throbber
Drug Design, Development and Therapy
`
`Open Access Full Text Article
`
`Preclinical profile of cabazitaxel
`
`Dovepress
`
`open access to scientific and medical research
`
`R e v i e w
`
`Patricia vrignaud 1
`Dorothée Semiond2
`veronique Benning2
`eric Beys 2
`Hervé Bouchard3
`Sunil Gupta4
`1Sanofi Oncology, Vitry-sur-Seine,
`France; 2Sanofi DSAR, Alfortville,
`France; 3Sanofi LGCR, Vitry-sur-
`Seine, France; 4Sanofi Oncology,
`Cambridge, MA, USA
`
`Correspondence: Patricia Vrignaud
`Sanofi Oncology, 13 quai Jules Guesde,
`Vitry-sur-Seine, 94403 Cedex, France
`Tel +33 1 58 93 36 29
`Fax +33 1 58 93 34 71
`email patricia.vrignaud77@gmail.com
`
`Abstract: First-generation taxanes have changed the treatment paradigm for a wide variety
`of cancers, but innate or acquired resistance frequently limits their use. Cabazitaxel is a novel
`second-generation taxane developed to overcome such resistance. In vitro, cabazitaxel showed
`similar antiproliferative activity to docetaxel in taxane-sensitive cell lines and markedly greater
`activity in cell lines resistant to taxanes. In vivo, cabazitaxel demonstrated excellent antitumor
`activity in a broad spectrum of docetaxel-sensitive tumor xenografts, including a castration-
`resistant prostate tumor xenograft, HID28, where cabazitaxel exhibited greater efficacy than
`docetaxel. Importantly, cabazitaxel was also active against tumors with innate or acquired
`resistance to docetaxel, suggesting therapeutic potential for patients progressing following
`taxane treatment and those with docetaxel-refractory tumors. In patients with tumors of the
`central nervous system (CNS), and in patients with pediatric tumors, therapeutic success with
`first-generation taxanes has been limited. Cabazitaxel demonstrated greater antitumor activity
`than docetaxel in xenograft models of CNS disease and pediatric tumors, suggesting potential
`clinical utility in these special patient populations. Based on therapeutic synergism observed
`in an in vivo tumor model, cabazitaxel is also being investigated clinically in combination
`with cisplatin. Nonclinical evaluation of the safety of cabazitaxel in a range of animal species
`showed largely reversible changes in the bone marrow, lymphoid system, gastrointestinal tract,
`and male reproductive system. Preclinical safety signals of cabazitaxel were consistent with
`the previously reported safety profiles of paclitaxel and docetaxel. Clinical observations with
`cabazitaxel were consistent with preclinical results, and cabazitaxel is indicated, in combination
`with prednisone, for the treatment of patients with hormone-refractory metastatic prostate cancer
`previously treated with docetaxel. In conclusion, the demonstrated activity of cabazitaxel in
`tumors with innate or acquired resistance to docetaxel, CNS tumors, and pediatric tumors made
`this agent a candidate for further clinical evaluation in a broader range of patient populations
`compared with first-generation taxanes.
`Keywords: XRP6258, CNS tumors, mCRPC, pediatric tumor, taxane resistance, xenograft
`
`Introduction
`Since the initial approval of paclitaxel (Taxol®; Bristol-Myers Squibb, New York City,
`NY, USA) in 1992,1,2 the first-generation taxanes paclitaxel and docetaxel (Taxotere®;
`Sanofi, Paris, France) have altered the treatment paradigm for a wide variety of solid
`tumors, including breast, lung, prostate, gastric, and ovarian cancers.3,4 Despite dem-
`onstrating significant antitumor activity as monotherapy or in combination regimens,
`clinical use of first-generation taxanes is frequently limited by innate or acquired
`resistance.5–7 In prostate cancer, the majority of patients will eventually acquire resis-
`tance to docetaxel therapy.8
`
`submit your manuscript | www.dovepress.com
`Dovepress
`http://dx.doi.org/10.2147/DDDT.S64940
`
`1851
`Drug Design, Development and Therapy 2014:8 1851–1867
`© 2014 Vrignaud et al. This work is published by Dove Medical Press Limited, and licensed under Creative Commons Attribution – Non Commercial (unported, v3.0)
`License. The full terms of the License are available at http://creativecommons.org/licenses/by-nc/3.0/. Non-commercial uses of the work are permitted without any further
`permission from Dove Medical Press Limited, provided the work is properly attributed. Permissions beyond the scope of the License are administered by Dove Medical Press Limited. Information on
`how to request permission may be found at: http://www.dovepress.com/permissions.php
`
`AVENTIS EXHIBIT 2010
`Mylan v. Aventis
`IPR2016-00627
`
`

`
`vrignaud et al
`
`Dovepress
`
`Cabazitaxel (Jevtana®, Sanofi) is a novel second-
`generation semisynthetic taxane that was identif ied
`through a preclinical screen of 450 molecules derived
`from 10- deacetylbaccatin-III, with the aim of identifying
`a compound with activity in both taxane-sensitive and
`taxane-resistant tumors.9 In the pivotal Phase III TROPIC
`study (NCT00417079), cabazitaxel combined with predni-
`sone significantly extended overall survival compared with
`mitoxantrone plus prednisone in patients with metastatic
`castration-resistant prostate cancer (mCRPC) previously
`treated with a docetaxel-containing regimen.10 This led to
`cabazitaxel’s approval in 2010, in combination with predni-
`sone, for the treatment of patients with hormone-refractory
`metastatic prostate cancer who have previously received
`docetaxel-based therapy.11,12
`This review article presents an overview of the preclinical
`properties of cabazitaxel, including its development, mecha-
`nism of action, antitumor activity in a range of in vitro and
`in vivo tumor models, pharmacokinetics (PK), and metabolic
`and toxicity profiles, as well as a summary of its clinical
`development.
