throbber
Review: Clinical Trial Outcomes
`
`Strategies to overcome endocrine therapy resistance in
`hormone receptor-positive advanced breast cancer
`
`Rowan T Chlebowski
`Chief of Medical Oncology & Hematology,
`Harbor-UCLA Medical Center, Torrance,
`CA 90502, USA
`Tel.: +1 310 222 2219
`Fax: +1 310 320 2564
`E-mail: rowanchlebowski@gmail.com
`
`Clin. Invest. (2014) 4(1), 19–33
`
`Use of endocrine therapies has made hormone receptor-positive breast
`cancer a manageable disease if diagnosed at an early stage. However,
`endocrine therapy resistance is a persistent problem in patients with
`advanced breast cancer. Ongoing research has identified a number
`of mechanisms that may mediate resistance, including estrogen-
`independent activation of the estrogen receptor; increased signaling
`through the RAS/MAPK, NF-kB, or PI3K/Akt/mTOR pathway; and
`maintenance of cyclin D1 expression. Based on such findings, various
`strategies to overcome endocrine resistance have been developed.
`Although some therapies are in early development, others are available
`in the clinic. The novel treatment strategies under evaluation in clinical
`trials for managing patients with endocrine-resistant advanced breast
`cancer will be reviewed in combination with endocrine therapy.
`
`Keywords: endocrine resistance • growth factor receptor
`• hormone receptor-positive • PI3K pathway • RAS/MAPK pathway • tyrosine kinases
`
`In 2013, approximately 232,000 women will be diagnosed with invasive breast
`cancer (BC) in the USA and 40,000 will die of the disease [1]. Approximately
`70% of BCs are hormone (estrogen and/or progesterone) receptor-positive (HR+)
`[2], and patients with HR+ BC have a better prognosis, in part, because of their
`responsiveness to endocrine therapies [3,4]. Tamoxifen, a selective estrogen recep-
`tor (ER) modulator, is a partial ER antagonist, initially used as first-line therapy
`in patients with HR+ advanced BC (ABC) [201]. Aromatase inhibitors (AIs) lower
`endogenous estrogen levels by inhibiting androgen to estrogen conversion [5]. The
`three third-generation AIs – anastrozole, letrozole and exemestane – were shown
`to be superior to tamoxifen in terms of efficacy and safety in postmenopausal
`women with ABC [6–12]. In addition, approximately 40% of patients treated with
`adjuvant endocrine therapy and almost all patients with metastatic BC (MBC)
`die of the disease [13].
`
`Overview of endocrine resistance & strategies to overcome resistance
`Endocrine resistance is a significant problem in treating BC. Patients can pres-
`ent with primary/acquired resistance (no response to initial endocrine therapy),
`or disease progression or recurrence can develop while the patient is receiving
`therapy (secondary/acquired resistance). In fact, approximately 30% of patients
`with MBC regress with initial endocrine therapy, whereas another 20% have pro-
`longed stable disease [14]; and the duration of response to second and subsequent
`lines of therapy is substantially lower [15,16]. Since current endocrine therapy is
`effective, it is important to identify those patients who respond and those who
`do not respond to therapy in order to improve treatment decisions. However,
`predicting response can be challenging. Molecular profiling of snap-frozen tumor
`
`10.4155/CLI.13.123 © 2014 Future Science Ltd
`
`ISSN 2041-6792
`
`19
`
`Ex. 1085-0001
`
`

`
`Review: Clinical Trial Outcomes Chlebowski
`
`biopsies has demonstrated that patients who recur early
`while on adjuvant tamoxifen therapy have a different
`molecular profile than those patients who experience
`a later recurrence while receiving tamoxifen [17]. In
`addition, molecular profiling analyses of patients who
`became resistant to aromatase inhibitors have found
`that patients who develop resistance to endocrine
`therapy have different gene expression patterns, sug-
`gesting that endocrine resistance is not a homogeneous
`phenomenon [18]. In addition, preclinical models have
`suggested that endocrine resistance results in upregu-
`lation of alternate signaling pathways that might be
`suitable targets for targeted therapies [19]. These data
`suggest that endocrine resistance could be a hetero-
`geneous phenomenon that might necessitate molecu-
`lar profiling to determine the appropriate course of
`action when resistance occurs. Additionally, because
`resistance might result in the upregulation of multiple
`pathways, the use of more than one therapy to target
`these pathways might be necessary.
`Ongoing research has provided insight into the
`causes of endocrine resistance and a number of mecha-
`nisms have been proposed, such as loss of ERa (one
`of the two types of ER) expression through methyla-
`tion; alterations in the expression of ER coactivators;
`and mutations in ERa [13,20]. However, no large-scale
`clinical data are available for most of these endocrine
`resistance mechanisms and they will not be discussed
`further.
