throbber
 
`
`Cancer Therapy Vol. 9, page 45
`
`
`
`Cancer Therapy Vol. 9, 45-54, 2013
`T-DM1: a giant step forwards in HER2 therapeutics
`Research Article
`
`Dey Nandini 1, 2, Carlson Jennifer1, Leyland-Jones Brian 1, 2, and De Pradip 1, 2*
`
`1 Edith Sanford Breast Cancer, Sanford Research/USD 60th Street N, Sioux Falls, SD, 57104
`2 Department of Internal Medicine, University of South Dakota, SD
`__________________________________________________________________________________________________
`*Correspondence: Pradip De, MS, PhD Associate Scientist, Edith Sanford Breast Cancer, Sanford Research/USD 60th Street N, Sioux
`Falls, SD, 57104 Tel: 0030-25510-74622, Fax: 605-312-6071, Email:  Pradip.De@SanfordHealth.org
`Keywords: Breast cancer, HER2, T-DM1, trastuzumab and pertuzumab
`
`
`Received: 15 August 2013; Revised: 17 September 2013
`Accepted: 18 September 2013; electronically published: 19 September 2013
`
`Summary
`
`Despite treatment advances, including the humanized anti-HER2 antibody trastuzumab and the dual EGFR/HER2
`tyrosine kinase small molecule inhibitor lapatinib, HER2+ breast cancer will eventually progress in most patients,
`highlighting the need for novel, alternative therapies. Moreover, HER2-targeted therapies are rarely given as a
`mono-therapy but are generally given in combination with other chemotherapy. It has been known that systemic
`toxicities are often associated with chemotherapies for cancer patients, therefore antibody drug conjugates with
`potential cytotoxic agent(s) are a promising therapeutic approach for HER2+ breast cancer patients. Trastuzumab
`emtansine (T-DM1) is an antibody drug conjugate that optimizes delivery of chemotherapy with an anti-HER2
`monoclonal antibody, trastuzumab. In HER2+ patients, T- DM1 delivers DM1 to the tumor and uses HER2 as the
`address. The mechanism of action does not rely on functional HER2 signaling and only requires high levels of
`HER2 on the cell surface. In addition, the presence of downstream alterations, such as PIK3CA mutation or the
`absence of PTEN, should not matter. T-DM1 has successfully passed through several clinical trials for the targeted
`delivery of chemotherapy and anti-HER2 monoclonal antibody therapy for patients with metastatic, HER2+ breast
`cancer. Recently, FDA gave approval to T-DM1, which will be marketed as Kadcyla, for patients with HER2+, late
`stage (metastatic) breast cancer. This is a novel approach to treating HER2+ breast cancer, the antibody drug
`conjugate (ADC) is seen as a significant advance for patients.
`
`I. Introduction
`The current view of breast cancer is that it is
`segmented into molecular subtypes defined by genomic
`alterations involved in tumor progression which then
`identify patient populations best treated with targeted
`inhibition. One such population is patients with HER2
`gene amplification and/or overexpression (approximately
`20-30% of human breast cancers) (O'Brien, Browne et al.
`2010). Before the development of HER2-targeted therapy,
`HER2-positivity was predictive of poor clinical outcomes
`(Slamon, Clark et al. 1987; Ross and Fletcher 1998). This
`group of proteins is comprised of EGFR (HER1), HER2,
`HER3 and HER4 (Yarden 2001) that are involved in cell
`growth and survival through activation of the PI3K-AKT-
`mTOR and the RAS-RAF-MEK-ERK pathways (Arteaga,
`Sliwkowski et al. 2012). Non-targeted breast cancer
`treatment options may
`include one or more of
`chemotherapy, radiation, and surgery, while HER2
`overexpressing breast cancers will typically involve
`trastuzumab-based therapy, with newer agents such as
`
`
`lapatinib providing a second line of treatment (Geyer,
`Forster et al. 2006; O'Brien, Browne et al. 2010). Single
`agent use of
`trastuzumab, a humanized anti-HER2
`monoclonal antibody, has shown objective responses in
`the range of 15–20% in phase II trials when used in first-
`line or refractory HER2-positive advanced breast cancer
`settings (Baselga, Tripathy et al. 1996; Cobleigh, Vogel et
`al. 1999; Vogel, Cobleigh et al. 2002). This benefit is
`magnified by the concomitant use of trastuzumab with
`traditional chemotherapy (anthracycline or epirubicin plus
`cyclophosphamide),
`leading
`to an
`improvement
`in
`objective response rates that range between 50–84% and
`median survivals that range between 25–37 months
`(Burstein, Kuter et al. 2001; Slamon, Leyland-Jones et al.
