throbber
The Chemistry and Pharmacology of Taxol and its Derivatives
`V Farina editor
`© 1995 Elsevier Science BV All rights reserved
`
`103
`
`3 P
`
`ACLITAXEL TAXOL®
`FORMULATION AND PRODRUGS
`
`Dolatrai M Vyas
`
`BristolMyers Squibb Company Pharmaceutical Research Institute
`5 Research Parkway Wallingford CT 064927660 USA
`
`31
`
`INTRODUCTION
`
`Interest
`
`Paclitaxel 111 Figure 11 a natural diterpene isolated from the bark of
`Taxus brevifolia Pacific yew 1 has been heralded as the antitumor agent of
`the 1990s because of its promising clinical activity against a variety of human
`solid tumors such as ovary breast lung head and neck and melanoma 24
`in various aspects of this drug has mushroomed due to the clinical
`significance of its antitumor profile in treating common human cancers 5 6
`is an FDA approved cancer agent
`in the United States for
`Currently paclitaxel
`second line treatment against cisplatinum refractory ovarian cancer and
`It has
`metastatic breast cancer which is refractory to anthracycline treatment
`for similar indications On the basis of
`
`also been approved in other countries
`
`first line chemotherapeutic
`
`broader ongoing clinical trials there is a good likelihood of paclitaxel becoming a
`in the very near future 7
`Within the current arsenal of cancer chemotherapeutics 8 paclitaxel is a
`
`agent
`
`unique tubulininteracting agent Unlike other clinical antimitotic agents such
`as the vinca alkaloids 9 101 which inhibit the microtubule assembly process
`
`Taxol® is a registered trademark of the BristolMyers Squibb corporation The generic name paclitaxel
`throughout
`this chapter
`
`is used
`
`Abraxis EX2003
`Actavis LLC v Abraxis Bioscience LLC
`1PR201701101 1PR201701103 1PR201701104
`
`

`

`104
`
`paclitaxel
`
`promotes tubulin polymerization and stabilizes the resulting
`microtubules toward depolymerization 11 This shift in the normal dynamics in
`the cellular tubulinmicrotubule system by paclitaxel
`is currently widely
`recognized as its mode of action for cell cytotoxicity
`Owing in part to the
`in 1979 11 of paclitaxels
`discovery by Susan Horwitz
`and coworkers
`mechanism of action the National Cancer Institute NCI decided to accelerate
`the human clinical
`trials for this drug Eventual marketing of paclitaxel
`for
`oncology clinical use was facilitated by a Cooperative Research and Development
`Agreement CRADA between the NCI and the Bristol Myers Squibb BMS
`Company in 1989 and the subsequent massive efforts by BMS to secure ample
`clinical supplies of paclitaxel 12
`
`BzNH 0
`
`Ph
`
`5H
`
`Ac0
`
`OH
`
`fee
`
`HO
`
`Bz0
`
`Aco
`
`Boc
`
`NH
`
`0
`
`Ph
`
`b
`
`OH
`
`111 Paclitaxel
`Taxol
`
`Figure 1 Clinically relevant
`
`taxanes
`
`HO
`
`OH
`
`HO
`
`= Ac0
`Bz0
`
`112 Docetaxel
`Taxotere
`
`In spite of paclitaxels promising clinical antitumor profile and the ongoing
`
`efforts to optimize its utility
`
`for curative cancer
`in multimodality treatment
`problems during its intravenous iv
`therapy the drug has presented
`administration to patients 13 These are formulation related problems
`stemming from the use of the excipient Cremophor EL® 14 as a solubilizing
`detergent for iv administration Interestingly a close analog of paclitaxel
`namely docetaxel 112Taxotere® 15 Figure 1 which is currently awaiting
`FDA approval
`for clinical use is devoid of such problems Docetaxel
`possesses a
`slightly better aqueous solubility than paclitaxel and is administered as a
`Tween80 polyoxyethylene sorbitan monooleateethanol
`formulation Several of
`formulation related patient care issues such
`as
`have been successfully addressed and managed in
`the clinic 16 However the pharmaceutical
`issues 17 related to Cremophor
`ELs use still persist during intravenous administration and demand precautions
`
`paclitaxels
`
`alleged
`
`hypersensitivity reactions
`
`

