-Year Distant Recurrence Rates (Postmenopausal, Node-Negative Breast Cancer) ER-Negative Fail Rate TOT ER-Positive Fail Rate TOT Overall Fail Rate TOT 5-Yr Tam 28 28.3% 99 58 13.2% 439 86 15.6% 538 2-Yr Tam 25 23.6% 106 72 17.7% 407 97 18.9% 513 Controls 30 29.1% 103 97 22.9% 423 127 24.1% 526 The relationships in Table 4 of the 1996 Swedish report can be used to estimate parallel trends for node-positive patients: B.12 Swedish Ten-Year Distant Recurrence Rates (Postmenopausal, Node-Positive Breast Cancer) ER-Negative Fail Rate TOT ER-Positive Fail Rate TOT Overall Fail Rate TOT 5-Yr Tam 62 55.9% 111 161 35.1% 458 223 39.2% 569 2-Yr Tam 50 47.6% 105 222 46.2% 481 272 46.4% 586 Controls 67 62.0% 108 254 54.2% 469 321 55.6% 577 Applying the trends in Tables B.07 and B.08 to the 1996 Swedish data in Tables B.11 and B.12 produces these HER-2 effects: 17 B.13 Swedish Ten-Year Distant Recurrence Rates (Postmenopausal, ER-Positive, Node-Negative Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 5-Yr Tam 31 8.3% 373 27 40.9% 66 58 13.2% 439 2-Yr Tam 51 14.8% 345 21 33.9% 62 72 17.7% 407 Controls 81 22.6% 359 16 25.0% 64 97 22.9% 423 B.14 Swedish Ten-Year Distant Recurrence Rates (Postmenopausal, ER-Negative, Node-Negative Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 5-Yr Tam 14 16.9% 83 14 87.5% 16 28 28.3% 99 2-Yr Tam 14 15.9% 88 11 61.1% 18 25 23.6% 106 Controls 23 26.7% 86 7 41.2% 17 30 29.1% 103 B.15 Swedish Ten-Year Distant Recurrence Rates (Postmenopausal, ER-Positive, Node-Positive Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 5-Yr Tam 72 19.7% 366 89 96.7% 92 161 35.1% 458 2-Yr Tam 151 39.2% 385 71 74.0% 96 222 46.2% 481 Controls 190 50.7% 375 63 67.0% 94 254 54.2% 469 B.16 Swedish Ten-Year Distant Recurrence Rates (Postmenopausal, ER-Negative, Node-Positive Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 5-Yr Tam 38 44.2% 86 24 96.0% 25 62 55.9% 111 2-Yr Tam 31 37.8% 82 19 82.6% 23 50 47.6% 105 Controls 50 59.5% 84 17 70.8% 24 67 62.0% 108 below, the In the consolidated table 18 0.70 risk ratio (5yr/2yr) in the HER-2 negative subgroup is slightly closer to parity than the 0.63 reported by Nordenskjold because of the addition of the shared first statistics for both arms. two years of tamoxifen to the The 1.26 risk ratio in the HER-2 positive subgroup is more affected by the shared two years of tamoxifen therapy because of the tumor stimulus which it produces; at this elevated level the maximum possible ratio is 1.63, so the sizeable influence of random factors on the Nordenskjold 1.90 ratio is more likely to have made the ratio too large than too small. B.17 Swedish Ten-Year Distant Recurrence Rates (Postmenopausal, Mixed ER- and Nodal-Status Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 5-Yr Tam 174 19.2% 908 154 77.4% 199 328 29.6% 1107 2-Yr Tam 247 27.4% 900 122 61.3% 199 369 33.6% 1099 Controls 344 38.1% 904 103 51.8% 199 447 40.5% 1103 The only adjuvant studies of tamoxifen therapy where this drug's impact has not been disastrous for HER-2 positive patients (the GUN-2 trial reported by Bianco et al, 2000 and the CALGB 8541 trial reported by Berry et al, 2000) have given an extensive amount of adjuvant chemotherapy to tamoxifen patients (which must have dealt a serious blow to the fast-growing tumor cells likely to be responsible for the tamoxifen-stimulus effect). It is entirely possible that extending tamoxifen therapy for ten or fifteen years wipes out all HER-2 positive patients, however good 19 their prognosis may have been without such therapy. Since no one has reported results subdivided according to HER-2 status for such extended tamoxifen therapy, and no one is likely to institute any such trial, one can only use indirect methods to estimate the HER2 influence on ten-year tamoxifen trials such as NSABP B-14 (Fisher et al, 2001) and the fifteen-year Scottish durational trial (Stewart et al, 2001). In the following table this is done for the estrogen-positive/node-negative category, where tamoxifen therapy has been shown to be most effective, by extrapolating the disparity between two years and five years of tamoxifen therapy. B.18 Composite Ten-Year Distant Recurrence Rates (Estrogen-Receptor Positive, Node-Negative Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 10-Yr Tam 16 7.3% 219 20 42.6% 47 36 13.5% 266 5-Yr Tam 20 9.1% 219 16 34.0% 47 36 13.5% 266 2-Yr Tam 34 15.5% 219 14 29.8% 47 48 18.0% 266 Controls 49 22.7% 216 12 26.1% 46 61 23.3% 262 Since there was no stimulation from continuous tamoxifen in this category after only five years of follow-up (Table A.01), the level of stimulation from two years of tamoxifen is assumed to be less after ten years than at fifteen years (Table B.05). There is not enough HER-2 status data in the other ER/nodal categories to justify individual tables, so a composite table is derived below by subtracting the ER+/N- trends trends. 