throbber
Endocrine-Related Cancer (2002) 9 75—85
`
`The development and clinical use of
`trastuzumab (Herceptin)
`
`M Harries and I Smith
`
`Breast Unit, The Royal Marsden Hospital and Institute of Cancer Research, Fulham Rd, London SW3 6JJ, UK
`
`(Requests for offprints should be addressed to M Harries; Email: mark.harries@rmh.nthames.nhs.uk)
`
`Abstract
`
`HER-2 is a member of the c-erbB family of receptor tyrosine kinases and is overexpressed by 20—
`30% of human breast cancers. HER-2 overexpression is an independent adverse prognostic factor
`and may also predict for response to both chemotherapy and endocrine agents. Trastuzumab is a
`humanised monoclonal antibody that binds with high affinity to the extracellular domain of HER-2. In
`HER-2—overexpressing preclinical models trastuzumab has been shown to have a marked
`antiproliferative effect and demonstrates synergy with a number of cytotoxic drugs. Several phase II
`and phase III clinical trials have now been performed in patients with advanced breast cancer that
`overexpress HER-2. Trastuzumab was initially shown to be active and well tolerated as a single agent
`in heavily pretreated women. Subsequently, studies of first-line treatment for metastatic breast cancer
`have demonstrated an improvement in survival for trastuzumab when used in combination with either
`paclitaxel or an anthracycline—cyclophosphamide regimen compared with chemotherapy alone.
`Unexpectedly,
`the combination of
`trastuzumab and the anthracycline-containing regimen was
`associated with a significant incidence of cardiac dysfunction. The benefit of trastuzumab is generally
`confined to patients whose tumours have gene amplification as detected by fluorescence in situ
`hybridisation (FISH) and this is tightly associated with immunohistochemical (IHC) staining at the
`highest (3+) level. A small number of patients have IHC 2+ tumours together with FISH evidence
`of gene amplification and may also derive benefit from treatment. Trastuzumab has also been shown
`to be effective when used as first-line monotherapy for advanced breast cancer. Trials to date have
`employed trastuzumab in a weekly schedule, but there is emerging evidence that a three-weekly
`regimen may be as effective. Trastuzumab has shown encouraging activity when used with other
`agents including docetaxel and vinorelbine. The combination of trastuzumab, docetaxel, and platinum
`salts also appears to be very active. The role of trastuzumab as adjuvant therapy for early breast
`cancer is being tested in a number of large randomised trials.
`
`Endocrine-Related Cancer (2002) 9 75—85
`
`Introduction
`
`and an assessment of current clinical trials to define further
`
`the background to
`the optimal usage of this agent. First,
`the discovery of the HER-2 target and the preclinical vali-
`dation of antibody therapies directed against
`it will be
`
`presented.
`
`In recent years a number of the molecular pathways respon-
`sible for tumorigenesis have been identified (Hanahan &
`Weinberg 2000). With this knowledge has come the possibil-
`_
`_
`_
`_
`1ty of effectlve anti-cancer treatments a1med at novel targets
`with few of the toxicities associated with conventional cyto-
`toxic therapies. This review focuses on the development and BaCKground
`use of trastuzumab (Herceptin), a humanised monoclonal
`HER-2 is a member of the erbB epidermal growth factor
`antibody specific for HER-2, a growth factor receptor found
`receptor tyrosine kinase family. In the early 1980s the erbB
`to be overexpressed in 20—30% of human breast cancers
`receptor tyrosine kinases became implicated in cancer when
`(Slamon et al. 1987, 1989). The clinical data that have led
`it was found that the avian erythroblastosis tumour Virus
`encoded an oncogene that was highly homologous to the
`human epidermal growth factor receptor (HER-1, also known
`as ErbBl and EGFR). Subsequently a gene called new was
`
`to the recent licensing of trastuzumab as a treatment for
`metastatic breast cancer will be presented together with a
`discussion of the controversial areas that surround its use
`
`Endocrine-Related Cancer (2002) 9 75—85
`1351—0088/02/009—075 © 2002 Society for Endocrinology Printed in Great Britain
`
`rg
`Online version via http://ngendpcrinolog
`enentech
`elltrion v.