`Some of the mechanisms of endocrine resistance
`that have been studied in clinical trials (Figure 1) [21]
`include ligand (estrogen)-independent activation of
`ER, increased signaling through the RAS/MAPK
`pathway, NF-kB pathway, or PI3K/Akt/mTOR path-
`way, increased growth factor (GF) expression and sig-
`naling, and loss of ERa-mediated signaling through
`chromatin remodeling [13,20].
`
` ■ Targeting the ER
`Unlike AIs and tamoxifen, fulvestrant, a selective
`ER down regulator, induces rapid degradation of ER
`[22]. In a Phase III trial in patients with previously
`untreated ABC comparing tamoxifen with fulves-
`trant at the initially evaluated dose of 250 mg every
`28 days, now considered low-dose, fulvestrant was as
`effective, but not superior to, tamoxifen [23]. Although
`steady-state drug concentrations are reached only after
`3–6 months with low-dose fulvestrant therapy, steady
`state levels were reached in less time using a loading
`dose consisting of 500 mg on day 1, 250 mg on days
`14 and 28, and 250 mg every 28 days thereafter, and a
`high-dose, consisting of 500 mg on days 1, 14, and 28
`of the first month and 500 mg every 28 days thereafter
`[24]. A Phase II, open-label trial (FIRST) in patients
`
`with ABC who may have had prior adjuvant endo-
`crine therapy for early disease more than 12 months
`before randomization but with no previous endocrine
`therapy exposure for advanced disease, evaluated the
`efficacy of high-dose fulvestrant versus anastrozole
`[25]. Although there was no difference in the primary
`end point of clinical benefit rate (CBR) in the FIRST
`trial, the median time to progression (TTP) was sub-
`stantially greater with high-dose fulvestrant (23.4 vs
`13.1 months; hazard ratio: 0.66; 95% CI: 0.47–0.92;
`p = 0.01) [25,26]. An ongoing Phase III randomized
`trial is attempting to provide definitive evaluation of
`those results.
`As AIs lower estrogen levels, endocrine resistance
`may result from development of estrogen-indepen-
`dent ER-mediated signaling [5]. Since fulvestrant
`promotes degradation of ERa through the ubiqui-
`tin–proteasome pathway in a preclinical setting [22],
`it was hypothesized that fulvestrant could overcome
`endocrine resistance. A combined ana lysis of two
`Phase III trials [27,28] in the second-line setting in
`postmenopausal women with locally ABC or MBC
`and disease progression during previous endocrine
`therapies, found low-dose fulvestrant as effective as
`anastrozole [29], leading to US FDA approval for use
`in postmenopausal women with HR+ ABC after pro-
`gression on previous anti-estrogen therapy. In another
`Phase III trial, low-dose fulvestrant and exemestane
`were equally effective in patients with HR+ ABC after
`previous nonsteroidal AI therapy [30]. Recently, the
`effectiveness of high- and low-dose fulvestrant in post-
`menopausal women with ABC or MBC progressing
`during previous endocrine therapy has been directly
`compared in the Phase III (CONFIRM) trial. In that
`setting, high-dose fulvestrant, compared with low-
`dose, significantly improved median progression-free
`survival (PFS; 6.5 vs 5.5 months; hazard ratio = 0.80;
`95% CI: 0.68–0.94; p = 0.006) [31] and overall sur-
`vival (OS; 26.4 vs 22.3 months; hazard ratio = 0.81;
`95% CI: 0.69–0.96; p = 0.016) [32]. Consequently, the
`FDA approved the high-dose fulvestrant schedule as a
`second-line therapy for postmenopausal women with
`HR+ metastatic disease [33].
`Although preclinical models suggested that combin-
`ing AI and fulvestrant may be an effective treatment
`option [34,35], clinical trial results have been conflicting.