`
`Taxanes’ mechanism of action
`and resistance mechanisms
`Mechanism of action
`Taxanes are microtubule inhibitors that induce cellular
`apoptosis through the stabilization of microtubules.7 Micro-
`tubules are major components of the cytoskeleton, with
`critical roles in a variety of cellular processes including
`maintenance of cell shape, intracellular transport, cell signal-
`ing, and cell division.7,13–15 It is this pivotal role in mitosis
`that makes microtubules a key cellular target for anticancer
`therapeutics.7
`Microtubules are highly dynamic polymers of tubulin,
`continually undergoing assembly and disassembly within the
`cell. Taxanes inhibit microtubule function by binding to tubu-
`lin molecules, promoting their polymerization, and stabilizing
`microtubules. Suppression of microtubule dynamics leads to
`a block in mitosis and, ultimately, tumor cell death.7,13,14
`
`Resistance mechanisms
`Innate or acquired resistance to first-generation taxanes is
`frequently observed in different tumor types, resulting in
`treatment failure. Multiple potential mechanisms of taxane
`resistance have been identified in preclinical studies, and
`it is likely that several of these contribute to a resistant
`phenotype.6,7,16–19
`Two mechanisms in particular have frequently been
`associated with the development of resistance to taxanes;
`
`1852
`
`submit your manuscript | www.dovepress.com
`Dovepress
`
`however, it is worth noting that these are yet to be validated
`in patient samples, and their clinical relevance is not fully
`understood.6,7 In preclinical studies, resistance commonly
`results from overexpression of members of the ATP-binding
`cassette family of transporters, of which P-glycoprotein,
`encoded by the multidrug resistance gene (ATP-binding
`cassette, sub-family B [MDR/TAP], member 1; ABCB1), is
`the best known.20 Docetaxel and paclitaxel are substrates of
`P-glycoprotein, which acts as a drug efflux pump, decreas-
`ing intracellular drug levels and limiting cytotoxicity.6,7,21,22
`Resistance may also arise from spontaneously acquired
`mutations in tubulin, the cellular target of taxanes, resulting
`in changes to the tubulin binding site or altered microtubule
`dynamics.6,7,23
`Clinical data suggest that additional mechanisms may
`contribute to taxane resistance in patients, including the
`altered expression of specific tubulin isotypes,17 and expres-
`sion or binding of microtubule-regulatory proteins,18 loss of
`functional p53,16 dysfunctional regulation of apoptotic and
`intracellular signaling (eg, HER2 overexpression),19 and
`decreased tumor cell permeability.24
`A number of potential predictive markers for taxane
` resistance have been identified, including the mitotic
`spindle checkpoint proteins Aurora A, BUBR1, MAD2, and
`synuclein-γ, and cell cycle proteins such as BRCA1;18 however,
`conflicting results have been reported clinically.24
`The development of alternative therapies able to over-
`come taxane resistance has been the focus of considerable
`attention.