`IPR2017—01122
`
`Genentech Exhibit 2016
`
`

`

`Harries and Smith: Trastuzumab
`
`identified from a chemically induced rat neuroblastoma that
`was able to transform fibroblast cell lines in culture and was
`
`shown to be related to but distinct from the HER—1 gene
`(Shih et al. 1981, Schechter et al. 1984). At about the same
`
`time two other groups independently isolated human erbB-
`related proto-oncogenes and named them HER-2 (Coussens
`et al. 1985) and c-erbB2 (Semba et al. 1985). These genes
`were then shown to be the same as new. King and colleagues
`also identified an EGFR-related gene that was overamplified
`in a human mammary carcinoma cell line; this gene was also
`found to be identical to the HER-2/neu/erbB2 gene (King et
`al. 1985).
`HER-1 and HER-2 differ in a number of ways: the HER-2
`gene is located on chromosome 17 whereas the HER-1 gene
`has been mapped to chromosome 7, and the HER-2 mRNA and
`protein are of different sizes from the HER-1 gene products.
`The erbB receptor tyrosine kinase family has two other mem-
`bers, HER-3 and HER-4 (erbB3 and erbB4), with the four
`receptors sharing an overall membrane spanning structure
`composed of extracellular and transmembrane components
`together with an intracellular region containing a kinase
`domain flanked by tyrosine autophosphorylation sites. There
`are a number of functional differences between the domains of
`
`the different family members. For example, HER-2 appears to
`have no direct ligand and HER-3 has no intrinsic kinase
`activity and therefore a number of complex interactions
`between the different family members involving dimerisation
`are required for signalling. The HER-2 receptor can signal by
`forming heterodimers with other members of the HER family
`that are bound to a ligand, or two HER-2 molecules can com-
`bine to form a homodimer which has intrinsic kinase activity.
`Overexpression of HER-2 favours the production of both acti-
`vated homo- and hetero-dimers. erbB receptor kinase acti-
`vation recruits a number of adaptor proteins to the cytoplasmic
`domains which in turn trigger a number of downstream signal-
`ling cascades. The end results of HER-2 activation are effects
`on cell growth, division, differentiation, migration and
`adhesion (reviewed in Yarden & Sliwkowski 2001).
`Slamon and colleagues initially reported that the HER-2
`receptor was overexpressed in 20—30% of human breast can-
`cers (Slamon et al. 1987). In the vast majority of cases over-
`expression is caused by amplification of the HER-2 gene
`(Pauletti et al. 1996). HER-2 gene amplification results in
`increased levels of mRNA as detected by Northern blot and
`of the HER-2 receptor as detected by imrnunohistochemistry
`(IHC) or Western blot analysis. Over amplification of the
`gene is most strikingly seen using fluorescence in situ hybrid-
`isation (FISH), when multiple copies of the HER-2 gene can
`be seen in the nuclei of affected cells. This technique has
`become a useful method of detecting HER-2 gene amplifica-
`tion in clinical samples.
`HER-2 gene amplification is seen early in the develop-
`ment of invasive breast carcinoma and can also be seen in
`
`ductal carcinoma in situ (Barnes et al. 1992). Overexpression
`
`of HER-2 in breast cancer cells correlates with a number
`
`of well recognised adverse histological prognostic features
`including tumour size, high grade, a high percentage of
`S-phase cells, aneuploidy and lack of steroid receptors
`(Slamon et al. 1987, Ross & Fletcher 1999). Overexpression
`of HER-2 correlates with poor breast cancer prognosis. In
`1987, Slamon and colleagues examined tumour samples from
`86 node-positive patients and found that overamplification of
`the HER-2 gene was a strong independent adverse prognostic
`factor (Slamon et al. 1987). In 1989, Slamon and co-workers
`
`collected a total of 668 human breast cancer specimens, 526
`of which had sufficient clinical information to examine a link
`
`between HER-2 expression and outcome. Of 345 node-
`positive patients, 27% had HER-2 gene amplification. This
`was found to be a significant predictor of disease-free and
`overall survival in multivariate analysis (P = 0.006 and 0.045
`respectively) and was superior to all other prognostic factors
`except nodal status. In this initial study there was no associa-
`tion found between gene amplification and disease outcome
`in the 181 node-negative patients (Slamon et al. 1989). In a
`subsequent retrospective series of 1506 patients from the
`Ludwig International Breast Cancer Study Group, HER-2
`overexpression was again found to be prognostically signifi-
`cant in node-positive but not in node-negative patients with
`breast cancer (Gusterson et al. 1992). However, more recent
`
`studies have demonstrated that HER-2 gene amplification is
`an independent prognostic factor even in node-negative
`patients (Seshadri et al. 1993, Press et al. 1997 and reviewed
`in Ross & Fletcher 1998).