`Results from the SWOG S0226 trial in post menopausal
`women with previously untreated MBC, showed that
`low-dose fulvestrant plus anastrozole significantly
`improved median PFS (15.0 vs 13.5 months; p = 0.007)
`and median OS (47.7 vs 41.3 months; p = 0.049) than
`anastrozole alone [36]. In contrast, results from another
`Phase III trial (FACT) in postmenopausal women with
`HR+ ABC in the same setting, comparing low-dose
`
`20
`
`www.future-science.com
`
`future science group
`
`Ex. 1085-0002
`
`

`
`Endocrine resistance in advanced breast cancer Review: Clinical Trial Outcomes
`
`Estrogen
`
`Growth factors
`
`A
`
`Plasma
`membrane
`
`PI3K
`
`B
`
`RTKs:
`EGFR,
`ERB2
`and IGFR
`
`C
`
`ER
`
`Ras
`
`Erk
`
`ER
`
`D
`
`PTEN
`
`Akt
`
`PI3K
`
`mTORC1
`
`ER
`
`Src
`
`M
`
`FAK
`
`E
`
`PELP1
`
`ER
`
`Src
`
`Src
`
`S6K1
`
`4E-BP1
`
`Akt
`
`S6K1
`
`elF4E
`
`Translation
`
`Erk
`
`PP
`
`ER
`
`P P
`
`Cytoplasm
`
`ER
`
`ER
`
`Nucleus
`
`CoA HAT
`
`ER ER
`
`CoA
`
`ER
`
`Ap1 Sp1
`
`TFs
`
`TFs
`
`TF
`
`TF
`
`ERE
`
`SRE
`
`RE
`
`mRNA
`
`AAAA
`
`MYC, cyclin D1,
`cyclin E1 and cyclin E2
`
`Growth proliferation and survival
`
`Clinical Investigation © Future Science Group (2014)
`
`Figure 1. Mechanisms that may mediate endocrine resistance. (A) Ligand-bound ER activates gene expression either directly or
`through protein–protein interactions. (B) RTKs such as the EGFR, ERBB2 (also known as HER2) and the IGFR, can activate downstream
`signaling events, thereby regulating translation and transcription. (C) ER localized at the cell membrane or in the cytoplasm can mediate
`nongenomic signaling. (D) Formation of an ER–PI3K–Src–FAK complex can activate Akt, resulting in activation of downstream signaling
`cascade. (E) Activation of Erk by ER–Src–PELP1 complexes can activate downstream signaling events. All together these complex
`signaling cascades regulate growth, proliferation and survival, resulting in endocrine therapy resistance.
`CoA: Coactivator; EGFR: EGF receptor; ER: Estrogen receptor; ERE: Estrogen response element; IGFR: IGF receptor; RE: Response element;
`RTK: Receptor tyrosine kinase; SRE: Serum response element; TF: Transcription factor.
`Reprinted with permission from [21].
`
`Clin. Invest. (2014) 4(1)
`
`21
`
`future science group
`
`Ex. 1085-0003
`
`

`
`Review: Clinical Trial Outcomes Chlebowski
`
`fulvestrant plus anastrozole with anastrozole alone,
`did not find statistically significant differences in TTP,
`CBR, objective response rate (ORR) or OS between the
`two treatments [37]. Results from another Phase III trial
`(SoFEA), comparing low-dose fulvestrant plus anas-
`trozole with anastrozole alone, also found no differ-
`ences in PFS, ORR, CBR or OS [38]. Therefore, current
`data are inconclusive for combining AI with low-dose
`fulvestrant to overcome endocrine resistance.
`
` ■ Inhibition of intracellular signaling cascade
`In preclinical models, activation of intra cellular signal-
`ing pathways (Figure 1) [21] – such as the PI3K/Akt/
`mTOR pathway, RAS/MAPK pathway, Src kinase sig-
`naling cascade, or NF-kB pathway – has been shown
`to mediate endocrine resistance. Strategies inhibiting
`one or more of these pathways in combination with
`endocrine therapies are under clinical evaluation.
`
`Inhibition of the PI3K/Akt/mTOR pathway
`Preclinical models of BC cells resistant to estrogen
`deprivation were found to have amplified PI3K/Akt/
`mTOR-mediated signaling [39], suggesting that acti-
`vation of the PI3K pathway may facilitate survival of
`these cells under hormone deprivation. Additionally,
`by using cell lysates from hormone receptor-positive
`primary breast tumors, PI3K pathway activation was
`associated with poor disease outcome after adjuvant
`therapy [39]. Activation of PI3K results in activation
`of its downstream target Akt and in cells expressing
`activated Akt, the efficacy of tamoxifen to induce
`growth inhibition was dramatically reduced [40]. Since
`treatment with mTOR inhibitors reversed tamoxifen
`resistance in cells over expressing activated Akt [40,41]
`and reduced tumor growth in xenograft models [40],
`mTOR inhibitors (sirolimus, temsirolimus and evero-
`limus) have been tested in clinical trials to overcome
`endocrine resistance.
`
`Sirolimus & temsirolimus
`Sirolimus, in combination with tamoxifen, has recently
`been shown to significantly improve median TTP and
`response rates compared with tamoxifen alone in a
`Phase II trial in postmenopausal women with HR +
`ABC in whom previous tamoxifen and/or AI therapy
`was ineffective [42]. Although temsirolimus was effec-
`tive in combination with letrozole in patients with
`ABC with disease progression during or after tamoxi-
`fen therapy in a Phase II trial [43], results from HORI-
`ZON, a Phase III trial in postmenopausal women with
`AI-naive, locally advanced or MBC, did not show ben-
`efit for the temsirolimus combination compared with
`letrozole alone in the first-line setting, with results
`perhaps limited by the substantial toxicity seen [44].