`2001; Esteva, Valero et al. 2002; Montemurro, Choa et al.
`2004; Andersson, Lidbrink et al. 2011; Valero, Forbes et
`al. 2011). Based on these and other data, the FDA and
`other regulatory agencies around the world approved the
`use of trastuzumab in the metastatic setting in 1998;
`
`  
`
`45
`
`IMMUNOGEN 2027, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Pradip De et al: T-DM1: a giant step forwards in HER2 therapeutics  
`
`an expansion of its use to the adjuvant setting was
`approved in 2006.
`Subsequently, two other HER2-targeted agents
`have been approved for the treatment of HER2-positive
`metastatic breast cancer,
`lapatinib and pertuzumab.
`Lapatinib is an oral small molecule dual receptor tyrosine
`kinase inhibitor that binds and inhibits both HER1 and
`HER2. Preclinical data demonstrated that lapatinib plus
`trastuzumab enhanced apoptosis (Konecny, Pegram et al.
`2006),
`improved
`tumor control
`in HER2-positive
`xenograft (Scaltriti, Verma et al. 2009) and resulted in a
`marketed accumulation of inactive HER2 on the cell
`surface in
` conjunction with enhanced cytotoxicity
`(Scaltriti, Verma et al. 2009). It was approved in 2007 for
`use in combination with capecitabine in patients whose
`disease progressed on or after an anthracycline, taxane
`and trastuzumab-based therapy. Data from a phase III trial
`showed an improved time to progression and response
`rate associated with lapatinib plus capecitabine compared
`to capecitabine alone (Geyer, Forster et al. 2006). In 2010,
`lapatinib also received FDA approval in combination with
`an aromatase inhibitor (letrozole) for post menopausal
`ER+/HER2+ metastatic breast cancer patients (Johnston,
`Pippen et al. 2009). The response rate associated with
`lapatinib monotherapy was 24% in trastuzumab-sensitive
`patients but the response rate was less than 10% in
`trastuzumab-resistant settings (Burstein, Storniolo et al.
`2008; Gomez, Doval et al. 2008; Blackwell, Burstein et
`al. 2010). Since each modality (trastuzumab and lapatinib)
`offers clinical benefit, combining
`trastuzumab plus
`lapatinib to target extracellular and intracellular domains
`of HER2 offers an attractive strategy. A phase III EGF
`104900 study demonstrated a 4.5 month median overall
`survival advantage with the lapatinib and trastuzumab
`combination (Blackwell, Burstein et al. 2012). The phase
`III NeoALTTO study randomly assigned 455 patients
`(HER2+ primary breast cancer with tumors greater than 2
`cm in diameter) to receive paclitaxel (T) with trastuzumab
`(TH), with lapatinib (TL), or with both HER2- targeting
`agents (THL). Pathological complete response (pCR) rate
`was significantly higher in the group given lapatinib plus
`trastuzumab plus paclitaxel (THL) (51%) than in the
`group given trastuzumab plus paclitaxel (TH) (29.5%) or
`lapatinib plus paclitaxel (TL) (24.7%) (Baselga, Bradbury
`et al. 2012). Similar results were observed from a
`randomized phase II CHER-LOB study. This is a non-
`comparative, randomized, phase II trial of preoperative
`taxane-anthracycline in combination with trastuzumab,
`lapatinib or combined trastuzumab plus lapatinib in
`patients with HER2-positive, stage II to IIIA operable
`breast cancer. The pCR rates were 25% for chemotherapy
`plus trastuzumab group, 26% for chemotherapy plus
`lapatinib group and 46%
`for chemotherapy plus
`trastuzumab plus
`lapatinib
`treated group (Guarneri,
`Frassoldati et al. 2012).
`In 2012, the third HER2-targeted therapy to
`receive
`regulatory approval was pertuzumab. Like
`trastuzumab, pertuzumab is a humanized monoclonal
`antibody that binds to the extra- cellular domain of HER2
`but binds to a different domain than that of trastuzumab
`(domain II instead of domain IV) (Franklin, Carey et al.
