throbber
Drug Evaluation
`
`Future Oncology
`
`Keywords
`
`n antisense oligonucleotide
`n cancer n chaperone protein
`n clusterin n custirsen
`n prostate cancer
`
`Custirsen (OGX-011): a second-
`generation antisense inhibitor of
`clusterin in development for the
`treatment of prostate cancer
`
`Robert Zielinski1 & Kim N Chi*2
`1Bristish Columbia Cancer Agency, 600 West 10th Avenue, Vancouver, British Columbia, V5Z 4E6, Canada
`2University of British Columbia, Vancouver, Canada
`*Author for correspondence: Tel.: +1 604 877 6000 ext. 2746 n Fax: +1 604 877 0585 n kchi@bccancer.bc.ca
`
`Clusterin is a stress-induced cytoprotective chaperone that confers broad-
`spectrum treatment resistance and is overexpressed across a number of cancers.
`custirsen (OGX-011) is a promising novel second-generation antisense inhibitor
`of clusterin in clinical development. This article describes the mechanism of
`action and safety profile of OGX-011 and details the Phase I and II results in human
`solid organ malignancies. Two Phase III registration trials are currently under
`recruitment evaluating OGX-011 in combination with chemotherapy in patients
`with metastatic castration-resistant prostate cancer. These studies not only have
`the potential to significantly alter the standard of care in prostate cancer, but
`would also endorse a new class of targets and targeted therapy approach for
`cancer.
`
`Custirsen (OGX-011, OncoGenex Pharma-
`ceuticals Inc., WA, USA and Teva Pharma-
`ceuticals Ltd, Petach Tikva, Israel) is a novel
`antisense oligonucleotide compound that tar-
`gets expression of the clusterin gene. This arti-
`cle describes clusterin’s structure, function and
`influential role in the development of treatment-
`resistant cancers. The rationale for employing
`antisense technology, and in particular OGX-
`011, to target gene overexpression is discussed.
`Finally, detailed efficacy and tolerability data
`are presented that establish the safety of OGX-
`011 in combination with chemotherapy and
`the potential role of OGX-011 in overcoming
`t reatment resistance in many malignancies.
`Clusterin is a secretable cytoprotective protein
`that is upregulated in response to cellular stress,
`which includes standard cancer treatments such
`as hormone-, radiation- and chemo-therapy, and
`is implicated in treatment resistance. Clusterin is
`expressed in many malignancies including pros-
`tate, breast, ovarian, non-small-cell lung, colon,
`renal, urothelial and pancreatic cancers, and
`anaplastic large-cell lymphoma and melanoma
`[1–7]. Since clusterin binds to a wide variety of cli-
`ent proteins involved in a diverse array of biologi-
`cal processes and regulated by HSF-1 it is viewed
`as a heat shock-like protein that chaperones and
`stabilizes proteins at times of cellular stress to
`promote cell survival. As clusterin isoforms are
`also secreted extracellularly, some investigators
`suggest that clusterin may be the first identified
`secreted mammalian chaperone [8].
`
`OGX-011 is a phosphorothioate antisense oli-
`gonucleotide (ASO) inhibitor of clusterin expres-
`sion that incorporates second-generation chemis-
`try in the form of a 2´-methoxyethyl (2´-MOE)
`modifications to increase tissue half-life, enhance
`potency and decrease nonsequence-specific tox-
`icity of the molecule. The clusterin gene was
`screened with a series of ASO sequences, and
`the most potent at inhibiting expression was the
`OGX-011 sequence targeting the translation ini-
`tiation site. Preclinical studies have demonstrated
`the potency of OGX-011 at inhibiting clusterin
`expression in vitro and in vivo, resulting in the
`therapeutic enhancement of s tandard anticancer
`treatments including hormone-, r adiation- and
`chemo-therapy [9].
`Phase I and II clinical trials with OGX-011 have
`demonstrated biologically active dosing and suc-
`cessful delivery of the drug to malignant tissues
`with inhibition of clusterin expression in both
`tissue and serum. A randomized Phase II study
`of OGX-011 in combination with docetaxel
`in patients with metastatic castration-resistant
`prostate cancer (CRPC) demonstrated a survival
`advantage for patients receiving the combination
`[10]. This led to the current registration pathway
`for OGX-011 in combination with standard
`therapies in patients with metastatic CRPC.
