throbber
Published OnlineFirst April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-3146
`
`Cancer Therapy: Preclinical
`
`Clinical
`Cancer
`Research
`
`Preclinical Antitumor Activity of Cabazitaxel, a Semisynthetic
`Taxane Active in Taxane-Resistant Tumors
`
`Patricia Vrignaud, Dorothee Semiond, Pascale Lejeune, Herve Bouchard, Loreley Calvet, Cecile Combeau,
`Jean-Fran¸cois Riou, Alain Commer¸con, Fran¸cois Lavelle, and Marie-Christine Bissery
`
`Abstract
`
`Purpose: Taxanes are important chemotherapeutic agents with proven efficacy in human cancers, but
`their use is limited by resistance development. We report here the preclinical characteristics of cabazitaxel
`(XRP6258), a semisynthetic taxane developed to overcome taxane resistance.
`Experimental Design: Cabazitaxel effects on purified tubulin and on taxane-sensitive or chemotherapy-
`resistant tumor cells were evaluated in vitro. Antitumor activity and pharmacokinetics of intravenously
`administered cabazitaxel were assessed in tumor-bearing mice.
`Results: In vitro, cabazitaxel stabilized microtubules as effectively as docetaxel but was 10-fold more
`potent than docetaxel in chemotherapy-resistant tumor cells (IC50 ranges: cabazitaxel, 0.013–0.414 mmol/L;
`docetaxel, 0.17–4.01 mmol/L). The active concentrations of cabazitaxel in these cell lines were achieved
`easily and maintained for up to 96 hours in the tumors of mice bearing MA16/C tumors treated with
`cabazitaxel at 40 mg/kg. Cabazitaxel exhibited antitumor efficacy in a broad spectrum of murine and human
`tumors (melanoma B16, colon C51, C38, HCT 116, and HT-29, mammary MA17/A and MA16/C, pancreas
`P03 and MIA PaCa-2, prostate DU 145, lung A549 and NCI-H460, gastric N87, head and neck SR475, and
`kidney Caki-1). Of particular note, cabazitaxel was active in tumors poorly sensitive or innately resistant to
`docetaxel (Lewis lung, pancreas P02, colon HCT-8, gastric GXF-209, mammary UISO BCA-1) or with
`acquired docetaxel resistance (melanoma B16/TXT).
`Conclusions: Cabazitaxel is as active as docetaxel in docetaxel-sensitive tumor models but is more potent
`than docetaxel in tumor models with innate or acquired resistance to taxanes and other chemotherapies. These
`studies were the basis for subsequent clinical evaluation. Clin Cancer Res; 19(11); 2973–83. Ó2013 AACR.
`
`Introduction
`Microtubules are highly dynamic cytoskeletal fibers com-
`posed of 2 tubulin subunits (a and b). The polymerization
`and depolymerization of these molecules are crucial pro-
`cesses, not only to mitosis but also to intracellular traffick-
`ing. Microtubules are the main target of taxanes, which bind
`to a specific binding site on the tubulin b-subunit (1, 2). The
`taxanes paclitaxel and docetaxel suppress microtubule
`dynamics by promoting tubulin assembly and stabilizing
`microtubules (3), blocking mitosis at the metaphase/ana-
`phase transition, which results in cell death (ref. 4; Supple-
`mentary Fig. S1A and S1B). By stabilizing microtubules,
`taxanes also impact intracellular trafficking. This was recent-
`ly reported as one of the main mechanisms of taxane action
`
`Authors' Affiliation: Sanofi, Vitry-sur-Seine, France
`
`Note: Supplementary data for this article are available at Clinical Cancer
`Research Online (http://clincancerres.aacrjournals.org/).
`
`Corresponding Author: Patricia Vrignaud, Sanofi Oncology, 13 quai Jules
`Guesde, Vitry-sur-Seine 94403, France. Phone: 33-1-58-93-36-29; Fax:
`33-1-58-93-34-71; E-mail: patricia.vrignaud@sanofi.com
`
`doi: 10.1158/1078-0432.CCR-12-3146
`Ó2013 American Association for Cancer Research.
`
`in prostate cancer, where taxanes were shown to inhibit
`nuclear translocation of the androgen receptor, thereby
`preventing androgen receptor transcriptional activity and
`leading to prostate cancer cell death (5).
`Paclitaxel and docetaxel form the backbone of both first-
`line and salvage chemotherapy regimens for patients with a
`wide variety of tumor types. Paclitaxel is indicated for first-
`line treatment of ovarian, breast, and lung cancer and for
`second-line treatment of AIDS-related Kaposi’s sarcoma (6).
`Docetaxel is indicated for first-line treatment of breast, head
`and neck, gastric, lung, and prostate cancer and for second-
`line treatment of breast cancer (7). However, the use of both
`paclitaxel and docetaxel is limited by the development of
`tumor resistance (8–10). During the last 2 decades, con-
`siderable efforts have been made to understand, and devel-
`op new agents to overcome, taxane resistance.