`
`  
`
`46
`
`2004; Sun Yuliang, Dey Nandini et al. 2013). In the
`NeoSphere study, 46% of
`the women who were
`administrated trastuzumab, pertuzumab, plus docetaxel
`(neoadjuvant) achieved a pCR (49 of 107 patients; 45.8%
`[95% CI 36.1—55.7]) 4 (Gianni, Pienkowski et al. 2012).
`The median progression free survival (PFS) in patients
`receiving trastuzumab, pertuzumab and docetaxel in the
`double-blind CLEOPATRA
`trial (808 patients with
`HER2-positive,
`locally
`recurrent, unresectable or
`metastatic breast cancer) was observed to be 18.5 months,
`as compared to 12.4 months in patients who received
`placebo, trastuzumab and docetaxel (Baselga, Bradbury et
`al. 2012). The results of these trials show that (1) in 54%
`of
`the patients enrolled
`in
`the NeoSphere
`trail,
`trastuzumab plus pertuzumab plus docetaxel combination
`fell short of achieving pCR, and (2) the median PFS in
`metastatic breast cancer patients receiving treatment with
`trastuzumab plus pertuzumab plus docetaxel in the
`CLEOPATRA trial was increased by not more than 6
`months. Given
`the
`limited benefits of
`these
`latter
`treatment regimens, there is clearly a room for additional
`improvement.
`The above
`findings suggest
`that currently
`available HER2-targeted therapies are rarely given as
`mono-therapy but are generally given in combination with
`other chemotherapy or chemotherapies. Since toxicities
`associated with chemotherapy can be a substantial source
`of comorbidity for patients with cancer, tumor cell-
`specific delivery of cytotoxic agents (via antibody drug
`conjugate, ADC) is a promising therapeutic approach for
`HER2-overexpressed patient populations. It is clear that
`HER2 remains an active/key target, even after multiple
`lines of treatment. It has been demonstrated from different
`neoadjuvant studies including GeparQuinto (Untch, Loibl
`et al. 2012), NeoALTTO (Baselga, Bradbury et al. 2012),
`CHER-LOB (Guarneri, Frassoldati et al. 2012) and
`NeoSphere
`(Gianni, Pienkowski et al. 2012)
`that
`continued HER2 targeting therapy works even after
`disease progression.
`II. Antibody Drug Conjugates (ADC)
`
`Antibody-drug conjugates (ADCs) represent a
`promising
`therapeutic modality
`for
`the
`clinical
`management of cancer. ADCs are composed of
`recombinant chimeric, humanized or human antibodies
`covalently bound by synthetic linkers to highly cytotoxic
`drugs. The primary objective
`is
`to combine
`the
`pharmacological potency of small (300 to 1000 Dalton)
`cytotoxic drugs with the high specificity of monoclonal
`antibodies that target tumor-associated antigens (Teicher
`and Chari 2011). Inside the cell, the cytotoxic agent is
`released from the antibody and kills the tumor cells.
`ADCs are designed to minimize the systemic toxicities of
`free drug and to augment the anti-tumor activity of the
`monoclonal antibody
`targeting
`tumor. ADCs offer
`significant advantages over conventional chemotherapy—
`they attach specifically to tumor cells with their target
`receptors while not affecting healthy cells that do not have
`sufficient number of those receptors. Patients would be
`expected to experience fewer and less severe adverse
`events than they would with systemic chemotherapy. The
`
`IMMUNOGEN 2027, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Cancer Therapy Vol. 9, page 47
`
`
`
`anti-tumor properties of trastuzumab with the cytotoxic
`activity of the microtubule-inhibitory agent DM1; the
`antibody and the cytotoxic agent are conjugated by means
`of a stable linker (Lewis Phillips, Li et al. 2008; Junttila,
`Li et al. 2011). T-DM1 allows HER2-overexpressing
`tumor cell-specific drug delivery, thereby improving
`therapeutic index and minimizing exposure of normal
`tissue to the toxicities of the DM component. The in vitro
`activity of maytansine ((DM part of the T-DM1) was
`highly substantial—100 fold more potent than vinca
`alkaloids (an anti-mitotic and anti-microtubule agent) and
`24 to 270 fold more potent than paclitaxel (Remillard,
`Rebhun et al. 1975; Cassady, Chan et al. 2004; Junttila, Li
`et al. 2011). It has been previously described by others
`that maytansine may bind to tubulin leading to a
`disassembly of microtubules which prevents tubulin
`spiralization (E H 2008). Furthermore, microtubule
`polymerization /depolymerization plays an important role
`in tumor-induced angiogenesis through the HIF1alpha-
`VEGF signaling axis (Mabjeesh, Escuin et al. 2003).