`For prostate cancer in 2012, standard treat-
`ments for the metastatic CRPC patient include
`chemotherapeutics (docetaxel [11], mitoxantrone
`[12] and cabazitaxel [13]), bone-targeting agents
`(zoledronic acid [14] and denosumab [15]),
`
`10.2217/FON.12.129 © 2012 Future Medicine Ltd
`
`Future Oncol. (2012) 8(10), 1239–1251
`
`ISSN 1479-6694
`
`1239
`
`part of
`
`For reprint orders, please contact: reprints@futuremedicine.com
`
`AVENTIS EXHIBIT 2046
`Mylan v. Aventis, IPR2016-00712
`
`

`
`immuno therapy (sipuleucel-T [16]) and a potent
`inhibitor of androgen biosynthesis (abiraterone
`acetate [17]). A novel, next-generation androgen
`receptor inhibitor, MDV3100 (enzalutamide)
`[18], and the infusional radioisotope radium-223
`[19], have also demonstrated improved over-
`all survival in patients after docetaxel or in
`docetaxel-ineligible patients (in the case of
`radium-223). Both agents remain investigational
`and are expected to enter the CRPC market in
`2012–2013. Despite these therapeutic advance-
`ments, treatment for metastatic CRPC remains
`palliative with a median overall survival of
`approximately 18 months [11]. Due to the role of
`clusterin in cell survival across multiple mecha-
`nisms and broad-spectrum treatment resistance,
`inhibition of clusterin with OGX-011 has the
`potential to improve outcomes across all these
`therapeutic modalities.
`
`Overview of the market
`Prostate cancer is the most frequently diagnosed
`cancer other than skin cancer and the second-
`leading cause of death from cancer in men in
`North America [20]. In 2011, prostate can-
`cer was diagnosed in 240,000 men and led to
`nearly 33,000 deaths in the USA. Worldwide,
`approximately 910,000 cases of prostate can-
`cer were recorded in 2008, accounting for
`approximately 14% of all new cancer cases in
`men [101]. CRPC is the lethal phenotype of the
`disease that may emerge after standard andro-
`gen ablation therapy for advanced disease. The
`term CRPC is commonly used when a patient’s
`cancer progresses despite castrate levels of testos-
`terone (<50 ng/ml). The therapeutic landscape
`has recently become enriched with multiple
`
`new agents that are either US FDA approved or
`recently reported in Phase III trials with positive
`results in terms of overall survival, progression-
`free survival (PFS) and symptomatic outcomes.
`Docetaxel was the first chemotherapeutic agent
`to demonstrate a survival advantage in CRPC
`[11], whereas mitoxantrone was only approved for
`palliation of symptoms in a similar patient group
`[12]. More recently cabazitaxel, a third-generation
`taxane, gained FDA approval by establishing a
`2.4-month survival advantage in patients pro-
`gressing after docetaxel [13]. Sipuleucel-T, the
`first FDA-approved vaccine-based therapy for
`any malignancy, established a 4.1-month sur-
`vival advantage in patients with asymptomatic
`or minimally symptomatic metastatic CRPC [16].
`New agents that target persistent androgen recep-
`tor signaling in CRPC through either inhibition
`of extragonadal steroidogenesis or direct inhi-
`bition of the androgen receptor have also been
`developed. Abiraterone acetate is currently the
`first approved drug in this class after establishing
`a 3.8-month median overall survival advantage
`in the postdocetaxel setting [17]. MDV3100 is a
`novel oral anti-androgen that directly inhibits
`the androgen receptor, which has also shown
`a survival advantage in the same patient group
`[18]. Lastly, the bone-targeting agents zoledronic
`acid and denosumab are also employed in the
`CRPC treatment model after demonstrating
`reduction in skeletal-related events and postpon-
`ing development of bone metastases [14,15]. Table 1
`summarizes both the currently approved agents
`and promising agents with unpublished positive
`Phase III results.
`The current approach to managing metastatic
`CRPC is under major transformation as the
`
`Table 1. Agents that are currently US FDA approved or with positive Phase III study
`data in castration-resistant prostate cancer.
`
`Agent
`
`Enzalutamide
`(MDV3100)
`
`Sipuleucel-T
`
`Mode of action
`
`Benefit
`
`Anti-androgen
`
`4.8-month median OS
`
`Immunotherapy
`
`4.1-month median OS
`
`Abiraterone acetate
`
`Anti-androgen
`
`3.9-month median OS
`
`Alpharadin
`
`Docetaxel
`
`Cabazitaxel
`
`Mitoxantrone
`
`Denosumab
`
`Radiopharmaceutical
`
`2.8-month medial OS
`
`Chemotherapeutic
`
`2.4-month median OS
`
`Chemotherapeutic
`
`2.4-month median OS
`
`Chemotherapeutic
`
`Pain palliation
`
`RANK ligand inhibitor
`
`4.2-month bone metastasis-free survival
`
`Inhibitor of osteoblasts Delayed time to SRE†
`Zoledronic acid
`†Pathologic bone fractures, spinal cord compression, surgery to bone, radiation therapy to bone (including the use of
`radioisotopes) or a change of antineoplastic therapy to treat bone pain.