`Cabazitaxel (RPR 116258; XRP6258; TXD258; Jevtana)
`is a new semisynthetic taxane derived from 10-deacetyl-
`baccatin III, which is extracted from European yew needles
`(ref. 11; Supplementary Fig. S1C). Cabazitaxel was identi-
`fied using a 3-step screening process, assessing activity
`against microtubule stabilization, in vitro activity in resistant
`cell lines, and in vivo activity in a tumor model in which
`docetaxel resistance had been induced in vivo. This article
`
`www.aacrjournals.org
`
`2973
`
`Downloaded from
`
`
`
`on January 8, 2014. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org
`
`AVENTIS EXHIBIT 2033
`Mylan v. Aventis
`IPR2016-00627
`
`

`
`Published OnlineFirst April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-3146
`
`Vrignaud et al.
`
`Translational Relevance
`Mechanisms of resistance to taxanes in patients have
`not been fully elucidated. In cell lines, overexpression of
`ATP-binding transporters, particularly P-glycoprotein,
`and alteration of microtubule dynamics are the most
`common mechanisms of taxane resistance. However,
`clinical data suggest that other mechanisms, including
`dysfunctional regulation of apoptotic and intracellular
`signaling, may operate in tumors escaping taxane ther-
`apy. To identify a docetaxel derivative with activity after
`taxane failure, we developed a clinically relevant doc-
`etaxel-resistant tumor model, mimicking tumor resis-
`tance development in patients who initially respond to
`docetaxel, but develop resistance over time. Cabazitaxel
`was selected from 450 derivatives based on activity in
`this model. Clinical proof-of-principle was achieved in a
`phase II study in patients with taxane-resistant metastatic
`breast cancer and a phase III study in metastatic hor-
`mone-refractory prostate cancer post-docetaxel therapy.
`The current study extends the characterization of caba-
`zitaxel, showing wide ranging in vitro and in vivo anti-
`tumor activity.
`
`describes the development and characterization of the in
`vivo–induced docetaxel-resistant tumor model, the mecha-
`nism of action of cabazitaxel on microtubules, and its
`preclinical evaluation in a wide range of taxane-sensitive
`and -resistant cell lines, both in vitro and in vivo.
`
`Materials and Methods
`Tubulin polymerization
`The effects of cabazitaxel on tubulin polymerization and
`cold-induced microtubule depolymerization were evaluat-
`ed using tubulin purified from porcine brain (12, 13).
`Tubulin was used at a concentration of 6 mmol/L for
`
`C) and 9 mmol/L for depo-
`polymerization assays (at 37
`
`lymerization assays (at 8
`C). Rates of polymerization/depo-
`lymerization were measured by optical density (OD) at
`350 nm and were expressed in DOD/min. Upper and lower
`limits for drug concentrations reducing polymerization lag
`time by 50% (LT50) and inhibiting cold-induced disassem-
`bly by 50% (dIC50) were determined.
`
`Microtubule and enzymatic parameters in tumors
`Microtubule parameters in B16 and B16/TXT tumors
`were characterized using real-time PCR (RT-PCR) analysis
`of at least 2 samples per tumor. PCR values in arbitrary units
`were obtained for the following genes: total a-tubulin
`(TUBA), total b-tubulin (TUBB), TUBB2, TUBB3, TUBB4A,
`TUBB4B, and TUBB7P.
`Glutathione S-transferase (GST) activity was assayed as
`previously described using 1-chloro-2,4-dinitrobenzene
`(CDNB) as the substrate (14). Formation of the CDNB
`glutathione (GSH) conjugate by cytosols was measured
`
`continuously in a spectrophotometer at 340 nm. The results
`were expressed as the quantity of CDNB conjugated per
`minute per milligram of cytosolic protein (nmol/min/mg).
`Total GSH concentration was determined as the sum of
`the reduced (GSH) and oxidized (GSSG) forms of GSH
`(15). In this assay, the sum of the reduced and oxidized
`forms of GSH is determined using a kinetic assay in which
`catalytic amounts of GSH or GSSG and GSH reductase bring
`0
`about the continuous reduction of 5,5
`-dithiobis(2-nitro-
`benzoic acid) by NADPH. The reaction rate is proportional
`to the concentration of GSH below 2 mmol/L. The forma-
`tion of 5-thio-2-nitrobenzoate was analyzed using a spec-
`trophotometer at 412 nm. The results were expressed as
`concentration per milligram of protein (nmol/mg).
`Cytochrome P450 3A (CYP3A) levels were determined
`using the Amersham ELISA system (code RPN 271; Amer-
`sham). This assay uses a rabbit primary antibody specific for
`rat CYP3A, a secondary conjugate of anti-rabbit immuno-
`globulin (Ig) and horseradish peroxidase antibody, and
`tetramethylbenzidine substrate. The horseradish peroxi-
`dase color that develops is proportional to CYP3A levels.
`This assay was validated against mouse CYP3A by the
`manufacturer. Protein concentrations of microsomes, cyto-
`sols, and homogenates were determined by the bicincho-
`ninic acid assay (16) using a commercial preparation
`(Pierce BCA Protein Assay Reagent).
`
`In vitro antiproliferative activity
`The HL60/TAX cell line (17) was a kind gift from Dr. K.