`
`
`
`
`
`
`Figure 1: Schematic illustrations of antibody drug conjugate
`(ADC): ADCs are unique combination of a precise and targeted
`monoclonal antibody, a stable linker and a potent cytotoxic
`agent.
`
`
`2.2 Pre-clinical and clinical efficacy
`Extensive analysis of T-DM1 in preclinical studies
`has shown that T-DM1 combines the distinct mechanisms
`of action of both DM1 and trastuzumab, and has
`antitumor activity in trastuzumab- and lapatinib-refractory
`experimental models (Barok, Tanner et al. 2011; Junttila,
`Li et al. 2011; Sun Yuliang, Dey Nandini et al. 2013).
`This is extremely important because trastuzumab and
`lapatinib are established for the treatment of HER2-
`postitive metastatic
`breast
`cancer
`(MBC). The
`mechanisms of action for trastuzumab include inhibition
`of the PI3K/AKT/mTOR signaling pathway, inhibition of
`HER2 shedding, and Fcγ receptor–mediated engagement
`of immune cells, which may result in antibody-dependent
`cellular cytotoxicity (Spector and Blackwell 2009; De,
`Hasmann et al. 2013).
`
`  
`level of acceptable target expression may depend on the
`target (e.g., HER2 needs high overexpression to serve as a
`good ADC target (Burris, Rugo et al. 2011) while an ADC
`targeting CD19 can be active on cell lines bearing only
`30,000 or so receptors (Blanc, Bousseau et al. 2011).
`However, delivery of a lethal quantity of a tubulin-acting
`payload into cancer cell via antibody-mediated uptake of
`an ADC may be difficult to achieve below ~10,000
`receptors/cell (Lapusan, Vidriales et al. 2012). Hence,
`HER2 protein is an ideal target for ADC as breast cancers
`with the amplification of HER2 gene have up to 1-2
`million receptors expressed per cell (Venter, Tuzi et al.
`1987).
`The leading ADC in the clinical arena is T-DM1, which is
`composed of Genentech/Roche’s HER2
`targeting,
`humanized IgG1 trastuzumab conjugated with DM1 via
`an non-cleavable thioether link formed using the non-
`reducable
`thioether crosslinker using
`ImmunoGen’s
`platform technology (Lewis Phillips, Li et al. 2008). This
`non-cleavable linker means the linker does not separate
`from the cytotoxin (maytansine) but uses a different
`mechanism to activate the cytotoxin inside the cancer
`cells. The rapid development of T-DM1 was greatly aided
`by the fact that trastuzumab was already marketed and
`tests were already in use for patient selection of likely
`better responders, such as those overexpressing HER2. In
`2000, gemtuzumab ozogamicin (Mylotarg) became the
`first ADC to be approved by the U.S. Food and Drug
`Administration
`(FDA)
`for
`the
`treatment of acute
`myelogenous leukemia (AML). However, this ADC was
`withdrawn from the market in 2010 because, in post-
`marketing follow-up clinical trials, it failed to meet
`prospective efficacy targets that were required as a
`condition of its accelerated approval by the FDA. T-DM1
`is not the first effective ADC, that distinction belongs to
`brentuximab vedotin (SGN-35, Adcetris), developed by
`Seattle Genetics, which was granted accelerated approval
`in 2011. Indicated for the treatment of Hodgkin’s
`lymphoma and systemic anaplastic large cell lymphoma,
`Adcetris is a CD30 antibody linked to the cytotoxin
`auristatin, which blocks cell division.
