throbber
8
`
`
`
`The Taxoids
`
`
`
`Marie-Christine Bissery, PhD and Francois Lavelle, PhD
`
`CONTENTS
`
`INTRODUCTION
`DISCOVERY
`
`MECHANISM OF ACTION OF TAXOIDs
`ANTITUMOR PROPERTIES
`ANIMAL PHARMACOLOGY
`ANIMAL TOXICOLOGY
`CONCLUSION
`
`1 . INTRODUCTION
`
`Paclitaxel and docetaxel belong to the taxoid family, a new class of antineoplastic
`drugs. The name taxoids refers to compounds, natural or modified, having a taxane
`skeleton.
`
`Paclitaxel (Taxol®, NSC 125973) was extracted in the late 19605 from the bark of
`the Pacific Yew, Taxus brevifolia. Because of the scarcity of the drug and the difficul-
`ties of formulation, the development was initially slow. Once these problems were
`solved, development accelerated. Docetaxel (Taxotere®, PR 56976) was obtained by
`hemisynthesis, using the starting material, 10-deacetyl baccatin III extracted from the
`needles of the European Yew tree, Taxus baccata (Fig. 1). This drug was more readily
`available because of the renewability of the source, and somewhat more soluble, and
`thus its development was rapid. Paclitaxel consists of an eight-member taxane ring
`with a four-member oxetane ring and a side chain at the C-13 position (Fig. l). Doce-
`taxel differs from paclitaxel in the 10-position on the baccatin ring and in the 3 '-posi-
`tion of the lateral chain (Fig. 1).
`This chapter will summarize the key steps in the development process of these two
`new exciting antitumor agents.
`
`2. DISCOVERY
`
`2. 1. Paclitaxel
`
`In 1960, a vast screening program for antitumor agents derived from plants was
`initiated by the Cancer Chemotherapy National Service Center under J. L. Hartwell
`(1—3). In 1962, a US Department of Agriculture botanist, A. S. Barclay, collected 650
`
`From: Cancer Therapeutics: Experimental and Clinical Agents
`Edited by: B. Teicher Humana Press Inc., Totowa, NJ
`
`175
`
`Genentech 2070
`
`Hospira v. Genentech
`|PR2017-00737
`
`Genentech 2070
`Hospira v. Genentech
`IPR2017-00737
`
`

`

`1 76
`
`Part I / Cytotoxic Agents
`
`HO
`
`0
`
`HO In.
`
` OH
`(530006»:5
`
`10-Deacetyl Baccatin Ill
`
`
`
`Fl1 = ‘CeHs
`
`R2 = —COCH3
`
`paclitaxel
`
`F!1 = -OC(CH3)3
`
`R2 = -H
`
`docetaxel
`
`Fig. 1. Structures of paclitaxel (Taxol®), docetaxel (Taxotere®) and 10-deacetyl-baccatin 111.
`
`samples in the West Coast States of the US, including samples of T. brevifolia, the
`Pacific Yew tree. Initial screening of crude extracts showed cytotoxicity toward 9KB
`cells derived from a nasopharyngeal tumor. M. Wall at Research Triangle Institute
`was particularly interested in plants with 9KB activity because of his previous
`experience with Camptotheca extracts. This is why he received among other plants,
`T. brevifolia. The isolation procedure of the drug substance was laborious and involved
`numerous steps (ethanol extraction; partition of the ethanolic residue between water
`and chloroform; followed by Craig countercurrent distribution), each of them being
`monitored by an in vivo assay using the rat Walker 256 carcinosarcoma (2—4) or/and
`the P1534 leukemia (1 ). Approximately 0.5 g of paclitaxel could be isolated starting
`with 12 kg of dried stem and bark (yield 0.004%). The isolation of the pure com-
`pound was achieved in 1966(1). In 1971 the structure of paclitaxel was published, and
`antitumor efficacy was reported in L1210, P388, and P15341eukemias, and in Walker
`carcinosarcoma 256 (5). However, the activity levels seen against the L1210 and P388
`leukemia models were very modest compared to that of other compounds. The only
`tumor system showing good efficacy, the P1534 leukemia, was not thought to be of
`predictive value (I ). Finally, the compound was poorly soluble (i.e., the in vivo evalu-
`
`