20 in Table B.18 from overall B.19 Composite Ten-Year Distant Recurrence Rates (ER-Negative and/or Node-Positive Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 10-Yr Tam 86 29.2% 295 86 81.9% 105 172 43.0% 400 5-Yr Tam 89 30.2% 295 77 73.3% 105 166 41.5% 400 2-Yr Tam 120 40.7% 295 74 70.5% 105 194 48.5% 400 Controls 148 51.0% 290 72 69.2% 104 220 55.8% 394 To estimate the impact of fifteen years of tamoxifen on HER-2 negative tumors breast which cancer, produce aggressive as continuous tamoxifen it is assumed detectable follow-up to distant continues. these that If patients the control-group metastases giving stops ten all are less years of but 1/4 (8.2%/22.2%) of distant metastases appearing in the ER+/N- control group during this decade, it is reasonable to assume that all but 1/6 of the next such failures are delayed by continuing tamoxifen for another five years. B.20 Composite Fifteen-Year Distant Recurrence Rates (Estrogen-Receptor Positive, Node-Negative Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 15-Yr Tam 20 9.1% 219 32 68.1% 47 52 19.5% 266 10-Yr Tam 26 11.9% 219 29 61.7% 47 55 20.7% 266 5-Yr Tam 33 15.1% 219 23 48.9% 47 56 21.1% 266 2-Yr Tam 52 23.7% 219 19 40.4% 47 71 26.7% 266 Controls 68 31.5% 216 16 34.8% 46 84 32.1% 262 If the GUN-1 2.23 risk ratio for two years of tamoxifen raised the control-group fifteen-year failure rate to 77.6%, the 21 Swedish 5yr/2yr risk ratio of 1.90 would easily raise the failure rate for five years of tamoxifen all the way to 100%. Where ten or fifteen years of tamoxifen therapy is given, the stimulation of the HER-2 positive tumors must account for such a large fraction of failures, that there is little reason to believe that such extended therapy is unsafe for HER-2 negative patients. In the "other" category the distant metastasis rate for HER-2 positive tumors at fifteen years is already 80% in the control group, so the rate approaches 100% with only a few years of tamoxifen therapy. B.21 Composite Fifteen-Year Distant Recurrence Rates (ER-Negative and/or Node-Positive Breast Cancer) HER-2 Negative Fail Rate TOT HER-2 Positive Fail Rate TOT Overall Fail Rate TOT 15-Yr Tam 131 44.4% 295 94 89.5% 105 225 56.2% 400 10-Yr Tam 132 44.7% 295 90 85.7% 105 222 55.5% 400 5-Yr Tam 133 45.1% 295 84 80.0% 105 217 54.2% 400 2-Yr Tam 148 50.2% 295 82 78.1% 105 230 57.5% 400 Controls 166 57.2% 290 79 76.0% 104 245 62.1% 394 Rutqvist et al (1987) report (at 261) an adverse 1.16 risk ratio for two years of tamoxifen versus controls among estrogenreceptor negative tumors at only 4 1/2 years of median follow-up (too soon for a substantial HER-2 effect). While other patient populations (such as in the NATO trial) show a substantial benefit for ER- tumors at this stage, this adverse tendency becomes strong enough with the longest durations to outweigh the benefit for 22 added therapy in the ER+/N+ segment of this consolidated category. C. Interplay between Host Defense and Body Tumor Burden Since the tamoxifen-stimulus effects documented above often reach detectable levels long after tamoxifen therapy has ended, there must be some sort of anti-tumor defense which slows down the rate of tumor growth. There are several studies which evidence such a tumor colony attrition mechanism. Veronesi et al (1990) cite findings of a Milan Tumor Institute breast-conservation trial that "local recurrences do not influence the appearance of distant metastases" and "survival in the two groups was identical (Fig. 1)." Id. at 673. al also note that NSABP Trial B-6, a Veronesi et comparison of total mastectomy with breast resection without radiotherapy by Fisher et al (1989e), "did not show any difference in the rate of distant metastases and in survival." Id. The lumpectomy operation in Trial B-6 left so much tumor tissue in the breast that Fisher (1992) notes: After more than 9 years of follow-up . . . 43% of the women treated by lumpectomy without breast irradiation . . . experienced a breast tumor recurrence. Id. at 2375. Since there is no reason to suppose that breast recurrences do not seed distant metastases, their contributions must have been swamped by the micrometastases that existed at the time of primary surgery. Obviously long-term survival from breast cancer is due to control of micrometastases by host defenses, with only rare cases actually lacking such tumor colonies. 23 (It would be a strange quirk of cancer biology if there existed a large group of node-positive patients with body tumor burdens which very soon become clinically apparent, while node-negative patients usually had few disseminated cancer cells, with relatively few cases in between.) It is likely that nodal status is such a powerful prognostic indicator because it reflects the "cleaning up" of tumor cells which reach the lymph nodes, and presumably other parts of the body. The influence on relapse rates of host defenses maintaining node-negative status, is clear in this data from Valagussa et al (1978) on 716 consecutive Milan mastectomy patients: C.01 Milan "Surgery Only" Recurrence Rates (Mixed Estrogen-Receptor and Nodal-Status Breast Cancer) 5-Yr Relapses Node- Node+ Total 10-Yr Relapses Node- Node+ Total < 2 cm 7.7% 36.8% 18.9% 19.9% 49.8% 31.5% 2-5 cm 24.3% 64.1% 45.9% 30.2% 75.9% 55.0% > 5 cm 18.7% 78.5% 55.3% 25.1% 82.4% 60.2% Total 21.0% 63.6% 43.9% 27.9% 75.5% 52.9% The much better prognosis of small tumors reflects the lesser time they have had to shed metastases into