`
`Cabazitaxel development
`Paclitaxel and docetaxel are semisynthetic derivatives of
`10-deacetylbaccatin-III,25,26 a natural paclitaxel precursor
`molecule that can be extracted easily and sustainably from
`the needles of the European yew tree (Figure 1).26 In light
`of the clinical limitations that result from taxane resistance,
`a large-scale preclinical screening process was undertaken
`that aimed to identify a taxane derivative with equivalent
`efficacy to docetaxel in docetaxel-sensitive tumors, but
`greater activity than docetaxel in tumors that are docetaxel-
`resistant.9
`In total, 450 candidate molecules were designed and
`generated for preclinical assessment, based on preclinical
`comparative structure–activity relationships of paclitaxel
`and docetaxel. Structural modifications initially focused on
`the side chain, as this was considered critical for potency,
`with subsequent modifications to other functional groups
`within the baccatin moiety.9 The antitumor potential of the
`taxane derivatives was assessed over three stages: in vitro
`
`Drug Design, Development and Therapy 2014:8
`
`

`
`Dovepress
`
`Preclinical profile of cabazitaxel
`
`A
`
`B
`
`C
`
`Side chain
`attachment
`at C-13
`
`HO
`
`13
`
`HO
`
`O
`
`O
`
`NH
`
`O
`
`O
`
`OH
`
`HO
`
`HO
`
`O
`
`OH
`
`10
`
`H
`
`O
`
`OCOCH3
`OCOC6H5
`
`HO
`
`O
`
`OH
`
`H
`
`O
`
`OCOCH3
`OCOC6H5
`
`Baccatin moiety
`
`Side chain
`
`O
`
`O
`
`NH
`
`O
`
`10
`
`H3C
`
`O
`
`O
`
`CH3
`
`O
`
`7
`
`O
`
`OH
`
`HO
`
`H
`
`O
`
`OCOCH3
`OCOC6H5
`
`Figure 1 Chemical structure of 10-deacetylbaccatin III, docetaxel, and cabazitaxel.
`Notes: (A) 10-deacetylbaccatin III. (B) Docetaxel. (C) Cabazitaxel.
`
`activity against microtubules; in vitro activity in resistant
`cell lines; and in vivo activity in a tumor model.24 The in
`vivo assessments included evaluation in a B16/TXT mela-
`noma resistance model, which was developed through repeat
`exposure to docetaxel in mice bearing the docetaxel-sensitive
`
`B16 tumor, to allow evaluation of the taxane derivatives in
`a clinically relevant setting. This model mimics the clinical
`development of docetaxel resistance, where tumors initially
`respond to treatment, but develop resistance progressively
`over time.24
`
`Drug Design, Development and Therapy 2014:8
`
`submit your manuscript | www.dovepress.com
`Dovepress
`
`1853
`
`

`
`vrignaud et al
`
`Dovepress
`
`Initial attempts to modify the C-3′N–Boc and C-3′ phenyl
`groups within the side chain of docetaxel resulted in derivatives
`demonstrating either reduced in vitro potency or failure to
`improve activity in docetaxel-resistant cell lines (Figure S1).9,27
`Alterations to the C-2/C-4 and oxetane ring regions of the bac-
`catin moiety were also evaluated, but failed to increase activity
`in the in vitro and/or in vivo resistance models.9
`Cabazitaxel is a dimethyl derivative of docetaxel, bear-
`ing methoxy groups in place of hydroxyl groups at posi-
`tions C-7 and C-10 (Figure 1).9 In both docetaxel-sensitive
`and docetaxel-resistant cell lines, these structural altera-
`tions resulted in the greatest increase in in vitro potency,
`without significantly increasing toxicity at the maximum
`tolerated dose, in contrast to other C-7/C-10 modifications
`(Figure S1).9 These modifications confer two advantages
`on cabazitaxel over docetaxel. Firstly, cabazitaxel, which is
`a P-glycoprotein substrate, has a higher lipophilicity than
`docetaxel (logP 3.9 versus 3.2),9,28 resulting from the con-
`version of two secondary alcohols to more lipophilic ethers.
`This may result in increased cell penetration through passive
`influx, consequently leading to better activity in resistant cell
`lines where permeability of the plasma membrane may be
`altered.24,28–30 This hypothesis was recently confirmed in an
`experiment in which drug uptake into MCF7 breast adeno-
`carcinoma cells, which do not overexpress P-glycoprotein,
`was faster for cabazitaxel than for docetaxel.31 Secondly,
`cabazitaxel has an improved ability to cross the blood–brain
`barrier (BBB) compared with docetaxel, offering potential
`benefit in patients with tumors of the central nervous system
`(CNS).9,29,32,33
`Accordingly, during the screening of taxane derivatives
`and subsequent preclinical evaluation, cabazitaxel has
`demonstrated equivalent efficacy to docetaxel for stabiliz-
`ing microtubules in vitro, greater potency than docetaxel in
`cell lines resistant to taxanes and other chemotherapeutics,
`activity superior to docetaxel in in vivo CNS disease models,
`broad-spectrum antitumor activity against a range of murine
`and human tumors, and in vivo activity in tumor models that
`are not sensitive, or are poorly sensitive, to docetaxel.9,24,33
`
`In vitro activity
`Microtubule stabilization
`Cabazitaxel has shown equivalent potency to docetaxel for
`stabilization of microtubules in vitro. Both cabazitaxel and
`docetaxel induced a similar reduction in lag time for tubulin
`assembly (lag time to 50% aggregation 0–0.1 µmol/L) and stabi-
`lization of microtubules against cold-induced depolymerization
`(concentration producing 50% cell killing 0.1–0.25 µmol/L),
`
`indicating that cabazitaxel has a cytotoxic mechanism of action
`similar to that of docetaxel.24
`
`Antiproliferative activity
`In cell lines sensitive to chemotherapy, cabazitaxel had similar
`antiproliferative activity to docetaxel, achieving comparable
`50% inhibitory concentration (IC50) values across a range of
`murine and human cell types (0.004–0.041 µmol/L for cabazi-
`taxel versus 0.008–0.079 µmol/L for docetaxel) (Table 1).24
`In a panel of cell lines bearing acquired resistance to
`taxanes or to the chemotherapeutic agents doxorubicin, vin-
`cristine, or vinblastine, cabazitaxel showed markedly greater
`antiproliferative activity than docetaxel (IC50 ranged from
`0.016–0.414 µmol/L for cabazitaxel versus 0.17–4.01 µmol/L
`for docetaxel).24 Resistance factors, an indication of the dif-
`ference in drug concentrations needed to inhibit resistant
`versus sensitive/parental cell lines, ranged from 2–10 for
`cabazitaxel and 5–59 for docetaxel in these P-glycoprotein-
`expressing cell lines (Table 1).24
`In murine and human cell lines with resistance mecha-
`nisms other than P-glycoprotein overexpression, no cross-
`resistance to cabazitaxel was observed.24
`
`In vivo activity
`Plasma pharmacokinetics
`The PK profile of cabazitaxel was evaluated in healthy and
`tumor-bearing mice, and healthy rats and dogs (Table 2 )
`(Sanofi, data on file, 2010).