`

`Chapter 8/ The Taxoids
`
`177
`
`ation was performed with drug suspension), and supplies were difficult to obtain.
`Because of this, paclitaxel was not selected for further preclinical development (1).
`Luckily in 1974—1975, extensive in vivo testing was conducted by the National Cancer
`Institute (NCI), and paclitaxel showed high activity against the murine B16 melanoma
`model, newly introduced to the NCI screen (1 ). This prompted the NCI to evaluate
`the compound further. Additional efficacy was noted in colon 26 carcinoma and in
`MX-l human breast carcinoma implanted under the renal capsule in nude mice (6).
`However, no efficacy was observed at that time in most of the other models tested,
`colon 38 adenocarcinoma, Lewis lung carcinoma, CD8F1 mammary, and human
`xenografts implanted subcutaneously (6). Preliminary studies of its mechanism of
`action indicated that it was a spindle poison that inhibited the cell proliferation at the
`GZ-M phase in the cell cycle and that it blocked mitosis (7). The turning point occurred
`with the demonstration that paclitaxel had a unique mechanism of action by Horwitz.
`It was established that paclitaxel stabilized microtubules and inhibited depolymeriza-
`tion back to tubulin. This differed from the mechanism of action of other spindle
`poisons, such as vinca-alkaloids, that bind to tubulin and inhibit its polymerization
`(8). On the basis of the in vivo B16 melanoma efficacy and the uniqueness of
`mechanism of action, the NCI initiated a very large effort to collect barks and wood
`to obtain enough material to initiate clinical trials (1—3).
`Formulation was also an issue, and in 1980, it was reported that toxicology studies
`would proceed if a satisfactory formulation was developed (6). These toxicology and
`clinical formulation development studies were completed by 1983. Clinical Phase I trials
`started in 1983. Progress of these trials was hampered by hypersensitivity reactions,
`which led to the premature closure of some Phase 1 studies (9, 10). Since these reactions
`were observed more commonly with infusions of shorter durations, a decision was
`made to pursue clinical trials using a 24-h continuous infusion, with premedication to
`lessen the reactions. The dose-limiting toxicity was neutropenia in seven out of the
`nine Phase I trial initiated (1,9,10).
`The final major step in paclitaxel’s development was the recognition of its activity
`against ovarian cancer with responses in approx 30% of patients, many of them hav-
`ing cisplatin or carboplatin refractory disease. These clinical results were reported in
`1989 (11) (i.e., 6 yr after first clinical entry). Even as evidence of paclitaxel’s activity
`increased, with the report of efficacy in breast and nonsmall—cell lung carcinoma
`(12,13), the clinical development was still prevented by the supply shortage. To address
`this issue, the NCI sought the assistance of the pharmaceutical industry. A coopera-
`tive research and development agreement (CRADA) was awarded to Bristol Myers
`Squibb in 1991 (3,9). In 1992, the company filed a New Drug Application. The Food
`and Drug Administration approved paclitaxel that same year for the treatment of
`patients whose ovarian carcinoma had progressed with other chemotherapy (3,9)
`and, in 1994, for metastatic breast cancer.
`
`2.2. Docetaxel
`
`The Institut de Chimie des Substances Naturelles (ICSN) of the Centre National de
`la Recherche Scientifique and thne-Poulenc were interested by the ongoing work on
`paclitaxel in the US and, in particular, of the newly described mechanism of action of
`paclitaxel (8). The ICSN had expertise in the chemistry and the biochemistry of anti-
`mitotic compounds, especially vinca-alkaloids, and it was using a test to measure the
`
`