the circulatory system. Among node-positive tumors the top size category has the highest relapse rate, which dissemination potential. is what one would expect from its However, tumors larger than 5 cm which have maintained their node-negative status have only 3/4 as many 5-year relapses and only 5/6 as many 10-year relapses as those in 24 the 2-5 cm group. This cannot be because the largest tumors have produced fewer metastases, and must mean that there is some ability to eradicate tumor colonies that does not exist in the counterpart category among node-positive tumors. This ability may be limited to small tumor burdens; as Hengst and Mitchell (1987) note (at 67), "the immune response by itself is able to overcome only a limited number of tumor cells, perhaps no more than 1000 to 10,000 cells in mice [citation].” Further analysis by Retsky et al (1997) of Milan data from surgical trials (where no chemotherapy was given) also supports the existence of a host-defense effect: Recent analysis of relapse data from 1173 untreated early stage breast cancer patients with 16-20 year follow-up shows that frequency of relapse has a double peaked distribution. There is a sharp peak at 18 months, a nadir at 50 months and a broad peak at 60 months. Patients with larger tumors more frequently relapse in the first peak while those with smaller tumors relapse equally in both peaks. Id. at 193. The timing of relapses is what one would expect from a low limit on the tumor cell burden which can be contained by host defenses, where a linear rate of relapses is bottled up for a while, and then released (as when a dam bursts). Retsky et al note, "No existing theory of tumor growth predicts this effect." This lends support to the idea that this is a matter of tumor colony attrition. Measurement of this effect is provided by Hamlin (1968), who uses histological appearance to analyze tumor grade and strength 25 of host immunological response. She found that death rates at ten years among those with 211 highly malignant tumors were strongly linked to immunological response; 43.6% (24/55) of those with a very strong defense died, as did 64.8% (57/88) in the intermediate category and 94.1% (64/68) with a weak defense. Id. at 390. D. Elevated Proliferation Rate of HER-2 Positive Tumors To manifest a substantial stimulus effect which persists after the source of the stimulus is removed, a variety of tumor cells must have a high intrinsic growth potential, which manifests itself after overcome. small the body's tumor-attrition mechanisms have been While HER-2 (also c-erbB-2) status accounts for only a fraction of SPF variability, tumors oncogene have a substantially elevated SPF. positive for this Borg et al (1991) report results (at 138) for a large panel of invasive breast tumors: ERBB2 amplification was strongly correlated to an increased rate of cell proliferation, measured as a high percentage of cells in SPF [S-Phase Fraction]. This connection was seen in both diploid and non-diploid tumors, though the former group had a significantly lower mean SPF than the latter (Table 3). D.01 South Swedish S-Phase Fraction vs HER-2 Amplification (Mixed Estrogen-Receptor and Nodal-Status Breast Cancer) SPF Non-Diploid cases % Ampl SPF Diploid cases % Ampl < 7% 68 5.9% < 7% 142 9.9% 7-12% 75 13.3% > 7% 43 27.9% >12% 159 32.7% Total 302 21.9% 185 14.1% 26 Borg et al (1991) report key growth-related attributes: ERBB2 amplification was significantly correlated to the presence of axillary lymph node metastases and, within the node-positive group, also to an increased number of involved nodes. ERBB2 amplification was also significantly correlated to larger size of primary tumor, to the presence of distant metastases at diagnosis and, consequently, also to stage of disease. Id. at 138. Gusterson et al (1992) analyzed links with c-erbB-2 status for 1506 patients entered into International Breast Cancer Study Group Trial V and found significant correlations between overexpression of HER-2 protein and growth indicators such as high tumor grade (in both nodal groups) and larger tumor size (for node-negative cases). Id. at 1050-1051. Slamon et al (1987) analyzed alterations of the HER-2/neu gene in 189 primary breast cancers, finding a significantly lower degree of gene amplification for 0 to 3 positive nodes than for >3 positive nodes. Id. at 179. Slamon et al (1987) also report: A strong and highly statistically significant correlation was found between the degree of gene amplification and both time to disease relapse (P < 0.0001) and survival (P = 0.0011) (Table 4). . . . It is easy to speculate that a gene encoding a putative growth factor receptor, when expressed in inappropriate amounts, may give a growth advantage to the cells expressing it. Id. at 180-181. O'Reilly et al (1991) analyze flow cytometry findings for 153 breast cancer patients treated at Guy's Hospital between 1980 and 1983: There was a significant association between c-erbB-2 staining and an SPF above the median using Chi-squared analysis (P = 0.003). In addition, the median SPF of c-erbB-2 positive tumors was significantly higher than the median SPF of cerbB-2 negative tumors (10.5 vs 7.9, P = 0.02). Id. at 445. 