`
`Current evidence suggests that there may be an associa-
`tion between the overexpression of HER-2 and response to
`therapy. For example, in endocrine therapy a recent neoadju-
`vant trial has suggested a significantly higher response rate
`to letrozole than to tamoxifen in a small subset of patients
`with both HER-l- and/or HER-2-positive and oestrogen
`receptor (ER)-positive cancers (15/17 88% compared with
`4/19 21%, odds ratio 28, P = 0.004) (Ellis et al. 2001). Like-
`
`that
`in chemotherapy circumstantial data suggest
`wise,
`anthracyclines may be more effective than cyclophospham—
`ide, methotrexate and 5-fluorouracil (CMF) schedules for
`
`patients with HER-2-overexpressing breast cancer. This com-
`plex area has recently been reviewed by Piccart et al. (2000).
`Although the evidence is intriguing, there are currently
`insufficient data to suggest that HER-2 status be used to
`select therapy for patients other than their suitability for
`HER-2-directed therapies. In 2000, the American Society of
`Clinical Oncology (ASCO) committee on the update of re-
`commendations for the use of tumour markers in breast and
`
`colorectal cancer stated that ‘levels of c-erbB2 expression
`should not be used to exclude patients from anthracycline
`treatment’ and ‘the use of c-erbB2 data to decide whether to
`
`prescribe endocrine therapy either in the adjuvant or meta-
`static setting is not recommended’ (Bast et al. 2001).
`
`76
`
`www.endocrinology.org
`
`

`

`Development of trastuzumab
`
`The discovery of HER-2 overexpression in a significant min-
`ority of human breast cancers and its adverse prognostic sig-
`nificance prompted investigators to develop agents using
`HER-2 as a target for treatments. Several groups including
`workers at Genentech Inc. raised murine monoclonal anti-
`bodies to the extracellular domain of HER-2 and showed that
`
`some of these antibodies were capable of inhibiting the
`growth of cell lines that overexpressed the receptor (Hudziak
`et al. 1989, Fendly et al. 1990). This effect was also seen in
`HER-2-overexpressing human breast
`cancer xenografts
`where the effects of the antibody were found to be synergistic
`to anti-neoplastic agents such as cisplatin (Pietras et al.
`1994).
`
`The Genentech researchers developed a panel of murine
`monoclonal antibodies capable of inhibiting HER-2+ cell
`lines; the most potent of these was muMAb 4D5. This anti-
`body was found markedly to inhibit proliferation of cell lines
`that overexpressed HER-2 but had little or no effect on cells
`without elevated levels of HER-2 (Sarup et al. 1991). 4D5
`was found to be a potent inhibitor of growth of human breast
`cancer xenografts (Baselga & Mendelsohn 1994) and was
`therefore selected for further clinical development.
`In order to reduce the potential for generating a human
`anti-mouse immune response the 4D5 murine monoclonal
`antibody was humanised. Carter and colleagues subcloned
`the hypervariable region of the antibody into plasmids en-
`coding a human K light chain and the IgG1 constant region
`to generate a vector encoding a chimeric antibody which was
`then further humanised by site-directed mutagenesis (Carter
`et al. 1992). The vector was transduced into Chinese hamster
`
`ovary (CHO) cells that then secrete the antibody into the
`culture medium from which it is purified. The chimeric anti-
`body called trastuzumab is 95% human and 5% murine and
`retains the high affinity for the HER-2 epitope of the parental
`antibody.