`

`
`
`1 78 Part I / Cytotoxic Agents
`
`o ozcnzca,
`
`o OZCHZCCI,
`
`a ozcuzca3
`
`o ozcnzcm,
`
`
`
` 8
`
`2 R=H
`
`Fig. 2. First hemi-synthesis of docetaxel 2 and lO—deacetyl-paclitaxel 8.
`
`inhibition of polymerization or depolymerization of tubulin, based on the Shelanski’s
`method (14). Finally, the ICSN had a large supply of T. baccata, the European
`species of yew tree that is widely dispersed in France and Europe. Therefore, in 1979,
`the ICSN decided to undertake research in this area. In 1980, Rhone-Poulenc decided
`to stop a 20-year period of research on anthracyclines and signed a research agree-
`ment concerning taxoids with the ICSN with three main objectives: to explore the
`chemistry of taxoids, bearing in mind the issue of supply, to build structure—activity
`relationships, and to select new and patentable antitumor compounds in these series.
`From the beginning, P. Potier and colleagues were convinced that semisynthesis
`was the only realistic approach for preparing paclitaxel and analogs in sufficient
`quantities for pharmaceutical research and clinical trials. They decided to explore
`systematically and extensively the different chemical components present in the Euro-
`pean yew tree T. baccata, in particular in the needles, a renewable source of biological
`material. The purification of the components present in the needles was monitored by
`measuring their interactions with tubulin purified from calf brain (14). This purifica-
`tion led to the isolation of an abundant precursor of paclitaxel, lO-deacetyl-baccatin
`III, with a yield of 1 g/kg of fresh needles (15) (Fig. 1). This yield was important
`especially in light of the 150 mg of paclitaxel that could be extracted from 1 kg of
`dried bark (16). It was considered an interesting precursor for hemisynthesis of pacli-
`taxel and other taxoids (17). It was 50- to 100-fold less active than paclitaxel in
`inhibiting microtubules depolymerization (18). The closer precursor, baccatin III,
`was also detected but at much lower concentrations.
`
`2.2.1. FIRST ACCESS TO PACLITAXEL: DISCOVERY OF DOCETAXEL (FIG. 2)
`lO-Deacetylbaccatin III, protected at both the C-7 and 010 positions (compound
`6), was converted into the cinnamoyl ester at the C-13 position (compound 7) with a
`90% yield. The cinnamic double bond was then oxyminated (19), leading to docetaxel
`(compound 2) and to lO-deacetylpaclitaxel (compound 8) after cleavage of the Boc
`
`

`

`Chapter 8 / The Taxoids
`
`179
`
`
`
`0H
`
`(m
`
`Ocom
`
`10-désacélylbaccatine Ill 3 (R: H)
`
`Baccaline Ill 5 (R: COCH3)
`
`
`
`Fig. 3. Convergent synthesis of docetaxel 2 and paclitaxel 1.
`
`and reacylation (20). Paclitaxel was obtained using the same pathway, starting from
`baccatin 111. Similar to all new derivatives of paclitaxel, docetaxel was tested for its
`interaction with tubulin/microtubules and, surprisingly, was found to be twice more
`potent than paclitaxel in inhibiting the cold-induced reaction of microtubule depoly-
`merization. At the end of 1985, a small batch of docetaxel was available, and the first
`demonstration of its in vitro cytotoxicity and in vivo antitumor properties was obtained
`in the Oncology Department of Rhéne-Poulenc, using P388 leukemia. Further in
`vivo evaluation revealed efficacy against L1210, Lewis lung carcinoma and B16 mel-
`anoma (16,21 ).
`However, although very successful, this first semisynthetic approach was not appli-
`cable at an industrial scale owing to the use of toxic and very expensive reagents, such
`as osmium tetraoxide.
`
`2.2.2. CONVERGENT SYNTHESIS OF TAXOIDs (FIG. 3)
`
`The convergent synthesis of paclitaxel and docetaxel was performed by direct
`esterification of lO-deacetyl-baccatin III or baccatin III, with the acids corresponding
`to the lateral chains present in paclitaxel and docetaxel, respectively. The first asy-
`metric synthesis of the C-13 phenylisoserine chain of paclitaxel was done by Denis
`and collaborators (22). The lateral chain of docetaxel was prepared using benzalde-
`hyde and terbutyl chloroacetate (23). Finally, esterification of the acids by baccatin
`III and 10-deacetyl-baccatin III yielded paclitaxel (24) and docetaxel (23), respec—
`tively. Since these first experiments, the yield of the esterification methods and of the
`synthesis of the lateral chain have been improved by different teams (25—28).
`
`