`Bhalla (Medical University of South Carolina, Charleston,
`SC). Calc18/TXT and P388/TXT were developed internally
`from Calc18 or P388 parental cell lines. The P388/TXT cell
`line was selected by mutagenesis with ethyl methane sul-
`fonate and soft agar cloning in the presence of 0.06 mmol/L
`docetaxel. The Calc18/TXT cell line was established by 6-
`month exposure to increasing concentrations of docetaxel
`(up to 0.019 mmol/L). The cross-resistance pattern of these 2
`cell lines is shown in Supplementary Table S1. The other
`tumor cell lines were obtained from the National Cancer
`Institute (NCI; Bethesda, MD).
`Parental and resistant tumor cells were incubated with
`
`different drug concentrations for 96 hours at 37
`C; cell
`viability was measured in quadruplicate using neutral red
`uptake (18). The resistance factor for each drug was calcu-
`lated by dividing the IC50 in resistant cells by the mean IC50
`in sensitive/parental cells, using data from at least 3 inde-
`pendent experiments. Relative expression of ABCB1 mRNA
`was determined by Northern blotting using a human
`ABCB1 gene probe.
`
`Antitumor activity in tumor-bearing mice
`All experimental procedures were approved by Sanofi,
`Southern Research Institute (SRI; Birmingham, AL), and
`Molecular Imaging Research (MIR) Preclinical Services Lab-
`oratory Animal Care and Use committees. Protocol design,
`chemotherapy techniques, and methods of data analysis
`have been described previously (19–21). Briefly, tumors
`were implanted subcutaneously and bilaterally on day 0.
`
`2974
`
`Clin Cancer Res; 19(11) June 1, 2013
`
`Clinical Cancer Research
`
`
`
`Downloaded from on January 8, 2014. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org
`
`
`
`

`
`Published OnlineFirst April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-3146
`
`Animals were randomly assigned to treatment (T) or control
`(C) groups. Tumors were measured using a caliper 2 to 5
`times weekly (according to tumor growth rate) until the
`tumor reached 2,000 mm3. Tumor volumes were estimated
`from 2-dimensional measurements using the formula:
`tumor volume (mm3) ¼ [length (mm)  width2
`(mm2)]/2. The day of death was recorded, and thoracic
`and abdominal cavities were examined macroscopically to
`assess probable cause of death.
`Mice. C57BL/6, B6D2F1 and Swiss nude mice were bred
`at Iffa Credo; C3H/HeN, BALB/c, BALB/c nude, and severe
`combined immunodeficient (SCID) mice were bred at
`Charles River; and ICR and NCR nude mice were bred at
`Taconic. All mice weighed more than 18 g at the start of
`treatment and had free access to food and water.
`Drugs. Cabazitaxel (RPR 116258; XRP6258; TXD258;
`Jevtana) and docetaxel (RP 56976) were prepared by mixing
`1 volume of ethanol stock solution, 1 volume of polysor-
`bate 80, and 18 volumes of 5% glucose in sterile water.
`Solutions were administered intravenously as a slow bolus
`(0.4 mL/mouse). Drug doses were adjusted on the basis of
`body weight at start of treatment. For cytotoxic compounds,
`such as docetaxel and cabazitaxel, a dose–response evalu-
`ation was conducted in each trial to determine the highest
`nontoxic dose (HNTD), defined as the highest drug dose
`inducing less than 20% body weight loss with no drug-
`related deaths. Animal body weights included the tumor
`weights.
`Tumor models. Murine tumors were obtained from Dr.
`Corbett (Wayne State University, Detroit, MI) and included
`colon C51 and C38 (19), pancreas P02 and P03 (22),
`mammary MA17/A and MA16/C (23), Lewis lung (24),
`and melanoma B16 (20). Tumors were maintained by serial
`passage in the mouse strain of origin. B16/TXT was isolated
`by treating C57BL/6 mice bearing docetaxel-sensitive B16
`melanoma at the HNTD of docetaxel (60 mg/kg) for 27
`passages, until the B16 tumor acquired full resistance to
`docetaxel. Human tumor cell lines were obtained from
`American Type Culture Collection and included prostate
`DU 145 (25), lung NCI-H460 (26) and A549 (27), pancreas
`MIA PaCa2 (28), and colon HT-29 (29), HCT 116 (30), and
`HCT-8 (31). Mammary UISO BCA-1 (32) and gastric GXF-
`209 (33) tumors were obtained from SRI and gastric N87
`(34) tumors from MIR Preclinical Services. Murine tumors
`were grafted into syngenic mice and human tumors were
`xenografted into immunocompromised mice.
`Plasma pharmacokinetics and tumor distribution. Caba-
`zitaxel concentrations in plasma and tumor tissue were
`evaluated in mice bearing advanced-stage (400 mm3)
`murine mammary adenocarcinoma MA16/C after admin-
`istration of the HNTD of cabazitaxel (40 mg/kg). Mice were
`treated on day 8 after subcutaneous tumor implantation
`with a single 45-second intravenous infusion of cabazitaxel
`in a polysorbate 80/ethanol/5% glucose solution, with a
`dosing volume of 25 mL/kg and a rate of infusion of
`1 mL/min. Blood and tumor samples were collected from
`3 animals per sampling time at 2, 5, and 15 minutes and 2,
`4, 8, 12, 24, 48, 96, and 168 hours after cabazitaxel treat-
`
`Preclinical Antitumor Activity of Cabazitaxel
`
`concentrations were analyzed by
`ment. Cabazitaxel
`liquid chromatography/tandem mass spectrometry, with
`limits of quantification of 2.5 ng/mL in plasma and 25
`ng/g in tumor tissue. Pharmacokinetic parameters were
`determined using WinNonLin software, Version 1.0 (Sci-
`entific Consulting Inc.), using a noncompartmental infu-
`sion model.