`
`Absorption
`Exposure to cabazitaxel increased with dose after single or
`repeated intravenous (IV) administration in all species. The
`increase in exposure was approximately dose-proportional
`in mice and more than dose-proportional in rats and dogs.
`No plasma accumulation was observed in mice, rats, or dogs
`after administration every 5 days, weekly, or every 3 weeks.
`No sex effect was observed in rats and dogs (Sanofi, data
`on file, 2010).
`
`Distribution
`Plasma protein binding of cabazitaxel was very high in mice
`(99.3%) and high in rats (95.5%), rabbits (91.4%), dogs
`(97.1%), and humans (91.9%) (Sanofi, data on file, 2010).
`Following a single IV administration, cabazitaxel exhibited
`a very large volume of distribution at steady state in both
`healthy (2.5–3.7 L/kg) and tumor-bearing mice (8.8 L/kg),
`in rats (22.7 L/kg), and in dogs (3.3–14.5 L/kg) (Sanofi, data
`on file, 2010).
`
`1854
`
`submit your manuscript | www.dovepress.com
`Dovepress
`
`Drug Design, Development and Therapy 2014:8
`
`

`
`Dovepress
`
`Preclinical profile of cabazitaxel
`
`Table 1 In vitro antiproliferative effects of cabazitaxel and docetaxel against sensitive and P-glycoprotein-expressing resistant cell
`lines
`
`Cell line
`
`ABCB1
`mRNA levelb
`
`Mean IC50, μmol/L ± SD
`Resistance factora
`Cabazitaxel
`Docetaxel
`Docetaxel
`Cabazitaxel
`-
`-
`-
`0.079±0.004
`0.041±0.017
`P388 murine leukemia
`+++
` 4.01±0.280
`0.414±0.036
`10
`51
`P388/DOX
`-
`-
`-
`0.039±0.012
`0.013±0.005
`P388 murine leukemia
`++
`0.188±0.022
`0.024±0.015
`2
`5
`P388/TXT
`-
`-
`-
`0.039±0.012
`0.013±0.005
`P388 murine leukemia
`++
`0.227±0.038
`0.024±0.003
`2
`6
`P388/VCR
`-
`-
`-
`0.031±0.004
`0.022±0.010
`HL60 human leukemia
`++
`0.250±0.110
`0.060±0.029
`3
`8
`HL60/TAX
`-
`-
`-
`0.008±0.002
`0.004±0.002
`Calc18 human breast adenocarcinoma
`++
`0.170±0.040
`0.016±0.004
`4
`21
`Calc18/TXT
`-
`-
`-
`0.042±0.021
`0.035±0.026
`KB human epidermoid carcinoma
`++++
`2.480±0.120
`0.270±0.013
`8
`59
`KBv1
`Notes: Cells were incubated for 96 hours at 37°C in liquid medium with drugs at different concentrations. Viability was assessed by neutral red, with the mean of at least
`three results obtained. aResistance factor = IC50 (resistant)/IC50 (parental) from the same experiment; brelative expression obtained from Northern blot experiments using
`the human ABCB1 gene as probe. Reprinted by permission from the American Association for Cancer Research: Vrignaud P, Sémiond D, Lejeune P, et al. Preclinical antitumor
`activity of cabazitaxel, a semi-synthetic taxane active in taxane-resistant tumors. Clin Cancer Res. 2013;19:2973–2983, doi: 10.1158/1078-0432.CCR-12-3146.24
`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; IC50, 50% inhibitory concentration; KBV1, 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; SD, standard deviation.