`

`180
`
`Part I / Cytotoxic Agents
`
`In 1989, the compound was obtained in sufficient amount to initiate extensive phar-
`macological and toxicological studies and a suitable formulation for iv evaluation was
`developed. Using the SC 316 melanoma murine model, it was found that iv docetaxel
`was more active than paclitaxel at an equitoxic dose (29).
`The compound was also found to be highly active against a large number of murine
`tumor models, most importantly when treated at an advanced stage (i.e., measurable
`disease), and schedule studies revealed that the compound was schedule-independent
`(29). Finally, a pharmacokinetic/distribution study in tumor-bearing mice showed
`that at optimal dosage, the area under the plasma and tumor concentration vs time
`curves (AUC) were much higher than the AUC required to kill human cancer cell
`lines in vitro. Toxicology studies were performed according to the NCI guidelines.
`Phase I clinical trials were initiated in 1990 in Europe and in the US (30). Five
`different schedules were investigated up front in record time. At the end of the Phase
`I trials, it was shown that neutropenia was the major dose-limiting toxicity, and
`responses were reported in different tumor types. Based on considerations, such as
`dose intensity, toxicity profile, and preclinical data, suggesting absence of schedule
`dependency, the recommended dose and schedule for Phase II studies were 100 mg/m2
`adminisetred as a 1-h infusion every 3 wk, without prophylactic measures. Broad
`Phase II testing was initiated in 1992 throughout Europe, North America, and Japan,
`and a CRADA was signed by the NCI and Rhone-Poulenc Rorer. A broad spectrum
`of efficacy was reported, including breast, nonsmall-cell lung, and ovarian cancers. A
`New Drug Application was filed in 1994, and docetaxel has now been approved in
`more than 30 countries.
`
`3. MECHANISM OF ACTION OF TAXOIDS
`
`3.1. The Cellular Target of Taxoids
`
`Together with actin microfilaments and intermediate filaments, microtubules form
`the cytoskeleton of eukaryotic cells. The microtubules are involved in a variety of
`cell functions, including chromosome movement and the regulation of cell shape and
`motility (31). These activities are modulated through associations with several bio-
`chemical components, such as guanosine triphosphate (GTP), and a wide range of
`proteins, the microtubule-associated proteins (MAPS). When a cell begins to divide,
`interphasic microtubules totally vanish and the mitotic spindle assembles. The
`depolymerization of mitotic spindle microtubules is essential for specific mitotic
`events, such as the movement of the chromosomes to the metaphase plate and their
`correct segregation during anaphase (32). Microtubules are long, hollow cylinders
`assembled from a heterodimeric (oz/B) globular protein called tubulin. They consist of
`13 aligned protofilaments within which the tubulin subunits interact through long-
`itudinal and lateral bonds (33). Not all the tubulin pool assembles into microtubules:
`a steady state is maintained between assembled tubulin and a concentration of free
`tubulin called the critical concentration.
`
`3.2. Taxoids Stabilize Microtubules
`
`The polymerization of tubulin purified from mammalian brain usually enhances
`the turbidity of the solution; thus, the degree of polymerization can be monitored
`simply by measuring turbidity (34). Figure 4 depicts, for example, the effects of doce-
`
`

`

`Chapter 8 / The Taxoids
`
`0.40
`
`181
`
`0.30 "
`
`0.20 “
`
`
`0.10
`
`CD.at350nm
`
`0.00
`
`.__-__L_—__L—_—_——L———J
`0
`1o
`20
`30
`40
`
`TIME (mln)
`
`Fig. 4. Effect of docetaxel on polymerization of tubulin and depolymerization of microtubules.
`Tubulin was polymerized by heating from 3 to 37 °C. Depolymerization of microtubules was obtained
`by cooling from 37 to 3 °C (arrow). Polymerization or depolymerization was monitored by following
`the turbidimetry at 350 nm. (A) 10 W porcine brain tubuline. (B) 10 “M tubulin and 3 uM docetaxel.
`
`taxel on the kinetics of tubulin assembly and disassembly. The lag time correspond-
`ing to the activation and nucleation of tubulin is notably reduced, and the rate of
`polymerization is increased (35). Finally, microtubules, stabilized by taxoids, do not
`depolymerize on cold treatment. In fact, paclitaxel and docetaxel analogs are usually
`evaluated on the basis of the drug concentration that inhibits half of the cold-induced
`depolymerization. Docetaxel is about twice as efficient as paclitaxel in this respect
`(36,3 7). The thermodynamic parameters of tubulin assembly are also modified by the
`taxoids, and the critical concentration is significantly reduced in the presence of pac-
`litaxel (8). Docetaxel is twice as efficient as paclitaxel in decreasing the critical con-
`centration of tubulin assembly (38).
`It should be pointed out that the mechanism of action of the taxoids is unique,
`since all other known mitotic spindle poisons, in particular, the vinca-alkaloids, shift
`the tubulin-microtubule equilibrium toward tubulin (39) (Fig. 5).
`
`3.3. Characterization of the Interaction Site
`
`Tritiated paclitaxel cosediments with microtubules and dissociates rapidly on the
`addition of paclitaxel. Thus, a rapid and reversible equilibrium exists between pac-
`litaxel and the microtubules. There is only one high-affinity binding site of paclitaxel
`per a/p tubulin subunit, indicating that the interaction between microtubules and
`paclitaxel is specific. The equilibrium dissociation constant was originally found to be
`870 nM (40). Docetaxel competes with paclitaxel for binding to the microtubules, but
`its equilibrium dissociation constant is two times less, i.e., it has better affinity (38).
`This difference could account for the higher efficiency of docetaxel to promote tubulin
`polymerization, to stabilize microtubules against cold-induced disassembly, and to
`
`