`Several endpoints
`Assessments of antitumor activity.
`were used. Tumor growth delay (TC) was defined as the
`difference between tumors in the T and C groups in the
`median time (days) to reach a predetermined volume (750–
`1,000 mm3). Tumor doubling time (Td) in days was esti-
`mated from log linear tumor growth during the exponential
`phase (range, 100–1,000 mm3). Log cell kill was calculated
`using the formula (TC)/(3.32  Td), with antitumor
`activity defined as a log cell kill value  0.7 (21). SRI score
`was used to categorize antitumor activity based on log cell
`kill values as follows: <0.7¼ (inactive); 0.7–1.2¼þ; 1.3–
`1.9 ¼ þþ; 2.0–2.8 ¼ þþþ; > 2.8 ¼ þþþþ (highly active).
`Complete tumor regression (CR) was defined as tumor
`regression below the limit of palpation (62 mm3). Animals
`without palpable tumors at the end of the study were
`declared tumor-free survivors (TFS) and were excluded from
`the TC value calculation.
`Statistical analysis was conducted using either a pairwise
`Wilcoxon rank-sum test, with P value adjustment by the
`Holm method (N87 study), or by log-rank multiple com-
`parisons test versus control (with Bonferroni–Holm correc-
`tion for multiplicity) on individual values for time to reach a
`prespecified tumor size for treated and control groups
`(UISO BCA-1 study). A P value of less than 5% (P <
`0.05) was considered significant.
`
`Results
`Isolation and characterization of B16/TXT, a docetaxel-
`resistant melanoma
`To identify taxane derivatives with activity following
`taxane failure, a docetaxel-resistant tumor model (B16/
`TXT) was developed to mimic the gradual development of
`resistance to docetaxel observed in some patients following
`an initial tumor response to the agent. Mice bearing the
`sensitive murine B16 melanoma were treated with docetaxel
`at the HNTD (60 mg/kg per passage; log cell kill 1.7; Table 1).
`Resistance occurred very slowly, with 27 passages over 17
`months needed to obtain a fully docetaxel-resistant tumor
`(log cell kill < 0.7). B16/TXT was found to have similar Td
`(1.3–2 days) and histologic characteristics to the parental
`B16 tumor. Cross-resistance (no antitumor activity) was
`observed to the tubulin-binding drugs paclitaxel, vincris-
`tine, and vinblastine, but not to cyclophosphamide (log cell
`kill 2.9 in B16 vs. 3.0 in B16/TXT), CCNU (log cell kill 3.7 in
`B16 vs. 4.7 in B16/TXT), and etoposide (log cell kill 1.2 in
`both). B16/TXT was partially cross-resistant to doxorubicin
`(log cell kill 2.4 in B16 vs. 0.9 in B16/TXT). There was no
`difference between the docetaxel-sensitive and -resistant
`B16 tumors either in factors involved in drug resistance,
`such as GST activity (B16, 0.42  0.03 mmol/min/mg
`
`www.aacrjournals.org
`
`Clin Cancer Res; 19(11) June 1, 2013
`
`2975
`
`
`
`Downloaded from on January 8, 2014. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org
`
`
`
`

`
`Published OnlineFirst April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-3146
`
`protein; B16/TXT, 0.39  0.04 mmol/min/mg protein) and
`GSH content (B16, 21.7  8.1 mmol/mg protein; B16/TXT,
`21.2  2.5 mmol/mg protein), or in activity of CYP3A,
`involved in TXT metabolism (B16, 2.4  0.8 mg/mg protein;
`B16/TXT, 2.9  0.06 mg/mg protein). Moreover, no over-
`expression of P-glycoprotein was found in B16/TXT, either
`by flow cytometry or Western blot analyses (data not
`shown). Analyses by RT-PCR of microtubule components
`revealed that B16/TXT expressed 3.13-fold higher levels of
`TUBB3 than the docetaxel-sensitive parental B16 tumor,
`whereas levels of other microtubule parameters were similar
`(Supplementary Table S2). As noted earlier, this model was
`pivotal in the selection of cabazitaxel, the characteristics of
`which are described hereafter.
`
`Microtubule stabilization
`Cabazitaxel had similar efficiency compared with doce-
`
`taxel for reducing the lag time for tubulin assembly (LT50 ¼
`0–0.1 mmol/L for both) and the rate of cold-induced micro-
`tubule depolymerization (dIC50 ¼ 0.1–0.25 mmol/L for
`both) in vitro (Table 2).
