throbber
European Journal of Cancer 37 (2001) 1590–1598
`
`www.ejconline.com
`
`Cremophor EL: the drawbacks and advantages of
`vehicle selection for drug formulation
`
`H. Gelderblom*, J. Verweij, K. Nooter, A. Sparreboom
`
`Department of Medical Oncology, Rotterdam Cancer Institute (Daniel den Hoed Kliniek) and University Hospital Rotterdam,
`3075 EA Rotterdam, The Netherlands
`
`Received 26 February 2001; received in revised form 26 March 2001; accepted 3 May 2001
`
`Abstract
`
`Cremophor EL (CrEL) is a formulation vehicle used for various poorly-water soluble drugs, including the anticancer agent
`paclitaxel (Taxol). In contrast to earlier reports, CrEL is not an inert vehicle, but exerts a range of biological effects, some of which
`have important clinical implications. Its use has been associated with severe anaphylactoid hypersensitivity reactions, hyperlipi-
`daemia, abnormal lipoprotein patterns, aggregation of erythrocytes and peripheral neuropathy. The pharmacokinetic behaviour of
`CrEL is dose-independent, although its clearance is highly influenced by duration of the infusion. This is particularly important
`since CrEL can affect the disposition of various drugs by changing the unbound drug concentration through micellar encapsulation.
`In addition, it has been shown that CrEL, as an integral component of paclitaxel chemotherapy, modifies the toxicity profile of
`certain anticancer agents given concomitantly, by mechanisms other than kinetic interference. A clear understanding of the biolo-
`gical and pharmacological role of CrEL is essential to help oncologists avoid side-effects associated with the use of paclitaxel or
`other agents using this vehicle. With the present development of various new anticancer agents, it is recommended that alternative
`formulation approaches should be pursued to allow a better control of the toxicity of the treatment and the pharmacological
`interactions related to the use of CrEL. # 2001 Elsevier Science Ltd. All rights reserved.
`
`Keywords: Cremophor EL; Formulation vehicles; Paclitaxel; Pharmacokinetics; Pharmacodynamics
`
`1. Introduction
`
`The choice of a suitable pharmaceutical formulation
`is an essential step in anticancer drug development. This
`development starts with the acquisition of a chemical
`entity from either natural sources or entirely synthetic
`routes. Subsequently, the compound is screened for
`cytotoxic activity in vitro and in vivo. Once the screening
`process has been completed the compound should be
`properly pharmaceutically-formulated and produced
`before entering animal toxicology and pharmacokinetic
`studies and subsequently human phase I, II and III
`studies [1,2]. In our opinion, pharmaceutical formula-
`tion is a seriously underrated aspect of anticancer drug
`development.
`With only a few exceptions, most new anticancer
`compounds are initially developed for intravenous (i.v.)
`
`* Corresponding author. Tel.: +31-10-439-1754; fax: +31-10-439-
`1003.
`E-mail address: gelderblom@onch.azr.nl (H. Gelderblom).
`
`use, despite some drawbacks such as the morbidity
`associated with gaining i.v. access, risk of i.v. catheter-
`related infection, thrombosis and extravasation, and
`patients’ preference for oral
`therapy when equally
`effective [3]. Important reasons for choosing i.v. use for
`initial drug development are the fact that usually less
`gastrointestinal
`toxicity occurs,
`there is
`immediate
`100% bioavailability and instantaneous pharmacody-
`namic effects and there is a possibility to modify the
`dosing rate or even halt the infusion if necessary. Solu-
`bility of the compound is a specific demand for i.v.
`administration, even for the newer chemotherapeutic
`agents which are known to be poorly water-soluble.
`Classical solubility approaches, which will be discussed
`subsequently, include the use of colloidal systems, pro-
`drug development or solubilisation techniques.
`Colloidal systems such as liposomes, microcapsules,
`microspheres, nanoparticles or macromolecule com-
`plexes may protect the anticancer drug from premature
`degradation or (chemical) inactivation within the sys-
`temic circulation. Prodrugs are inactive derivatives that
`
`0959-8049/01/$ - see front matter # 2001 Elsevier Science Ltd. All rights reserved.