`
`
`
`2.1 T-DM1
`
`Chemistry
`
`Preclinical studies demonstrated synergistic or
`
`additive interactions of trastuzumab with a variety of anti-
`microtubulin agents, including maytansines (Baselga,
`Norton et al. 1998; Pegram, Hsu et al. 1999; Burstein,
`Keshaviah et al. 2007). Because the initial studies with
`maytansine proved to be associated with intolerable
`toxicities such as nausea and peripheral neuropathy,
`laboratory-based translational cancer research was geared
`to the development of various trastuzumab-maytansinoid
`conjugates (Lewis Phillips, Li et al. 2008). This strategy
`culminated in the development of an antibody drug
`conjugate (ADC), trastuzumab linked to DM1 (derivative
`of maytansine) (Figure.1). Trastuzumab emtansine (T-
`DM1) is an ADC that incorporates the HER2-targeted
`
`  
`
`47
`
`IMMUNOGEN 2027, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Pradip De et al: T-DM1: a giant step forwards in HER2 therapeutics  
`
`same
`these
`retains
`importance, T-DM1
`Of
`trastuzumab
`mechanisms of action of unconjugated
`(Junttila, Li et al. 2011). The cytotoxic antitubulin agent
`DM1 remains attached to trastuzumab outside the cell
`because of the non- cleavable linker (nonreduciable
`thioether) molecule until the entire ADC is transported
`into the cytoplasm through endocytosis. It is assumed that
`
`intra-lysosomal proteolytic
`then undergoes
`T-DM1
`degradation resulting in the release of nonreduciable
`thioether
`linked-DM1 and consequent antitubulin-
`associated cell death (Lewis Phillips, Li et al. 2008)
`(Figure. 2).
`
`
`
`
`
`
`Figure 2: Schematic representation of mechanism of action of T-DM1: T-DM1 locks onto HER2 receptor in HER2+ breast tumor
`cells with drug in tow. Once internalization, the antibody and receptor begins to break each other. Then antibody breaks into pieces,
`thereby release the drug. The cytotoxic drug begins to disrupt the cell, killing the cell and eventually augments antitumor activity.
`It has been reported that T-DM1 has been shown
`months and the clinical benefit rate (CBR; objective
`to induce apoptotic cell death in a trastuzumab-resistant
`response rate [ORR] plus stable disease at 6 months) was
`xenograft tumor model (Barok, Tanner et al. 2011). In our
`73% (Krop, Beeram et al. 2010). Most commonly
`cell based model systems, we clearly demonstrated that T-
`reported grade 1 or 2 adverse events at MTD were
`DM1 treatment associated cell death is significantly
`thrombocytopenia
`(54.2%),
`elevated
`transaminases
`higher (by annexinV staining) in HER2+/trastuzumab-
`(41.7%), fatigue (37.5%), anemia (29.2%), and nausea
`sensitive,
`trastuzumab-resistant and HER2+/PIK3CA
`(25%). Cardiac toxicity was not reported. This was the
`mutated cells when compared to trastuzumab alone
`first phase I breast cancer trial with T-DM1 and
`treated cells (data not shown).
`established MTD of 3.6 mg/kg every 3 weeks. The
`Initially T-DM1 was evaluated as a single agent
`original phase I study also evaluated a weekly schedule of
`in a dose escalation phase I trial in patients with HER2+
`T-DM1 with 1.2 mg/kg, 1.6 mg/kg, and 2 mg/kg. In this
`metastatic breast cancer (MBC) who previously received
`weekly schedule, partial responses were noted in four
`a trastuzumab- containing chemotherapy. T-DM1 was
`patients (57%). Grade 3 or higher adverse events were
`administered at various doses on a weekly (23 weeks) or
`limited to rapidly reversible thrombocytopenia in one
`every 3 weeks schedule. The maximum tolerated dose
`patient. No dose limiting toxicity was observed (S. N.
`(MTD) was 3.6 mg/kg every 3 weeks, based on the dose-
`Holden, M. Beeram et al. 2008).
`limiting toxicity (DLT) of grade 4 thrombocytopenia at
`A proof-of-concept phase II study by Burris et al
`4.8 mg/kg every 3 weeks (Krop, Beeram et al. 2010). In a
`showed that single agent T-DM1 (3.6 mg/kg given every
`group of 15 patients receiving 3.6 mg/kg every 3 weeks,
`three weeks) in 112 patients with HER2-positive MBC,
`the median progression free survival (PFS) was 10.4
`who had progressed with previous chemotherapy and
`
`  
`
`48
`
`IMMUNOGEN 2027, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Cancer Therapy Vol. 9, page 49
`
`
`
`rates of grade 3 or 4 bleeding events were low in both
`groups (1.4% and 0.8% respectively). In the majority of
`patients, left ventricular ejection fraction of 45% or more
`was maintained during the study treatment (in 97% of
`patients in the T-DM1 group and 93% of patients in the
`lapatinib plus capcetabine group). The incidence of
`diarrhea,
`nausea,
`vomiting
`and
`palmar-planter
`erythrodysesthesia were much higher with lapatinib plus
`capecitabine group. Tumor biomarkers data from the
`EMILIA trial also revealed that PIK3CA mutations in
`HER2-positive
`tumors did not
`compromise
`the
`effectiveness of T-DM1, but led to poorer responses to
`conventional HER2-
`targeted
`therapies
`such
`as
`trastuzumab and lapatinib. T-DM1 patients with mutated
`PIK3CA had 10.9 months progression-free survival, and
`9.8 months for those with wild-type PIK3CA. However,
`PIK3CA status did make a difference for the capecitabine-
`lapatinib treated patients: progression-free survival was
`4.3 months for patients with mutated PIK3CA and 6.4
`months for those with the wild type PIK3CA (Baselga J,
`Verma S et al. 2013). The two major lessons we learned
`from the above mentioned trials with T-DM1 are: a) there
`is a relative lack of side-effects following the treatment
`with T-DM1 and b) T-DM1 obviates the need for
`chemotherapy.