27 Muss et al (1994) analyze flow cytometry data on proliferation rates for 302 primary breast cancers from CALGB trial 8869 and find (at 1262) a similarly significant association (P = 0.03). Allred et al (1992) note that higher growth rates are usually found in those most likely to receive adjuvant tamoxifen therapy: Borg et al (1990) have recently shown that in overexpression of HER-2/neu, a putative growth factor receptor is associated with high proliferation rates (flow cytometric S-Phase) in ER- positive tumors, but not ER-negative tumors, providing circumstantial evidence that HER-2/neu may be functioning as a growth factor receptor primarily in ER-positive tumors. This finding may partially explain the apparently aggressive behavior of ER-positive, node-negative lesions overexpressing HER-2/neu. Id. at 604. E. Immune Surveillance Effects on Early-Stage HER-2+ Tumors There is direct evidence of retardation of the growth of HER2 positive tumors by the body's anti-tumor defenses: Liu et al (1992) describe their HER-2/neu evaluation process: Amplification and overexpression of the HER-2 (c-erbB-2) oncogene was assessed in paraffin-embedded specimens from 27 in situ carcinomas of the breast and from 122 stage II breast cancers. . . . Coded sections of the paraffin-embedded tissues were distributed such that the immunohistochemical and molecular analyses were performed in a blinded fashion. Id. at 1027. Liu et al (1992) note HER-2/neu findings from prior studies: In large studies, 15-40% of node-positive (stage II) cases exhibit HER2 gene amplification, which is associated with a poorer prognosis. Analysis of node-negative (stage I) breast lesions reveals a similar 10-30% incidence of HER2 amplification [6 citations].Id. at 1029-1030. If one were to extrapolate this trend back to stage 0 (DCIS) tumors, one might expect to find 5-20% in the HER-2/neu+ group. 28 However, Liu et al (1992) find HER-2/neu+ is much more common in in situ tumors than in node-negative invasive tumors, and even more common than in node-positive tumors: To extend this analysis to earlier forms of breast cancer, 27 cases of in situ breast cancers without invasive disease were subjected to detailed molecular and immunohistochemical analyses. The results showed that nearly twice as many in situ carcinomas harbored increased HER2 gene copies as seen in the node-positive carcinomas (48% vs 21%, p< 0.01). Earlier studies have reported 42-61% of ductal CIS tumors with HER2 overexpression as compared with 14-21% of stage II breast cancers [5 citations]. Id. at 1030. To explore this situation further, Liu et al (1992) check on the possibility that HER2 expressiveness changes an individual patients breast cancer as it proceeds toward a more advanced stage, and found that: there was excellent concordance between the level of HER2 protein staining in the ductal CIS component and that of the invasive component in the subset of 77 node-positive tumors with coexisting in situ and invasive lesions (r = 0.86, p < 0.0001). Id. at 1028. When one compares (at 1028 and 1029) the tumors with at least some HER2 gene amplification at either end of the invasiveness continuum, it becomes apparent (chi-square = 17.3 p < 0.001) that degree of gene amplification increases with invasiveness: E.01 CALGB Levels of HER-2 Gene Amplification (Mixed Estrogen-Receptor and Nodal-Status Breast Cancer) Tumors Total Degree of Gene two-fourfold Ductal CIS 9 (69%) N+ Tumors 11 (43%) Amplification in four-eightfold 4 (31%) 10 29 (38%) HER-2/neu-Positive eightfold & more 0 (0%) 13 5 (19%) 26 Liu et al (1992) are perplexed by their findings: A widely accepted model of breast tumor progression suggests that stage II carcinomas arise from stage I carcinomas, which in turn arise from in situ carcinomas [citation]. Within this conceptual framework, the only explanation for our molecular data is that in situ lesions bearing HER2 gene amplification lose their excess gene copies as the tumor progresses from preinvasive to invasive cancer. Though the loss of an amplicon containing an oncogene has been described in vitro [citation], such a process has not been documented in vivo. . . . These findings suggest that there is considerable stability in the pattern of HER2 expression as tumors progress from in situ to invasive disease. Since HER2 correlates with the degree of gene amplification for both in situ and invasive breast cancer, we suspect that the property of HER2 gene amplification as seen in CIS lesions represents a stable genetic alteration that is maintained during the progression to invasive breast cancer. Id. at 1030-1031. One explanation for this data is that the immunoreactivity which Liu et al and other researchers use to recognize HER2 gene amplification is also used by the human body to target and destroy breast cancer cells. If this host defense is a) less effective against tumors with only normal HER2 characteristics, and b) less effective against the faster-growing HER2+ tumors with the most highly amplified oncogenes; this would account for a) the observed large reduction in the fraction of invasive tumors which