`

`current clinical
`
`alleviate or circumvent
`
`105
`
`to
`
`and adherence to strict pharmacy protocols The main intent of this chapter is to
`document and highlight all of the formulation related issues stemming from the
`formulation and briefly discuss possible solutions sought
`them The quest
`toward the development
`of a widely
`formulation devoid of Cremophor EL is
`and safe intravenous
`continuing in several research laboratories and is discussed herein
`
`acceptable
`
`32
`
`PHARMACEUTICAL DEVELOPMENT SUMMARY
`
`collection of
`
`Paclitaxels early pharmaceutical development history 12 18 19 is
`mainly confined to the NCI laboratories since it was through a joint NCI USDA
`United States Department of Agriculture initiative in the early 1960s to screen
`plant material for novel cytotoxic agents that paclitaxel was first discovered
`The isolation and identification of this agent were accomplished in 1966 at the
`NCI contract
`laboratories of the Research Triangle Institute North Carolina by
`M Wani and M Wall approximately four years after the initial
`Taxus Brevifolia plant material 18 The isolation of pure paclitaxel was
`facilitated through a bioassay guided fractionation protocol
`following in vitro
`cytotoxicity in KB cells and in vivo antitumor activity against murine tumor
`models such as leukemia P1534 and carcinosarcoma Walker 256 During the
`in several of the
`next few years the predinical antitumor profile of paclitaxel
`NCIs murine hematological
`tumor models namely leukemias L1210 P388 and
`P1354 was established Also activity against the Walker 256 sarcoma model
`was demonstrated
`These efficacy evaluations were carried out
`in mice by
`administering paclitaxel as a suspension in the peritoneal ip cavity against
`ipimplanted tumors Due to the extremely hydrophobic character of paclitaxel
`20 it was difficult
`to obtain a purely water based formulation for iv or ip
`administration Consequently the vehicles employed for ip administration
`included steroidal suspensions vegetable oils peanut sesame olive normal
`cellulose CMC It was not until 1974 that the first
`saline and carbomethoxy
`evidence of paditaxels efficacy in an ipip murine solid tumor B16 melanoma
`was obtained Until this time paclitaxel was considered an unexciting cytotoxic
`agent with solubility problems and in vivo efficacy confined mainly to ipip
`localized tumor models Consequently there was little enthusiasm in pushing
`this drug further toward human clinical trials However by 1980
`several new
`findings had made paclitaxel a prime candidate for human clinical
`
`trials
`
`In
`
`

`

`106
`
`the introduction of human tumor xenograft models to the NCI
`1978 after
`screening program paclitaxel was shown for the first
`time to possess distal
`the LX 1 lung the MX1 breast and the CX1 colon
`tumor efficacy against
`xenografts 18 In all
`these tumor models the drug was administered
`to the human tumor implanted in the sub renal capsule kidney
`subcutaneously
`of mice More importantly the excitement and interest about
`this drug was
`in 1979 when a report by SHorwitz and coworkers 11 disclosed
`heightened
`that paclitaxel acted via a novel tubulininteracting mechanism Unfortunately
`
`at this juncture a suitable formulation for intravenous administration of the
`the outset 21 it was
`drug was lacking and remained to be developed
`determined at the NCI
`that paclitaxel was totally devoid of any activity when
`administered orally to mice up to a dosage of 160 mgKg
`
`At
`
`321 Early Formulation Studies
`the NCI was directed at developing
`an
`Since 1978 a major effort at
`intravenous formulation for paclitaxel in order to initiate human clinical studies
`Paclitaxels extremely low water solubility < 001mgm1 coupled with the
`functionality amine or carboxylic acid for salt
`absence of a suitable chemical
`formation led to the evaluation of cosolvents and excipients as the first strategy
`for developing an iv formulation Much of this effort is described in detail in a
`recent report 21 The approximate solubility of paclitaxel
`and certain organic solvents are reported in Table 1
`
`in aqueous vehicles
`
`Table 1 Solubility of Paclitaxel
`
`in Various Solventsa
`
`Solvent
`
`Solubility mgml
`
`Methylene Chloride
`
`Ethanol
`75 Propylene glycol
`75 Polyethylene glycol 400 PEG 400
`35 PEG 400
`Soybean oil
`
`a adapted from ref 21
`
`Triacetin
`
`> 19
`
`ca 39
`<14
`
`31
`
`003
`03
`
`75
`
`