`
`In vitro antiproliferative activity in chemotherapy-
`sensitive and -resistant cell lines
`Cabazitaxel showed similar antiproliferative activity
`compared with docetaxel in cell lines sensitive to chemo-
`therapy (murine leukemia P388, human tumor HL60 and
`KB, and breast Calc18), as shown by the similar IC50 ranges
`across different cell types (cabazitaxel, 0.004–0.041 mmol/L;
`docetaxel, 0.008–0.079 mmol/L; Table 3). In P-glycopro-
`tein–expressing cell lines with in vitro–acquired resistance to
`taxanes (P388/TXT, Calc18/TXT, and HL60/TAX) or to
`other chemotherapy agents (P388/DOX, P388/VCR, and
`KBV1), cabazitaxel was found to be more active than doc-
`etaxel (IC50 ranges: cabazitaxel, 0.013–0.414 mmol/L; doc-
`etaxel, 0.17–4.01 mmol/L). Resistance factors (an indication
`of the difference in drug concentrations needed to inhibit
`resistant vs. sensitive/parental cell lines) were 2 to 10 for
`cabazitaxel and 5 to 59 for docetaxel. Cell lines expressing
`moderate levels of P-glycoprotein (P388/TXT, P388/VCR,
`HL60/TAX, and Calc18/TXT), which may be more clinically
`representative, had minimal cross-resistance to cabazitaxel
`(resistance factors ¼ 2–4).
`
`Plasma pharmacokinetics and drug distribution in
`tumors
`The pharmacokinetic profile of cabazitaxel was evaluated
`in mice bearing docetaxel-sensitive murine mammary
`MA16/C adenocarcinoma tumors. Cabazitaxel was highly
`active in this tumor model, inducing CRs in 80% of mice and
`having a log cell kill of 3.7 at the HNTD of 40 mg/kg (Table
`1). This antitumor activity was consistent with drug uptake
`into the tumor, which was both rapid (maximum drug
`concentrations were reached 15 minutes after dosing) and
`sustained (at 48 hours post-dose, cabazitaxel concentrations
`were 40-fold higher in the tumor vs. plasma; Fig. 1). Ratios of
`cabazitaxel exposure in tumors versus plasma were 1.6 from
`0 to 48 hours and 2.9 over the entire experimental period.
`
`Vrignaud et al.
`
`bDefinitionofantitumoractivity:logcellkilltotal<0.7¼inactive;>2.8¼highlyactive.
`aMediantumorburdenatstartoftherapy:290,310,and400mm3forC38,P03,andMA16/Cstudies,respectively.
`Abbreviations:N/A,notavailableastreatmentconductedonearly-stagedisease;ND,notdeterminedinthesamestudy.
`(B16,B16/TXT,C38,P03,P02,and3LL).
`NOTE:MurinetumorsweregraftedinthesyngenicstrainofmiceoforiginofthetumorforMA17/A(C3H/HeN)andC51(BALB/c)andinB6D2F1micefortheC57BL/6syngenictumors
`
`ND
`ND
`ND
`ND
`ND
`0/5
`0/5
`0/5
`0/5
`
`TFS
`
`ND
`ND
`ND
`ND
`ND
`N/A
`0/5
`N/A
`N/A
`
`CR
`
`ND
`ND
`ND
`ND
`ND
`3.1
`3.1
`0.6
`1.7
`
`killb
`Logcell
`
`Docetaxel
`
`ND
`ND
`ND
`ND
`ND
`31.1
`29.1
`2.8
`6.6
`
`days
`TC,
`
`ND
`ND
`ND
`ND
`ND
`45
`60
`60
`60
`
`mg/kg
`HNTD,
`Total
`
`0/5
`0/5
`0/5
`0/5
`4/5
`0/5
`5/5
`0/5
`0/5
`
`TFS
`
`N/A
`N/A
`4/5
`N/A
`5/5
`N/A
`5/5
`N/A
`N/A
`
`CR
`
`1.2
`3.9
`3.7
`0.8
`
`—
`
`2.6
`
`—
`
`1.3
`2.1
`
`cellkillb
`log
`
`4.6
`15.7
`13.4
`6.6
`
`—
`
`25.8
`
`—
`
`5.5
`8.5
`
`days
`TC,
`
`Cabazitaxel
`
`58.5
`36
`40
`60
`60
`45
`60
`60
`60
`
`31.5,19.5,12.1,7.5(3,5,7)
`19.4,12,7.4,4.6(3,5,7)
`64.5,40,24.8,15.4(8a)
`32.2,20,12.4,7.7(3,5,7)
`32.2,20,12.4,7.7(17a,19,21)
`24.2,15,9.3,5.8(4,6,8)
`32.2,20,12.4,7.7(14a,17,20)
`32.2,20,12.4,7.7(3,5,7)
`32.2,20,12.4,7.7(3,5,7)
`
`mg/kg
`TotalHNTD,
`
`injection(scheduledays)
`Dose,mg/kgper
`
`Lung3LL(Td:1.2d)
`MammaryMA17/A(Td:1.2d)
`MammaryMA16/C(Td:1.1d)
`PancreasP02(Td:2.5d)
`PancreasP03(Td:3.5d)
`ColonC51(Td:3d)
`ColonC38(Td:2.8d)
`B16/TXTmelanoma(Td:1.3d)
`B16melanoma(Td:1.2d)
`Tumortype
`