`
`Preclinical pharmacology
`
`Trastuzumab has a binding affinity for HER-2 that is three
`times greater than that of its parent murine antibody 4D5.
`Like 4D5,
`it has been shown to have a marked anti-
`proliferative effect on HER-2-overexpressing cell lines and
`very little effect on cells not expressing HER-2 (Carter et al.
`1992). This anti-proliferative effect has also been demonstra-
`ted in viva in breast cancer xenograft experiments by Baselga
`and colleagues in which established BT-474 tumour xeno-
`grafts were inhibited from growing by trastuzumab. In doses
`of less than 1 mg/kg growth was inhibited in a dose-
`dependent fashion and no growth at all was seen at higher
`doses
`(Baselga et al.
`1998).
`In the same study,
`the
`researchers explored the addition of trastuzumab to either
`paclitaxel or doxorubicin. Chemotherapy alone was shown
`
`Endocrine-Related Cancer (2002) 9 75—85
`
`to have only modest anti-tumour activity, whereas combined
`treatment with trastuzumab resulted in a marked enhance-
`
`ment of the effect of chemotherapy with the greatest growth
`inhibition being seen with paclitaxel and trastuzumab
`(Baselga et al. 1998).
`Pegram and colleagues examined the effect of trastu-
`zumab on a number of other chemotherapeutic agents in a
`HER-2 transfected MCF7 xenograft model. Synergistic inter-
`actions were seen with cisplatin, docetaxel, thiotepa, cyclo-
`phosphamide, vinorelbine and etoposide. Additive effects
`were seen with doxorubicin, paclitaxel, vinblastine and
`methotrexate and the combination of trastuzumab with
`
`S-fluorouracil (5-FU) was found to be antagonistic (Pegram
`et al. 1999, Konecny et al. 2001 and reviewed in Pegram et
`al. 2000). The synergy seen in these in viva models has led
`to the exploration in clinical trials of trastuzumab in combi-
`nation with chemotherapy.
`Trastuzumab was shown to be safe when administered
`
`chronically to a range of animals
`(reviewed in Roche 2001).
`
`including primates
`
`Mechanism of action
`
`The mechanism by which trastuzumab inhibits the growth
`of HER-2-expressing breast cancer cells and interacts with
`cytotoxic drugs is uncertain. It has been shown that trastu-
`zumab downregulates the HER-2 receptor by accelerating
`endocytic degradation (De Santes et al. 1992). The reduction
`in available HER-2 receptor results in fewer homo- and hetero-
`dimers capable of signalling. Either as a result of reduced
`numbers of receptors or by a direct inhibitory effect, trastu-
`zumab antagonises growth and cell division signals associ-
`ated with HER-2 signalling (reviewed in Sliwkowski et al.
`1999). In addition to cell cycle effects, trastuzumab may also
`antagonise the induction of angiogenic factors such as vascu-
`lar endothelial growth factor by HER-2 (Petit et al. 1997).
`The ability of cells to repair DNA damage after radiation or
`chemotherapy damage may also be impeded by trastuzumab
`(Pietras et al. 1999). In addition to direct effects on HER-2
`
`signalling, trastuzumab may induce a host tumour response
`via antibody-dependent cell cytotoxicity (ADCC) mechan-
`isms and complement activation (Hotaling et al. 1996).
`
`Early clinical trials
`
`The first single agent phase I studies of trastuzumab began
`in 1992. In the first study, 16 patients with HER-2-positive
`metastatic breast cancer were treated with fixed doses of 10—
`
`500mg as a single dose. Subsequently, two phase I studies
`evaluated a weekly schedule of trastuzumab either alone or
`in combination with cisplatin. In neither study was dose-
`limiting toxicity reached with trastuzumab doses of 10—
`500 mg, and 100 mg was taken forward as the recommended
`phase H dose. Pharmacokinetic data showed a half-life of 8.3
`
`www.endocrinology.org
`
`77
`
`

`

`Harries and Smith: Trastuzumab
`
`thus suggesting a weekly schedule (summarised in
`days,
`Roche 2001).