`

`107
`
`Paclitaxel has substantial solubility in organic solvents such as ethanol
`and methylene chloride Highly concentrated millimolar paclitaxel solutions
`in some of these solvents were attainable However diluting paclitaxel solutions
`of water miscible solvents such as ethanol with water presented problems of
`precipitation The solubility of paclitaxel
`
`in lipids such as soybean oil
`
`intralipid
`
`was also not quite adequate for formulation considerations
`The two intravenous formulations which received indepth evaluation at
`the NCI were those involving cosolvent PEGpolyethylene glycol 400 75 and
`surfactant Cremophor EL® 19 21 The 75 PEG 400 solution in water
`containing 16 mgml paclitaxel was found to be chemically stable by HPLC
`analysis and free of particulate material for up to 14 days at 25°C However
`infusion precipitation cloudiness was discernible
`upon dilution for
`
`Formulation with solubilizing surfactant Cremophor EL® 5 in ethanol 5
`and 09 saline under equilibrium conditions
`gave a solution with paclitaxel
`concentration of about 01 mgml With this formulation concentrations greater
`than 06 mgml were achievable by dilution of a 6 mgml solution of paclitaxel in
`11 Cremophor ELEt0H Fortunately this solution had adequate physical and
`chemical stability over periods as long as 24 h >96 of drug was found in
`solution over this time period Thus the Cremophor formulation provided some
`
`assurance against
`
`the possibility of drug precipitating during the infusion
`
`period Also this formulation was the best on an efficacy basis since paclitaxel
`administered ip as an aqueous suspension containing Cremophor EL® was
`found to be more efficacious against an ipimplanted B16 melanoma tumor than
`paclitaxel administered ip in PEG 400 Due to all these considerations in 1980
`the Cremophor formulation was selected for clinical trials The current clinical
`dosage form of paclitaxel consists of a 5m1 size vial containing 30 mg of
`paclitaxel 264 g of Cremophor EL® and 497 Et0H 11 vv This
`concentrated solution required further dilution with injectable fluids such as 5
`dextrose 09 sodium chloride and 5 dextrose in Ringers solution The intact
`vial shelf life is estimated to be 5 years under refrigeration 21
`
`322 Formulation Issues
`With the launching
`into clinical
`trials
`of paclitaxel
`pharmaceutical and patient care issues surfaced at the very outset
`These
`apparently stemmed from the use of Cremophor EL® as an excipient in the
`intravenous formulation Cremophor EL® is a chemically and physiologically
`
`several
`
`

`

`108
`
`active surfactant 14 Thus the formulation is associated with potential medical
`depending upon the amount of Cremophor EL® present and the route
`by which this formulation is administered to patients As it currently stands
`
`liabilities
`
`formulation has the highest amount of this surfactant
`the paclitaxel clinical
`among all currently marketed drugs 14 Other drugs with Cremophor in their
`formulations are teniposide 22 and cyclosporine 23 The potential for harmful
`that
`effects of Cremophor to patients is further augmented by the fact
`quantities of this solubilizing surfactant are required for intravenous delivery of
`doses of paclitaxel ranging from 150 to 300 mgm2
`issues 17 stemming from the
`Pharmaceutical
`Issues Pharmaceutical
`intravenous administration of the Cremophor ELObased formulation have
`mainly affected hospital health care professionals such as pharmacists and
`nurses who are required to exercise caution and care during iv infusion
`Chemical stability studies have shown that the paclitaxel
`
`therapeutic
`
`large
`
`formulation diluted
`
`with standard infusion fluids is stable for up to 27 hours at room temperature
`This has facilitated the preparation and storage of iv solutions in hospital
`pharmacies
`However due to the leaching properties of Cremophor toward
`phthalate plasticizers from PVC infusion bags and intravenous administration
`set tubings use of polyolefin plastic containers glass bottles or vinyl acetate
`type bags has been mandated
`
`Also to guard against
`precipitation during infusion paclitaxel is administered with an in line filtration
`device containing a 022 µm membrane filter eg IVEX2 and IVEXHP
`The issue of paclitaxel compatibility with other medications has also been
`studied in some detail 24 25 An examination of visual and turbidimetric
`compatibility of paclitaxel with 59 other drugs was carried out The list of drugs
`included antimicrobials anticancer agents analgesics antiemetics and
`involved the mixing of 12 mgml
`The experiment
`antiallergy medications
`solution 4 ml of paclitaxel
`in D5W with a 4 ml sample of the test drug at
`clinically relevant concentrations
`All of the drugs but four were compatible with
`
`the perils of drug
`
`paclitaxel based on the turbidity measurement criteria These included two
`cancer drugs mitoxantrone hydrochloride and doxorubicin
`In the case of
`doxorubicin a loss of 12 paclitaxel
`in 96 hours was determined by HPLC
`measurements It was stressed in this study that the absence of turbidity effects
`does not automatically imply chemical compatibility with paclitaxel and caution
`
`should be exercised particularly when need arises for combination therapy with
`
`other drugs
`
`