`P I I : S 0 9 5 9 - 8 0 4 9 ( 0 1 ) 0 0 1 7 1 - X
`
`Abraxis EX2090
`Cipla Ltd. v. Abraxis Bioscience, LLC
`IPR2018-00162; IPR2018-00163; IPR2018-00164
`
`

`

`H. Gelderblom et al. / European Journal of Cancer 37 (2001) 1590–1598
`
`1591
`
`release the active drug following spontaneous degrada-
`tion or enzymatic reactions. Solubilisation is the process
`of uptake of drugs through complex formation into for,
`e.g. oligomers of dextrose and fatty acids, through co-
`solvent systems (such as ethanol, polyethyleneglycol and
`glycerol), or through surfactant systems. The surfactant
`systems consist of either amphoteric compounds (e.g.
`lecithin or gelatin), ionic surfactants (e.g. sodium pal-
`mitate) or non-ionic surfactants (e.g. Tween 80 and
`Cremophor EL (CrEL)) [4,5]. This review focuses on
`biological and pharmacological properties of CrEL,
`which is the formulation vehicle of various hydrophobic
`drugs, including the anticancer agent paclitaxel.
`
`2. Paclitaxel formulation
`
`After the identification of paclitaxel as the active
`ingredient in crude ethanolic extracts of the bark of the
`Western Yew tree, Taxus brevifolia, against several
`murine tumours [6], the development of the drug was
`suspended for more than a decade due to problems
`associated with the solubilisation of the drug. Paclitaxel
`is insoluble in water (less than 0.03 mg/ml), slightly
`soluble in octanol, propylene glycol and butanol, solu-
`ble in CrEL, ethanol, methanol, chloroform, acetone
`and ether, and freely soluble in dimethyl acetamide. The
`formulation approach using 50% CrEL and 50% dehy-
`drated ethanol United States Pharmacopeia (USP) was
`chosen for further development [7]. The pharmaceutical
`formulation of paclitaxel
`(paclitaxel; Bristol-Myers
`Squibb) contains 30 mg paclitaxel dissolved in 5 ml of
`this (1:1, v/v) mixture.
`
`The heterogeneous non-ionic surfactant CrEL is a
`white to off-white viscous liquid with an approximate
`molecular weight of  3 kDa and a specific gravity
`(25C/25C) of 1.05–1.06, and it is produced by the
`reaction of castor oil with ethylene oxide at a molar
`ratio of 1:35 [4]. Castor oil is a colourless or pale yellow
`fixed oil obtained from the seeds of Ricinus communis,
`with an extremely high viscosity, and consists mainly of
`the glycerides of ricinoleic,
`isoricinoleic, stearic and
`dihydroxystearic acids. CrEL is usually of highly vari-
`able composition, with the major component identified
`as oxylated triglycerides of ricinoleic acid (i.e. poly-
`oxyethylene glycerol triricinoleate 35) (Fig. 1). Polyvinyl
`chloride (PVC)-free equipment for CrEL administration
`is obligatory, since CrEL is known to leach plasticizers
`from PVC infusion bags and polyethylene-lined tubing
`sets which can cause severe hepatic toxicity [5].
`CrEL is being used as a vehicle for the solubilisation
`of a wide variety of hydrophobic drugs,
`including
`anaesthetics, photosensitisers, sedatives,
`immunosup-
`pressive agents and (experimental) anticancer drugs
`(Table 1). The amount of CrEL administered with these
`drugs averages 5 ml (range, 1.5–10.3 ml), although
`paclitaxel is an exception as the amount of CrEL is
`much higher per administration, approximately 26 ml.
`Therefore, it is important to understand the biological
`and pharmacological behaviour of CrEL, especially in
`the formulation of paclitaxel.
`
`3. Biological effects of Cremophor EL (CrEL)
`
`3.1. Anaphylactic hypersensitivity reactions
`
`The most well known biological effect of paclitaxel
`formulated with CrEL is a clinical acute hypersensitivity
`reaction, characterised by dyspnoea, flushing, rash,
`chest pain, tachycardia, hypotension, angio-oedema,
`and generalised urticaria. Despite premedication, con-
`sisting of high-dose corticosteroids, H1 and H2 antago-
`nists, minor reactions (flushing and rash) still occur in
`
`Table 1
`Examples of surfactant systems using CrEL
`
`Agent
`
`Therapeutic class
`
`Aplidine
`C8KC
`Clanfenur
`Cyclosporin A
`Diazepam
`Didemnin B
`Paclitaxel
`Propofol
`Teniposide
`
`Antineoplastics
`Photosensitisers
`Antineoplastics
`Immunosuppressives
`Sedatives
`Antineoplastics
`Antineoplastics
`Anaesthetics
`Antineoplastics
`
`Amount administereda (ml)
` 1.5
`5.5
`10.3
`3.5
`1.5
`2.0
`25.8
` 7.0
`1.5
`
`structures of paclitaxel
`Fig. 1. Chemical
`oxyethyleneglycerol triricinoleate 35) (b).