`Trastuzumab-induced
`cardio-toxicity
`(symptomatic or asymptomatic) is a major concern.
`Although, the cardio-toxicity varies according to the
`definition used in different studies, it has been reported to
`be as high as 30% when associated with anthracyclines
`(Telli, Hunt et al. 2007; Guglin, Hartlage et al. 2009).
`T-DM1 had a better safety profile compared to
`trastuzumab in this context and no significant dose-
`limiting cardio-toxicity was observed with T-DM1 in
`heavily pre-treated patients. T-DM1 has exhibited a
`unique capacity to discriminate between cancer cells and
`normal cells in terms of cytotoxic drug delivery. EMILIA
`study also showed that patients receiving T-DM1 whose
`tumors had above-median levels of HER2 had a median
`overall survival of 34.1 months, vs. 26.5 months for
`patients with lower levels. Progression-free survival with
`T-DM1 was 10.6 months for patients with higher HER2
`levels vs. 8.2 months for lower levels (Baselga J, Verma S
`et al. 2013). Data suggest
`that
`the greater
`the
`overexpression of HER2 in a breast tumor, the more
`sensitive the tumor is to anti-HER2 therapy. Overall these
`results take targeted therapy for HER2-positive breast
`cancer to a newer level.
`MARIANNE, is a phase III randomized study of
`T-DM1 with or without pertuzumab (T- DM1 and
`pertuzumab bind to different epitopes of HER2) compared
`with trastuzuamb plus taxane for the first line treatment of
`HER2-positive, progressive or recurrent locally advanced
`or metastatic breast cancer patients. With 1,092 enrolled
`patients, MARIANNE will compare the efficacy of T-
`DM1 plus pertuzumab, T-DM1 plus placebo, and the
`combination of
`trastuzumab plus a
`taxane. The
`independent Data Monitoring Committee which assesses
`safety data has recently recommended continuation of the
`study without any modification. Brisk completion of
`
`  
`HER2-directed therapy, had anti-tumor activity (Burris,
`Rugo et al. 2011). In this study, the overall response rate
`by
`independent review was 25.9% and 37.5% by
`investigator assessment, including 4 complete responses.
`The median PFS was 4.6 months. In the following study,
`Krop and colleagues gave T-DM1
`(3.6 mg/kg
`intravenously every three weeks) to patients (n=110) with
`HER2-positive MBC who had prior treatment with
`trastuzumab, lapatinib, an antracycline, a taxane and
`capecitabine. The overall response rate was 34.5%, CBR
`was 48.2%, median PFS was 6.9 months and median
`duration of response was 7.2 months. In patients with
`confirmed HER2-positivity
`(n=80) by
`retrospective
`central testing, the response rate was 41.3% and median
`PFS was 7.3 months (Krop, LoRusso et al. 2012). Most
`adverse events were grades 1 or 2; the most frequent
`grade 3 events were thrombocytopenia (9%), fatigue
`(4.5%) and cellulitis (4.6%). Sara Hurvitz and group
`recently published (Hurvitz, Dirix et al. 2013) their phase
`II
`randomized study of T-DM1
`(n= 67) versus
`trastuzumab plus docetaxel (n= 70) in patients with
`HER2-positive MBC. The objective response rate was
`64% on T-DM1 arm versus 58% for patients received
`trastuzumab plus docetaxel. Furthermore, the progression
`free survival in the T-DM1 arm was 14.2 months versus
`9.2 months for patients receiving standard- of-care, an
`improvement in favor of T-DM1 that was significant
`despite the relatively small number of patients. T-DM1
`had a favorable safety profile, with fewer grade 3 adverse
`events
`(AEs),
`and AEs
`leading
`to
`treatment
`discontinuations was 7.2% (for T-DM1) versus 40.9%
`(tarstuzumab plus docetaxel).