`

`
`182
`Part I / Cytotoxic Agents
`
`MICROTUBULES
`
`VI NCA
`ALCA LOI DS
`
`POLYMEHIZATION
`
`DEPOLYMERIZATION
`
`TAX O. D S
`
`TUBULIN
`
`Fig. 5. Mechanism of action of vinca alkaloids and taxoids.
`
`decrease the critical concentration of tubulin assembly. To acquire further insight into
`the taxoid—microtubule interaction at the molecular level, it is essential to locate the
`taxoid binding site. So far, it is known that the binding of taxoids is linked to the
`polymerization process. Furthermore, no binding of taxoids to dimeric tubulin has
`been detected, indicating that the site is located on assembled tubulin (38). This site
`does not overlap those of other known ligands, such as colchicine, podophyllotoxin,
`vinblastine, or GTP (41,42).
`
`3.4. Models of the Mechanism of Action
`
`Paclitaxel-bound microtubules, the structure of which has been resolved at 3 nm
`employing X-ray scattering, appear to be constituted of 12 protofilaments instead of
`the 13 protofilaments usually observed (43,44). The solution structure of microtu-
`bules induced by docetaxel has been also characterized using the same technique (45).
`The substructures of the microtubule walls are identical in paclitaxel- and docetaxel-
`induced microtubules; however, the population of docetaxel microtubules has an
`average of 13 protofilaments like the control microtubules. It is proposed that the
`chemical substitutions present in docetaxel side chain in its binding site increases
`slightly the contact angle between adjacent protofilaments. The simple working
`hypothesis is that taxoids bind between adjacent tubulin molecules, and such a hypoth-
`esis is fully compatible with the observed thermodynamic behavior of the taxoid-
`induced microtubule assembly system (43).
`
`4. ANTITUMOR PROPERTIES
`
`4.1. In Vitro Activities
`
`4. 1 . 1 . CELLULAR CYTOTOXICITY
`
`Both taxoids have been found extremely potent against a wide variety of murine
`and human cancer cell lines. Using the COMPARE computer program, it was con-
`
`

`

`Chapter 8/ The Taxoids
`
`183
`
`eluded that docetaxel response profile on 50 human tumor cell lines in the new NCI
`screening panel, correlated with the data pattern of test agents acting on the tubulin/
`microtubule system, the closest compound being paclitaxel (NCI, unpublished results).
`Several in vitro studies have been done comparing their activities under various exper-
`imental conditions (liquid medium, semisolid medium, short- and long-term expo-
`sures). The cytotoxicity of paclitaxel and docetaxel at submicromolar concentrations
`was compared in several murine, P388, SVras, and human tumor cell lines, breast
`Calc18, colon HCT 116, bladder T24, and nasopharyngeal KB (46). Docetaxel was
`found to be 1.3- to 12-fold more potent than paclitaxel. The cytotoxic properties of
`paclitaxel and docetaxel were also compared against nine cell lines established from
`human ovarian tumors and having intrinsic or acquired resistance to cisplatin. These
`cell lines were not crossresistant to the taxoids, and docetaxel was found to be active
`at a twofold lower concentrations than paclitaxel (47).
`In addition, the activities of docetaxel and paclitaxel, in a human tumor cloning
`stem cell assay (starting from fresh human tumor biopsies), were compared at con-
`centrations of taxoids similar to the plasma levels obtained after treatment of patients
`(48,49). Melanoma, breast, lung, ovarian, and colon tumors cells were significantly
`inhibited, regardless of the schedule of incubation (1-h exposure or continuous expo-
`sure for 14—28 (1). Interestingly, 29 samples were found to be more sensitive to doce-
`taxel, whereas 13 were more sensitive to paclitaxel, suggesting partial crossresistance
`between these two drugs (49). Finally, the in vitro cellular effects of docetaxel and
`paclitaxel have been recently assessed against a wide range of human normal and
`tumor samples, including tumor cell lines, primary cultures from tumor biopsies and
`normal bone marrow samples (50). ICSO (50°70 inhibitory concentrations) values of the
`two taxoids were in the nanomolar range and docetaxel appeared to be two- to four-
`fold more cytotoxic than paclitaxel (50).
`
`4.1.2. MECHANISM OF CYTOTOXICITY AND CELLULAR EFFECTS
`
`Uptake and efflux studies were performed on P388 leukemia cells in vitro with
`radiolabeled docetaxel and paclitaxel. Uptake experiments revealed that a threefold
`higher intracellular concentration of docetaxel was obtained as compared to pacli-
`taxel, for the same initial extracellular concentration (0.1 M4) (51). Efflux studies
`revealed that the half-time of efflux of docetaxel from P388 cells was at least three
`
`times slower than that of paclitaxel (150 vs 45 min, respectively).
`Thus, the higher potency of docetaxel observed in vitro may be explained by the
`combination of its higher affinity for microtubules, its higher achievable intracellular
`concentration, and the slower cellular efflux.
`Cell-cycle studies revealed that paclitaxel was mainly cytotoxic during mitosis (M
`phase), as demonstrated by experiments on CHO and A 2780 ovarian tumor cell lines
`(52). Inhibition of cytokinesis has been observed, but some cells can progress through
`new cell cycles, leading to the formation of polyploid cells (53,54). Using synchronized
`HCT116 cells,
`it was demonstrated that paclitaxel inhibits formation of mitotic
`spindles in cells without affecting function of preformed spindles and without arrest-
`ing cells in mitosis (54). Docetaxel has been found to be more active on proliferating
`than on nonproliferating KB cells (46) and to inhibit mitosis in several cell lines, such
`as J82 and KB (55). Surprisingly, using synchronized HeLa cells, it has been shown
`that docetaxel exerts cell killing specifically during the S phase of the cell cycle; no
`
`