are HER2+ compared to their antecedents, and b) the large increase in the degree of HER2 overexpression among invasive HER2+ tumors compared to the in situ tumors which are HER2+. Another factor in favor of this explanation is the fact that many breast cancers become quite large without overcoming the host defenses which confine them to the mammary duct system. Thus Silverstein et al (1996) give data for 425 DCIS cases, very few of which ever produce nodal or 30 distant metastases, where about one-fourth were larger than 3 cm. Allred et al (1992) discuss their immunohistochemistry study of 613 breast cancer patients with long-term clinical follow-up enrolled in Intergroup Study 0011: Several recent studies have demonstrated that HER-2/neu is amplified and/or overexpressed in 10% and 35% of human breast carcinomas, and that these manifestations of oncogene activation are associated with poor prognosis in patients with node-positive disease [12 citations]. However, studies evaluating the prognostic significance of HER-2/neu in nodenegative patients have yielded conflicting results [14 citations]. The present study was undertaken to help clarify the latter issue by studying overexpression of HER-2/neu in a large series of node-negative breast cancer patients with long-term clinical follow-up. Id. at 600. Just as human host defenses employ antibodies, Allred et al (1992) used a polyconal primary antibody (21N-SAT) to identify breast cancer cells overexpressing HER-2/neu: While all [649] tumors evaluated were composed of invasive carcinoma, they were heterogeneous with regard to the concurrent presence of noninvasive carcinoma. The rate of overexpression was higher in [237] tumors containing significant in situ carcinoma (estimated as at least 10% total tumor cellularity) as compared with [412] without a significant in situ component (estimated as < 10% tumor cellularity). These values were 21.5% and 11.2%, respectively (P <.001). Id. at 601. Allred et al (1992) analyze these node-negative breast cancers in terms of the interplay between HER-2/neu status and an in situ component (of at least 10%) where about half of high-risk patients received adjuvant chemotherapy: In this context, there were no associations between overexpression and clinical outcome in the low-risk, high-risk, or combined-risk patient groups with tumors containing significant amounts of in situ carcinoma (data not shown). Id. at 601-602. 31 In all of these circumstances host defenses are still able to exert some including tumors. restraining the enhanced influence on tumor proliferative growth, tendency of apparently HER-2/neu+ However, in the low-risk group (ER-positive tumors < 3 cm in size) Allred et al find a very different situation (where no adjuvant therapy was given): In contrast, a strong association emerged between overexpression and outcome in an analysis of low-risk patients with predominantly invasive tumors (n = 179, Fig 3), where the possible contributions from having both a significant in situ component and high risk [ER- and/or >3 cm] features were eliminated. The 5-year DFS for patients of this group with HER-2/neu-positive tumors (n = 14) was only 41%, compared with over 80% for those with HER-2/neu-negative tumors (n = 165; P < .0001 by log-rank test; P = .0002 by Peto and Peto exact test). A similar decrease was observed in the OS of low-risk patients with HER-2/neu-positive tumors composed of predominantly invasive carcinoma (P = .0001 by log-rank test; P = .0004 by Peto and Peto exact test). Id. at 602. The overall trend for the low-risk group in Figure 4 and the trend for its predominantly invasive component in Figure 3, allow the data not shown for its invasive-plus-in-situ component to be found by subtraction: E.02 Intergroup Trial 0011 Five-Year Recurrence Rates (ER-Positive, Node-Negative, < 3 cm Breast Cancer) No Therapy In Situ+Invasive Fail Rate TOT Invasive Only Fail Rate TOT Overall Fail Rate TOT HER-2 Pos. 5 19% 26 8 57% 14 13 32% 40 HER-2 Neg. 19 19% 102 31 19% 165 50 19% 267 HER+/HER- 1.00 3.00 1.68 Allred et al comment on possible explanations for these findings: The biologic basis behind our observation that overexpression 32 of HER-2/neu influences prognosis in node-negative invasive carcinomas without a significant in situ component is unknown. These lesions may represent a relatively late stage in tumor progression compared with tumors containing significant amounts of in situ carcinoma and, therefore, may be more closely related to node-positive breast cancer, where there is a strong (but poorly understood) correlation between overexpression and unfavorable prognosis. [12 citations.] Id. at 604. The lesions where there is a clear in situ component are those where host defenses can latch onto HER-2 features on cell membranes; here the prognosis is the same is for breast cancer with normal HER-2 status. The tumors which have become completely invasive are those where the body tumor burden has overwhelmed host defenses (as is the case with node-positive tumors) and like them, the prognosis of the patient tends to deteriorate. F. Adjuvant Chemotherapy Avoids HER-2 Stimulation by Tamoxifen