`
`(a) of CrEL (poly-
`
`CrEL, Cremophor EL.
`a For an average patient for a single administration of dose.
`
`Abraxis EX2090
`Cipla Ltd. v. Abraxis Bioscience, LLC
`IPR2018-00162; IPR2018-00163; IPR2018-00164
`
`

`

`1592
`
`H. Gelderblom et al. / European Journal of Cancer 37 (2001) 1590–1598
`
`41–44% of all patients and major, potentially life-
`threatening, reactions in 1.5–3% [8–10]. Mostly, the
`hypersensitivity reaction occurs within the first two
`courses of paclitaxel and can be prevented by reducing
`the infusion rate. Various (pre)clinical observations,
`discussed below, point to CrEL as the main contributor
`to the hypersensitivity reactions:
`
`and a change in shape of leucocytes in blood smears
`[24]. This suggests that manual blood-cell count analysis
`is warranted after the administration of preparations
`containing CrEL. Whether the observed hyperlipidae-
`mia after CrEL administration increases the risk of
`vascular accidents is, as yet, unknown.
`
`(a) Using an elegant series of in vitro studies, com-
`plement activation by CrEL was found to cause
`the hypersensitivity reaction associated with
`paclitaxel chemotherapy [11], as well as reactions
`seen to other drugs where CrEL was used as a
`vehicle [12,13]. Recently, it was shown that the
`CrEL-induced complement activation in human
`serum was clearly concentration-dependent with
`a minimum activating CrEL level in the order of
`2 ml/ml, a concentration readily achieved clini-
`cally in the plasma following standard doses of
`paclitaxel [14].
`(b) Histamine release in dogs by CrEL was mainly
`caused by one of its (minor) constituents, oleic
`acid [15], whereas the cardiac toxicity attributed
`to paclitaxel, mainly asymptomatic rhythm dis-
`turbances, might also be caused by CrEL
`through a mechanism of histamine release [16].
`(c) Improper mixing of high-dose cyclosporin A
`infusions caused (non-solubilised) CrEL to sink
`to the bottom of vials, producing anaphylactoid
`responses because of highly concentrated CrEL
`at the initial i.v. bolus [17]. All patients allergic to
`i.v. cyclosporin A, tolerated the CrEL-free oral
`formulation [18].
`(d) Finally, the fact that CrEL concentrations are
`lower with prolonged paclitaxel infusion schemes
`(see below) may be an explanation for the lower
`incidence of hypersensitivity reactions with these
`schemes. Collectively, these findings indicate that
`CrEL plays a crucial role in the occurrence of
`hypersensitivity reactions of paclitaxel and other
`drugs using CrEL as a formulation vehicle.
`
`3.2. Lipoprotein patterns and hyperlipidaemia
`
`Lipoprotein alterations accompanying the adminis-
`tration of miconazole formulated with CrEL were
`reported as early as 1977 by Bagnarello and colleagues
`[19]. Later, CrEL was found to alter the buoyant density
`of high-density lipoproteins (HDL) [20] and shift the
`electrophoretic and density gradient HDL to low den-
`sity lipoproteins (LDLs) [21–23]. These authors showed
`that paclitaxel had a strong affinity for the serum lipo-
`protein dissociation products, potentially affecting the
`biodistribution and clearance of the drug. High con-
`centrations of CrEL may also cause hyperlipidaemia,
`possibly resulting in Roulaux formation of erythrocytes
`
`3.3. Neurotoxicity
`
`Axonal degeneration and demyelination, one of the
`principal side-effects of paclitaxel resulting in peripheral
`neuropathy, is also supposedly a biological effect caused
`by CrEL. The plasma levels of CrEL achieved after
`therapeutic doses of paclitaxel and i.v. cyclosporin A
`(also formulated in CrEL), have been shown to produce
`axonal swelling, vesicular degeneration and demyelina-
`tion in rat dorsal ganglion neurones [25]. Interestingly,
`in rats treated with CrEL-free [3H]-paclitaxel, paclitaxel
`was not detectable in the peripheral nervous system,
`indicating that the anticancer drug itself might not be
`responsible for the observed toxicity [26]. The hypoth-
`esis of CrEL-induced neurotoxicity is further supported
`by the fact that i.v. cyclosporin A causes neurotoxicity
`in approximately 25% of patients [27], while this side-
`effect is rarely seen with oral administration. This is also
`consistent with a previous finding that CrEL is not
`absorbed when given orally as a result of intestinal
`degradation [28]. It is also noteworthy that neurological
`symptoms are 10 times less common after treatment
`with docetaxel, a semi-synthetic taxane chemically simi-
`lar to paclitaxel, than with paclitaxel [29]. This could be
`because unlike paclitaxel, docetaxel
`is formulated in
`Tween 80 (i.e. polyoxyethylenesorbitan monooletate),
`again rendering CrEL as the likely cause of the clinical
`observations of neuropathy. The neurotoxic properties
`of CrEL are most likely induced by residual unsaturated
`fatty acids, possibly due to the appearance of peroxida-
`tion products [30]. Therefore, it is suggested that the
`ethoxylated derivatives of castor oil account for most of
`the neuronal damage observed [31].