`OS: Overall survival; SRE: Skeletal-related event.
`
`Ref.
`[18]
`
`[16]
`
`[17]
`
`[19]
`
`[11]
`
`[13]
`
`[12]
`
`[15]
`
`[14]
`
`1240
`
`Future Oncol. (2012) 8(10)
`
`future science group
`
`Drug Evaluation Zielinski & Chi
`
`

`
`optimal sequencing of these new agents contin-
`ues to evolve. Yet perhaps the greater challenge is
`how best to manage the emergence of universal
`treatment resistance. Targeting the fundamental
`effectors of treatment resistance is an appealing
`approach; inhibition of heat-shock and cellular
`chaperone proteins fit into this category.
`
`The target: clusterin
`Clusterin is a single-copy gene, organized into
`nine exons (eight introns) and a 5´-untranslated
`region, located on chromosome 8p21–p12 and
`extending more than 16 kb [21,22]. It is highly
`conserved across species and constitutively
`expressed in almost all mammalian tissues. In
`humans, the clusterin gene codes for two secre-
`tory isoforms (sCLU-1 and sCLU-2), originat-
`ing from transcriptional start sites in exons 1
`and 2, respectively; only sCLU-2 is expressed
`in subprimates. Secreted clusterin (sCLU) is an
`endoplasmic reticulum-targeted, 449-amino
`acid polypeptide that represents the predomi-
`nant translation product of the human gene.
`Although sCLU is cytoprotective and anti-apop-
`totic, a pro-apoptotic activity ~55-kDa nuclear
`(nCLU) splice variant lacking exon 2 and the
`endoplasmic reticulum signal peptide has been
`described [23,24]. Analysis of the promoter has
`revealed numerous transcription factor-binding
`sites and also a conserved clusterin element, rec-
`ognized by HSF-1/HSF-2 heterocomplexes [25].
`Clusterin is transcriptionally activated by HSF-1
`in response to cellular stress [26,27].
`In malignancy, clusterin has largely been
`defined as an inhibitor of apoptosis [28–31]. Its
`anti-apoptotic actions have been described as
`functioning through a variety of mechanisms
`including inhibition of activated Bax [32] and
`enhanced survival signaling through upregu-
`lation of PI3k/Akt pathway signaling [33].
`Clusterin has also been implicated in tumo-
`rigenesis through loss of the tumor suppres-
`sor gene Nkx3.1 [34], and its overexpression in
`response to hormonal ablation, chemotherapy
`and radiotherapy contributes to treatment
`resistance [6,29,35]. This clusterin-induced treat-
`ment-resistant phenotype has been much stud-
`ied in prostate cancer and docetaxel-resistant
`prostate tumor cells have been resensitized to
`docetaxel following exposure to OGX-011
`[36,37]. However, drug resistance induced by
`clusterin has been observed within other tumor
`types and with chemotherapy agents includ-
`ing doxorubicin, camptothecin, cisplatin,
`5-fluorouracil, gemcitabine, dacarbazine and
`etoposide [2,32,38–42].
`
`Introduction to the compound
`OGX-011 is a novel second-generation 2´-MOE-
`modified phosphorothioate ASO complementary
`to clusterin mRNA. OGX-011 is a potent inhibi-
`tor of clusterin expression in vitro, in vivo and in
`humans in clinical trials. As a single agent and
`in combination with chemotherapy, OGX-011
`has been well-tolerated in Phase I trials and with
`promising activity in Phase II clinical trials.
`Registration Phase III trials are underway with
`OGX-011 in combination with chemotherapy in
`patients with metastatic CRPC.
`
`Chemistry
`ASO therapy is one strategy to specifically target
`functionally relevant genes. ASOs are chemically
`modified stretches of single-strand DNA com-
`plementary to mRNA regions of a target gene
`that inhibit translation by forming RNA/DNA
`duplexes, thereby reducing mRNA and pro-
`tein levels of the target gene [43]. OGX-011 is
`a 21-nucleoside ASO complementary to the
`clusterin exon 2 mRNA AUG translation ini-
`tiation site, with one CpG motif. The custirsen
`sequence was identified as the most potent to
`inhibit clusterin expression after the gene was
`‘walked’ with a series of antisense sequences.