`
`The PK of cabazitaxel was also evaluated in mice
`bearing advanced-stage ( 400 mm 3) mammar y
`MA16/C adenocarcinomas.24 Cabazitaxel was adminis-
`tered at the highest nontoxic dose (HNTD) of 40 mg/kg.
`Drug uptake into the tumor was both rapid and sustained,
`with maximum drug concentrations in tumor tissue reached
`within 15 minutes, and a 40-fold greater concentration of
`cabazitaxel within the tumor versus plasma after 48 hours
`(Figure 2).24
`In this model, exposure to cabazitaxel was 1.6-times greater
`in the tumor compared with plasma during the 48 hours fol-
`lowing treatment administration, and 2.9-times greater over
`
`the entire experimental period (168 hours). Concentrations
`of cabazitaxel above the cellular antiproliferative IC50 were
`sustained for 24 hours in the plasma and for 96 hours in
`tumor tissue.24
`Brain distribution of cabazitaxel was assessed in mice,
`rats, and dogs. Cabazitaxel penetrated rapidly in the brain,
`with similar relative exposure between brain and blood across
`the different species.33
`
`Metabolism
`Cabazitaxel metabolism has been compared across mul-
`tiple species. In vivo, cabazitaxel was the major circulating
`
`Normal mice
`
`F
`
`Tumor-bearing mouse
`Rat
`Dog
`
`Table 2 Pharmacokinetic parameters of cabazitaxel in normal and tumor-bearing mice, rats, and dogs
`Species
`Sex
`Dose
`Number of
`Infusion
`AUC
`Cmax
`(ng ⋅ h/mL)
`(M/F)
`(mg/kg)
`administrations
`duration
`(ng/mL)
`F
`5
`1
`1 h
`2,728
`4,468
`10
`1
`1 h
`4,805
`11,211
`15
`1
`1 h
`6,072
`13,460
`5
`5a
`1 h
`4,421
`6,881
`10
`5a
`1 h
`6,478
`17,497
`15
`5a
`1 h
`6,504
`13,489
`40
`1
`45 s
`23,784
`24,113
`F
`2.5
`1
`1 h
`477
`522
`M
`0.5
`1
`72–91 m
`65
`95
`M
`0.5
`1
`72–91 m
`97
`101
`F
`1
`1
`72–91 m
`164
`230
`M
`1
`1
`72–91 m
`360
`417
`F
`Notes: aevery 3 weeks. Sanofi, data on file, 2013.
`Abbreviations: AUC, area under the concentration–time curve; CL, clearance; Cmax, maximum plasma concentration; F, female; M, male; t1/2, half-life; Vss, steady-state volume
`of distribution.
`
`CL
`(L/hr/kg)
`1.1
`0.9
`1.1
`0.7
`0.6
`1.1
`1.7
`4.8
`5.3
`5.2
`4.4
`2.5
`
`Vss
`(L/kg)
`2.5
`2.7
`3.7
`2.1
`1.1
`2.8
`8.8
`22.7
`14.5
`12.8
`9.5
`3.3
`
`t1/2
`(hr)
`5.1
`7.4
`7.6
`4.9
`5.5
`7.5
`26.0
`10.1
`4.3
`3.2
`3.8
`3.0
`
`Drug Design, Development and Therapy 2014:8
`
`submit your manuscript | www.dovepress.com
`Dovepress
`
`1855
`
`

`
`vrignaud et al
`
`Dovepress
`
`Plasma
`Tumor
`
`the dose). In contrast, unchanged cabazitaxel was the major
`circulating compound in plasma in all species, including
`humans, accounting for 65% of the total radioactivity area
`under the curve.34
`
`105
`
`104
`
`103
`
`102
`
`101
`
`100
`
`(ng/mL or ng/g)
`
`Cabazitaxel concentration
`
`0
`
`10
`
`20
`
`30
`
`50
`40
`60
`Time (h)
`
`70
`
`80
`
`90 100
`
`Figure 2 Pharmacokinetics of 40 mg/kg cabazitaxel (highest nontoxic dose) in
`plasma and tumor tissues in mice.
`Note: Reprinted by permission from the American Association for Cancer
`Research: Vrignaud P, Sémiond D, Lejeune P, et al. Preclinical antitumor activity of
`cabazitaxel, a semi-synthetic taxane active in taxane-resistant tumors. Clin Cancer
`Res. 2013;19:2973–2983, doi: 10.1158/1078-0432.CCR-12-3146.24
`compound in all species including humans (65% of the
`total radioactivity area under the curve). Cabazitaxel under-
`goes extensive biotransformation, with less than 2.5% of the
`administered dose excreted unchanged in studies of mice,
`rats, dogs, and humans. The two main metabolic pathways,
`accounting for 40%, 54%, and 45% of the dose excreted in
`humans, dogs, and mice, respectively, corresponded to two
`O-demethylations at the C-7 and C-10 positions, one leading
`to 7-O-demethyl-cabazitaxel (pathway B) and the other to
`10-O-demethyl-cabazitaxel (pathway A) (Figure 3; Table 3).