`
`3.4. Reversal of P-glycoprotein activity
`
`In the early 1990s, several groups independently
`observed that CrEL was able in vitro to modulate the
`activity of P-glycoprotein, a drug-transporting mem-
`brane protein that is elevated in tumour cells having a
`multidrug resistance phenotype [32–35]. More recently,
`similar phenomena have also been described for various
`other non-ionic surfactants, including Tween 80 [36],
`Solutol HS 15 [37] and Triton X-100 [38]. Surprisingly,
`however, multidrug resistance has never been success-
`fully modified in vivo by any non-ionic surfactant,
`including CrEL [39–41]. A possible explanation for this
`lack of in vivo efficacy is the extremely low volume of
`distribution of CrEL, approximately equal
`to the
`
`Abraxis EX2090
`Cipla Ltd. v. Abraxis Bioscience, LLC
`IPR2018-00162; IPR2018-00163; IPR2018-00164
`
`

`

`H. Gelderblom et al. / European Journal of Cancer 37 (2001) 1590–1598
`
`1593
`
`volume of the blood compartment, suggesting that con-
`centrations necessary to affect reversal of multidrug
`resistance in vitro are probably not attained in vivo in
`(solid) tumours [42]. Recent pharmacokinetic experi-
`ments conducted in mdr1a P-glycoprotein knockout
`mice support this lack of efficacy, despite high peak
`plasma levels of CrEL [28]. In contrast to treatment in
`solid tumours, the pharmacokinetic selectivity of CrEL
`for the central blood/bone marrow compartment can be
`an advantage in the treatment of haematological malig-
`nancies, in which the expression of P-glycoprotein is
`known to be a principal factor contributing to resistance
`to chemotherapy [43].
`
`3.5. In vitro cytotoxicity
`
`Cytotoxic properties of CrEL in doxorubicin-resistant
`human breast-cancer cell lines were first reported by
`Fja¨ llskog and colleagues [44], and confirmed in various
`human tumour samples [45,46]. It was postulated that
`formation of free radicals by peroxidation of poly-
`unsaturated fatty acids and/or a direct perturbing effect
`in the cell membrane causing fluidity and leakage are
`possible mechanisms contributing to this type of cyto-
`toxicity [47–49]. Using clonogenic assays, however, it
`has been demonstrated that CrEL can antagonise the
`cytotoxicity of paclitaxel by blocking the cell-cycle that
`results in the inhibition of cytokinesis [50]. Thus,
`although CrEL in itself might have some potential to
`affect cell survival, the concentrations required to mod-
`ulate cell growth will also change paclitaxel-mediated
`(and overall) cytotoxicity. In addition, the pharmacoki-
`netic selectivity of CrEL most likely precludes any
`vehicle-mediated change in (solid) tumour cell kill in
`vivo.