`Table1.Dose–responseantitumoractivityofcabazitaxelanddocetaxelinmicebearingmurinetumors
`
`2976
`
`Clin Cancer Res; 19(11) June 1, 2013
`
`Clinical Cancer Research
`
`
`
`Downloaded from on January 8, 2014. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org
`
`
`
`

`
`Published OnlineFirst April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-3146
`
`Preclinical Antitumor Activity of Cabazitaxel
`
`Table 2. Effects of taxoids on the assembly–disassembly process of pure tubulin
`
`Rate of cold-induced microtubule
`disassembly, DOD/min
`
`Lag time, min
`
`Docetaxel
`Cabazitaxel
`2 (n ¼ 5)
`7.42  10
`5.36  10
`2 (n ¼ 4)
`
`6  102 (n ¼ 3)
`0.89
`2 (n ¼ 5)
`
`3.05  102 (n ¼ 5)
`2.82  10
`0.92
`1.8  10
`2 (n ¼ 6)
`
`1.95  102 (n ¼ 5)
`0.92
`1.2  10
`2 (n ¼ 5)
`
`1.25  102 (n ¼ 4)
`0.96
`2 (n ¼ 4)
`
`0.5  102 (n ¼ 6)
`0.61  10
`1.22
`0.56  10
`2 (n ¼ 3)
`
`0.38  102 (n ¼ 3)
`1.47
`LT50: 0–0.1 mmol/L
`LT50: 0–0.1 mmol/L
`dIC50: 0.1–0.25 mmol/L
`dIC50: 0.1–0.25 mmol/L Mean: 1.06
`NOTE: Tubulin was used at a concentration of 6 mmol/L for polymerization (at 37C) and 9 mmol/L for depolymerization (at 8C). OD was
`measured at 350 nm. Rates of depolymerization were expressed in DOD/min. Ratios between depolymerization rates were calculated
`for each drug concentration. Boundaries of drug concentrations for dIC50 and LT50 are given.
`
`Drug concentration,
`mmol/L
`Control
`0.1
`0.25
`0.5
`1
`2.5
`5
`
`Ratio
`
`Cabazitaxel
`
`Docetaxel
`
`40
`
`4.18
`1.57
`1.0
`0.5
`
`9.16
`2.61
`1.04
`0.65
`
`Cabazitaxel concentrations were maintained above the range
`of cellular antiproliferative IC50 values [0.004–0.041 mmol/L
`(see Table 3), corresponding to 3–29 ng/mL, 4-day exposure]
`for 24 hours in plasma and 96 hours in the tumor.
`
`Schedule of administration
`The optimal schedule of cabazitaxel administration in
`vivo was initially determined by assessing the total dose that
`could be injected without undue toxicity for different sche-
`
`dules in nontumor-bearing B6D2F1 female mice (Supple-
`mentary Table S3). Three schedules of intravenous cabazi-
`taxel were administered: intermittent [days 1 and 5 (A1)],
`daily [days 1–5 (A2)] and split-dose [days 1–5, 3 times daily
`(A3)]. HNTDs were 58 mg/kg (A1), 29 mg/kg (A2), and 12
`mg/kg (A3), suggesting a trend for schedule dependency.
`These results indicate that, compared with intermittent
`treatment (A1) of the same duration, the daily (A2) and
`split-dose (A3) schedules require 2-fold and 4.8-fold dose
`
`Table 3. In vitro antiproliferative effects of cabazitaxel and docetaxel against sensitive and P-glycoprotein–
`expressing resistant cell lines
`
`Mean IC50, mmol/L,  SD
`
`Resistance factora
`
`ABCB1 mRNA levelb
`Cabazitaxel
`Docetaxel
`Docetaxel
`Cell line
`Cabazitaxel
`
`0.041  0.017
`0.079  0.004
`
`P388 murine leukemia
`þþþ
`0.414  0.036
`4.01  0.28
`51
`P388/DOX
`10
`
`0.013  0.005
`0.039  0.012
`
`P388 murine leukemia
`þþ
`0.024  0.015
`0.188  0.022
`5
`P388/TXT
`2
`
`0.013  0.005
`0.039  0.012
`
`P388 murine leukemia
`þþ
`0.024  0.003
`0.227  0.038
`6
`P388/VCR
`2
`
`0.022  0.010
`0.031  0.004
`
`HL60 human leukemia
`þþ
`0.060  0.029
`0.25  0.11
`8
`HL60/TAX
`3
`
`0.004  0.002
`0.008  0.002
`
`Calc18 human breast adenocarcinoma
`þþ
`0.016  0.004
`0.17  0.04
`21
`Calc18/TXT
`4
`
`0.035  0.026
`0.042  0.0212
`
`KB human epidermoid carcinoma
`þþþþ
`0.27  0.013
`2.48  0.12
`59
`KB V1
`8
`NOTE: Cells were incubated for 96 hours at 37C in liquid medium with drugs at different concentrations. Viability was assessed by
`neutral red, with the mean of at least 3 results obtained.