`In male and female rats, the main pathway corresponded
`to t-butyl-hydroxylation on the lateral chain (pathway C),
`accounting for 49%–55% of the dose excreted. This pathway
`was also abundant in mice, representing 41% of the dose, and
`was found in humans (21% of the dose). Another pathway
`consisting of the cleavage of the parent drug (pathway D)
`was very minor in all species, including humans (0.3% of
`
`excretion
`The PK of cabazitaxel following a single IV infusion in
`normal mice, tumor-bearing mice, and in dogs is generally
`characterized by a biphasic elimination (Sanofi, data on file,
`2010). Plasma clearance was high in rats (4.8 L/h/kg) and dogs
`(2.5–5.3 L/h/kg) and moderate in normal (0.9–1.1 L/h/kg)
`and tumor-bearing mice (1.7 L/h/kg), compared with respec-
`tive hepatic blood flow. The terminal half-life was moderately
`long in dogs (3.0 to 4.3 hours), long in normal mice (5.1–7.6
`hours) and rats (10 hours), and extremely long in tumor-
`bearing mice (26 hours). It should be noted that a higher
`dose was given to tumor-bearing mice, which was reflected
`in quantifiable levels occurring at later sampling times than
`in normal mice (Sanofi, data on file, 2010).
`Cabazitaxel excretion was nearly complete (91%–95%
`of the administered dose) in mice, rats, and dogs, and radio-
`activity was largely excreted in the feces in all three species
`(87%–91% of the administered dose), with minimal excretion
`via the urinary route (1%–4%). In bile-duct-cannulated male
`rats, radioactivity was mainly excreted via the biliary route,
`accounting for 65% of the administered dose within 48 hours.
`Similarly, in humans the majority of the radioactivity was
`recovered in feces (around 76% of the administered dose),
`with 4% of the dose found in urine.34
`In summary, the disposition of cabazitaxel was assessed
`in a range of animal species, and the distribution, metabolic
`pathways, and elimination processes documented in ani-
`mals are consistent with those observed in humans (Sanofi,
`
`Pathway D cleavage
`
`Pathway A 10-O demethylation
`Pathway B 7-O demethylation
`
`Pathway C hydroxylation
`
`O
`
`H3C
`
`O
`
`O
`
`O
`
`NH
`
`O
`
`10
`
`CH3
`
`O
`
`7
`
`O
`
`OH
`
`HO
`
`Lateral chain
`
`H
`
`O
`OCOCH3
`OCOC6H5
`Taxane ring
`
`Figure 3 Proposed schematic of the principal metabolic pathways of cabazitaxel.
`
`1856
`
`submit your manuscript | www.dovepress.com
`Dovepress
`
`Drug Design, Development and Therapy 2014:8
`
`

`
`Dovepress
`
`Preclinical profile of cabazitaxel
`
`Table 3 Relative contribution of different metabolic pathways to
`cabazitaxel metabolism across species
`
`Pathway
`
`% of administered dose
`Dog Human
`Mouse
`Rat
`Rat
`Female Male Female Male Male and
`female
`16.2
`
`9.8
`
`5.7
`
`19.7
`
`16.4
`
`(complete response [CR]) and 83% long-term tumor-free
`survival (TFS).24
`Cabazitaxel was compared with docetaxel and abiraterone
`acetate (a specific inhibitor of CYP17) in a patient-derived
`prostate tumor xenograft with induced resistance to castration
`(HID28). At equivalent dose levels (20 mg/kg), cabazitaxel
`demonstrated greater antitumor efficacy than docetaxel.38
`Cabazitaxel inhibited tumor growth with a percentage tumor
`volume change (calculated as the percentage ratio between
`the mean tumor volume of a treated and a control group)
`of 1.4% at Day 35 compared with 16.7% for docetaxel
`(Figure 4).38 Complete and partial remissions, respectively,
`were achieved in six out of ten and four out of ten mice
`receiving cabazitaxel, and in one out of ten and two out
`of ten mice treated with docetaxel.38 No antitumor activity
`was demonstrated with abiraterone acetate (50 mg/kg orally
`administered daily for 21 days).38 These data provide support
`for the clinical development of cabazitaxel for the first-line
`treatment of mCRPC.