`
`Everolimus
`In a Phase II neoadjuvant trial in patients with oper-
`able ER+ BC, everolimus in combination with letro-
`zole improved the clinical response rate compared with
`letrozole alone [45]. In another Phase II trial, everolimus
`plus tamoxifen was compared with tamoxifen alone in
`postmenopausal patients with HR+, HER2– MBC with
`previous exposure to AIs, and the combination signifi-
`cantly improved CBR (61 vs 42%; p < 0.045), TTP
`(8.6 vs 4.5 months; p < 0.002), and OS (hazard ratio:
`0.45; 95% CI: 0.24–0.81; exploratory p = 0.007) [46].
`In the BOLERO-2 Phase III trial evaluating everoli-
`mus plus exemestane versus exemestane alone in post-
`menopausal women with HR+, HER2– ABC after previ-
`ous letrozole or anastrozole, median PFS at 7.1 months
`was significantly improved with the everolimus combi-
`nation (6.9 vs 2.8 months by local assessment; hazard
`ratio 0.43; 95% CI: 0.35–0.54; p < 0.001) [47]. The
`Kaplan–Meier plot shows substantial visual separa-
`tion of the PFS curves at the 6-week reimaging period.
`Analy ses from the 12.5- and 18-month follow-up data
`from BOLERO-2 produced similar results [48,49]. Based
`on the BOLERO-2 trial, everolimus (10 mg/day) in
`combination with exemestane (25 mg/day) was FDA
`approved for managing postmenopausal women with
`HR+ ABC and is currently the only approved mTOR
`inhibitor for managing patients with ABC after failure
`with letrozole or anastrozole [50]. In additional analy-
`ses from BOLERO-2, the everolimus combination was
`associated with a longer time to definitive deterioration
`of health-related quality of life (8.3 vs 5.8 months; haz-
`ard ratio: 0.74; p = 0.0084) [51] and was effective regard-
`less of whether patients had visceral metastases [52].
`Finally, exploratory analyses in the study found favorable
`effects on bone turnover, and less progression in bone
`metastases was seen for the everolimus combination [53].
`Of potential clinical relevance are findings from
`the Phase III BOLERO-3 trial, in which everolimus
`(5 mg/day) addition was compared with trastuzumab
`and weekly vinorelbine alone in patients with HER2+
`ABC [54]. Everolimus addition improved median PFS
`(hazard ratio: 0.78; p = 0.0067), but subgroup analyses
`suggested an effect limited to women with ER– dis-
`ease (hazard ratio: 0.65; 95% CI: 0.49–0.86). These
`findings raise the hypothesis that combination therapy
`targeting both ER and HER2 pathways may be needed
`to optimize outcome in ER+, HER2+ ABC.
`
`Ongoing trials
`A Phase III adjuvant trial evaluating 1-year therapy
`with everolimus in addition to adjuvant endocrine
`therapy in high-risk patients with HR+ HER2– invasive
`BC is ongoing (Table 1) [202]. Other ongoing BC stud-
`ies evaluating everolimus are also outlined in Table 1.
`
`22
`
`www.future-science.com
`
`future science group
`
`Ex. 1085-0004
`
`

`
`Table 1. Ongoing clinical trials using PI3K/Akt/mTOR inhibitors to overcome endocrine resistance in patients with
`advanced breast cancer.
`Clinical trial
`Intervention
`identifier
`NCT01231659
`
`Expected
`enrollment (n)
`70
`
`Select primary and secondary
`outcomes
`Primary: ORR
`Secondary: PFS, OS, DCR, safety
`
`Patient population
`
`Phase
`
`II
`
`Postmenopausal women
`with locally advanced BC
`or MBC after recurrence
`or progression on
`tamoxifen, anastrozole
`or exemestane
`Postmenopausal women
`with endocrine-resistant
`advanced BC
`Postmenopausal women
`with AI-resistant MBC
`
`Letrozole +
`everolimus
`
`NCT01499160
`
`NCT01797120
`
`NCT01698918
`
`NCT01437566
`
`NCT01783444
`
`Letrozole +
`lapatinib +
`everolimus
`Fulvestrant +
`everolimus
`
`Everolimus +
`letrozole
`
`Fulvestrant ±
`GDC-0941
`fulvestrant ±
`GDC-0980
`Everolimus
`Capecitabine
`Everolimus +
`exemestane
`
`NCT01626222
`
`Exemestane
`+ everolimus
`
`NCT01633060
`
`Fulvestrant ±
`BKM120
`
`NCT01610284
`
`Fulvestrant ±
`BKM120
`
`NCT01674140
`
`Adjuvant
`Primary: IDFS assessed up to
`hormone
`10 years
`therapy ±
`Secondary: OS, DRFS assessed
`everolimus
`up to 10 years, toxicity
`AI: Aromatase inhibitor; BC: Breast cancer; CBR: Clinical-benefit rate; DCR: Disease-control rate; DOR: Duration of response; DRFS: Distant recurrence-free survival;
`ECOG: Eastern Cooperative Oncology Group; ER: Estrogren receptor; HR: Hormone receptor; HR-QoL: Health-related QoL; IDFS: Invasive disease-free survival;
`MBC: Metastatic breast cancer; ORR: Overall-response rate; OS: Overall survival; PFS: Progression-free survival; PK: Pharmacokinetic; QoL: Quality of life; TTP: Time to
`progression.