Fortunately, beneficial for adjuvant HER-2 tamoxifen positive therapy patients, because can the be made original minority of HER-2 overexpressing tumor cells which are stimulated by tamoxifen is vulnerable to attrition by chemotherapy. Berns et al (1995) discuss their post-relapse chemotherapy: We used the Gehan-Wilcoxon test for detection of early differences for progression-free survival, and observed that patients with HER-2/neu-amplified primary tumors showed a better response to chemotherapy when compared to those having normal HER-2/neu gene copy numbers (p=0.03; Fig. 2B). The median length of tumor progression after chemotherapy was 90 days in patients whose primary tumors were not amplified, compared to 287 days in patients with HER-2/neu-amplified primary tumors, i.e., a difference of 6.5 months. Id. at 16. Due to the lack of response to endocrine therapy, chemotherapy could be considered to be offered to patients with HER-2/neu-positive tumors, whereas endocrine therapy could be preferably offered to patients with [other types of 33 breast cancer]. Id. at 17. When Bianco et al (2000) separated out the premenopausal women with node-positive tumors given 9 cycles of CMF in GUN-2 they found that two years of tamoxifen benefitted both HER-2 categories. Among HER-2 negative patients the (TAM+CMF/CMF only) mortality risk ratio was 0.76 (CI = 0.32-1.79); for HER-2 positive patients it was 0.65 (CI = 0.29-1.42). Apparently the nine months of chemotherapy given shortly after primary surgery depleted the more aggressive tumor cells in HER-2 positive tumors to such an extent that two years of tamoxifen was insufficient to create a net stimulative effect. (2000) all In a CALGB study reported by Berry et al node-positive patients received cyclophosphamide, doxorubicin, and fluorouracil shortly after primary surgery; the reduction in the risk of disease recurrence or death for 155 patients given tamoxifen was 39% with normal expression of HER2/neu and 32% with overexpression. Menard et al (2001) give 20-year data for 386 Milan patients: Bayesian analysis treatment . . . ratios were equal specific survival for HER2-positive . . . indicated a clinical benefit from CMF estimates of relapse-free survival hazard to 0.484 and 0.641 and estimates of cancer hazard ratios were equal to 0.495 and 0.730 and -negative tumors, respectively. Moliterni et al (2001) cite another Milan clinical trial: HER2 overexpression was found to predict a good response of breast carcinoma patients treated with doxorubicin (DOX). We recently put into evidence that node positive patients respond to cyclophosphamide, methotrexate and fluorouracil (CMF) regardless to HER2 status (ASCO 1999). We address now the issue of whether therapy regimens including CMF and DOX vs CMF alone, have the same therapeutic effects in patients with HER2+ and HER2- tumors, in terms of relapse free (RFS) and overall survival (OS). Methods: archival specimens of 34 primary tumors from 506 of the 552 patients (92%) enrolled in a prospective clinical trial were stained with anti HER2 monoclonal antibody CB11 and 19% of the specimens were classified as HER2+. Originally, patients were randomly allocated to receive either 12 courses of intravenous CMF or 8 courses of the same regimen followed by 4 cycles of DOX. RFS and OS were analyzed by a Cox model considering treatment, HER2 status and the interaction between treatment and HER2 status, adjusting for the effect of other known clinical and bio-pathological factors (T stage, age, PgR, ER, p53). Results: an apparent benefit from DOX was observed in HER2+ tumors. At a median follow-up of 15 years, the Hazard Ratio (HR) with 95% confidence interval (CI) for RFS was 0.83 (CI 0.46-1.49) in HER2+ tumors and 1.22 (CI 0.91-1.64 in HER2- tumors. The effect of treatment appeared more evident on OS in HER2+ patients (HR = 0.61; CI 0.32-1.16) with respect to HER2- (HR = 1.26; CI 0.89-1.79). Present data suggest that HER2+ tumors are sensitive to doxorubicin and that, in this patient subset, doxorubicin-containing regimens can improve long-term treatment outcome compared to CMF-alone chemotherapy. This advantage from the substitution of doxorubicin for onethird of the rounds of CMF did not manifest itself at all until many years after chemotherapy was completed. This is what one would expect if the stronger therapeutic agent reduced mediumsized tumor burdens to below the threshold where they could be contained by host defenses, with the advantage becoming apparent only when such cases in the other arm produced widespread metastases and death. Pegram et al (1998) observe that the bad results of tamoxifen when used to treat HER-2/neu positive tumors may be counteracted by a more powerful drug (herceptin) which is tailored to this specific mutation. Furthermore, the demonstration that HER-2/neu-associated tamoxifen resistance may be at least partially reversed by anti-HER-2/neu antibodies suggests this may be an important therapeutic strategy that warrants future study in the 35 clinic. Id. at 72. In view of the catastrophic rates of recurrence and death resulting from giving tamoxifen therapy to patients with HER-2 amplification, the