`OGX-011 is a second-generation phospho-
`rothioate and incorporates the 2´-O-2´-MOE
`modification with four 2´-MOE-modified nucl-
`eosides at the 3´ side, four 2´-MOE-modified
`nucleosides at the 5´ side and 13 2´-deoxyribo-
`nucleosides in between (referred to as a 4-13-4
`MOE gapmer).
`Phosphorothioate ASOs are water-soluble, sta-
`ble agents resistant to nuclease digestion through
`substitution of a nonbridging phosphoryl oxy-
`gen of DNA with sulfur [44]. In clinical trials, a
`major technical limitation with the first genera-
`tion of phosphorothioate ASOs was the require-
`ment for continuous or frequent intravenous
`infusions owing to the short tissue half-life of
`these agents. The approach to overcome this has
`been by a 2’-MOE modification to the 2´-posi-
`tion of the carbohydrate moiety. This forms
`ASO–RNA duplexes with a significantly higher
`affinity relative to unmodified phosphorothioate
`ASOs. This increased affinity has been shown to
`result in improved antisense potency in vitro and
`in vivo. In addition, 2´-MOE second-generation
`ASOs display significantly improved resistance
`against nuclease-mediated metabolism resulting
`in an improved tissue half-life in vivo, which
`produces a longer duration of action and allows
`for an intermittent dosing regimen [45]. Finally,
`these second-generation phosphorothioate
`
`1241
`
`future science group
`
`www.futuremedicine.com
`
`Custirsen (OGX-011): a second-generation antisense inhibitor of clusterin Drug Evaluation
`
`

`
`ASOs have the potential for an improved safety
`profile relative to u nmodified phosphorothioate
`ASOs [46].
`
`Pharmacokinetics
`The disposition and metabolism of OGX-011
`was measured in mice and monkeys. Consistent
`with pharmacokinetic (PK) studies with other
`ASOs, plasma was rapidly cleared of OGX-011
`in both species [47]. Plasma concentrations of
`OGX-011 generally peaked at the end of the
`60-min intravenous infusion period and then
`decreased in an apparent biexponential fash-
`ion that included two distinct half-lives. Mean
`concentrations increased with dose in all tissues
`examined, except for brain where no measur-
`able drug could be detected. Approximately
`90% of the compound was found in the par-
`ent form in all tissues at all time points. PK
`parameters were similar at the beginning and
`end of the treatment period in both species,
`suggesting no plasma accumulation or changes
`in plasma kinetics after multiple dosing in
`either species.
`In humans, plasma PK parameters for
`OGX-011 have followed predictions from pre-
`clinical studies. The first-in-man Phase I trial
`[48] was uniquely designed to permit a dose-
`dependent analysis of OGX-011 levels in the
`plasma and prostatic tissue. The mean plasma
`distribution half-life was 3.3 h at the 640-mg
`dose. Average peak concentrations and area
`under the curve were dose dependent and dis-
`played proportional and predictable increases in
`a linear fashion. This study also demonstrated
`concentrations in prostate cancer cells of full-
`length OGX-011 at levels sufficient for inhibi-
`tion of clusterin expression. Furthermore, the
`prostate tissue concentrations were proven to
`be dose dependent: 320 mg delivered 223 nM
`tissue concentration and 640 mg delivered
`644 nM into the prostate tissue. PK plasma
`parameters for humans are detailed in Table 2.
`As ASOs are degraded by both serum and intra-
`cellular nucleases [45] and only 7% of the drug is
`excreted unchanged in the urine, tissue distribu-
`tion of OGX-011 dominates plasma clearance.
`
`Pharmacodynamics
`OGX-011 is a potent inhibitor of clusterin
`expression in in vitro and in vivo laboratory mod-
`els [45]. Furthermore, because of the targeting to
`exon 2 of clusterin RNA there is a specificity for
`OGX-011 to inhibit only the expression of the
`anti-apoptotic secreted form of clusterin, with
`no effect on the pro-apoptotic nuclear form [49].
`In vitro activity of ASOs and documented
`overexpression of tissue clusterin has been
`demonstrated in multiple chemotherapy resist-
`ant tumor cell lines [35,50]. In prostate cancer, a
`docetaxel-resistant human prostate cancer cell
`line (PC-3dR) had significantly higher secreted
`clusterin levels compared with controls [37].