`
`Docetaxel-sensitive models
`Cabazitaxel demonstrated broad-spectrum activity in murine
`and human xenograft models (Table 4). Excellent antitumor
`activity was observed in murine B16 melanoma, colon adeno-
`carcinoma C51, and mammary adenocarcinoma MA16/C and
`MA17/A tumors.24 In a range of advanced disease models,
`including human colon HCT 116 and HT-29, lung A549 and
`NCI-H460, pancreas MIA PaCa-2, head and neck SR475,
`and kidney Caki-1 xenografts, administration of cabazitaxel
`
`Vehicle (once every 3 weeks)
`Docetaxel 20 mg/kg (once every 3 weeks)
`Cabazitaxel 20 mg/kg (once every 3 weeks)
`Cabazitaxel 15 mg/kg (once every 3 weeks)
`Abiraterone 50 mg/kg (daily for 21 days)
`
`Pathway A
`(10-O-demethylation)
`Pathway B
`(7-O-demethylation)
`Pathway C
`(hydroxylation on
`lateral chain)
`0.1
`Pathway D (cleavage)
`Note: Sanofi, data on file, 2010.
`
`28.4
`
`40.5
`
`15.1
`
`12.7
`
`34.7
`
`24.2
`
`49.3
`
`54.8
`
`12.6
`
`20.9
`
`0.1
`
`0.3
`
`0.3
`
`Trace
`
`data on file, 2010). Thus, the animals studied are likely to
`represent good models for the disposition of cabazitaxel in
`humans. The finding that unchanged cabazitaxel is the main
`circulating compound in plasma indicates that analysis of
`the parent drug is appropriate for PK and pharmacodynam-
`ics studies of cabazitaxel.
`
`Antitumor activity
`Prostate cancer models
`The efficacy of taxanes in prostate cancer, in addition to
`their impact on cell division, may in part relate to inhibitory
`effects on androgen receptor signaling.35–37 In mice bear-
`ing a docetaxel-sensitive cell line-derived castrate-resistant
`prostate cancer xenograft (DU145), cabazitaxel was found to
`be highly active, inducing 100% complete tumor regressions
`
`2,000
`
`1,500
`
`1,000
`
`500
`
`(mean ± SEM)
`
`Tumor volume (mm3)
`
`0
`
`0
`
`7
`
`14
`
`21
`
`28
`Days
`
`35
`
`42
`
`49
`
`Vehicle, cabazitaxel 15 and 20 mg/kg, and docetaxel 20 mg/kg
`treatments (Day 0 and Day 21)
`Abiraterone 50 mg/kg treatments (Day 0 to Day 20)
`
`Figure 4 Antitumor activity of cabazitaxel, docetaxel, and abiraterone in a docetaxel-sensitive hormone-refractory prostate cancer xenograft model.
`Note: Sanofi, data on file, 2012.
`Abbreviation: SEM, standard error of the mean.
`
`Drug Design, Development and Therapy 2014:8
`
`submit your manuscript | www.dovepress.com
`Dovepress
`
`1857
`
`

`
`vrignaud et al
`
`Dovepress
`
`Table 4 Dose–response antitumor activity of cabazitaxel and docetaxel in mice bearing murine and human docetaxel-sensitive and
`-resistant tumors
`
`Tumor type
`
`Cabazitaxel
`Total HNTD,
`mg/kg
`
`Log
`cell killa
`
`CR
`
`TFS
`
`Docetaxel
`Total HNTD,
`mg/kg
`
`Log
`cell killa
`
`CR
`
`TFS
`
`NA
`NA
`5/5
`NA
`5/5
`NA
`4/5
`NA
`NA
`
`0/5
`0/5
`5/5
`0/5
`4/5
`0/5
`0/5
`0/5
`0/5
`
`60
`60
`60
`45
`ND
`ND
`ND
`ND
`ND
`
`1.7
`0.6
`3.1
`3.1
`ND
`ND
`ND
`ND
`ND
`
`NA
`NA
`0/5
`NA
`ND
`ND
`ND
`ND
`ND
`
`0/5
`0/5
`0/5
`0/5
`ND
`ND
`ND
`ND
`ND
`
`60
`60
`60
`45
`60
`60
`40
`36
`58.5
`
`2.1
`1.3
`–
`2.6
`–
`0.8
`3.7
`3.9
`1.2
`
`Murine tumors
` B16 melanoma
` B16/TXT melanoma
` Colon C38
` Colon C51
` Pancreas P03
` Pancreas P02
` Mammary MA16/C
` Mammary MA17/A
` Lung 3LL
`Human tumors
`ND
`ND
`ND
`ND
`5/6
`6/6
`–
`48.0
` Prostate DU 145
`ND
`ND
`ND
`ND
`2/7
`7/7
`3.4
`36.0
` Colon HCT 116
`0/6
`6/6
`3.4
`96.6b
`0/6
`6/6
`2.0
`22.2
` Colon HT-29
`0/5
`0/5
`0.8
`50b
`0/5
`0/5
`1.9
`28.0
` Colon HCT-8
`6/6
`6/6
`–
`75b
`6/6
`6/6
`–
`48.0
` Pancreas MIA PaCa-2
`ND
`ND
`ND
`ND
`5/8
`NA
`3.4
`61.5
` Breast Calc18
`ND
`ND
`ND
`ND
`0/8
`NA
`0.5
`38.1
` Breast Calc18/TXT
`0/5
`NA
`0.6
`45
`0/5
`NA
`45.0
` Breast UISO BCA-1
`6
`ND
`ND
`ND
`ND
`0/6
`2/6
`2.7
`24.0
` Lung NCI-H460
`0/5
`0/5
`1.9
`96.6b
`0/6
`2/6
`2.2
`36.0
` Lung A549
`1/8
`NA
`4.5
`73.2
`1/8
`NA
`73.2
` Gastric N87
`6
`0/8
`0/8
`0.5
`23.1
`0/8
`0/8
`1.4
`37.2
` Gastric GXF-209
`0/6
`1/6
`2.5
`45b