`
`4. Pharmacokinetics of CrEL
`
`4.1. Analytical methods
`
`In view of the contribution of CrEL to clinically
`observed effects, and to enable further assessment of the
`impact of its use on paclitaxel pharmacology, the kinetic
`behaviour of CrEL has been studied extensively in
`recent years. For this purpose, a variety of analytical
`methods have been developed. The first assay developed
`for the measurement of CrEL concentrations in plasma
`was based on the ability of CrEL to modulate daunor-
`ubicin efflux in multidrug resistant T-cell
`leukaemia
`VLB100 cells [51]. Later, a more sensitive and reliable
`method was developed, which required only micro-
`volumes (20 ml) of plasma [52]. This method is based on
`the measurement of ricinoleic acid after saponification
`of CrEL followed by precolumn derivatisation and
`reversed-phase high-performance
`liquid chromato-
`
`graphy. Because of the high costs, and time consuming
`nature of both assays a new method, based on a selec-
`tive binding of CrEL to the Coomassie brilliant blue G-
`250 dye in protein-free extracts, was developed [53,54].
`Most recently, an electrochemical detection method was
`developed [55]. Large-scale pharmacokinetic studies
`have only recently been possible with the development
`of these newer methods.
`
`4.2. CrEL Disposition
`
`Clinical pharmacokinetic studies with CrEL following
`3-h paclitaxel
`infusions indicate a dose-independent
`behaviour with a terminal disposition half-life of
`approximately 80 h, with a large range depending on the
`method used for measurement [56,57]. Interestingly,
`with prolongation of the duration of infusion from 1 to
`3 and to 24 h, the CrEL clearance increased from
`approximately 160 to 300 to 400 ml/h/m2, respectively
`[14] (Fig. 2). It thus appears that CrEL shows a linear
`and dose-independent, but schedule-dependent phar-
`macokinetic behaviour, possibly related to the satura-
`tion of serum esterase-mediated metabolic degradation.
`This schedule dependency leads to an increase in sys-
`temic exposure, and thus an increase in the possible
`CrEL-related biological (side-)effects with a shortening
`of the infusion duration. An example of this phenom-
`enon is the higher risk of allergic reactions in the 1-h
`versus 3-h or 24-h infusions of paclitaxel.
`As mentioned earlier, the volume of distribution of
`CrEL is extremely low, implying that the tissue (and
`tumour) delivery of CrEL is probably insignificant. This
`is in line with observations that CrEL levels in normal
`and tumour tissue were not detectable in mice [58].
`Not much is known about the elimination routes of
`CrEL. CrEL may be largely degraded in the blood
`compartment
`by
`serum carboxylesterase-induced
`degradation, similar to that described for Tween 80 [59],
`causing a gradual release of the ricinoleic acid residues
`
`Fig. 2. Schedule-dependency for CrEL clearance (CL) as a function of
`the duration of infusion in cancer patients treated with paclitaxel at
`dose of 135 (CrEL: 11.3 ml/m2), 175 (CrEL: 14.6 ml/m2) and 225
`(CrEL: 18.8 ml/m2) mg/m2 [14].
`
`Abraxis EX2090
`Cipla Ltd. v. Abraxis Bioscience, LLC
`IPR2018-00162; IPR2018-00163; IPR2018-00164
`
`

`

`1594
`
`H. Gelderblom et al. / European Journal of Cancer 37 (2001) 1590–1598
`
`attached to the triglyceride structure. It has been shown
`that hepatobiliary elimination of CrEL is a minor elim-
`ination pathway [60]. In addition, the urinary excretion
`of CrEL accounted for less than 0.1% of the adminis-
`tered dose, in spite of its relatively hydrophilic nature
`[61].
`
`4.3. Effects of CrEL on drug disposition
`
`Various studies have shown that CrEL alters the
`pharmacokinetics of many drugs including cyclosporin
`A, etoposide, doxorubicin, a number of photosensitisers
`(e.g. C8KC) and paclitaxel. Initially, the effect of CrEL
`on the disposition of paclitaxel was studied in mice that
`received the drug by i.v. injection at doses of 2, 10 and
`20 mg/kg in the presence of various amounts of CrEL
`[58] (Fig. 3). The paclitaxel clearance of 2.4 l/h/kg at the
`lowest dose level was reduced to 0.33 and 0.15 l/h/kg at
`the 10 and 20 mg/kg dose levels. It was also shown that
`the area under the concentration time curve (AUC) of
`paclitaxel is higher when it is formulated in CrEL com-
`pared with formulation in Tween 80, suggesting that
`CrEL is responsible for the non-linearity of paclitaxel
`disposition. Despite the fact that much higher plasma
`levels of paclitaxel are reached when given in the CrEL-
`containing formulation, the tissue levels of paclitaxel
`were essentially similar with all tested preparations,
`indicating that the profound influence of CrEL is only
`taking place in the central blood compartment. In sub-
`sequent years, numerous causes of this apparent non-
`linear pharmacokinetic behaviour were proposed. It has
`been suggested that CrEL might interfere with P-glyco-
`protein-mediated biliary secretion,
`thereby reducing
`paclitaxel elimination [62]. In the isolated perfused rat
`liver, CrEL inhibited the hepatic elimination of pacli-
`taxel, primarily preventing the drug from reaching the
`sites of metabolism and excretion [63]. However, recent
`studies indicate that drug-transporting P-glycoproteins
`are not essential per se for normal hepatobiliary secre-
`tion of paclitaxel [28,64] and that, as discussed, the dis-
`
`position of CrEL itself limits the potential to modulate
`P-glycoprotein activity in vivo [42].