`
`Endocrine resistance in advanced breast cancer Review: Clinical Trial Outcomes
`
`Ref.
`
`[214]
`
`[215]
`
`[216]
`
`[217]
`
`[218]
`
`[219]
`
`[220]
`
`[221]
`
`[222]
`
`[202]
`
`II
`
`II
`
`II
`
`II
`
`II
`
`76
`
`130
`
`200
`
`270
`
`300
`
`Primary: CBR
`Secondary: PFS
`
`Primary: PFS
`Secondary: ORR, TTP, OS
`toxicity
`Primary: PFS
`Secondary: ORR, OS, reduction
`in severity and duration of
`stomatitis, safety
`Primary: PFS, safety
`Secondary: ORR, DOR,
`PK parameters
`
`Primary: PFS
`Secondary: OS, ORR, CBR,
`change in ECOG status, QoL,
`safety
`
`IIIb
`
`300
`
`Primary: ORR
`Secondary: PFS, OS, safety, HR-
`QoL, resource use
`
`III
`
`III
`
`615
`
`842
`
`Primary: PFS
`Secondary: OS, ORR, CBR,
`safety, PK, QoL
`
`Primary: PFS
`Secondary: OS, ORR, CBR,
`safety, PK, QoL
`
`III
`
`3500
`
`Postmenopausal women
`with ER+ MBC as first-
`line therapy
`
`AI-resistant patients
`with advanced BC or
`MBC
`
`Postmenopausal women
`with locally advanced
`BC, recurrent BC or
`MBC after recurrence
`or progression on prior
`letrozole or anastrozole
`Postmenopausal
`women with ER+, locally
`advanced BC or MBC
`progressing on prior
`therapies
`Postmenopausal women
`with HR+ HER2– AI
`treated with locally
`advanced BC or MBC
`progressing on or after
`mTOR inhibitor therapy
`Postemenopausal
`women with HR+ HER2–
`AI treated with locally
`advanced BC or MBC
`refractory to AI therapy
`High-risk patients with
`HR+, HER2– BC
`
`Clin. Invest. (2014) 4(1)
`
`23
`
`future science group
`
`Ex. 1085-0005
`
`

`
`Review: Clinical Trial Outcomes Chlebowski
`
` ■ Identifying biomarkers of response to everolimus
`A key aspect of administering everolimus therapy is deter-
`mining which patients would benefit most from treat-
`ment. However, identification of appropriate biomark-
`ers has been challenging. Although PIC3AK-activating
`mutations are common in BC [55], data have suggested
`that activation of PIK3CA mutations is not predictive
`of clinical benefit to mTOR inhibitors. An ana lysis of
`primary ER+ BC tumor samples found that high levels
`of the PIK3CA-GS gene signature expression (indicative
`of a PIK3CA mutant phenotype) is indicative of low
`mTOR-pathway activation [56]. Additionally, an explor-
`atory ana lysis of the BOLERO-2 study that evaluated
`the mutational status of key genes (PIK3CA, CCND1
`or FGFR1/2) found that, when examining these genes
`individually, patients whose genes were altered derived
`similar benefit to everolimus, compared with the overall
`trial population [57]. Also, patients who had wild-type
`or a single genetic alteration in the PIK3CA, CCND1,
`or FGFR1/2 genes seemed to derive greater benefit from
`everolimus than the overall BOLERO-2 population
`[57]. Another method to evaluate the potential benefit
`of mTOR inhibitors is the evaluation of the pattern of
`protein expression with mTOR pathway activation. In
`an exploratory translational ana lysis of the TAMRAD
`data, high p4EBP, low LKB1 and low PI3K seemed to be
`associated with everolimus efficacy [58]. However, these
`data must be validated in larger studies.