`

`184
`
`Part I / Cytotoxic Agents
`
`In Vivo Antitumor Activity of Paclitaxel Against Human Tumor Xenografts
`
`Table 1
`
`sc Human tumor
`
`A 2780 ovarian
`LX-l lung
`H 2981 lung
`L 2987 lung
`RCA colon
`HGT-116 colon
`A431 vulva
`
`Highest nontoxic
`iv dosage
`mg/kg/dose
`
`'
`
`18
`24
`24
`36
`36
`36
`36
`
`Schedule
`days
`
`7, 9, 11, 13, 15
`5, 7, 9,11,13
`5, 7, 9,11,13
`1
`, 16, 18, 20, 22
`4, 6, 8, 10, 12
`3, 5, 7, 9, 11
`3, 5, 7, 9, 11
`
`Activity“
`rating
`
`+ + +
`+ + + +
`+ +
`+ + +
`> + +
`> + + +
`+ + +
`
`‘1 Activity rating: + + + + = highly active (log cell kill > 2.8), + + + = highly active (log cell
`kill = 2.0 to 2.8), + + = active (log cell kill = 1.3—1.9; T/C 2 150% for L1210, + = active (log
`cell kill = 0.7-1.2 for s.c. tumors, T/C = 125—174% for P388), — = inactive.
`
`cytotoxicity was observed during mitosis, a different situation from what is observed
`with paclitaxel (56).
`Finally, it was found that paclitaxel greatly increases the pool of polymerized tubu-
`lin in cells, and new short microtubules free in the cytoplasm were observed (57). In
`addition, at high concentration, it induced the formation of microtubules bundles (58).
`Using J82 human bladder and KB 3-1 human carcinoma cells, it was shown that pac-
`litaxel and docetaxel lead to the formation of bundles and asters in a dose- and time-
`
`dependent manner (55). Asters were observed in mitotic cells, and bundles were seen
`in interphase cells. The effects of docetaxel as compared to paclitaxel appeared at a
`twofold lower concentration (55 ).
`
`Paclitaxel and even more docetaxel have been studied in many murine tumor
`models and human tumor xenografts.
`
`4.2. In Vivo Activity
`
`4.2. 1. PACLITAXEL
`
`The development of an adequate formulation for paclitaxel led to a re-evaluation
`of its in vivo antitumor efficacy, using better experimental conditions, i.e., avoiding
`the previous ip/ip evaluation and administering the drug iv at a site different from the
`tumor site. The formulation used was 10% Cremophor®, 10% ethanol, 80% NaCl
`0.9%. These studies have been recently reviewed, and most of them were performed
`after initial clinical trials (59,60). Indeed, these studies demonstrated that paclitaxel
`delivered iv was active against several tumors implanted in distal sites and treated at an
`early stage: sc Madison 109 murine lung carcinoma and A 431 vulva, A 2780 ovarian,
`H 2981 and LX-l lung, and RCA and HGT-116 colon human tumor xenografts
`implanted under the renal capsule of nude mice (Table 1) (60). When administered
`sc five times weekly for three consecutive weeks, paclitaxel caused the complete regres-
`sion of a human breast tumor xenograft, and significantly delayed the growth of
`endometrial, ovarian, brain, tongue, and lung human tumor xenografts (61). Pacli-
`taxel was also evaluated against ovarian carcinoma xenografts HOC8, HOC18, and
`HOC22, and was found to have similar efficacy to docetaxel (62).
`
`