`6/6
`6/6
`–
`42.0
` Head and neck SR475
`0/5
`2/5
`1.4
`64.4b
`0/6
`5/6
`1.7
`24.0
` Kidney Caki-1
`Notes: Murine tumors were originally obtained and then maintained in the same syngenic mouse strain for MA17/A (C3H/HeN) and C51 (BALB/c), and in B6D2F1 mice for
`the C57BL/6 syngenic tumors. Human tumors were grafted in immunocompromised mice. aDefinition of antitumor activity (log cell kill = tumor growth delay/3.32 × tumor
`doubling time): log cell kill total 0.7= inactive, 2.8= highly active; bthe dose–response pattern for docetaxel was different from that of cabazitaxel in the following studies:
`HT-29, A549, and Caki-1 studies – 51.9, 32.2, 20, and 12.4 mg/kg per injection; HCT-8, MIA PaCa-2, and SR475 studies – 41.7, 25, and 15 mg/kg per injection. Adapted by
`permission from the American Association for Cancer Research: Vrignaud P, Sémiond D, Lejeune P, et al. Preclinical antitumor activity of cabazitaxel, a semi-synthetic taxane
`active in taxane-resistant tumors. Clin Cancer Res. 2013;19:2973–2983, doi: 10.1158/1078-0432.CCR-12-3146.24
`Abbreviations: CR, complete regression; HNTD, highest nontoxic dose; NA, not available as treatment conducted on early-stage disease; ND, not determined in the same
`study; TFS, long-term tumor-free survivors.
`
`achieved CR. In seven of eleven such models, CR levels were
`100%. In seven models, CR resulted in TFS at study comple-
`tion, with 80 to 100% TFS in five of them.24
`In most cases, the antitumor activity of cabazitaxel was
`similar to that of docetaxel, as assessed by log cell kill values
`derived from measurements of tumor growth (log cell kill =
`tumor growth delay/3.32× tumor doubling time).24 Despite
`this, docetaxel induced CR in just four of seven evaluable
`xenograft models (human colon HCT-29, pancreas MIA
`PaCa-2, head and neck SR475, and kidney Caki-1), and
`TFS at study completion was only observed in mice bearing
`human pancreas MIA PaCa-2 xenografts.
`
`Models resistant or poorly sensitive to docetaxel
`Cabazitaxel was evaluated in the B16/TXT acquired-
` resistance model, which was developed by repeatedly
`exposing mice bearing the docetaxel-sensitive murine B16
`
`melanoma to docetaxel at the HNTD (60 mg/kg per passage),
`with a total of 27 passages over 17 months required to obtain
`a fully docetaxel-resistant tumor. Cabazitaxel demonstrated
`antitumor activity against B16/TXT, which does not over-
`express P-glycoprotein, but was not active against the
`P-glycoprotein-overexpressing tumor Calc18/TXT in which
`docetaxel resistance was induced in vitro.24
`Interestingly, cabazitaxel showed activity against a number of
`murine and human xenograft models bearing innate resistance to
`docetaxel, including the human breast tumor UISO BCA-1. In
`this early-stage model, cabazitaxel achieved a log cell kill value
`of greater than 6, compared with 0.6 with docetaxel. Moreover,
`docetaxel did not delay tumor growth at its HNTD (15 mg/kg
`per injection; log cell kill 0.6, P0.5), whereas cabazitaxel was
`highly active both at its HNTD (15 mg/kg per injection; log cell
`kill 6, P=0.0016) and at the dose level below (9.3 mg/kg per
`injection; log cell kill 4.4, P=0.0016).24
`
`1858
`
`submit your manuscript | www.dovepress.com
`Dovepress
`
`Drug Design, Development and Therapy 2014:8
`
`

`
`Dovepress
`
`Preclinical profile of cabazitaxel
`
`Taken together, these findings made cabazitaxel a can-
`didate for further clinical evaluation in patients who have
`relapsed following taxane treatment, as well as in those with
`innately docetaxel-refractory tumors.24
`
`Special populations
`CNS tumors
`The activity of first-generation taxanes in patients with
`tumors of the CNS is limited,39–43 due to poor penetration
`of these drugs across the BBB under normal physiologic
`conditions.44,

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