`
` ■ Investigational strategies to inhibit PI3K/Akt/
`mTOR pathway
`Additional PI3K/Akt/mTOR pathway inhibitors are being
`evaluated in clinical trials. For example, NVP-BEZ235 (a
`dual pan-PI3K/mTOR inhibitor) and BKM120 (a pan-
`PI3K inhibitor) are being evaluated in combination with
`letrozole in a Phase I trial in patients with HR+ MBC [203].
`
`Inhibition of the RAS/MAPK pathway
`The proto-oncoprotein RAS is a central mediator of many
`GF receptor-mediated signals, and activation of GF-medi-
`ated signaling results in ligand-independent activation of
`ER-mediated signaling through phosphorylation of ER
`or its coactivators [59]. Farnesyltransferase inhibitors that
`inhibit RAS have been shown to synergize with endocrine
`therapies to inhibit cell growth and induce apoptosis in a
`preclinical setting [60]. Despite promising preclinical stud-
`ies, results from Phase II clinical trials evaluating tipifa-
`rinib, a farnesyltransferase inhibitor, with tamoxifen [61]
`or fulvestrant [62] have been disappointing.
`
`Inhibition of Src-Kinase signaling cascade
`In preclinical studies, Src-kinase activity was increased in
`tamoxifen-resistant cells, resulting in increased migration,
`and inhibition of Src was found to reverse this aggressive
`
`phenotype [63] and restore sensitivity to tamoxifen [64].
`Dasatinib, an Src-kinase inhibitor, blocked the Src-
`induced proliferation of tamoxifen-resistant cells [65].
`However, in the clinic, dasatinib addition to exemestane
`in a Phase II trial did not increase PFS, compared with
`exemestane alone [204]. Nonetheless, several additional
`Phase II combination studies are ongoing (Table 2).
`
`NF-kB pathway & proteasome inhibition
`Active NF-kB in BC tissue identified a high-risk subset of
`ER+ BC patients [66]. In preclinical studies, inhibition of
`NF-kB activation with proteasome inhibitors stimulated
`the growth inhibitory effect of tamoxifen [67,68]. Bortezo-
`mib, a proteasome inhibitor that blocks NF-kB activation,
`was studied in combination with endocrine therapy in a
`small Phase II trial [69]. Although no clinical response was
`observed, a Phase II trial in combination with fulvestrant
`is ongoing (Table 2) [205].
`
` ■ Inhibition of aberrant GF receptor activation
`Endocrine resistance has been linked to aberrant expres-
`sion, activation or signaling through GF RTKs. Aber-
`rant activation of GF RTKs have been shown to activate
`a number of intracellular signal transduction cascades
`(Figure 1) [21], including the PI3K/Akt/mTOR, Src-kinase,
`and RAS/MAPK pathways [70,71]. Hence, a number of GF
`receptor inhibitors are being studied in clinical trials to
`overcome endocrine resistance.
`
`HER2 inhibition
`Approximately 10% of HR+ BC are also HER2+ [72].
`Using archival tumor blocks from the ATAC trial, time
`to recurrence was shorter in patients with HER2+ BC
`who were treated with either anastrozole or tamoxifen,
`suggesting that the effectiveness of tamoxifen might
`be impeded by HER2 positivity [73]. Additionally, pre-
`clinical evidence has suggested that crosstalk between
`HER2 and ER might lead to endocrine resistance [74,75].
`In a Phase III trial comparing letrozole plus lapatinib, a
`dual HER2 and EGF receptor (EGFR) inhibitor, with
`letrozole alone as first-line therapy, the combination sig-
`nificantly improved CBR and PFS in postmenopausal
`women with HER2+ HR+ MBC but not in patients with
`HR+ HER2– MBC [76]. Currently, lapatinib in combi-
`nation with letrozole is FDA approved for patients with
`HR+, HER2+ MBC [77]. Two trials exploring the effi-
`cacy and safety of fulvestrant with lapatinib in patients
`with previous exposure to endocrine therapy are ongoing
`(Table 3) [206,207].
`Trastuzumab, a HER2 inhibitor, in combination
`with anastrozole, was studied in a Phase III trial in
`postmenopausal patients with HER2+ HR+ MBC,
`some of whom had previous exposure to endocrine
`therapy [78]. Although the combination significantly
`
`24
`
`www.future-science.com
`
`future science group
`
`Ex. 1085-0006
`
`

`
`Endocrine resistance in advanced breast cancer Review: Clinical Trial Outcomes
`
`improved PFS by 2.4 months, the PFS for patients
`receiving the combination was only 4.8 months [78].