only hope for many of them is probably adjuvant herceptin. One hopes that the future study envisioned in 1998 has been done by now, and that adjuvant herceptin will soon become available to this imperiled group of breast cancer patients. REFERENCES Allred DC, Clark GM, Tandon AK, Molina R, Tormey DC, Osborne CK, et al (1992): HER-2/neu in Node-Negative Breast Cancer: Prognostic Significance of Overexpression Influenced by the Presence of In Situ Carcinoma. J Clin Oncol 10:599-605. Benz CC, Scott GK, Sarup JC, Johnson RM, Tripathy D, Coronado E, et al (1992): Estrogen-Dependent, Tamoxifen-Resistant Tumorigenic Growth of MCF-7 Cells Transfected with HER2/neu. Breast Cancer Res Treat 24:85-95. Berns PMJJ, Klijn JGM, Van Staveren IL, Portengen H, Noordegraaf E, Fockens JA (1991): Frequency and Relevance of Oncogene Amplification in Human Breast Tumors (abstract). Proc Am Assoc Cancer Res 32:291. Berry DA, Muss HB, Thor AD, Dressler L, Liu ET, Broadwater G, et al (2000): HER-2/neu and p53 Expression Versus Tamoxifen Resistance in Estrogen Receptor-Positive, Node-Positive Breast Cancer. J Clin Oncol 18:3471-3479. Bianco AR, De Laurentiis M, Carlomagno C, Lauria R, Petrella G, Panico L, et al (1998): 20 Year Update of the Naples GUN Trial of Adjuvant Breast Cancer Therapy: Evidence of Interaction Between cerbB-2 Expression and Tamoxifen Efficacy (abstract 373). Proc ASCO 17:97a. Bianco AR, De Laurentiis M, Carlomagno C, Gallo C, Panico L, De Placido S (2000): HER2 Overexpression Predicts Adjuvant Tamoxifen (TAM) Failure for Early Breast Cancer (EBC): Complete Data at 20 Years of the Naples GUN Randomized Trial. (Abstract 289) Proc ASCO 19:75a. Borg A, Tandon AK, Sigurdsson H, Clark GM, Ferno M, Fuqua SAW, et al (1990): HER-2/neu Amplification Predicts Poor Prognosis in 36 Node-Positive Breast Cancer. Cancer Res 50:4332-4337. Borg A, Baldetorp B, Ferno M, Killander D, Olsson H, Sigurdsson H (1991): erbB-2 Amplification in Breast Cancer with a High Rate of Proliferation. Oncogene 6: 137-143. Borg A, Baldetorp B, Ferno M, Killander D, Olsson H, Ryden S, et al (1994): erbB-2 Amplification is Associated with Tamoxifen Resistance in Steroid-Receptor Positive Breast Cancer. Cancer Letters 81:137-144. Carlomagno C, Perrone F, Gallo C, De Laurentiis M, Lauria R, Morabito A, et al (1996): c-erbB2 Overexpression in Early-Stage Breast Cancer Without Axillary Lymph Node Metastases. J Clin Oncol 14:2702-2708. Costantino J, Fisher B, Gunduz N, Fisher E, Mamounas E, Paik S, et al (1994): Tumor Size, Ploidy, S-Phase, and erbB-2 Markers in Patients with Node-Negative, ER-Positive Tumors: Findings from NSABP B-14 (abstract 59). Proc ASCO 13:64. De Laurentiis M, Arpino G, Massarelli E, Carlomagno C, Ciardiello F, Tortora G, et al (2000): A Metanalysis of the Interaction Between HER2 and the Response to Endocrine Therapy (ET) in Metastatic Breast Cancer (MBC) (abstract 300) Proc ASCO 19: 78a. De Placido S, Carlomagno C, De Laurentiis M, and Bianco AR (1998): c-erbB2 Expression Predicts Tamoxifen Efficacy in Breast Cancer Patients. Breast Cancer Res Treat 52:55-64. Fisher B (1992): The Evolution of Paradigms for the Management of Breast Cancer: A Personal Perspective. Cancer Res 52: 2371-2383. Fisher B, Costantino J, Redmond C, et al (1989b): A Randomized Clinical Trial Evaluating Tamoxifen in the Treatment of Patients with Node-Negative Breast Cancer Who Have Estrogen-Receptor Positive Tumors. N Engl J Med 320: 479-484. Fisher B, Redmond C, Poisson R, et al (1989e): Eight-Year Results at a Randomized Clinical Trial Comparing Total Mastectomy and Lumpectomy with or without Irradiation in the Treatment of Breast Cancer. N Engl J Med 320: 822-828. Fisher B, Dignam J, Bryant J, Wolmark N (2001): Five Versus More Than Five Years of Tamoxifen for Lymph-Node Negative Breast Cancer: Updated Findings From the National Surgical Adjuvant Breast and Bowel Project B-14 Randomized Trial. J Natl Cancer Inst 93: 684-690. Gusterson BA, Gelber RD, Goldhirsch A, Price KN, Save-Soderborgh 37 J, Anbazhagan R, et al (1992): Prognostic Importance of c-erbB-2 Expression in Breast Cancer. J Clin Oncol 10:1049-1056. Hamlin IM (1968): Possible Host Resistance in Carcinoma of the Breast: a Histological Study. Br J Cancer 22: 383-401. Hengst JC and Mitchell MS (1987): Principles of Combining Biological Response Modifiers with Cancer Chemotherapy. Pp. 64-76 in Fundamentals of Cancer Chemotherapy (Hellmann K and Carter SK eds.) New York, McGraw-Hill. Horwitz KB (1993): Can Hormone "Resistant" Breast Cancer Cells Be Inappropriately Stimulated by Tamoxifen? Annals N.Y. Academy of Sciences 684:63-74. Liu E, Thor A, He M, Barcos M, Ljung BM, Benz C (1992): The HER (c-erbB-2) Oncogene is Frequently Amplified in In Situ Carcinomas of the Breast. Oncogene 7:1027-1032. McCann AH, Dervan PA, O'Reagan M, Codd MB, Gullick WJ, Brendan M, et al (1991): Prognostic Significance of c-erbB-2 and Estrogen Receptor Status in Human Breast Cancer. Cancer Res 51:3296-3303. Menard S, Valagussa P, Pilotti S, Gianni L, Biganzoli E, Boracchi P, et al (2001): Response to Cyclophosphamide, Methotrexate, and Fluorouracil in Lymph Node-Positive Breast Cancer According to HER2 