`The above phase I and II studies led to the
`landmark phase III EMILIA trial, which was published
`recently in New England Journal of Medicine by Verma,
`Blackwell and the EMILIA study group (Verma, Miles et
`al. 2012) and
`subsequently published associated
`biomarker analysis by Baselga and group at the AACR
`2013 annual meeting (Baselga J, Verma S et al. 2013). In
`this trial, 991 patients with advanced HER2-positive
`breast cancer whose disease had progressed through
`treatment with trastuzumab plus taxane were assigned to
`T-DM1 or lapatinib plus capecitabine, an FDA approved
`and standard treatment option in this setting. The median
`PFS as assessed by independent review was 9.6 months
`with T-DM1 versus 6.4 months with lapatinib plus
`capecitabine (p<0.001, HR 0.65) and median overall
`survival at the second interim analysis was significantly
`improved in the T-DM1 arm (30.9 months versus 25.1
`months, HR 90.68). The objective response rate was also
`significantly higher with T-DM1 (43.6%) than lapatinib
`plus capecitabine (30.8%). Rates of grade 3 or 4 AEs were
`higher with lapatinib plus capecitabine than within the T-
`DM1 arm (57% versus 41%). The
`incidences of
`thrombocytopenia were higher
`in
`the T-DM1 arm;
`although the majority of these patients were able to
`continue treatment (2% discontinued T-DM1 due to
`thrombocytopenia). The overall incidence of bleeding
`events was higher with T-DM1 (29.8% versus 15.8%);
`
`  
`
`49
`
`IMMUNOGEN 2027, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Pradip De et al: T-DM1: a giant step forwards in HER2 therapeutics  
`
`accrual and data analyses are eagerly awaited (Ellis EA,
`CH et al. 2011). Roche/Genentech also is actively
`recruiting metastatic HER2+ breast cancer patients into
`the TH3RESA
`trial
`(NCT01419197) at 210 sites,
`including 90 within the US. Women entering this trial will
`be randomized at a 2 to 1 ratio to receive T-DM1 (3.6
`mg/kg intravenously for 3 weeks) or treatment of the
`physician's choice. Anticipated time on study treatment is
`until disease progression or unacceptable toxicity occurs.
`A
`randomized, multicenter, open-label phase
`III
`KATHERINE study (NCT01772472) of T-DM1 vs.
`trastuzumab as adjuvant therapy for patients with HER2+
`
`primary breast cancer who have residual tumor present
`pathologically in the breast or axillary lymph nodes
`following preoperative therapy, which was initiated in
`April 2013. Other ongoing phase II trials are currently
`exploring the feasibility and/or antitumor potency of T-
`DM1 in the adjuvant (NCT01196052) and neoadjuvant
`setting (NCT01745965) of HER2-positive BC (see Table
`1).
`
`
`
`
`
`
`Table 1: Ongoing trials with T-DM1 in HER2 + breast cancer
`MBC: Metastatic breast cancer HT: Hormone treatment AEs: Adverse events DLTs: Dose limiting toxicities PFS:
`Progression free survival pCR: Pathological complete response IDFS: Invasive disease free survival
`
`
`
`2.3 Pharmacokinetic and metabolic
`profile of T-DM1
`The pharmacokinetics of T-DM1 has been assessed
`in preclinical and clinical studies. The results showed that
`T-DM1 exhibits dose-proportional pharmacokinetics in
`non– trastuzumab-binding species (i.e., mice and rats)
`(LoRusso, Weiss et al. 2011) and a dose- dependent
`decrease in clearance associated with increasing dose in
`trastuzumab binding species (i.e., cynomolgus monkeys
`and humans) (Girish S, Gupta M et al. 2011). Results
`from a preclinical absorption, distribution, metabolism
`and excretion study of T-DM1 in rats suggest that T-DM1
`nonspecifically
`distributes
`to
`tissues
`without
`accumulation. The major elimination routes of DM1-
`containing metabolites are through fecal/biliary (~80%)
`and urine (<10%) (Shen, Bumbaca et al. 2012).