`More recently, it has been proposed that the effect of
`CrEL on paclitaxel pharmacokinetics is associated with
`encapsulation of the drug within CrEL micelles, causing
`(concentration-dependent) changes in cellular partition-
`ing and blood:plasma concentration ratios of paclitaxel
`[64] (Fig. 4). It was shown that the affinity of paclitaxel
`was
`(in decreasing order) CrEL>plasma>human
`serum albumin, with CrEL present above the critical
`micellar concentration (i.e.  0.01%). Since this effect
`was also observed in the absence of plasma proteins, it
`could not have been caused by altered protein binding
`or by an increased affinity of paclitaxel for protein dis-
`sociation products that are produced by the action of
`CrEL on native lipoproteins [22]. These findings are
`consistent with the hypothesis that paclitaxel can be
`entrapped within micelles (composed primarily of poly-
`ethyleneglycerol triricinoleate) and that these micelles
`act as the principal carrier of paclitaxel in the systemic
`circulation. The percentage of total paclitaxel trapped in
`micelles increases disproportionately with higher doses
`of CrEL administered.
`The hypothesis that the non-linear pharmacokinetics
`of paclitaxel is related to time-varying CrEL concentra-
`tions was recently confirmed in a group of cancer
`patients all receiving increasing doses of 135, 175 and
`225 mg/m2 [65]. Again, the plasma clearance of pacli-
`taxel turned out to be dose-dependent with the slowest
`clearance at the highest dose level (Fig. 5). In line with
`the in vitro data, the non-linear disposition of paclitaxel
`in the plasma appeared to be an artifact caused by dose-
`related levels of CrEL in blood [65]. Therefore, the non-
`micellar bound or unbound fraction of paclitaxel
`in
`plasma might be a better pharmacokinetic parameter to
`predict toxicity and to guide dosing of paclitaxel, since it
`is generally acknowledged that the unbound fraction of
`a drug is capable of diffusing across biological barriers
`and interacting with essential structures in (tumour) tis-
`sues [66].
`
`Fig. 3. Effect of the formulation vehicle (CrEL, Tween 80 or dime-
`thylacetamide (DMA)) on paclitaxel concentration in female FVB
`mice receiving paclitaxel at a dose of 10 mg/kg [58].
`
`Fig. 4. Effect of the CrEL concentration on the fraction of unbound
`paclitaxel (fu) in human plasma [56].
`
`Abraxis EX2090
`Cipla Ltd. v. Abraxis Bioscience, LLC
`IPR2018-00162; IPR2018-00163; IPR2018-00164
`
`

`

`H. Gelderblom et al. / European Journal of Cancer 37 (2001) 1590–1598
`
`1595
`
`4.4. Effects of CrEL on anticancer drug
`pharmacodynamics
`
`Besides its effect on (anticancer) drug disposition, the
`use of CrEL has also been associated with alterations in
`the pharmacodynamic outcome of a number of drugs
`co-administered with paclitaxel. For example, clinical
`combination chemotherapy studies with paclitaxel and
`cisplatin revealed important sequence-dependent differ-
`ences in toxicity, with less myelotoxicity when the tax-
`ane was given before cisplatin, that could not be linked
`to consistent changes
`in pharmacokinetics
`[67,68].
`Interestingly, no toxicity- or pharmacokinetic-sequence
`interaction was seen in the combination of carboplatin
`and paclitaxel [69]. One of the mechanisms underlying
`these clinically important interactions appears to be
`related to a selective inhibition of cisplatin accumula-
`tion in peripheral blood and bone marrow cells by
`CrEL, without affecting antitumour activity [70–72]
`(Fig. 6). In addition,
`it has been demonstrated that
`CrEL markedly reduced the level of serum haemato-
`poietic inhibitory activity, which resulted in a decrease
`in femoral bone marrow cellularity and an upregulation
`of B and T cells, as well as a transiently elevated inci-
`dence of both primitive and committed haematopoietic
`progenitor cells within several hours after injection [73].