`A Phase III trial comparing letrozole plus trastuzumab
`with letrozole alone in patients with HER2+ HR+ MBC,
`when approximately 50% of patients had received pre-
`vious tamoxifen therapy, showed that the median TTP
`with the combination therapy was 14.1 months, com-
`pared with 3.3 months with letrozole alone [79]. All
`together, these observations indicate that combining
`a HER2 inhibitor with an AI may be clinically effec-
`tive. A Phase II trial in patients with HR+ HER2– ABC
`that progressed during previous endocrine therapy is
`currently evaluating the efficacy of MM-21, an inhibi-
`tor of HER3 ligand-stimulated dimerization between
`HER2 and HER3, in combination with exemestane
`(Table 3) [208].
`
`gefitinib in addition to anastrozole significantly improved
`median PFS by 6.3 months, compared with anastro-
`zole alone [84]; however, another Phase II trial failed
`to find significant benefit for tamoxifen plus gefitinib
`[85]. Gefitinib is currently being studied in a Phase II
`trial in patients with MBC who progressed during first-
`line endocrine therapy in combination with fulvestrant
`(Table 3) [209]. Overall, the results for gefitinib addition to
`endocrine therapy are inconclusive. In addition to gefi-
`tinib, vandetanib, a novel tyrosine kinase inhibitor with
`activity against a number of RTKs, including EGFR
`and the VEGF receptor (VEGFR) [86], is currently being
`studied in a Phase II trial in combination with fulves-
`trant in patients with predominantly bone metastasis
`HR+ MBC who progressed during previous endocrine
`therapy (Table 3) [210].
`
`EGF-receptor inhibition
`Approximately 30% of HR+ BC is EGFR+ [80], and pre-
`clinical studies have suggested that EGFR pathway acti-
`vation may mediate endocrine resistance [81]. In a Phase II
`trial, CBR was improved with gefitinib, an EGFR inhibi-
`tor, in patients with HR+ tamoxifen-resistant tumors,
`compared with HR– tamoxifen-resistant tumors [82].
`However, results from another Phase II trial failed to
`support those findings and reported higher toxicity with
`the combination [83]. In another Phase II trial in patients
`with HR+ MBC with no previous endocrine therapy for
`MBC or who progressed on adjuvant tamoxifen therapy,
`
`VEGFR inhibition
`Preclinical studies have shown that estradiol stimu-
`lates proliferation of human endothelial cells and that
`anti-estrogen inhibits these effects [87]. ER was shown
`to bind to the promoter of VEGF and activate its
`transcription, resulting in increased angiogenesis [88].
`Retrospective studies of tumor samples have associ-
`ated higher levels of VEGF with decreased response
`to endocrine therapy [89]. Bevacizumab, a monoclonal
`antibody that prevents VEGF and VEGFR interaction,
`in combination with letrozole, resulted in a median PFS
`of 17.1 months [90]. The combination of fulvestrant and
`
`Table 2. Ongoing clinical trials using other signal transduction inhibitors to overcome endocrine resistance in patients
`with advanced breast cancer.
`Clinical trial
`Intervention
`identifier
`Src-kinase inhibitors
`NCT00754325
`Fulvestrant ± dasatinib
`
`Patient population
`
`Phase
`
`Enrollment
`expected (n)
`
`Select primary and
`secondary outcomes
`
`II
`
`100
`
`Primary: PFS
`Secondary: ORR, TTF,
`bone markers, toxicity,
`bone pain, BMD
`
`Men and
`postmenopausal
`women with advanced
`BC previously treated
`with AI
`Postmenopausal
`women with
`unresectable, locally
`recurrent, or MBC – as
`first- and second-line
`therapy
`
`NCT00696072
`
`Letrozole ± dasatinib
`
`II
`
`120
`
`Primary: CBR
`Secondary: ORR, PFS,
`TTF, bone markers,
`toxicity, bone pain,
`BMD
`
`NF-kB pathway inhibitors
`NCT01142401
`Fulvestrant ± bortezomib Postmenopausal
`Primary: CBR
`women with locally
`Secondary: OS, PFS,
`advanced BC or MBC
`CBR at 12 and 24
`resistant to AI therapy
`weeks
`AI: Aromatase inhibitor; BC: Breast cancer; BMD: Bone mineral density; CBR: Clinical-benefit rate; MBC: Metastatic breast cancer; ORR: Overall-response rate;
`OS: Overall survival; PFS: Progression-free survival; TTF: Time to failure.
`
`118
`
`II
`
`Clin. Invest. (2014) 4(1)
`
`Ref.
`
`[223]
`
`[224]
`
`[225]
`
`25
`
`future science group
`
`Ex. 1085-0007
`
`

`
`Table 3. Ongoing clinical trials using growth factor-receptor inhibitors to overcome endocrine resistance in patients with
`advanced breast cancer.