`
`
`
`
`
`
`The clinical PK of T-DM1 shows that while the
`conjugate is quite stable in circulation, it nevertheless
`appears to show a slow rate of maytansiniod loss over
`time (Krop, Beeram et al. 2010; Burris, Rugo et al. 2011)
`and very low levels of free DM1 were reported to be
`present in plasma samples from patients treated with T-
`DM1 (Burris, Rugo et al. 2011). The median half-life of
`T-DM1 is 4.5 days and steady state is achieved in cycle 2
`(Gupta, Lorusso et al. 2012). Pharmacokinetics-based
`drug interactions between T-DM1 and the HER2-targeted
`monoclonal antibody pertuzumab, or T-DM1 and
`paclitaxel showed that the combination had no effect on
`the pharmacokinetics of the individual agents and had a
`low risk for drug interactions (Lu D, Krop I et al. 2010;
`Lu, Burris et al. 2012).
`
`
`
`
`  
`
`50
`
`IMMUNOGEN 2027, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`  
`
`2.4 T-DM1 and resistance mechanism
`The design of cancer
`therapy has become
`increasingly sophisticated, yet there is no cancer treatment
`that is 100% effective against disseminated cancer.
`Resistance to treatment with anticancer drugs results from
`a variety of factors including individual variations in
`patients, somatic cell genetic differences in tumors, and
`signaling pathway alteration/upregulation. Frequently,
`resistance is intrinsic to the cancer but as therapy becomes
`more and more effective, acquired resistance has also
`become common. Trastuzumab-containing therapy is now
`an established standard of care across all HER2-positive
`breast cancer disease stages. However, despite the robust
`clinical efficacy of trastuzumab in HER2-positive MBC,
`primary and secondary resistance remains a clinical
`challenge. Alterations of
`signal
`transducers
`lying
`downstream of HER2, which
`facilitate
`signaling
`independently of the HER2 kinase, have been extensively
`studied as potential mechanisms of
`trastuzumab
`resistance. T-DM1 is a relatively new drug in the horizon
`and the EMILIA trial revealed that PI3K pathway
`upregulation (via activating mutation of PIK3CA) did not
`hinder T-DM1’s efficacy. Pegram and group recently
`reported that despite inactivation of the PI3K-AKT
`signaling pathway, HER2 overexpressed
`lapatinib-
`resistant cells exhibited activation of mTORC1 and its
`downstream signaling pathway (Jegg, Ward et al. 2012).
`Recently, Elkabets and group also reported that breast
`cancer cells (containing PIK3CA mutation) that were
`resistant to p110α- isoform-specific inhibitor (BYL719)
`had persistently active pS6 expression, although AKT
`phosphorylation was inhibited (Elkabets, Vora et al.
`2013). Along the same line, our cell-based preclinical data
`showed that T-DM1 blocked AKT activation in HER2-
`positive breast cancer cells (BT474) but failed to block
`downstream
`of mTORC1
`signaling molecule
`(phosphorylation of P70S6K) (Sun, Dey et al. 2013). This
`data suggest that T-DM1 alone may not be sufficient to
`completely inactivate the PI3K-AKT-mTOR signaling
`
`References:
`Andersson, M., E. Lidbrink, et al. (2011). "Phase III
`randomized
`study
`comparing
`docetaxel
`plustrastuzumab
`with
`vinorelbine
`plus
`trastuzumab as first-line therapy of metastatic
`or13locally advanced human epidermal growth
`factor receptor 2-positive breast cancer:
`the
`HERNATA study." J Clin Oncol 29(3): 264-271.
`Arteaga, C. L., M. X. Sliwkowski, et al. (2012).
`"Treatment of HER2-positive breast cancer:
`current status and future perspectives." Nat Rev
`Clin Oncol 9(1): 16-32.
`Barok, M., M. Tanner, et al. (2011). "Trastuzumab-
`DM1 causes tumour growth inhibition by mitotic
`catastrophe in trastuzumab-resistant breast cancer
`cells in vivo." Breast Cancer Res 13(2): R46.
` Barok, M., M. Tanner, et al. (2011). "Trastuzumab-
`DM1 is highly effective in preclinical models of
`
`Cancer Therapy Vol. 9, page 51
`
`
`
`pathway. Literature references suggest that activation of
`the PI3K-AKT-mTOR pathway is one of the major causes
`for drug
`resistance
`including
`chemotherapy
`and
`trastuzumab
`therapy
`in different
`cancer mod

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