`Taken together, these findings suggest that the unusual
`clinical observations noted with cisplatin-paclitaxel
`combination therapy are due to a (myelo)protective
`interaction with cisplatin by CrEL. Since CrEL doses of
`up to 30 ml/m2 as a 3-h infusion can be safely admi-
`nistered [74], it is anticipated that an improvement in
`the therapeutic window for cisplatin could be obtained
`by re-formulation of this agent with CrEL. This con-
`cept is currently under further investigation (data not
`shown).
`
`In contrast to the potential benefits obtained from the
`use of CrEL in the case of cisplatin chemotherapy,
`combination studies conducted with the combination of
`paclitaxel and doxorubicin have shown that CrEL can
`be linked to clinically important kinetic and dynamic
`interactions that greatly impact upon the overall toxi-
`city profiles. Two independent studies have shown pro-
`found decreases in doxorubicin clearance, particularly
`when paclitaxel was given prior to the anthracycline
`[62,75], resulting in substantially worse mucositis and
`haematological
`toxicity. Therefore,
`it
`is advised to
`administer doxorubicin first in this combination. In
`addition, it is expected that even minor modifications in
`the duration of the infusion or dose may lead to unpre-
`dictable pharmacodynamic consequences. Similar inter-
`actions with CrEL have also been described for
`etoposide, epirubicin and the irinotecan metabolite SN-
`38 [76]. Clearly, the magnitude of these interactions
`depend largely on the combination drug involved, the
`paclitaxel and CrEL dose administered, and the dura-
`tion of infusion administration sequence used for the
`combined agents.
`
`5. Conclusions and future perspectives
`
`Recent investigations have revealed that CrEL, a
`widely used formulation vehicle, is a biologically and
`pharmacologically active ingredient of various com-
`mercially available drugs. For example, when used in
`paclitaxel
`administrations,
`an exceptionally
`large
`amount of CrEL is inevitably co-administered with the
`i.v. infusions, causing important biological events that
`can lead to serious acute hypersensitivity reactions and
`neurological toxicity, depending on the dose and dura-
`tion of infusion. In addition, the substantial effects of
`
`Fig. 5. Effect of dose on paclitaxel plasma (circles) and whole blood
`area under the concentration curve (AUC) (triangles)
`in cancer
`patients treated with paclitaxel at three different dose levels [65]. Please
`note that the higher the AUC, the slower the clearance of a drug.
`
`Fig. 6. Influence of CrEL on anticancer drug pharmacodynamics.
`Bars indicate the amount of drug accumulated in peripheral blood
`leucocytes (PBL) (expressed as percentage relative to a 100% control)
`for 2-h incubations with cisplatin (CDDP), carboplatin (Carbo),
`doxorubicin (dox), and epirubicin (Epi) ([70], data not shown). NS,
`non significant.
`
`Abraxis EX2090
`Cipla Ltd. v. Abraxis Bioscience, LLC
`IPR2018-00162; IPR2018-00163; IPR2018-00164
`
`

`

`1596
`
`H. Gelderblom et al. / European Journal of Cancer 37 (2001) 1590–1598
`
`CrEL on the disposition of other co-administered drugs
`(e.g. anthracyclines) with a narrow therapeutic window
`in poly-chemotherapeutic regimens also can be poten-
`tially hazardous to the patient. In contrast, the in vitro
`and in vivo observations of myeloprotective effects rela-
`ted to the use of CrEL in combination with some
`agents, such as cisplatin, might be used to re-formulate
`such agents with CrEL in order to achieve an optimisa-
`tion of their therapeutic window.
`In view of the inherent problems associated with the
`use of CrEL, it can be anticipated that there may be a
`therapeutic advantage from using paclitaxel formula-
`tions in which CrEL is absent. Such new formulation
`approaches should clearly be pursued to allow a better
`control of the systemic (CrEL-mediated) toxicity and
`pharmacokinetic interactions observed with numerous
`agents given in combination with the taxane. Obviously,
`alternative paclitaxel
`formulations should allow the
`drug to be delivered at adequate doses, and the pre-
`paration should be stable for several hours to allow
`handling in the clinical setting. Currently, a large variety
`of new (CrEL-free) formulation vehicles for paclitaxel
`are in (pre)clinical development,
`including co-solvent
`systems (Tween 80/ethanol/Pluronic L64), water-soluble
`polymers (e.g. polyethylene glycols), emulsions (e.g.