`

`109
`
`Patient Care Issues In clinical
`
`trials paclitaxel has manifested some of the
`classical drug related toxicities associated with an anticancer agent 16 These
`The
`include neutropenia neurotoxicity mucositis GI toxicities and alopecia
`
`cumulative dose paclitaxel
`
`major dose limiting toxicity was demonstrated to be neutropenia which was
`usually severe at doses >200 mgm2 The incidence of severe neutropenia was
`similar at all dose levels and recovery was complete after 510 days Since
`hematopoietic growth factors eg GCSF are now commonly used to ameliorate
`remains a major concern in high single and
`neutropenia neurotoxicity
`therapy The neurotoxicity manifested is sensory
`neuropathy which is common when paclitaxel is administered by a 24 h infusion
`and exceeds a dose of 200 mgm2
`signs and
`Hypersensitivity reactions particularly
`anaphylactoid
`liability and patient care issue in the clinical
`symptoms remain a major medical
`use of paclitaxel 26 The hypersensitivity
`reaction is alleged to be due to
`Cremophor EL® contained
`in the formulation The symptoms include rapid
`onset of hypotension respiratory distress eg bronchospasms urticaria and
`rash These appear to be classical histamine release mediated reactions which
`have also been observed in dogs treated with Cremophor EL® alone 27 These
`allergic reactions due to the Cremophor threatened the continuation of earlier
`paclitaxel clinical trials due to the death of a patient 7 However
`they were
`infusion and emergency treatment of
`controlled by discontinuation of paclitaxel
`patients with antihistamines H2blockers and oral steroids 13
`and the severity of
`Currently in order to minimize the occurrence
`hypersensitivity reactions in patients two routine measures have been
`instituted These include the extension of the infusion duration time to 6 or 24 h
`and the prophylactic use of antihistamines and corticosteroids prior to paclitaxel
`administration 7 These measures do not completely eliminate the histamine
`
`they certainly reduce the
`release effects of paclitaxel administration but
`episodes and severity of hypersensitivity reactions In spite of the introduction
`
`of these safeguards working with paclitaxel
`in the hospital setting remains a
`challenge A nonCremophor based intravenous formulation should alleviate the
`above pharmaceutical and patient care concerns However a suitable clinical
`substitute has not yet been identified Several NCI funded labs and independent
`
`research groups are currently actively engaged in research
`
`to either develop a
`
`safer parenteral administration of paclitaxel through watersoluble prodrugs or
`to identify a suitable carrier system for iv administration
`
`

`

`110
`
`33 PRODRUGS OF PACLITAXEL
`
`A thoroughly explored preclinical strategy for iv administration of
`paclitaxel has involved the use of prodrugs in a nonCremophorcontaining
`preferably 100 aqueous formulation 28 Initially this effort has involved the
`synthesis and evaluation of prodrugs with moieties carrying a solubilizing group
`In this context an ideal handle for prodrug synthesis has been the C2 hydroxyl
`functionality since derivatives masked at this position are devoid of activity
`until unmasked in vivo through hydrolysis by enzymatic or chemical means A
`free hydroxyl group at C2 seems to be a prerequisite for tubulin polymerization
`and consequently for cytotoxicity 29 In contrast
`the C7 hydroxyl
`is not such an ideal site for prodrug design since masked derivatives eg esters
`at this position are usually much more stable than their C2 counterparts to in
`vivo cleavage 30 Consequently only special C7 derivatives with a rapid in
`vivo unmasking pathway have the potential
`to serve adequately
`as a prodrug
`Pro moieties which are slow to unmask in vivo may simply act as paclitaxel
`analogs 31
`
`functionality
`
`as Prodrugs
`
`331 Acvl Derivatives
`In 1984 Mellado et al 32 during their investigation on the biological
`activity of paclitaxel acetates established that the C2 hydroxyl can be readily
`the C7 hydroxyl
`and selectively acetylated over
`functionality Furthermore
`they found the C2 acetate to be more labile toward hydrolysis than its C7
`counterpart These earlier observations formed the basis for considering the C2
`for future research efforts on
`hydroxyl group as the preferred functionality
`watersoluble prodrugs 28
`Paclitaxel C2 Esters The basic strategy in designing C2 esters as water
`soluble prodrugs has been the incorporation of ionizable groups such as amines
`amino acids and sulfonic acid groups In this vein a plethora of C2 derivatives
`have been synthesized and evaluated
`for their suitability as watersoluble
`prodrugs of paclitaxel These include Figure 2 succinate eg 311 and
`glutarate derivatives eg 312 33 34 sulfonic acid derivatives eg 313 and
`314 35 and amino acid derivatives eg 315 and 316 30 34 Although
`these derivatives eg 311 and 312
`possess adequate water
`solubility up to 1 for an iv formulation and good biological activity in vivo
`
`several of
`
`