`
`Baselga and colleagues reported on 46 patients with
`metastatic breast cancer who had received extensive prior
`chemotherapy treated with a 250 mg loading dose of trastu-
`zumab followed by weekly doses of 100 mg until disease
`progression. Five out of 43 assessable patients had clinical
`responses (12%) and 16 additional patients also had minor
`responses or stable disease with a median time to progression
`of 5.1 months (Baselga et al. 1996). A separate study evalua-
`ted the same dose of trastuzumab in combination with cispla-
`tin in a similar group of heavily pretreated patients. Of 37
`patients evaluable for response, 9 patients had a partial
`response (24%). No excessive toxicity was seen above that
`expected for cisplatin alone (Pegram et al. 1998).
`
`Pivotal trials
`
`In parallel with these early trials a large multinational phase
`II study was initiated to confirm the efficacy and safety of
`trastuzumab. Two hundred and twenty-two women were
`enrolled into the study between April 1995 and September
`1996. All women had previously received one or two chemo-
`therapy regimens for metastatic breast cancer and had
`tumours that overexpressed HER-2 as determined by IHC in
`a central reference laboratory by weak to strong membrane
`staining in >10% of cells (IHC 2 + or 3 + ). Patients received
`trastuzumab 4mg/kg as a loading dose and then 2 mg/kg
`weekly thereafter until disease progression. The primary
`endpoint was response rate and secondary endpoints were
`duration of response, time to disease progression, time to
`treatment failure, and survival. In an intent to treat analysis
`the overall response rate was found to be 15% (95%, confi-
`dence interval (CI) 11—21%) as determined by an indepen-
`dent response evaluation comrnittee. The median duration of
`response was 9.1 months
`(95%, CI 6.5—10.5 months).
`Amongst all treated patients median survival was 13 months,
`median time to progression was 3.1 months and median time
`to treatment failure was 11 months. In the subgroup of
`patients whose tumours had IHC staining that was 3 + , the
`response rate was 18% and median survival 16.4 months,
`suggesting that this group of patients derived greatest benefit
`from trastuzumab. A retrospective analysis of gene expres-
`sion by FISH showed that the response rate was 19% in
`patients with gene amplification (FISH+) and was 0% in
`the FISH-negative group. The median number of infusions
`received was 12 (range 1—96) and treatment was generally
`found to be well tolerated, with the most common adverse
`
`event being mild to moderate infusion reactions that usually
`only occurred with the first treatment and were managed suc-
`cessfully with paracetamol and/or antihistamines. The inci-
`dence of cardiac dysfunction, defined as congestive heart
`failure, cardiomyopathy and/or a decrease in ejection fraction
`of >10% was examined by retrospective analysis and was
`
`identified in 4.7% of patients. This trial confirmed that trastu-
`zumab was active and generally well tolerated as a single
`agent in heavily pretreated patients (Cobleigh et al. 1999).
`The synergy seen in the preclinical studies between tras-
`tuzumab and cytotoxic agents encouraged investigators to
`evaluate combination therapy in the clinic. A large multi-
`national phase III study was performed to compare chemo-
`therapy in combination with trastuzumab to chemotherapy
`alone as first-line therapy in patients with metastatic breast
`cancer whose tumours were found to overexpress HER-2
`(again as determined to be IHC 2 + or 3 + in a central refer-
`ence laboratory). Four hundred and sixty-nine patients were
`randomised to receive chemotherapy alone or chemotherapy
`plus trastuzumab. Patients who had received an anthracycline
`in an adjuvant setting received paclitaxel 175 mg/m2 three-
`weekly and for other patients chemotherapy was an anthra-
`cycline (the majority receiving doxorubicin 60 mg/m2) and
`cyclophoshamide 600 mg/m2 three-weekly. Chemotherapy
`was given every three weeks for 6 cycles and then further
`chemotherapy was allowed to be given at the discretion of
`the investigator. Trastuzumab was given as a loading dose
`of 4 mg/kg and then 2 mg/kg weekly thereafter until disease
`progression. Patients had measurable disease and were of
`good performance status. The primary study endpoint was
`time to progression, with secondary endpoints of objective
`response rate, duration of response, time to treatment failure
`and 1-year survival. At a median of 30 months of follow-up
`the time to progression for patients receiving both trastu-
`zumab and chemotherapy was 7.4 months compared with 4.6
`months for patients who received chemotherapy alone. The
`overall
`response rate and response duration were also
`improved in patients receiving the combination treatment.