`Clusterin protein expression was significantly
`decreased upon exposure to OGX-011 compared
`with a control oligonucleotide. Chemotherapy
`resensitization was demonstrated by combin-
`ing the docetaxel-resistant PC-3dR cells with
`OGX-011 resulting in a fall of the IC50 of
`docetaxel from >1000 nM to 125 nM.
`Downregulation of sCLU was established in
`multiple in vivo mice models (lung [35], bladder
`[50] and prostate [37,45]). In prostate cancer, treat-
`ment with OGX-011 decreased sCLU expression
`in both PC-3 and PC-3dR xenografts. Western
`blot analysis of protein harvested from tumors
`obtained from mice at the end of the study indi-
`cated that OGX-011 decreased sCLU expression
`levels to 32 and 27% of the sham ASO-treated
`controls in PC-3 and PC-3dR tumors, respec-
`tively. Resembling the in vitro data, baseline
`sCLU expression were 2.25-fold higher in the
`PC-3dR tumors compared with the wild-type
`PC-3 tumors (p < 0.001).
`Antitumor effect was also confirmed across
`several mouse models. In the prostate cancer mice
`xenografts, OGX-011 treatment significantly
`enhanced the antitumor effects of paclitaxel
`compared with the sham ASO control, reduc-
`ing the mean PC-3 tumor volume to 33% of
`controls at week 9 (p < 0.01). More importantly,
`OGX-011 was also able to significantly enhance
`cytotoxicity of paclitaxel in PC-3dR xenografts,
`decreasing mean tumor volume by 76% com-
`pared with the MOE gapmer mismatch control
`
`Table 2. Plasma and tissue pharmacokinetic data for OGX-011.
`
`Dose
`
`AUC
`(μg/h/ml)
`
`Plasma
`Clearance
`(ml/h/kg)
`
`Cmax
`(μg/ml)
`
`Half-life
`(h)
`
`Tissue
`Mean prostate tissue
`concentration (μg/g)
`
`69.85
`640 mg† 290.15
`†Recommended dose in humans.
`AUC: Area under the curve.
`
`32.9
`
`7.63
`
`4.82
`
`1242
`
`Future Oncol. (2012) 8(10)
`
`future science group
`
`Drug Evaluation Zielinski & Chi
`
`

`
`Historical controls
`
`
`
`Ptrend = 0.008
`
`600
`
`500
`
`400
`
`300
`
`200
`
`100
`
`Relative % clusterin mRNA
`
`
`
`
`
`
`
`0
`OGX-011 dose (mg) =
`
`NT <2M NHT 40/80
`
`
`160
`
`n =
`
`8
`
`10
`
`4
`
`3
`
`320
`
`6
`
`480
`
`6
`
`640
`
`
`
` 6
`
`Ptrend < 0.001
`
`Historical controls
`
`3.00
`
`2.50
`
`2.00
`
`1.50
`
`1.00
`
`0.50
`
`Overall IHC score
`
`
`
`
`
`0.00
`OGX-011 dose (mg) =
`n =
`
`NT <2M NHT 40/80
`10
`15
`4
`
`160
`3
`
`320
`6
`
`480
`6
`
`640
`6
`
`Figure 1. OGX-011 inhibits clusterin expression in human prostate tissue.
`(A) Box-plot of clusterin mRNA expression in prostate cancer cells of men with
`prostate cancer who were treated with OGX-011 prior to prostatectomy and in
`historical control subjects. (B) Box-plot of the overall score of
`immunohistochemistry staining. Three slides per patient and ten fields per slide
`were evaluated for staining intensity from 0 to 3 (representing negative to strong
`staining, respectively) and graded independently by two pathologists.
`<2M NHT: Less than 2 months of neoadjuvant hormone therapy historical controls;
`IHC: Immunohistochemistry; NT: No treatment.
`Reproduced with permission from [48] © JNCI Oxford University Press.
`
`and is due for completion in December 2013 with
`a planned enrollment of 1000 men and a pri-
`mary outcome of overall survival. The trial has
`been designed for 90% power to detect a hazard
`ratio of 0.75 assuming an 18-month expected
`overall survival in the control arm. A second
`Phase III trial in patients with metastatic CRPC
`previously treated with docetaxel, SATURN
`[103], aimed to assess pain response as a primary
`end point with OGX-011 in combination with
`docetaxel–prednisone retreatment versus pla-
`cebo and docetaxel–prednisone. The study was
`
`at week 9 (p < 0.01). Collectively, these results
`show that sCLU knockdown using OGX-011
`not only sensitizes PC-3 tumors to chemotherapy
`in vivo, but also reverses resistance and enhances
`chemosensitivity in PC-3dR xenografts.