`Abbreviations: ABCB1, ATP-binding cassette, sub-family B, member 1; Calc18/TXT, Calc18 human breast adenocarcinoma resistant
`to docetaxel; HL60/TAX, HL60 human leukemia resistant to paclitaxel; KB V1, KB human epidermoid carcinoma resistant to vinblastine;
`P388/DOX, P388 murine leukemia resistant to doxorubicin; P388/TXT, P388 murine leukemia resistant to docetaxel; P388/VCR, P388
`murine leukemia resistant to vincristine.
`aResistance factor ¼ IC50 (resistant)/IC50 (parental) from the same experiment.
`bRelative expression obtained from Northern blot experiments using the human ABCB1 gene as probe.
`
`www.aacrjournals.org
`
`Clin Cancer Res; 19(11) June 1, 2013
`
`2977
`
`
`
`Downloaded from on January 8, 2014. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org
`
`
`
`

`
`Published OnlineFirst April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-3146
`
`Plasma
`Tumor
`
`Figure 1. Pharmacokinetics of
`cabazitaxel in plasma and tumor
`tissue in mice. Cabazitaxel
`concentrations in plasma and
`tumor tissue were measured after a
`single intravenous dose of
`cabazitaxel at its HNTD of 40
`mg/kg in C3H/HeN female mice
`bearing advanced-stage
`(400 mm3) mammary
`adenocarcinoma MA16/C. Mean
`concentration  SD from 3 animals
`was determined at each sampling
`time.
`
`Vrignaud et al.
`
`105
`
`104
`
`103
`
`102
`
`101
`
`100
`
`Cabazitaxel concentration (ng/mL or ng/g)
`
`0
`
`10
`
`20
`
`30
`
`40
`
`50
`Time (h)
`
`60
`
`70
`
`80
`
`90
`
`100
`
`reductions, respectively. In addition, host recovery time
`(time from last treatment to recovery of initial body weight)
`was shorter with the intermittent schedule (A1; 8 days)
`compared with the daily (A2; 12 days) and split-dose (A3;
`15 days) schedules. Results of these dose-scheduling studies
`were consistent with further evaluations conducted in C3H/
`HeN mice bearing mammary adenocarcinoma MA17/A
`tumors, which showed a 4.5-fold lower HNTD for a
`split-dose schedule (days 3–7, 3 times daily; HNTD 42
`mg/kg) compared with an intermittent schedule (days 3
`and 7; HNTD 9.3 mg/kg; Supplementary Table S4). Caba-
`zitaxel antitumor activity against MA17/A tumors was also
`lower with the split-dose schedule compared with the inter-
`mittent schedule (log cell kill 1.3 vs. 4.6, respectively). These
`differences between split-dose and intermittent schedules
`were confirmed in a second tumor model, murine mam-
`mary adenocarcinoma MA16/C, which showed a 4.2-fold
`reduction in HNTD (11 vs. 46 mg/kg, respectively) and
`decreased antitumor activity (log cell kill 1.6 vs. 5.4, respec-
`tively). As the intermittent schedule allowed the highest drug
`dose to be administered, with the best host recovery, and had
`the greatest antitumor activity of the schedules tested, inter-
`mittent dosing was selected for further evaluation.
`
`Antitumor activity in docetaxel-sensitive tumors
`The antitumor properties of cabazitaxel in vivo were eval-
`uated using murine tumors grafted in syngenic mice and
`human tumors xenografted in immunocompromised mice
`(Tables 1 and 4). Cabazitaxel was found to be very active
`against murine B16 melanoma (log cell kill 2.1 at HNTD of
`20 mg/kg per injection on days 3, 5, and 7), murine colon
`adenocarcinoma C51 (log cell kill 2.6 at HNTD of 9.3 mg/kg
`per injection on days 4, 6, and 8), and mammary adenocarci-
`nomas MA16/C and MA17/A (reported earlier). Cabazitaxel
`
`was highly active, with CRs observed against the advanced-
`stage murine tumors colon C38 (5 of 5 CR; 5 of 5 TFS) and
`pancreas P03 (5 of 5 CR; 4 of 5 TFS) as well as human tumor
`xenografts including: prostate DU 145 (6 of 6 CR; 5 of 6 TFS);
`colon HCT 116 (7 of 7 CR; 2 of 7 TFS) and HT-29 (6 of 6 CR);
`pancreas MIA PaCa2 (6 of 6 CR; 6 of 6 TFS); breast Calc18
`(5 of 8 TFS); lung NCI-H460 (2 of 6 CR) and A549 (2 of 6 CR);
`head and neck SR475 (6 of 6 CR; 6 of 6 TFS); and kidney Caki-
`1 (5 of 6 CRs). In most of the above models, cabazitaxel and
`docetaxel exhibited similar antitumor activity.
`Dose–response effects were examined in the advanced
`human gastric carcinoma N87 model, a tumor expressing
`HER2 (ref. 34; Fig. 2A). At the HNTD (24.4 mg/kg on days
`27, 31, and 35), cabazitaxel was highly active and delayed
`tumor growth by 101 days (log cell kill > 6, 1 of 8 TFS; P <
`0.0001). The 2 dose levels below the HNTD (15 and 9.3 mg/
`kg per injection) also had a high level of antitumor activity
`(4.5 and 2.5 log cell kill; P < 0.0001 and 0.091, respectively).