`

`111
`
`their chemical
`
`This was attributed to
`they were found unsuitable as prodrugs of paclitaxel
`instability in aqueous solution a property deemed unacceptable
`for intravenous administration of a highly insoluble agent such as paclitaxel To
`date among this class of prodrugs 243NNdiethylaminopropionylpaclitaxel
`316 prepared by Stella and coworkers 30 appears to be best suited for
`It has acceptable solution stability at pH 35
`half life >400 h However the half life at physiological
`pH 74 was <30 mm
`life was even shorter < 5 min The aqueous
`In human plasma the half
`In vivo 316 showed
`solubility of this prodrug was determined to be >10 mgml
`complete tumor remission in the MX1 mammary tumor model
`
`prodrug delivery of paclitaxel
`
`Ac0
`
`OH
`
`BzNH 0
`Ph
`
`OR
`
`HO
`
`E Ac0
`OBz
`
`311 R= COCH22COOH HOCH2CH23N
`312 R= COCH23COONa
`313 R= COCH22SO3Na
`314 R= COCH22CONHCH2SO3Na
`315 R= COCH22NH2 HCO2H
`316 R= COCH22NEt2 CH3S03H
`317 R= COCH2OCH2CO2H
`318 R= COCH2SCH2CO2H
`319 R= COCH2S02CH2CO2H
`3110 R= COCH22CONHPEG
`
`Figure 2 Watersoluble C2 esters as paclitaxel prodrugs
`
`More recently a Scripps group led by Nicolaou has reported another series
`of C2 esters named protaxols 36 These are monoesters 317 318 and
`319 of dicarboxylic acids incorporating a heteroatom functionality oxygen or
`sulfur for the purpose of imparting further water solubility These compounds
`have been reported to have acceptable water solubility ca 1 mgml and stability
`at neutral pH and room temperature Under basic conditions 319 rapidly
`
`

`

`112
`
`The authors envisage that such a mechanism may be
`generated paclitaxel
`relevant in vivo in the basic microenvironment of certain tumor cells However
`in vivo antitumor activity of these protaxols in murine tumor models have not
`
`yet been reported by the Scripps group
`Thus far all of the above approaches have only yielded prodrugs with
`moderate up to 10 mgml water solubility However
`conjugates of hydrophobic
`glycol PEG of molecular weight of 2kD or greater
`molecules with polyethylene
`are known to impart even greater water solubility
`In this context Greenwald and coworkers 37 have recently reported the
`synthesis and limited biological evaluation of a variety of C2 polyethyleneglycol
`esters as highly watersoluble prodrugs of paclitaxel 3110 The PEG esters
`reported are claimed to have water solubility of >666 mgml at ambient
`temperature The half life of 3310 was determined to be approximately 40 h
`and 11 h in pH 74 buffer and rat plasma respectively The authors have not
`reported any in vivo antitumor activity but the approach seems very promising
`Paclitaxel C7 Esters In general C7 esters have found little utility as water
`To date several C7 derivatives have been
`soluble prodrugs of paclitaxel
`reported 30 and in most cases
`they have been the counterparts of their
`corresponding C2 derivatives
`Their poor prodrug properties have
`attributed to enhanced in vivo stability
`toward esterases and hydrolytic
`
`been
`
`cleavage
`For example the cationic watersoluble derivative 3111Figure 3 had a
`life of 378 h at pH 38 compared with a half
`life of of 96 h for its
`half
`corresponding C2 ester derivative A similar trend was observed
`physiological pH 74 where 3111 had a half life of 34 h compared to 6 h for its
`C2 counterpart Half lives in human plasma for 3111 and its C2 counterpart
`were reported to be 3 h and <30 mm respectively
`The enhanced hydrolytic chemical and enzymatic stability of C7 esters
`over their C2 counterparts
`can be ascribed to the steric congestion surrounding
`this position Biologically unlike the C2 esters cationic watersoluble C7
`esters such as 3111 were shown to promote microtubule assembly as effectively
`as paclitaxel but have been found to be poorly bioactive in whole cell assays
`This was attributed to poor cell membrane penetration
`
`at
`
`