`The first-in-man study combined OGX-011
`with neoadjuvant hormone ablation for patients
`with localized prostate carcinoma prior to radi-
`cal prostatectomy (RP). Prostate specimens and
`historical controls were then evaluated for clus-
`terin expression. Changes in expression of tis-
`sue clusterin were correlated to the administered
`plasma dose and the dose delivered to the pros-
`tate. OGX-011 produced statistically significant
`dose-dependent suppression of clusterin mRNA
`and protein expression in normal and tumor tis-
`sue. Approximately double the amount of tumor
`cell death (as determined by apoptotic index)
`occurred in the prostate of patients receiving the
`highest dose of OGX-011 (640 mg) compared
`with hormone ablation therapy alone. With this
`novel design and the use of PK and pharmaco-
`dynamic end points, an effective biologic dose of
`640 mg was established for OGX-011 based on
`its ability to suppress clusterin mRNA by >90%
`in prostate cancer tissue (Figure 1) [51].
`In the historical control specimens treated
`with and without neoadjuvant hormone therapy
`(NHT), the mean apoptotic indices were 9.0%
`(95% CI: 5.1–13.0) and 7.0% (95% CI: 4.2–9.9),
`respectively. The apoptotic index from patients
`treated at the lower two dose levels of OGX-011
`was 7.1% (95% CI: 2.4–11.8), but at the 640-mg
`dose level, the mean apoptotic index was 21.2%
`(95% CI: 18.1–24.2). These data provide proof-
`of-principle for the biologic activity of OGX-011
`and reinforce the dose–response effect initially
`demonstrated in preclinical testing.
`
`Clinical efficacy
`To date, two Phase I and five Phase II trials
`have been completed with a total of 313 patients
`enrolled: 213 with prostate cancer and 95 in lung
`and breast cancer. In prostate cancer, OGX-011
`has been evaluated in multiple settings and in
`combination with both hormonal and chemo-
`therapy agents (see Table 3). In advanced lung and
`breast malignancies, OGX-011 has been com-
`bined with chemotherapy agents (platinums,
`gemcitabine and docetaxel). Three Phase III
`trials have been initiated and two are currently
`recruiting patients with CRPC. The SYNERGY
`study is randomizing chemotherapy-naive
`patients with metastatic CRPC to receive stand-
`ard docetaxel and prednisone with or without
`OGX-011 [102]. The study commenced in 2010
`
`1243
`
`future science group
`
`www.futuremedicine.com
`
`Custirsen (OGX-011): a second-generation antisense inhibitor of clusterin Drug Evaluation
`
`

`
`Table 3. List of completed and open clinical trials with OGX-011.
`
`Phase Experimental arm
`
`Comparator arm
`
`Random n
`
`Clinical setting
`
`I
`
`I
`
`II
`
`II
`
`II
`
`II
`
`II
`
`III
`
`III
`
`OGX-011 + LHRH + anti-androgen None
`
`OGX-011 + docetaxel
`
`None
`
`OGX-011 + docetaxel + prednisone Placebo
`
`OGX-011 + docetaxel + prednisone Mitoxantrone
`+ prednisone
`
`OGX-011 + LHRH + anti-androgen None
`
`OGX-011 + platinum†
`+ gemcitabine
`
`OGX-011 + docetaxel
`
`OGX-011 + docetaxel
`
`OGX-011 + docetaxel
`
`Platinum†
`+ gemcitabine
`
`Docetaxel
`
`Docetaxel
`
`Docetaxel
`
`No
`
`No
`
`Yes
`
`Yes
`
`No
`
`No
`
`No
`
`Yes
`
`Yes
`
`25
`
`40
`
`82
`
`42
`
`24
`
`85
`
`15
`
`Neoadjuvant in localized PrCa
`
`Advanced solid organ malignancies
`
`mCRPC
`
`mCRPC
`
`Neoadjuvant in localized, high-risk PrCa
`
`Treatment-naive stage IIIb or IV NSCLC
`
`mBrCa first- or second-line
`chemotherapy
`
`1000‡ mCRPC first-line chemotherapy
`
`300‡ mCRPC second-line chemotherapy
`
`Ref.