`

`Chapter 8 / The Taxoids
`
`185
`
`In Vivo Antitumor Activity of Docetaxel Against Human Tumor Xenografts
`
`Table 2
`
`sc Human tumor
`
`Calc 18 mammary
`MX-l mammary
`LX-l lung
`SKMEL-Z melanoma
`CX-l ovarian
`KM20L2 ovarian
`OVCAR-3
`
`Highest nontoxic
`iv dosage
`mg/kg/dose
`
`32.2
`22
`22
`33
`15
`33
`33
`
`Schedule
`days
`
`11, 15, 19
`11, 15, 19
`9, 13, 17
`27, 31, 35
`12,16, 20
`14,18,22
`3, 7, 11
`
`Activitya
`rating
`
`+ + +
`+ + + +
`+ +
`+ + + +
`+ + +
`+ +
`+ + + +
`
`“For activity rating, see Table 1.
`
`Further schedule-dependency studies were performed and showed that daily injec-
`tion for 7 d was the best schedule as opposed to longer spaced administration (60).
`
`4.2.2. DOCETAXEL
`
`The experimental antitumor activity of docetaxel has been evaluated against a
`panel of 30 tumors of mice and human tumors xenografted in nude mice, represent-
`ing a variety of tissue types and chemosensitivity patterns. The tumors were grafted in
`distal sites, and several tumors were treated at advanced and metastatic stages. Dose
`response was evaluated in all trials to determine accurately the maximum tolerated
`dose. The formulation used was a 1:1 ethanol polysorbate 80 solution, administered
`after a 1:10 dilution in glucose 5% in water (29). Docetaxel had a broad spectrum of
`antitumor activity, since 28/30 models responded to this agent (29, 63—65 ) (Tables 2
`and 3). The experimental antitumor activities of paclitaxel and docetaxel were com-
`pared by testing these drugs against B16 melanoma, a tumor sensitive to taxoids using
`an intermittent schedule, every 2 d x 3. Antitumor activity was expressed by the
`tumor growth delay (T—C) and by the log cell kill (LCK) of tumor cells obtained at the
`maximal tolerated dose (MTD) of each drug. According to these criteria, docetaxel
`was approximately two times more active and potent than paclitaxel (docetaxel:
`T-C = 12.2 d, LCK = 2.9, MTD = 11.3 mg/kg/d; paclitaxel: T-C = 4.7 d, LCK =
`1.1, MTD = 21.7 mg/kg/d) (29) (Fig. 6). Among the murine models tested, good
`activity was observed, with in some cases cures of early stage tumors. However, the
`most meaningful data were those obtained against advanced stage tumors (i.e.,
`tumors at least 200 mg at start of therapy) where complete tumor regressions could be
`observed. This occurred with the murine mammary adenocarcinomas 16/C and
`13/C, pancreatic ductal carinoma O3, colon 38 adenocarcinoma, and the human
`xenografts MX—l mammary and SK-MEL-Z melanoma (63,64). Prolonged tumor
`growth delays were also observed with Calc-18 breast, LX-l lung, CX-l colon, head
`and neck HNX-14C, and HNX-22B xenografts (64,66). Since the clinical activity of
`paclitaxel against ovarian tumors was impressive, five human ovarian xenografts hav-
`ing different sensitivities to the reference drug cisplatin were included in this study.
`Docetaxel was active against the three tumors sensitive to cisplatin; interestingly, it
`was also active against OV-Pe, which is resistant to cisplatin (65).
`
`