`triacetin), liposomes, cyclodextrines, nanocapsules and
`microspheres (reviewed in Ref. [76]). Although one of
`these preparations might eventually replace the current
`paclitaxel formulation, for the coming years we will
`have to cope with the drawbacks of CrEL in the clinical
`setting. It is also important to keep in mind that CrEL is
`widely used not only by oncologists, but also by anaes-
`thetists and in transplantation medicine, and that few
`physicians are aware of the biological and pharmacolo-
`gical activity of the compound. It is thus essential to
`find other ways to solubilise the drugs formulated in
`CrEL but also, in some cases, to explore the potential
`positive effects of this castor oil derivative. It is of note
`that a proper understanding of all the biological effects
`of the formulation would presumably have avoided the
`major delays in development of paclitaxel as an antic-
`ancer agent. This underscores the importance of the
`choice of vehicle in drug development in general.
`
`References
`
`1. Jonkman-de Vries JD, Flora KP, Bult A, Beijnen JH. Pharma-
`ceutical development of (investigational) anticancer agents for
`parenteral use — a review. ECC Newsletter 1994, 3, 11–14.
`2. Pinedo HM. Review: Development of new anticancer drugs. Med
`Oncol Tumor Pharmacother 1986, 3, 63–69.
`3. Liu G, Fransen E, Fitch MI, Warner El. Patient preferences for
`oral versus intravenous palliative chemotherapy. J Clin Oncol
`1997, 15, 110–115.
`4. Hoffman H. Polyoxythylenglycerol triricinoleat 35 DAC 1979.
`Pharm Zeit 1984, 129, 1730–1733.
`
`5. Goldspiel BR. Guidelines for administration. In: McGuire WP,
`Rowinsky EK, eds. Paclitaxel in Cancer Treatment. New York,
`Marcel Dekker, 1995, 175–186.
`6. Wani MC, Taylor HL, Wall ME, Coggon P, McPhail AT. Plant
`antitumor agents. VI: the isolation and structure of Taxol, a
`novel antileukemic and antitumor agent from Taxus brevifolia. J
`Am Chem Soc 1971, 93, 2325–2327.
`7. Adams JD, Flora KP, Goldspiel BR, Wilson JW, Arbuck SGI.
`Taxol: a history of pharmaceutical development and current
`pharmaceutical concerns. J Natl Cancer Inst Monogr 1993, 15,
`141–147.
`8. Rowinsky EK, Eisenhauer EA, Chaudhry V, Chaudrhy V,
`Arbuck SG. Clinical
`toxicities encountered with paclitaxel
`(Taxol). Semin Oncol 1993, 20, 1–15.
`9. Weiss RB, Donehower RC, Wiernik PH, et al. Hypersensitivity
`reactions from Taxol. J Clin Oncol 1990, 8, 1263–1268.
`10. Eisenhauer EA, ten Bokkel Huinink WW, Swenerton KD, et al.
`European-Canadian randomized trial of paclitaxel in relapsed
`ovarian cancer: high dose versus low-dose and long versus short
`infusion. J Clin Oncol 1994, 12, 2654–2666.
`11. Szebeni J, Muggia FM, Alving CR. Complement activation by
`Cremophor EL as a possible contributer to hypersensitivity to
`paclitaxel: an in vitro study. J Natl Cancer Inst 1998, 90, 300–306.
`12. Watkins J, Ward AM, Appleyard TN. Adverse reactions to
`intravenous anaesthetic induction agents. Br Med J 1997, 2,
`1084–1085.
`13. Hu¨ ttel MS, Schou Olesen A, Stoffersen E. Complement-mediated
`reactions to diazepam with Cremophor as solvent (stesolid MR).
`Br J Anaesth 1980, 52, 77–79.
`14. van Zuylen L, Gianni L, Verweij J, et al. Interrelationships of
`paclitaxel disposition,
`infusion duration and Cremophor EL
`kinetics in cancer patients. Anticancer Drugs 2000, 11, 331–337.
`15. Lorenz WM, Reimann HJ, Schmal A, et al. Histamine release

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