`

`113
`
`Bz NHO
`
`Ph
`
`OH
`
`Figure 3 Typical C7 ester prodrug of paclitaxel
`
`Paclitaxel C2 Carbonates Recently the BMS group and the Scripps group
`have demonstrated that C2 carbonates like their ester counterparts behave as
`paclitaxel prodrugs in vivo Carbonates and carbamates have so far found little
`
`utility as prodrugs because they are more stable than the corresponding esters
`toward hydrolytic and enzymatic cleavage Notable exceptions in this area are
`38 and
`carbamate derivatives of drugs such as 10hydroxycamptothecin
`CC1065 39 which have been shown to be effective prodrugs in vivo
`The BMS group has recently disclosed the synthesis and in vivo biological
`profile of C2 carbonates derivatives 311217 Figure 4401
`
`Ac0
`
`OH
`
`H 0
`
`Bz
`
`Ph
`
`0
`
`HO
`
`CO
`
`E
`
`OBz
`
`RO
`
`3112 R= CH3
`3113 R= CH2CH3
`3114 R= CHCH32
`3115 R= CH2CI
`3116 R= CH2Ph
`3117 R=CH=CH2
`
`Figure 4 C2 carbonate derivatives synthesized by the BMS group
`
`When evaluated in the cytotoxicity assay paclitaxel2carbonates were 2
`line HCT
`10 times less cytotoxic than paclitaxel against human colon cancer cell
`116 Also they were found to be inactive in the tubulin polymerization assay
`However after incubation in rat plasma at 37 °C for 18 hrs some of these
`carbonate derivatives particularly 3112 and 3113 were found to promote
`microtubule assembly indicating the generation of
`the parent compound
`
`paclitaxel
`
`in rat plasma All of the above observations were fully substantiated
`
`

`

`114
`
`by their in vivo antitumor activity in the Madison 109 murine lung carcinoma
`M109 tumor model 41 as summarized in Table 2 In this tumor model both
`the prodrug and the tumor were localized intraperitoneally ip and as such
`represented an in vivo testtube model All carbonates were effective TC >
`125 is considered to be active in increasing life span The methyl carbonate
`3112 was less active than paclitaxel but all
`the other carbonates exhibited
`comparable in vivo antitumor activity with respect
`results indicated that paclitaxel2carbonates
`are converted to the parent drug
`
`These in vivo
`
`to paclitaxel
`
`under the in vivo conditions and act as true prodrugs of paclitaxel
`
`Table 2 In vivo Antitumor Activity of Paclitaxel2Carbonates 311217
`
`Compound
`
`3112
`3113
`3114
`3115
`3116
`3117
`
`In vivo Antitumor Activitya
`TC mgKginjection
`Carbonate Deny
`
`Paclitaxel
`
`162 90 mgKgb
`>475 60c
`247 100b
`275 60c
`310 50c
`>475 60c
`
`276 75 mgKgb
`275 30c
`197 50b
`275 30c
`270 50c
`275 30c
`
`a Madison 109 murine lung carcinoma M109 ipimplant model Drugs administered ip in 10
`in 10 DMSOsaline 311317 or in 10 DMSOH20
`Tween 80 in saline paclitaxel
`80 3112 TC refers to the percentage
`few drops of Tween
`of the median survival
`drug treated mice six per dose vs saline treated control The TC values are determined at
`the maximum tolerated dose shown in brackets TC >125 is defined as active in this tumor
`model b Dose administered ip on days 5 and 8c Dose administered ip on days 1 5 and 9
`
`plus a
`
`time of
`
`One obvious drawback of the above carbonate derivatives is their extreme
`
`release in vivo
`
`insolubility in water As will be discussed later
`a watersoluble prodrug
`strategy for one of these carbonates was successfully developed by attaching
`the C7 hydroxyl functionality
`In a similar vein
`soluble pro moieties at
`have recently reported novel watersoluble C2
`Nicolaou and coworkers
`arylsulfonyl ethylcarbonates 311820 Figure 5 36 These compounds were
`prepared in order to evaluate a novel mechanism of paclitaxel
`in vivo through a base
`
`Accordingly they were conceived
`
`to generate paclitaxel
`
`