`

`186
`
`Part I / Cytotoxic Agents
`
`In Vivo Antitumor Activity of Docetaxel Against Murine Tumors
`
`Table 3
`
`Tumor
`
`Solid tumors sc
`
`Melanoma B16 early
`Pancreas
`
`P02
`P03 early
`P03 advanced
`
`Mammary
`MA16/C early
`MA16/C advanced
`MA13/C early
`MA13/C advanced
`MA44 early
`Colon
`
`C26 early
`C38 early
`C38 advanced
`C51 early
`C51 advanced
`
`Lewis lung early
`
`Osteosarcoma GOS early
`
`Hystiocytosarcoma M5076 early
`
`Leukemias ip
`P388 106 cells
`L1210 10‘ cells
`
`a For activity rating, see Table 1.
`b CR = complete regressions.
`
`Highest nontoxic
`iv dosage
`mg/kg/dose
`
`Schedule
`days
`
`Activity”
`rating
`
`24
`
`32.2
`20.5
`18.3
`
`15
`10.8
`14.2
`15
`22
`
`5
`23.5
`26.8
`12.7
`15.2
`
`23.2
`
`18.6
`
`8.6
`
`23.2
`21.7
`
`3, 5, 7, 9
`
`+ + + +
`
`3, 5, 7
`3, 5, 7, 9
`22, 24, 26, 28
`
`i
`+ + + +
`5/6 CR
`
`3, 5, 7
`7, 9, 11
`3, 5, 7
`24, 27, 30
`3, 5, 7
`
`1—4
`3, 5, 7
`14, 16, 18
`3, 5, 7
`10, 12, 14
`
`3—7
`
`3—7
`
`3—7
`
`1-4
`1—4
`
`+ + +
`5/5 CR
`+ + + +
`3/5 CR”
`i
`
`+
`+ + + +
`5/5 CR
`+ + +
`+ +
`
`+
`
`+
`
`—
`
`+
`+ +
`
`Scheduling studies were performed against advanced colon 38 adenocarcinoma.
`Docetaxel was tested using three different schedules comparing the effect of 2, 3, and
`10 administrations over the same duration of treatment. Overall, the administration
`schedule did not influence markedly the total dosage that can be administered and,
`thus, the compound was considered schedule-independent for the MTD (29).
`
`4.3. Combination Chemotherapy
`
`Since taxoids have clinical activity in ovarian, breast, and lung tumors, most of the
`experimental studies have been done with drugs active in these diseases: doxorubicin,
`5-fluorouracil, cyclophosphamide, cisplatin, and etoposide.
`
`4.3. 1. PACLITAXEL
`
`Both in vitro and in vivo studies were performed. The efficacy of combination
`therapy consisting of paclitaxel plus a topoisomerase II inhibitor, doxorubicin or
`
`

`

`
`
`Chapter 8 / The Taxoids 187
`10000
`
`CONTROL
`
`/
`PAQLITAXEL
`
`DOCETAXEL
`
`1000
`
`100
`
`
`
`
`
`TUMORWEIGHT(mg)
`
`:
`— — — L.
`,' 12 2 dayss‘
`Limit of palpation
`
`21.7
`
`L.C.K.
`
`COMPOUND
`
`DOCETAXEL
`PACLITAXEL
`
`M.T.D.
`mglkg/day
`13.4
`
`0
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`DAYS POST TUMOR IMPLANTATION
`
`Fig. 6. Comparative in vivo activities of paclitaxel and docetaxel against 316 melanoma. Detailed
`experimental conditions were described in ref. (29). Briefly, BGDZFl mice (7 mice/group) were
`grafted sc on day 0 with 30 mg 816 tumors fragments. Drugs were injected iv on days 4, 6, 8, and 10
`at the MTD (21.7 mg/kg/d for paclitaxel; 13.4 mg/kg/d for docetaxel). Tumor growths were mea-
`sured biweekly. Activity is expressed by the TC (where T and C are the median time in days neces-
`sary for the tumors of the treated group '1‘ and the control group C to reach a size of 1 g). Activity is
`also expressed by the LCK, which quantifies the number of tumor cells killed by the chemotherapy.
`
`etoposide, against various cell lines has been studied in vitro; better results were ob-
`tained when cells were first incubated with paclitaxel (67). Cisplatin—paclitaxel com-
`bination was evaluated using L1210 leukemic cells: maximal effects were observed
`when cells were incubated for 24 h with paclitaxel, and then treated for 30 min with
`cisplatin (68). Combinations of taxoids and tubulin-interactive agents are of interest
`because of their complementary mechanism of action. Paclitaxel—estramustine was
`found to give supra-additive cytotoxic effects on several lines of human prostatic
`adenocarcinoma. No additive properties were noted when taxoids were combined
`with vinblastine (69).
`In vivo combination chemotherapy studies have also been performed with pac-
`litaxel using the M109 tumor model (60). The combined agents included cisplatin,
`etoposide, doxorubicin, cyclophosphamide, methotrexate, pentamethylmelamine,
`and bleomycin. Taxol-cisplatin and Taxol-bleomycin were the two

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