Overexpression and Other Tumor Biologic Variables. J Clin Oncol 19:329-335. Moliterni A, Menard S, Valagussa P, Biganzoli E, Boracchi P, Marubini E, et al (2001): HER2 Overexpression and Doxorubicin in the Adjuvant Chemotherapy of Resectable Breast Cancer (abstract 89). Proc ASCO 20:23a Muss HB, Thor AD, Berry D, Kute T, Liu ET, Koerner F, et al (1994): c-erbB-2 Expression and Response to Adjuvant Therapy in Women with Node-Positive Early Breast Cancer. N Engl J Med 330:1260-1266. Nicholson S, Wright C, Sainsbury JR, et al (1990): Epidermal Growth Factor Receptor (EGFR) is a Marker is a Poor Prognosis in Node-Negative Breast Cancer Patients: Neu and Tamoxifen Failure. J Steroid Biochem Molecular Biol 37:811-814. Nolvadex Adjuvant Trial Organization (1988): Controlled Trial of Tamoxifen as a Single Agent in the Management of Early Breast Cancer. Br J Cancer 57:608-611. Nordenskjold B, Hatschek T, Kallstrom AC, et al (1999) Results of Prolonged Adjuvant Tamoxifen Therapy of Breast Cancer Correlated 38 to Steroid Receptor, S-Phase and ERBB2 Levels (abstract 263). Proc ASCO 18:70a. O'Reilly SM, Barnes DM, Camplejohn RS, Bartkova J, Gregory WM, Richards MA (1991): The Relationship between c-erbB-2 Expression, S-Phase Fraction and Prognosis in Breast Cancer. Br J Cancer 63:444-446. Paik S, Hazan R, Fisher ER, Sass RE, Fisher B, Redmond C, et al (1990): Pathological Findings form the National Surgical Adjuvant Breast and Bowel Project: Prognostic Significance of erbB-2 Protein Overexpression in Primary Breast Cancer. J Clin Oncol 8:103-112. Patterson AHG, Lees AW, Jamil N, Dietrich KD, Hanson J, Forney RM, et al (1989): Comparison of HER-2/neu Oncogene Amplification with Other Prognostic Factors in Node-Negative Patients with Breast Cancer (abstract 90). Proc ASCO 8:24. Pegram MD, Pauletti G, and Slamon DJ (1998): HER-2/neu as a Predictive Marker of Response to Breast Cancer Therapy. Breast Cancer Res Treat 52:65-77. Perren TJ (1991): c-erbB-2 Oncogene as a Prognostic Marker in Breast Cancer (editorial). Br J Cancer 63:328-332. Ravdin PM and Chamness GC (1995): The c-erbB-2 Proto-Oncogene as a Prognostic and Predictive Marker in Breast Cancer: a Paradigm for the Development of Other Macromolecular Markers -- a Review. Gene 159:19-27. Retsky MW, Demicheli R, Swartzendruber DE, Bame PD, Wardell RH, Bonadonna G, Speer JF and Valagussa P (1997): Computer Simulation of a Breast Cancer Metastasis Model. Breast Cancer Res Treat 45(2): 193-202. Richner J, Gerber HA, Locher GW, et al (1990): c-erbB-2 Protein Expression in Node Negative Breast Cancer. Ann Oncol 1:263. Ro J, El-Naggar A, Ro JY, Blick M, Frye D, Fraschini G, et al (1989): c-erbB-2 Amplification in Node-Negative Human Breast Cancer. Cancer Res 49:6941-6944. Rutqvist LE, Cedermark B, Glas U, et al (1987): The Stockholm Trial on Adjuvant Tamoxifen in Early Breast Cancer. Correlation between Estrogen Receptor Level and Treatment Effect. Breast Cancer Res Treat 10: 255-266. Silverstein MJ, Poller DN, Barth A, Waisman JR, Jensen JA, Masetti R, et al (1996): Intraductal Breast Carcinoma: Experiences from 39 The Breast Center in Van Nuys, California. Recent Results Cancer Res 140:139-153. Sjogren S, Inganas M, Lindgren A, Holmberg L, and Bergh J (1998): Prognostic and Predictive Value of c-erbB-2 Overexpression in Primary Breast Cancer, Alone and in Combination with Other Prognostic Markers. J Clin Oncol 16:462-469. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL (1987): Human Breast Cancer: Correlation of Relapse and Survival with Amplification of the HER-2/neu Oncogene. Science 235:177-182. Slamon DJ, Gondolphin W, Jones LA, Holt JA, Wong SG, Keith DE, et al (1989): Studies of the HER-2/neu Proto-oncogene in Human Breast and Ovarian Cancer. Science 244:707-712. Soble MB, Oleske DM, Cobleigh MA (1997): HER-2/c-erbB2 (HER-2) Overexpression is a Biomarker of Tamoxifen Resistance in Breast Cancer (abstract 493). Proc ASCO 16:140a. Swedish Breast Cancer Cooperative Group (1996): Randomized Trial of Two Versus Five Years of Adjuvant Tamoxifen for Postmenopausal Early Stage Breast Cancer. J Natl Cancer Inst 88:1543-1549. Stewart HJ, Prescott RJ, Forrest APM (2001): Scottish Adjuvant Tamoxifen Trial: a Randomized Study Updated to 15 Years. J Natl Cancer Inst 93: 456-462. Tandon AK, Clark GM, Chamness GC, Ulrich A, McGuire WL (1989): HER-2/neu Oncogene Protein and Prognosis in Breast Cancer. J Clin Oncol 7: 1120-1128. Tetu B and Brisson J (1994): Prognostic Significance of HER-2/neu Oncoprotein Expression in Node-Positive Breast Cancer: The Influence of the Pattern of Immunostaining and Adjuvant Therapy. Cancer 73:2359-2365. Valagussa P, Bonadonna G and Veronesi U (1978): Patterns of Relapse and Survival Following Radical Mastectomy: Analysis of 716 Consecutive Patients. Cancer 41: 1170-1178. Veronesi U, Volterriani F, Luini A, et al (1990): Quandrantectomy versus Lumpectomy for Small Size Breast Cancer. Eur J Cancer 26: 671-673. Wright C, Nicholson S, Angus B, Cairns J, Sainsbury JRC, Gullick WJ, et al (1989a): Association of c-erbB-2 Oncoprotein Expression with Lack of Response to Endocrine Therapy in Recurrent Breast Cancer (abstract). J Pathol 158:350A. 40