`Clinical trial
`Intervention
`identifier
`HER2 inhibitors
`NCT01151046
`
`Exemestane ±
`MM-121
`
`NCT00390455
`
`NCT00688194
`
`Fulvestrant ±
`lapatinib
`
`Fulvestrant ±
`lapatinib ± AI
`
`Postmenopausal women with
`HR+ HER2– locally advanced
`BC or MBC
`Postmenopausal women with
`HR+ advanced BC
`
`Postmenopausal women with
`MBC who progressed after
`prior AI therapy
`
`EGFR inhibitors
`NCT00811369
`
`Fulvestrant ±
`vandetanib
`
`Postmenopausal women with
`bone-predominant HR+ MBC
`
`NCT00570258
`
`Fulvestrant ±
`erlotinib
`
`Patients with HR+ MBC who
`progressed on first-line
`hormonal therapy
`
`VEGFR inhibitors
`NCT01466972
`
`NCT00545077
`
`Anastrozole
`or letrozole +
`pazopanib
`Fulvestrant
`or letrozole ±
`bevacizumab
`
`Patients with HR+ advanced
`BC progressing on NSAI
`therapy
`Postmenopausal women with
`advanced BC or MBC; as first-
`line therapy
`
`II
`
`131
`
`Primary: PFS
`Secondary: NR
`
`III
`
`III
`
`II
`
`II
`
`II
`
`324
`
`396
`
`126
`
`130
`
`30
`
`III
`
`338
`
`Primary: PFS
`Secondary: OS, DOR, ORR,
`safety, QoL
`Primary: PFS
`Secondary: OS, TTP, RR, CBR
`
`Primary: Decrease in bone
`marker (NTx)
`Secondary: PFS, response,
`improvement in pain
`Primary: TTP
`Secondary: CBR, RR
`
`Primary: CBR
`Secondary: TTP, safety
`
`Primary: PFS
`Secondary: OS, TTF, RR, DOR,
`clinical benefit proportion,
`safety
`
`Ref.
`
`[208]
`
`[206]
`
`[207]
`
`[210]
`
`[209
`
`[226]
`
`[211]
`
`[212]
`
`Review: Clinical Trial Outcomes Chlebowski
`
`Patient population
`
`Phase
`
`Enrollment
`expected (n)
`
`Select primary and
`secondary outcomes
`
`FGFR inhibitors
`NCT01528345
`
`Fulvestrant ±
`dovitinib
`
`Postmenopausal women with
`locally advanced BC or MBC
`not amenable to curative
`surgery or radiotherapy and
`progressing on or after prior
`endocrine therapy
`AI: Aromatase inhibitor; BC: Breast cancer; CBR: Clinical benefit rate; DOR: Duration of response; EGFR: EGF receptor; FGFR: FGF receptor; HR: Hormone receptor;
`MBC: Metastatic breast cancer; NR: Not reported; NSAI: Nonsteroidal aromatase inhibitor; ORR: Overall-response rate; OS: Overall survival; PFS: Progression-free
`survival; QoL: Quality of life; RR: Response rate; TTF: Time to failure; TTP: Time to progression; VEGFR: VEGF receptor.
`
`II
`
`150
`
`Primary: PFS
`Secondary: OS, ORR, DOR,
`safety
`
`bevacizumab was tested in a Phase II trial in postmeno-
`pausal women with newly diagnosed MBC who were
`intolerant to an AI or who progressed while receiving
`an AI [91]. In this same trial, anastrozole plus bevaci-
`zumab was studied in patients with earlier stage disease.
`The median TTP was 21 months with anastrozole plus
`bevacizumab, essentially as first-line therapy, whereas
`it was 9 months with fulvestrant and bevacizumab as
`second-line therapy [91]. Although these results are dif-
`ficult to interpret, bevacizumab, in combination with
`endocrine therapy, is currently being tested as a first-line
`therapy in a Phase III trial (Table 3) [211].
`
`FGF receptor inhibition
`Aberrant expression of FGF receptor (FGFR)-1, -2,
`-3, and -4 have all been shown to result in BC devel-
`opment and to mediate endocrine resistance [92,93].
`Brivanib, a dual FGF and VEGF RTK inhibitor, was
`shown in preclinical studies to inhibit FGF-stimulated
`growth in FGFR-1-amplified BC cells [94]. Dovitinib,
`an inhibitor of FGFR, VEGFR, and the PDGF recep-
`tor, was shown in a Phase II trial to have some activity
`in patients who were heavily pretreated [95]. Dovitinib
`is currently undergoing another Phase II trial in com-
`bination with fulvestrant (Table 3) in postmenopausal
`
`26
`
`www.future-science.com
`
`future science group
`
`Ex. 1085-0008
`
`

`
`Endocrine

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