`[48]
`
`[51]
`
`[10]
`
`[61]
`
`[52]
`
`[53]
`
`[60]
`
`[102]
`
`– §
`
`Cabazitaxel
`OGX-011 + cabazitaxel
`III
`†Platinum agents in this trial used cisplatin or carboplatin.
`‡Planned enrollment.
`§Trial terminated.
`LHRH: Luteinizing hormone-releasing hormone; mBrCa: Metastatic breast cancer; mCRPC: Metastatic castrate-resistant prostate cancer; NSCLC: Non-small-cell lung
`cancer; PrCa: Prostate cancer.
`
`Yes
`
`630
`
`mCRPC second-line chemotherapy
`
`[104]
`
`opened in March 2010 and planned to enroll 292
`men; however, it was closed prematurely given
`the shifting landscape of new therapies with the
`introduction of abiraterone and cabazitaxel in the
`post-docetaxel indication. With the approval of
`cabazitaxel for CRPC patients previously treated
`with docetaxel, a replacement Phase III trial [104]
`has instead been planned that is combining
`OGX-011 with cabazitaxel versus cabazitaxel
`alone with overall survival as the primary end
`point. Table 3 outlines the clinical trial details that
`are completed or recruiting.
`
`Phase I trial with neoadjuvant OGX-011
`prior to prostatectomy in patients with
`localized prostate cancer
`A uniquely designed first-in-man study adminis-
`tered OGX-011 in escalating doses in combina-
`tion with NHT to men with localized prostate
`carcinoma prior to RP [48]. The design of the trial
`was novel for two reasons; first, it demonstrated a
`dose-dependent relationship between tissue lev-
`els of OGX-011 and its target, prostatic tissue
`clusterin. Secondly, it established a relationship
`between decreased prostatic tissue clusterin (the
`target) and an increased apoptotic index (the
`biological effect). This allowed a biologically
`active dose (640 mg), rather than a dose-limiting
`toxic dose, to be selected as the recommended
`Phase II dose. The Phase II schedule requiredpa-
`tients to receive three loading doses of OGX-011
`(640 mg) prior to chemotherapy followed by
`weekly administration of OGX-011 (640 mg).
`
`Phase I trials with OGX-011 in
`combination with chemotherapy
`The next Phase I trial of OGX-011 examined
`its safety profile in combination with two dif-
`fering schedules of docetaxel [51]. Patients with
`solid organ malignancies were administered
`escalating doses of OGX-011 (40–640 mg) with
`weekly (30 mg/m2) or 3 weekly (75 mg/m2) doc-
`etaxel. Adverse events close to dose limiting were
`observed at 640 mg dosing of OGX-011 with
`docetaxel at 75 mg/m2 and this was declared as
`the recommended dose. These adverse events
`included grade 3–4 fatigue, stomatitis, granu-
`locytopenia, anemia and elevated alkaline
`phosphatase. Antitumor activity was observed
`with two patients (6%) experiencing a partial
`radiological response, 40% of the prostate cancer
`cohort experiencing a prostate-specific antigen
`(PSA) response (defined as post-treatment PSA
`decline of ≥50%) and 11 patients (34%) expe-
`riencing stable disease. Importantly, toxicity of
`the combination was mild or moderate for the
`most part, with toxicities being qualitatively
`and quantitatively in keeping with what would
`be expected with docetaxel alone. Finally, there
`was no apparent effect of OGX-011 on the PK
`behavior of docetaxel.
`
`Phase II trials of OGX-011 in combination
`with NHT
`A two-stage Phase II study combined weekly
`OGX-011 with NHT in previously untreated
`patients with high-risk, localized prostate cancer
`
`1244
`
`Future Oncol. (2012) 8(10)
`
`future science group
`
`Drug Evaluation Zielinski & Chi
`
`

`
`prior to RP [52]. Hormone therapy consisted of
`subcutaneous lutenizing hormone-releasing hor-
`mone agonist every 3 months with either daily
`flutamide or bicalutamide. A total of 12 weeks of
`combined therapy was administered prior to sur-
`gery. The study did not progress from stage 1 as
`it failed to meet its primary end point of at least
`one pathological complete response. However,
`the apoptotic index in prostate tumors was sign-
`ficantly higher in patients given OGX-011 and
`NHT compared with both untreated controls
`and NHT-treated controls.