`The addition of trastuzumab to paclitaxel
`increased the
`response rate from 17% to 41% and median response dura-
`tion from 4.5 to 10.5 months. The addition of trastuzumab to
`
`the anthracycline regimens increased response rates from 42
`to 56% and median duration of response from 6.7 to 9.1
`months compared with chemotherapy alone.
`Taking both groups of patients together, overall survival
`was significantly improved with the addition of trastuzumab
`to chemotherapy from 20.3 to 25.1 months (P = 0.046). Sev-
`enty-two percent of patients who had been randomised to
`receive chemotherapy alone subsequently received trastu-
`zumab and therefore the magnitude of the survival advantage
`may be underrepresented due to this crossover effect. As with
`the multicentre phase II trial, the benefit for the addition of
`trastuzumab was particularly marked for patients whose
`tumours were 3 + on IHC staining. In addition, the benefit
`of trastuzumab was only seen in patients whose tumours had
`amplification of the HER-2 gene as determined by FISH. In
`patients without gene amplification, response rates were not
`significantly improved in the trastuzumab-containing regi-
`mens. In contrast, for patients with HER-2 amplification the
`response rate to chemotherapy alone was 31% and this
`
`78
`
`www.endocrinology.org
`
`

`

`increased to 54% in the trastuzumab-containing regimens. In
`this group median survival was increased from 20 to 26.2
`months (P = 0.007) (Mass et al. 2001a, Slamon et al. 2001).
`
`were mild to moderate infusion reactions that were easily
`manageable and usually only associated with the first treat-
`ment.
`
`Endocrine-Related Cancer (2002) 9 75—85
`
`Trastuzumab as first-line monotherapy
`
`Subsequent to the two pivotal studies discussed above, Vogel
`and colleagues (2002) have reported the results of an impor-
`tant phase II study of single agent trastuzumab as first-line
`treatment for 114 patients with metastatic breast cancer. In
`this study HER-2-overexpressing patients were randomised
`to receive either stande dose (4 mg/kg loading then 2 mg/
`kg weekly) or a higher dose regimen (8 mg/kg loading dose
`and 4mg/kg weekly thereafter) until disease progression.
`The overall response rate was 24% for the stande trastu-
`zumab dose arm, 28% for those receiving the higher dose
`(non-significantly different) and was 26% for all patients in
`the study. A further 13 patients had stable disease that per-
`sisted for greater than 6 months. As in the previous studies,
`higher response rates were seen in IHC 3+ patients and
`FISH+ patients (35% and 41% respectively).
`In FISH-
`negative patients, the response rate was only 7%. Median
`survival for all patients was 24.4 months but follow-up is
`short and at the time of censor 49 patients were alive or lost
`to follow-up. In this group of patients single agent trastu-
`zumab was extremely well
`tolerated with only 2 (2%)
`patients discontinuing therapy due to adverse events, both of
`whom had a history of cardiac disease and were withdrawn
`due to cardiac dysfunction. Furthermore, there was an overall
`improvement
`in global quality of life and fatigue scores
`(Vogel et al. 2002).
`
`Measuring HER-2 expression
`
`These clinical studies have shown a strong correlation
`between the efficacy of trastuzumab and the strength of IHC
`staining, with patients with IHC 3+ tumours responding
`better than those with 2+ tumours. Trastuzumab efficacy
`was also highly correlated with gene amplification with
`almost no benefit seen in tumours that had normal copy num-
`bers of the gene. FISH has been shown to select almost all
`IHC 3 + HER-2-expressing tumours. In addition, in these
`studies and others about 24% of patients whose tumours
`stained 2+ by IHC were found to be FISH+ (Mass et al.