`

`induced 13 elimination reaction However these authors have not yet reported
`
`whether these putative prodrugs are actually effective in vivo
`
`115
`
`NH
`
`0
`
`Ph
`
`0
`
`RO
`
`3118 R
`
`3119 R =
`
`3120 R =
`
`0
`
`0
`
`0
`
`0
`
`0
`
`0
`
`NO2
`
`NH2
`
`Figure 5 Paclitaxel 2 carbonates prepared by the Scripps group
`
`332 Phosphate Esters Derivatives
`The Bristol Myers Squibb BMS group elected for reasons of novelty to
`focus on the synthesis and evaluation of watersoluble phosphatasecleavable
`prodrugs of paclitaxel 28 This strategy mandated the synthesis and
`evaluation of hitherto unknown phosphate esters of paclitaxel Use of phosphate
`
`as Prodrugs
`
`derivatives at
`
`derivatives as water solubleprodrugs of clinically useful drugs such as etoposide
`42 clindamycin 43 and mustards 44 has been well documented
`The
`rationale behind their use as prodrugs stems from the ubiquitous nature of
`phosphatase enzymes in mammalian systems 45 Interestingly certain tumors
`are shown to express high levels of membrane bound alkaline phosphatases and
`and accumulation of these
`thus provide an opportunity for selective cleavage
`the tumor site 46 In the BMS prodrug program the initial
`targets synthesized were the obvious prototypic C2 and C7 phosphate
`derivatives 321 and 322 Figure 6 47
`the sodium salts of 321 and 322 were endowed with
`Although
`impressive water solubility 10 mgml their in vitro and in vivo evaluation
`indicated that they were poor prodrugs of paclitaxel The first
`indication of this
`was evident when both derivatives failed to generate paclitaxel
`in vitro upon
`treatment with purified preparations of bovine intestinal alkaline phosphatase
`
`

`

`116
`
`BzNH 0
`
`Ph
`
`OR1
`
`Ac0
`
`9 R2
`
`321 R1=P0OH2 R2=H
`322 Ri=H R2=P0OH2
`
`=
`
`HO
`
`Ac0
`0 Bz
`Figure 6 Prototypical paclitaxel phosphates
`
`This was further corroborated by their extreme stability and failure to generate
`in rat plasma Also both were inactive in promoting microtubule
`paclitaxel
`assembly in vitro In vivo evaluation of sodium salts of 321 and 322 against
`the intraperitoneal ip Madison 109 murine M109 lung tumor model
`in
`comparison with paclitaxel demonstrated see Table 3 that
`these analogs
`possessed marginal antitumor activity at best in this model TC 125 is
`
`considered active
`
`Table 3 In vivo Activity of Sodium Salts of 321 and 322 in the M109 Tumor
`Modela
`
`Maximum TCb mgkginj
`
`Experiment
`
`Compound
`
`Phosphate
`
`Paclitaxelc
`
`id
`
`2d
`
`321 Na salt
`322 Na salt
`
`14025
`123 30
`
`27050e
`19010
`
`aMurine lung carcinoma ip implant model bTC refers to the percent of the median survival
`time of drug treated mice 6 per dose us saline treated controls at the maximum tolerated dose
`in brackets cAdministered in 10 Tween 80 in saline dDose administered ip on days 1 5 and
`9 eAlso a dose of 25 mgkginj achieved a TC of 240 with 16 mice cured
`
`The lack of in vitro enzymatic cleavage and inferior in vivo activity of 321
`and 322 indicated that
`these phosphates
`are poor substrates for alkaline
`phosphatase Sterically both the C2 phosphate and the C7 phosphate moieties
`in 321 and 322 are quite close to the congested
`taxane core Consequently it
`enzymes are not able to easily bind these
`is likely that
`the phosphatase
`functionalities and process them effectively
`In order to
`
`to generate paclitaxel
`
`