`In comparison, docetaxel was also highly active at the
`HNTD, but to a lesser extent (TC ¼ 67 days; log cell kill
`4.5; 1 of 8 TFS; P < 0.0001), and the activity was observed
`only at one dose below the HNTD. Thus, cabazitaxel had a
`greater therapeutic index (3 active dose levels) than doc-
`etaxel (2 active dose levels) in this gastric tumor model.
`
`Antitumor activity in tumor models poorly or not
`sensitive to docetaxel
`Cabazitaxel showed antitumor activity against the fully
`docetaxel-resistant B16/TXT tumor model (log cell kill 2.1
`in B16 vs. 1.3 in B16/TXT), but no antitumor activity was
`obtained against the P-glycoprotein–overexpressing tumor
`Calc18/TXT in which docetaxel resistance was induced
`in vitro (log cell kill 3.4 in Calc18 vs. 0.5 in Calc18/TXT).
`Cabazitaxel was also found to be active against 2 aggressive
`
`2978
`
`Clin Cancer Res; 19(11) June 1, 2013
`
`Clinical Cancer Research
`
`
`
`Downloaded from on January 8, 2014. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org
`
`
`
`

`
`Published OnlineFirst April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-3146
`
`Preclinical Antitumor Activity of Cabazitaxel
`
`2/50/5
`1/60/6
`0/80/8
`N/A1/8
`0/50/5
`NDND
`N/A0/5
`NDND
`NDND
`6/66/6
`8.8
`0.8
`0/50/5
`45.8
`3.4
`6/60/6
`ND
`ND
`NDND
`ND
`ND
`NDND
`days
`cellkillaCRTFS
`Log
`TC,
`Docetaxel
`
`21.3
`40.5
`6.8
`66.8
`40
`ND
`4.1
`ND
`ND
`
`—
`
`1.4
`2.5
`0.5
`4.5
`1.9
`ND
`0.6
`ND
`ND
`
`—
`
`cDocetaxelgroupswerenottreatedonday27.
`HCT-8,MIAPaCa-2andSR475studies:41.7,25and15mg/kgperinjection.
`bThedose–responsepatternfordocetaxelwasdifferentfromthatofcabazitaxelinthefollowingstudies:HT-29,A549andCaki-1studies:51.9,32.2,20and12.4mg/kgperinjection;
`aDefinitionofantitumoractivity:logcellkilltotal<0.7¼inactive;>2.8¼highlyactive.
`Abbreviations:N/A,notavailableastreatmentconductedonearly-stagedisease;ND,notdeterminedinthesamestudy.
`
`5/60/6
`6/66/6
`0/80/8
`N/A1/8
`2/60/6
`2/60/6ND
`N/A0/5
`45
`N/A0/8ND
`N/A5/8ND
`75b
`6/66/6
`50b
`0/50/5
`96.6b
`6/60/6
`7/72/7ND
`6/65/6ND
`
`64.4b
`45b
`23.1
`73.2
`96.6b
`
`1.7
`
`—
`
`25.8
`
`—
`
`18.0
`1.4
`100.9>6
`2.2
`46.0
`2.7
`17.8
`>6
`75.0
`0.5
`7.3
`3.4
`50.2
`
`—
`
`1.9
`2.0
`3.4
`
`—
`
`—
`
`21.5
`27.2
`48.2
`
`—
`
`mg/kg
`TotalHNTD,
`
`cellkillaCRTFS
`log
`
`days
`TC,
`Cabazitaxel
`
`24.0
`42.0
`37.2
`73.2
`36.0
`24.0
`45.0
`38.1
`61.5
`48.0
`28.0
`22.2
`36.0
`48.0
`mg/kg
`TotalHNTD,
`
`KidneyCaki-1(Td:4.6d)
`140
`HeadandneckSR475(Td:4.9d)22.5,14,8.6,5.4(16,20,24)
`250
`GastricGXF-209(Td:4d)
`32.3,20,12.4,7.7(14,17,20)
`130
`GastricN87(Td:4.5d)
`39.4,24.4,15,9.3(27,31,35)
`140
`LungA549(Td:6.4d)
`19.4,12,7.4,4.6(21,27,33)
`130
`LungNCI-H460(Td:2d)
`19.4,12,7.4,4.6(10,13)
`130
`BreastUISOBCA-1(Td:2.1d)
`24.2,15,9.3,5.8(13,16,19)
`70
`BreastCalc18/TXT(Td:4d)
`33,20.5,12.7,7.9(5,7,9)
`N/A
`BreastCalc18(Td:4.5d)
`33,20.5,12.7,7.9(5,7,9)
`N/A
`PancreasMIAPaCa-2(Td:3d)
`19.4,12,7.5,4.6(15,19,23,27c)310
`ColonHCT-8(Td:3.5d)
`250
`22.4,14,8.6,5.4(13,17)
`ColonHT-29(Td:4d)
`140
`19.4,12,7.5,4.6(8,12,16)
`ColonHCT116(Td:4.3d)
`220
`19.4,12,7.5,4.6(16,19,22)
`ProstateDU145(Td:4.5d)
`210
`19.4,12,7.4,4.6(24,30,36,42)
`(scheduleindays)
`Tum

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