`2001b). It is therefore becoming the practice of many clin-
`icians to select patients for trastuzumab on the basis of IHC
`3 + reactivity and if staining is 2+ then treatment is only
`given if gene amplification is present on FISH analysis.
`Unfortunately FISH is not so far available in all centres.
`
`Clinical safety of trastuzumab
`
`The large studies of trastuzumab have shown it to be gener-
`ally a well tolerated treatment. The commonest side effects
`
`Unexpectedly, cardiac dysfunction that had not been
`seen or predicted from the preclinical studies occurred in a
`number of patients. In the single agent phase II study for
`pretreated patients, the incidence of symptomatic heart fail-
`ure was 6—8.8% and all but one of these patients had received
`prior anthracyclines (Cobleigh et al. 1999). In the first-line
`phase II study of single agent
`trastuzumab only 2% of
`patients had to stop treatment due to cardiac dysfunction
`(Vogel et al. 2002). In the trastuzumab—chemotherapy com-
`bination study there was a marked increase in the incidence
`of heart failure for patients receiving trastuzumab plus an
`anthracycline, with an incidence of cardiac dysfunction (all
`grades) of 26—28% compared with 6—9.6% for patients who
`had received the anthracycline regimen alone. The incidence
`of marked symptomatic dysfunction (New York Heart Asso-
`ciation (NYHA) grade III—IV) was 16% and 3% respectively.
`Only 1—4% of patients treated with paclitaxel alone devel-
`oped cardiac dysfunction (1% at NYHA grade III/IV) com-
`pared with 8.8—1 1% (2% at NYHA grade III/IV) in patients
`treated with the trastuzumab and paclitaxel combination. In
`the small numbers of patients who had not received prior or
`concurrent anthracycline the incidence of cardiac dysfunction
`was 4%, all of whom were found to have had risk factors for
`
`cardiac disease. Most of the patients with symptomatic heart
`failure continued to receive trastuzumab and 75% improved
`with stande medical management. Only in the anthracyc-
`line/trastuzumab-treated patients were a small minority of
`patients (6%) left with NYHA grade III/IV toxicity at the
`end of treatment. Deaths related to cardiac dysfunction were
`very rare with no significant difference in incidence between
`the subgroups (Slamon et al. 2001, Tripathy et al. 2001). It
`should be noted that these data were collected retrospectively
`but prospective monitoring of cardiac function is an integral
`part of all current trastuzumab studies.
`Thus experience with trastuzumab to date suggests that
`cardiac toxicity is particularly related to the concurrent use
`of anthracyclines. In addition, the incidence of cardiac dys-
`function seems greatest in patients with pre-existing risk fac-
`tors for heart failure.
`
`The mechanism of cardiotoxicity is unclear. It appears
`unlikely that
`trastuzumab-associated heart damage occurs
`directly as a result of HER-2 expression on heart muscle. In
`60 patients with evidence of cardiac dysfunction, including
`25 patients with previous anthracycline exposure, Fuchs and
`colleagues found only faint membrane staining in less than
`10% of cardiac biopsies and no evidence of gene amplifica-
`tion by FISH (Fuchs et al. 2001). It also seems unlikely that
`trastuzumab alters the pharmacokinetic properties of anthra-
`cyclines and one study has failed to demonstrate any differ-
`ence in the pharmacokinetics of doxorubicin when it was
`administered before or after trastuzumab (Gianni et al. 2001).
`
`www.endocrinology.org
`
`79
`
`

`

`Harries and Smith: Trastuzumab
`
`One possible hypothesis, partly born out by preclinical data,
`is that trastuzumab impairs growth factor-mediated repair of
`anthracycline-induced cardiac damage (reviewed in Chien
`2000).