`
`Phase II trials of OGX-011 in combination
`with chemotherapy
`A multicenter noncomparative Phase II trial,
`randomized chemotherapy-naive metastatic
`CRPC patients to daily prednisone and doc-
`etaxel every 3 weeks with or without weekly
`OGX-011 (640 mg) [10]. A total of 82 patients
`were enrolled and the primary end point was
`PSA response. The trial was insufficiently
`powered for statistical analysis of observed
`differences; however, the experimental arm
`demonstrated an unadjusted HR of 0.61 (95%
`CI: 0.36–1.02; p = 0.06). The OGX-011 plus
`docetaxel arm obtained median PFS and OS of
`7.3 months (95% CI: 5.3–8.8) and 23.8 months
`(95% CI: 16.2–not reached), respectively while
`the docetaxel-alone arm achieved PFS and OS of
`6.1 (95% CI: 3.7–8.6) and 16.9 months (95%
`CI: 12.8–25.8). A multivariate analysis using
`pre-identified prognostic factors identified per-
`formance status Eastern Cooperative Oncology
`Group of 0 (p < 0.0001), no visceral metasta-
`sis (p = 0.01) and treatment assignment to
`OGX-011 (HR = 0.50, 95% CI: 0.29–0.87; p =
`0.01) associated with an improved overall sur-
`vival. This trial led to the Phase III registration
`path currently underway.
`Another multicenter Phase II lung cancer trial
`used OGX-011 in combination with cisplatin
`(75 mg/m2)/gemcitabine (1250 mg/m2) every 3
`weeks in a treatment-naive cohort of advanced
`non-small-cell lung cancer (NSCLC) patients
`(stage IIIb/IV) for a maximum of six cycles [53].
`The objective response rate was 31% with one
`patient reporting a complete response. A total
`of 95% of patients experienced serum clusterin
`falls compared with baseline. There was a 50%
`reduction in risk of death in patients experienc-
`ing a serum clusterin response, defined as less
`than 38 µg/ml. Finally, overall survival was
`14.1 months, which compares favorably with
`published median survivals of 8–10.8 months
`in similar patients treated with gemcitabine/
`
`platinum doublets [54–58]. The overall survival
`data also compare favorably with the 12.3-month
`OS from newer regimens of platinum doublets in
`combination with bevacizumab [59].
`A multicenter, Phase II breast cancer trial
`used OGX-011 with docetaxel (75 mg/m2)
`every 3 weeks in patients with metastatic and/or
`locally advanced breast cancer as first- or second-
`line chemotherapy [60]. A partial response rate
`of 33% was observed in the experimental arm,
`which was similar to the response rate expected
`of single-agent docetaxel. An additional 60%
`showed stable disease and the median duration
`of response was 4.9 months. Serum clusterin
`decreased by 23–32% from baseline values yet
`no relationship between response rate and serum
`clusterin levels was shown. Although clinical
`activity was demonstrated, the study failed to
`progress to the second stage of accrual due to
`lack of prespecified objective responses.
`The final clinical study sought to address
`the issue of reversing docetaxel resistance or
`improving mitoxantrone efficacy in the second-
`line setting. An open-label, randomized, multi-
`center study evaluated weekly administration
`of OGX-011 in combination with second-line
`chemotherapy in patients with metastatic CRPC
`who were previously treated with a minimum of
`two cycles of a docetaxel-based regimen and pro-
`gressed on or within 6 months of discontinua-
`tion of treatment [61]. Safety and tolerability were
`the primary objectives; however, the secondary
`objective was to assess for reversal of docetaxel
`resistance in this pretreated cohort. A total of
`45 patients were randomized to OGX-011 in
`combination with mitoxantrone 12 mg/m2
`intravenously every 3 weeks with 5 mg pred-
`nisone twice daily or docetaxel retreatment at
`75 mg/m2 every 3 weeks with 5 mg prednisone
`twice daily. An additional 20 patients were ran-
`domized to the docetaxel retreatment following
`preliminary safety and efficacy analysis favoring
`docetaxel. The overall PSA response rate in the
`docetaxel arm was 42%, with 91% experiencing
`a PSA decline of 30% or more. In the mitox-
`antrone arm, the PSA response rate was 27%
`with 59% experiencing a PSA decline of 30%
`or more. The estimated median overall survival
`duration for the OGX-011 plus mitoxantrone
`arm was 11.5 months (95% CI: 6.1–15.2). For
`the OGX-011 plus docetaxel retreatment arm,
`the median overall survival was 15.8 months
`(95% CI: 9.9–23.3) for the 20 randomized
`patients and 12.8 months (95% CI: 9.9–17.0)
`for the combined 45 patients. Serum clusterin
`

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