`

`in prototypes 321 and 322 toward
`address the steric barrier encountered
`phosphatase cleavage the BMS chemists pursued a more complex strategy ie
`the synthesis of proprodrugs of paclitaxel which can be activated by
`phosphatase Figure 7
`
`117
`
`Bz
`
`NH
`
`0cLPh
`
`OH
`
`Bz
`
`Ph
`
`NH 0
`
`oH
`
`spont
`
`00P0OH
`
`phosphatase
`
`0
`
`Ac0
`
`HO
`
`= Ac0
`OBz
`
`323
`
`Ac0
`
`OH
`
`= Ac0
`OBz
`
`HO
`
`111
`
`OH
`
`324
`
`4= self immolating
`
`linker
`
`Figure 7 Proprodrug strategy based on phosphatase
`
`enzymes
`
`Proprodrug strategies have been sucessfully applied in medicinal chemistry
`48 The two most widely employed concepts in the design of proprodrugs have
`been the exploitation of the fragmentation cascade 49 and of the lactonization
`reaction 50 Adaptation of these strategies to paclitaxel entails the design and
`use of appropriate selfimmolating linkers at either C2 or C7 as depicted in
`Figure 7 The concept
`involves dephosphorylation of the proprodrug 323 by
`phosphatases in vivo followed by the rapid collapse through a fragmentation
`cascade reaction
`step of
`transient dephosphorylated
`lactonization
`intermediate 324 to yield paclitaxel 111 The lactone approach
`previously exploited by medicinal chemists in prodrug design since the factors
`
`or
`
`has been
`
`

`

`118
`
`that influence the rates of lactonization reactions are generally well understood
`51
`
`NH2
`
`OMe
`
`327
`
`325
`
`326
`
`Figure 8 A selfimmolative linker approach to drug delivery
`
`A prototypic proprodrug design to demonstrate the proof of principle was
`lock linker recently reported by Borchardt and co
`based on the ltrimethyl
`workers 52 in the design of an esterase cleavable proprodrug of an amine
`Figure 8 It was elegantly demonstrated by this group through in vitro
`experiments with isolated enzyme preparations and plasma studies that 325
`was an effective proprodrug of pmethoxy aniline 327 The success
`of this
`lactonization rate enhancement of the
`approach was ascribed to the phenomenal
`order of >105 achieved by the presence of methyl substituents on the alkyl chain
`and the aromatic ring The estimated half life of lactonization for the desacetyl
`analog of 325 is of the order of 1 min
`Extension of this strategy to phosphate derivatives
`required the synthesis
`of acid linker 328 Figure 9 This was then utilized in the synthesis of the
`desired targets 329 and 3210 53 The sodium salts of 329 and 3210 were
`found to possess acceptable water solubility ca 10 mgml for iv evaluation of
`these prodrugs in murine tumor models Solution stability determinations in pH
`74 Tris buffer 50mM at 37 °C were substantially different Tgo values defined
`as the time required for the compound to undergo 10 degradation were
`determined to be ca 20 h for 329 and >100 h for 3210 Thus 329 may lack
`the stability necessary for intravenous administration without encountering
`precipitation problems during iv infusion An indepth in vitro evaluation of
`these two proprodrugs in biological
`fluids and with pure enzyme was
`undertaken
`the necessary parameters Also kinetic
`the cleavage process using bovine intestinal AP and p
`
`investigations
`
`of
`
`to establish
`
`all
`
`

`

`119
`
`nitrophenol phosphate PNP as a reference standard were performed
`The
`VmaxKm ratio for the prodrugs was determined and compared with that of PNP
`It was found that
`as a measure of enzyme efficiency with these substrates
`3210 had VmaxKm = 06 vs PNP=1 whereas VmaxKm for 329 was only
`013 This may indicate that the enzyme is confronted with lesser steric
`congestion around C7 than C2 Interestingly the selfimmolation process for
`by the AP in both cases
`the linker to generate paclitaxel after dephosphorylation
`was rapid t12<5 min in agreement with literature precedents
`
`Ph26
`
`NH
`
`0
`
`Bz
`
`OH20P9
`
`9
`
`OH
`
`328
`
`Bz
`
`NH
`
`0
`
`1111
`
`6H
`
`329
`
`Figure 9 Phosphate proprodrugs of paclitaxel
`
`The above in vitro observations for the two phosphate derivatives were
`further substantiated by their in vivo antitumor performance When evaluated
`the C2 esterphosphate 329 was found
`against the ipip M109 tumor model
`to be marginally active whereas the C7 esterphosphate 3210 was as active as
`paclitaxel Table 4
`The above results led to the exploration of other

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