`
`Therefore, it is current policy not to recommend concur-
`rent treatment with trastuzumab and doxorubicin. In patients
`with previous exposure to anthracyclines or with a predis-
`position to heart disease careful monitoring of cardiac func-
`tion either with echocardiography or multigated radionucleo-
`tide angiography (MUGA) should be performed to pick up
`early evidence of cardiac dysfunction that may necessitate
`withdrawal of trastuzumab. If symptomatic cardiac dysfunc-
`tion develops whilst on trastuzumab then stande medical
`therapy should be initiated and is generally effective.
`Post-marketing safety analysis has indicated that there is
`also a rare incidence of severe pulmonary events associated
`with trastuzumab that may or may not follow severe infusion
`reactions. The clinical picture can include dyspnoea, pulmon-
`ary infiltrates, pleural effusions, non-cardiogenic pulmonary
`oedema, and adult respiratory distress syndrome. Patients
`with symptomatic intrinsic lung disease or with extensive
`tumour involvement of the lungs resulting in dyspnoea at rest
`may be at greater risk of such events (Roche 2001).
`
`Summary of the pivotal trials and
`licensing of trastuzumab
`
`The two large pivotal trials discussed above demonstrated
`that trastuzumab was active as a single agent in patients with
`metastatic breast cancer who had previously received chemo-
`therapy and when used in combination with paclitaxel or
`anthracycline—cyclophosphamide regimens was associated
`with improved survival
`in first-line therapy of metastatic
`breast cancer compared with chemotherapy alone. The ben-
`efit of trastuzumab was greatest for patients whose tumours
`expressed HER-2 at the 3 + level by IHC and was generally
`only seen in patients who had evidence of gene amplification
`as detected by FISH. Although trastuzumab was generally
`well tolerated, cardiac dysfunction was seen in some patients
`who received an anthracycline—cyclophosphamide regimen
`together with trastuzumab.
`As a result of these trials trastuzumab is now licensed in
`
`many countries for use as first-line therapy in combination
`with paclitaxel
`for patients whose tumours overexpress
`HER-2 and as a single agent in paclitaxel pretreated patients.
`It is our view that overexpression of HER-2 in this context
`should be defined as IHC 3 + or IHC 2 + and FISH + .
`
`at progression, or in combination with chemotherapy, or as
`single agent therapy after failure of first-line chemotherapy.
`As discussed above the pivotal phase III trial demonstrated
`a survival advantage for the trastuzumab and chemotherapy
`combination compared with chemotherapy alone despite a
`large number of the patients on the chemotherapy alone arms
`receiving trastuzumab on disease progression. These data and
`the synergy seen in preclinical xenografts would suggest that
`combination therapy should be superior to sequential therapy
`with chemotherapy followed by trastuzumab.
`On the other hand, for patients whose tumours were
`FISH+ the phase II study of first-line single agent trastu-
`zumab reported by Vogel and colleagues reported an overall
`survival on a par with the combination of chemotherapy and
`trastuzumab seen in the large phase HI studies (23 months
`compared with 26.8 months). This suggests that there may
`be no survival disadvantage for sequential therapy compared
`with combination treatment for this group (Slamon et al.
`2001, Vogel et al. 2002). Furthermore, some patients relaps-
`ing after adjuvant chemotherapy might prefer the improved
`quality of life associated with trastuzumab alone compared
`with trastuzumab in combination with more chemotherapy.
`Trials addressing this issue are ongoing: for example, a Euro-
`pean study is comparing first-line trastuzumab followed at
`disease progression by combination trastuzumab and pacli-
`taxel compared with a first line combination of trastuzumab
`and paclitaxel.
`
`Duration of therapy for metastatic
`disease
`
`The trials discussed above have all continued the use of tras-
`
`tuzumab until disease progression based on the preclinical
`tumour xenograft models that demonstrated tumour regrowth
`could occur after trastuzumab withdrawal
`(Pietras et al.
`1998). Nevertheless, clinical trials to determine the optimum
`duration of therapy are necessary on both clinical and cost
`grounds.
`For patients being treated with chemotherapy and trastu-
`zumab conventional practice in cancer therapy would suggest
`tha

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket