throbber
PCT
`INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT)
`WO 00/06152
`
`WORLD INTELLECTUAL PROPERTY ORGANIZATION
`International Bureau
`
`(51) International Patent Classification 7 :
`A61K 31/337, 47/48, 47/42
`
`Al
`
`(11) International Publication Number:
`
`(43) International Publication Date:
`
`10 Febrnary 2000 ( 10.02.00)
`
`(21) International Application Number:
`
`PCT/US99/17179
`
`(22) International Filing Date:
`
`29 July 1999 (29.07.99)
`
`(30) Priority Data:
`60/094,687
`
`30 July 1998 (30.07.98)
`
`us
`
`(71) Applicant
`except US):
`States
`designated
`all
`(for
`NOVOPHARM BIOTECH, INC. [CA/CA]; 30 Novopharm
`Court, Toronto, Ontario MIB 2K9 (CA).
`
`(72) Inventors; and
`(75) Inventors/Applicants (for US only): KADIMA, Tenshuk, A.
`[CA/CA]; 7 Woodington Bay, Winnipeg, Manitoba R3P
`1M6 (CA). KAPLAN, Howard, A. [CA/CA]; 18 Hillhouse
`Road, Winnipeg, Manitoba R2V 2V9 (CA). TUTTLE,
`Robert, C. [US/CA]; 782 Allegheny Drive, Winnipeg,
`Manitoba R3T 5L2 (CA).
`
`(74) Agents: WU, Frank et al.; Morrison & Foerster LLP, 755 Page
`Mill Road, Palo Alto, CA 94304-1018 (US).
`
`(81) Designated States: AE, AL, AM, AT, AU, AZ, BA, BB, BG,
`BR, BY, CA, CH, CN, CU, CZ, DE, DK, EE, ES, FI, GB,
`GE, GH, GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KP,
`KR, KZ, LC, LK, LR, LS, LT, LU, LV, MD, MG, MK,
`MN, MW, MX, NO, NZ, PL, PT, RO, RU, SD, SE, SG, SI,
`SK, SL, TJ, TM, TR, TT, UA, UG, US, UZ, VN, YU, ZA,
`ZW, ARIPO patent (GH, GM, KE, LS, MW, SD, SL, SZ,
`UG, ZW), Eurasian patent (AM, AZ, BY, KG, KZ, MD,
`RU, TJ, TM), European patent (AT, BE, CH, CY, DE, DK,
`ES, FI, FR, GB, GR, IE, IT, LU, MC, NL, PT, SE), OAPI
`patent (BF, BJ, CF, CG, CI, CM, GA, GN, GW, ML, MR,
`NE, SN, TD, TG).
`
`Published
`With international search report.
`Before the expiration of the time limit for amending the
`claims and to be republished in the event of the receipt of
`amendments.
`
`(54) Title: PHARMACEUTICALLY ACCEPTABLE COMPOSITION COMPRISING AN AQUEOUS SOLUTION OF PACLITAXEL
`AND ALBUMIN
`
`(57) Abstract
`
`An optically clear, pharmaceutically acceptable aqueous composition comprising paclitaxel or a derivative thereof, sernm albumin
`and a pharmaceutically acceptable vehicle, wherein the composition comprises no more than 10 % organic solvent and has a pH of about
`3.0 to about 4.8, is described. The sernm albumin can be fatted or defatted, and the composition can optionally be lyophilized or optionally
`lyophilized and reconstituted. At least 70 % of the paclitaxel is bound to sernm albumin, the ratio of paclitaxel to albumin is at least about
`I :5, and the concentration of paclitaxel is at least about 25 µg!ml. Methods of making and using this composition are also provided.
`
`Actavis - IPR2017-01103, Ex. 1004,
`p. 1 of 162
`
`

`

`FOR THE PURPOSES OF INFORMATION ONLY
`
`Codes used to identify States party to the PCT on the front pages of pamphlets publishing international applications under the PCT.
`
`AL
`AM
`AT
`AU
`AZ
`BA
`BB
`BE
`BF
`BG
`BJ
`BR
`BY
`CA
`CF
`CG
`CH
`CI
`CM
`CN
`cu
`CZ
`DE
`DK
`EE
`
`Albania
`Armenia
`Austria
`Australia
`Azerbaijan
`Bosnia and Herzegovina
`Barbados
`Belgium
`Burkina Faso
`Bulgaria
`Benin
`Brazil
`Belarus
`Canada
`Central African Republic
`Congo
`Switzerland
`Ci\te d'Ivoire
`Cameroon
`China
`Cuba
`Czech Republic
`Germany
`Denmark
`Estonia
`
`ES
`FI
`FR
`GA
`GB
`GE
`GH
`GN
`GR
`HU
`IE
`IL
`IS
`IT
`JP
`KE
`KG
`KP
`
`KR
`KZ
`LC
`LI
`LK
`LR
`
`Spain
`Finland
`France
`Gabon
`United Kingdom
`Georgia
`Ghana
`Guinea
`Greece
`Hungary
`Ireland
`Israel
`Iceland
`Italy
`Japan
`Kenya
`Kyrgyzstan
`Democratic People's
`Republic of Korea
`Republic of Korea
`Kazakstan
`Saint Lucia
`Liechtenstein
`Sri Lanka
`Liberia
`
`LS
`LT
`LU
`LV
`MC
`MD
`MG
`MK
`
`ML
`MN
`MR
`MW
`MX
`NE
`NL
`NO
`NZ
`PL
`PT
`RO
`RU
`SD
`SE
`SG
`
`Lesotho
`Lithuania
`Luxembourg
`Latvia
`Monaco
`Republic of Moldova
`Madagascar
`The former Yugoslav
`Republic of Macedonia
`Mali
`Mongolia
`Mauritania
`Malawi
`Mexico
`Niger
`Netherlands
`Norway
`New Zealand
`Poland
`Portugal
`Romania
`Russian Federation
`Sudan
`Sweden
`Singapore
`
`SI
`SK
`SN
`sz
`TD
`TG
`TJ
`TM
`TR
`TT
`UA
`VG
`us
`uz
`VN
`YU
`zw
`
`Slovenia
`Slovakia
`Senegal
`Swaziland
`Chad
`Togo
`Tajikistan
`Turkmenistan
`Turkey
`Trinidad and Tobago
`Ukraine
`Uganda
`United States of America
`Uzbekistan
`Viet Nam
`Yugoslavia
`Zimbabwe
`
`Actavis - IPR2017-01103, Ex. 1004,
`p. 2 of 162
`
`

`

`WO 00/06152
`
`PCT/US99/17179
`
`PHARMACEUTICALLY ACCEPTABLE COMPOSITION COMPRISING AN AQUEOUS SOLUTION OF PACLITAXEL
`AND ALBUMIN
`
`5
`
`10
`
`CROSS-REFERENCE TO RELATED APPLICATIONS
`
`(Not Applicable)
`
`STATEMENT OF RIGHTS TO INVENTIONS
`
`MADE UNDER FEDERALLY-SPONSORED RESEARCH
`
`(Not Applicable)
`
`TECHNICAL FIELD
`
`The present invention relates generally to aqueous formulations of paclitaxel and
`
`methods of use thereof. More specifically, it pertains to pharmaceutical compositions
`
`15
`
`comprising paclitaxel (Ptx) or a derivative thereof and serum albumin or a fragment
`
`thereof, particularly human serum albumin, and more particularly recombinant human
`
`serum albumin, and a physiologically acceptable vehicle; methods of preparation of such
`
`pharmaceutical compositions; and methods of use thereof. The vehicle can comprise an
`
`organic solvent, and the composition lacks a toxic emulsifier such as Cremophor EL®
`
`20
`
`(polyoxyethylated castor oil).
`
`BACKGROUND OF THE INVENTION
`
`Paclitaxel, a structurally complex natural plant product, has demonstrated efficacy
`
`25
`
`in the treatment of a wide variety of human malignancies. This drug shows strong
`
`cytotoxicity in KB cell structures and in several of the National Cancer Institute's in vivo
`
`screens, including the P-388, L-1210, and P-1534 mouse leukemias, the B-16
`
`melanocarcinoma, the CX-1 colon xenograft, the LX-1 lung xenograft, and the MX-1
`
`breast xenograft. Further, studies by McGuire et al. [(1989) Ann. Int. Med. 111 :273-279]
`
`30
`
`found paclitaxel to be active against drug-refractory ovarian cancer. Positive results were
`
`also seen with paclitaxel treatment of patients with other cancers, including melanoma.
`
`Actavis - IPR2017-01103, Ex. 1004,
`p. 3 of 162
`
`

`

`WO 00/06152
`
`PCT/US99/17179
`
`Einsig et al. (1988) Proc. Am. Soc. Clin. Oneal. 7:249; Holmes (1991) J Natl. Cancer Inst.
`
`83:1797-1805; and Kohn et al. (1994) J Natl. Cancer Inst. 86:18-24.
`
`In addition to various cancers, paclitaxel has been used in treating several other
`
`diseases, including malaria and babesiosis. U.S. Patent Nos. 5,356,927 and 5,631,278.
`
`5
`
`Paclitaxel can be used to treat indications characterized by chronic inflammation such as
`
`rheumatoid arthritis and auto-immune disease. U.S. Patent No. 5,583,153; and Song et al.
`
`(1996) Arthritis Rheum. 39:S 178. Paclitaxel can impair chronic inflammation by inhibiting
`
`the activity of white blood cells involved in the inflammatory response; reducing the
`
`production of matrix metalloproteinases that permanently damage tissues; blocking the
`
`10
`
`cancer-like growth of previously normal cells which respond to chronic inflammation by
`
`proliferating; and inhibiting the growth of blood vessels which lead to the formation of scar
`
`tissue. Paclitaxel is also a potent inhibitor of angiogenesis and other processes involved in
`
`the development of chronic inflammation. This activity is due, in part, to paclitaxel's
`
`ability to inhibit the transcription factor AP-1. AP-1 is a key regulator of genes involved in
`
`15
`
`the production of (i) matrix metalloproteinases, (ii) cytokines associated with chronic
`
`inflammation, and (iii) proteins necessary for cell proliferation. Therefore, paclitaxel
`
`inhibits a regulator which plays an important role in chronic inflammation and conditions
`
`that are dependent on angiogenesis (new blood vessel formation), including tumor growth.
`
`Paclitaxel has shown strong anti-angiogenic activity when tested in the chorioallantoic
`
`20
`
`membrane of the developing chick embryo. The drug is a more potent angiogenesis
`
`inhibitor than approved anti-arthritic agents such as methotrexate, penicillamine, and
`
`steroids.
`
`Atherosclerosis and restenosis have also been treated with low paclitaxel dosages.
`
`U.S. Patent No. 5,616,608. Paclitaxel can alter several aspects of the process leading to
`
`25
`
`restenosis, including inhibition of vascular smooth muscle cell ("VSMC") migration,
`
`inhibition of VSMC proliferation, and inhibition of the effects of certain growth factors on
`
`these cells. Paclitaxel also inhibits synoviocyte proliferation. Paclitaxel is capable of
`
`inhibiting proliferation of synoviocytes in vitro and inducing apoptosis (programmed cell
`death) at concentrations as low as 10-7 M, and is cytotoxic to the synoviocytes at slightly
`higher concentrations of 1 o-6 to 10-5 M. Paclitaxel inhibits collagenase production by
`chondrocytes in vitro, but is not toxic to normal chondrocytes. A concentration of 10-7 M
`
`30
`
`paclitaxel, for example, reduced collagenase expression by over 50% in cultured
`
`2
`
`Actavis - IPR2017-01103, Ex. 1004,
`p. 4 of 162
`
`

`

`WO 00/06152
`
`PCT/US99/17179
`
`chondrocytes stimulated by tumor necrosis factor and interleukin- I. This inhibition occurs
`
`downstream from the transcription factor activity of c-fos and c-jun, apparently by
`
`disrupting the normal functioning of the AP- I molecule, resulting in inhibition of
`
`transcription of the collagenase gene. As such, inhibition of collagenase secretion by
`
`5
`
`paclitaxel is not strictly due to interruption of the protein secretory pathway, which is
`
`dependent upon microtubule function for the movement of secretory granules. Paclitaxel
`
`also appears to act at the level of the genetic response to stimuli directing the cell to
`
`produce collagenase.
`
`The drug is also known to be effective in treating a number of other indications.
`
`I 0
`
`Paclitaxel is useful for treating surgical adhesions and post-surgical hyperplasias. In
`
`Alzheimer's disease treatment, paclitaxel has been used to stabilize microtubules
`
`destabilized by insufficient tau protein levels. U.S. Patent No. 5,580,898. Paclitaxel is also
`
`thought to be effective against polycystic kidney disease (PKD). Sommardahl et al. (1997)
`
`Pediatr. Nephrol. I I :728-33. Paclitaxel derivatives are also effective in treating psoriasis.
`
`I5
`
`EP 747385 and WO 96I3494.
`
`Other therapeutic agents have been successfully co-administered with paclitaxel.
`
`For example, Vitamin C can be used to increase the efficacy of paclitaxel. Kurbacher et al.
`
`(1996) Cancer Lett. 103: I83-I89. EP 78I552 and EP 7877I6 describe additional
`
`compounds that enhance paclitaxel activity. U.S. Patent No. 5,565,478 describes
`
`20
`
`combinational therapy of paclitaxel with signal transduction inhibitors for cancer treatment
`
`In treatment of autoimmune arthritis, paclitaxel has been administered with other
`
`antiarthritic drugs, such as an angiogenesis inhibitor. U.S. Patent No. 5,583,I53. Anilide
`
`derivatives have also been administered to sensitize multidrug-resistant cancer cells to
`
`paclitaxel. EP 6494IO. Paclitaxel can also be administered with antibodies specific to
`
`25
`
`cancerous cells. U.S. Patent No. 5,489,525. In breast cancer treatment, paclitaxel has been
`
`administered in combination with estramustine phosphate. Keren-Rosenberg et al. (1997)
`
`Sem. Oneal. 24 (Suppl. 3):S3-26-29. Paclitaxel and IGF-I (Insulin-like growth factor I)
`
`have been used together to treat peripheral neuropathy. U.S. Patent Nos. 5,648,335,
`
`5,569,648 and 5,633,228. Paclitaxel has also been successfully administered along with
`
`30
`
`doxorubicin, cyclophosphamide, and cisplatin. O'Shaughnessy et al. (1995) Breast Cancer
`
`Res. Treat. 33:27-37. P-glycoprotein blocker SDZ PSC 833, a cyclosporin derivative, has
`
`demonstrated a I 0-fold increase in oral bioavailability of paclitaxel in mice. Asperen et al.
`
`3
`
`Actavis - IPR2017-01103, Ex. 1004,
`p. 5 of 162
`
`

`

`WO 00/06152
`
`PCT/US99/l 7179
`
`(1997) Brit. J Cancer 76:1181-1183. Essential oils have also been suggested to increase
`
`paclitaxel's bioavailability. U.S. Patent No. 5,716,928.
`
`The mechanism of paclitaxel action has been extensively studied and is summarized
`
`by Horwitz (1984) Pharm. Ther. 25:83-125. Paclitaxel can act by promoting tubulin
`
`5
`
`assembly into stable aggregated structures which resist depolymerization by dilution,
`
`calcium ion, cold, and several microtubule-disrupting drugs. Tubulin depolymerization is
`
`essential for cell division, and thus paclitaxel causes this process to cease. Schiff et al.
`
`(1979) Nature 277:665-667. Paclitaxel is unique in promoting tubulin polymer formation,
`
`whereas other anti-cancer drugs, such as vinblastine and colchicine, prevent this process.
`
`10
`
`As originally described in Wani et al. [(1971) J Amer. Chem. Soc. 93:2325-2327],
`
`paclitaxel can be purified via alcohol extraction from the Pacific yew tree, Taxus brevifolia.
`
`It is also present in other Taxus species, such as T baccata and T cuspidata. However,
`
`paclitaxel is found only in minute quantities in the bark of these slow-growing trees,
`
`causing concern that the limited paclitaxel supply will not meet the demand. Consequently,
`
`15
`
`chemists in recent years have attempted to find alternative or synthetic routes for producing
`
`paclitaxel. U.S. Patent No. 5,019,504 describes the purification of paclitaxel from tissues
`
`of T brevifolia grown in vitro. U.S. Patent No. 5,322,779 describes the production of
`
`paclitaxel from a fungus, Taxomyces andreanae, found in association with the yew tree.
`
`More recently, novel compounds have been suggested for use in enhancing plant
`
`20
`
`production of paclitaxel. U.S. Patent No. 5,710,099.
`
`Paclitaxel has also been synthesized from related compounds found in higher
`
`quantities in Taxus trees. These compounds include baccatin III, obtained from Taxus
`
`wood, and 10-deacetyl baccatin III, from Taxus leaves. Methods of preparing paclitaxel
`
`from these precursor compounds, which themselves lack anti tumor activity, have been
`
`25
`
`described. Greene et al. (1988) JACS 110:5917-5919; U.S. Patent Nos. 5,717,103,
`
`4,857,653, and 4,924,011 (Re. 34,277).
`
`Various synthetic routes and intermediates in paclitaxel synthesis have been
`
`described, including a route directed to the synthesis of the tricyclic taxane nucleus from
`
`commodity chemicals. Holton et al. (1994) J Am. Chem. Soc. 116:1597-1598, 1599-1600;
`
`30
`
`Nicolaou et al. (1994) Nature 367:630-634; and Danishefsky et al. (1996) J Am. Chem.
`
`Soc. 118:2843-59; and U.S. Patent Nos. 5,723,635 and 5,726,318. Additional compounds
`
`useful in paclitaxel synthesis have also been described. U.S. Patent No. 5,015,744
`
`4
`
`Actavis - IPR2017-01103, Ex. 1004,
`p. 6 of 162
`
`

`

`WO 00/06152
`
`PCT/US99/17179
`
`describes the use of an oxazinone as a side-chain precursor for paclitaxel synthesis. U.S.
`
`Patent No. 4,876,399 describes an intermediate, 2,5-dihydroxy-2-patchoulene. U.S. Patent
`
`Nos. 5,523,219 and 5,705,671 describe additional intermediates.
`
`Paclitaxel itself has been chemically modified, sometimes producing compounds
`
`5
`
`with even greater antitumor activity than paclitaxel itself. U.S. Patent No. 4, 814,470.
`
`Cephalomannine, which differs from paclitaxel and baccatin III in the C-13 ester
`
`functionality, demonstrates activity against leukemia in animals. U.S. Patent No.
`
`4,206,221. Other paclitaxel derivatives include prodrug forms, in which paclitaxel is
`
`conjugated to cleavage spacer and sugar groups. EP 781778.
`
`10
`
`Some paclitaxel derivatives have been produced in attempts to address a significant
`
`problem limiting the utility of paclitaxel: paclitaxel is largely insoluble in water. This has
`
`created significant problems in developing suitable pharmaceutical formulations for human
`
`therapy both in terms of formulation and side effects. The problem is also a serious
`
`impediment for experimental research on paclitaxel and its clinical effectiveness.
`
`15
`
`Derivatives of paclitaxel, designed to have increased water solubility, include 2' - and/or 7-
`
`position paclitaxel esters, as described in U.S. Patent No. 4,960,790. Additional
`
`substitutions at the C-2' and C-7 positions were described by Magri et al. (1988) J Natural
`
`Products 51:298-306. 2' -succinyl paclitaxels are described in U.S. Patent No. 4,942,184;
`
`and sulfonated 2' -acryloyltaxol and sulfonated 2 '-0-acyl acid paclitaxel derivatives, in
`
`20
`
`U.S. Patent No. 5,059,699.
`
`Unfortunately, many of these more soluble derivatives reduce paclitaxel antitumor
`
`activity. A 2' -succinyltaxol, prepared by the treatment of paclitaxel with succinic
`
`anhydride, had decreased in vivo activity compared with paclitaxel, and a 2'-(t(cid:173)
`
`butyldimethylsilyl)taxol was essentially inactive. Magri et al. (1988). Other derivatives,
`
`25
`
`such as 2' -(~-alanyl)taxol, are unstable. Magri et al. (1988). Attempts to derivatize
`
`paclitaxel generally increase the molecule's size, which decreases its ability to passively
`
`diffuse through the cellular and nuclear membranes of cancerous cells.
`
`The insolubility of paclitaxel itself has yielded a further complication: it has elicited
`
`the widespread use of a toxic carrier. Paclitaxel is generally supplied through CTEP
`
`30
`
`(Cancer Therapy Evaluation Program), DCT (Division of Cancer Treatment), and NCI
`
`(National Cancer Institute, IND#2280) as a concentrated solution in 50% polyoxyethylated
`
`castor oil [Cremophor EL® (BASF)] and 50% dehydrated alcohol. This is then mixed with
`
`5
`
`Actavis - IPR2017-01103, Ex. 1004,
`p. 7 of 162
`
`

`

`WO 00/06152
`
`PCT/US99/1717'J
`
`either a dextrose or sodium chloride solution prior to administration. Although Cremophor
`
`EL® is the industry-standard administration vehicle for paclitaxel, Cremophor EL® is
`
`itself toxic, causing idiosyncratic histamine release and anaphylactoid-like response.
`
`Cremophor EL® is also likely to be the cause of several side effects associated with
`
`5
`
`paclitaxel treatment, including cutaneous flushing, urticaria, dyspnea, bronchospasm, and
`
`hypotension. Runowicz et al. (1993) Cancer 71: 1591-1596; and Weiss et al. (1990) J
`
`Clin. Oneal. 8:1263-126. In studies with dogs, Cremophor EL® and its fatty acid
`
`constituents induced histamine release and hypotension within 10 minutes of
`
`administration. Lorenz et al. (1977) Agents Actions 7:63-67. Some tested animals died as a
`
`10
`
`result of this hypotension.
`
`Other organic carriers have been proposed for paclitaxel administration or used in in
`
`vitro paclitaxel preparations. Polyethylene glycol (PEG) has been suggested as a substitute
`
`emulsifier for paclitaxel, but PEG decreases the anti tumor activity of paclitaxel in murine
`
`tumor studies. Weiss et al. (1990) J Clin. Oneal. 8:1263-1268. Paclitaxel has also been
`
`15
`
`prepared in solution with dimethylsulfoxide [Kumar et al. (1993) Res. Comm. Chem. Path.
`
`Pharm. 80:337-344], which is itself toxic [Kamiya et al. (1967) Nippon Ganka Kiyo
`
`18:387-9; Sperling et al. (1979) Acta Ophthalmol. 57:891-8]. Polysorbate-80 was used in
`
`in vitro mixtures containing very low concentrations of docetaxel [Urien et al. (1996)
`
`Invest. New Drugs 14:147-151], but polysorbates are toxic, reducing locomotor activity,
`
`20
`
`inducing ataxia and hypotension, and increasing the activity of various carcinogens. Pesce
`
`et al. (1989) Ann. Clin. Lab. Sci. 19:70-3; (1984) J Am. Coll. Toxicol. 3/5: 1-82; and Varma
`
`et al. (1985) Arzneimittelforschung 35:804-8. Therefore, the sole use of these carriers to
`
`solubilize paclitaxel is not a desirable solution to the problem of developing therapeutically
`
`effective paclitaxel formulations.
`
`25
`
`In the absence of workable alternatives, and despite its toxicity, Cremophor EL®
`
`remains the standard vehicle used for paclitaxel administration to human patients.
`
`Documents demonstrating the universal use of Cremophor EL® in paclitaxel preparations
`
`and paclitaxel administration include: Einzig et al. (1991) Cancer Invest. 9:133-136;
`
`O'Shaughnessy et al. (1994) Breast Cancer Res. Treat. 33:27-37; Kawano et al. (1994) J
`
`30
`
`Toxicol. Sci. 19(suppl.1):113-122; Asperen et al. (1997) Brit. J Cancer 76:1181-1183;
`
`Sparreboom et al. ( 1998) Anti-Cancer Drugs 9: 1-1 7; Runowicz et al. ( 1993) Cancer
`
`71: 1591-1596; Sparreboom et al. (1998) Anal. Biochemistry 255: 171-175; Plasswilm et al.
`
`6
`
`Actavis - IPR2017-01103, Ex. 1004,
`p. 8 of 162
`
`

`

`WO 00/06152
`
`PCT/US99/17179
`
`(1998) Strahlentherapie und Onkologie 174:37-42; Xu et al. (1997) Hospital Pharmacy
`
`32: 1635-1638; Khan et al. (1997) Ann. Pharmacotherapy 31: 1471-1474; Michaud et al.
`
`(1997) Ann. Pharmacotherapy 31: 1402-1404; Zhang et al. (1997) Anti-Cancer Drugs
`
`8:696-701; Wilson et al. (1997)Ann. Pharmacotherapy 31:873-875; Reinecke et al. (1997)
`
`5
`
`Eur. J Cancer Part A :United Kingdom 33:1122-1129; Kuangjing Shao et al. (1997) Anal.
`
`Chemistry 69:2008-2016; Bonfrer et al. (1997) Tumor Biology; Switzerland 18:232-240;
`
`Decorti et al. (1997) Cancer Chemother. Pharmacology 40:363-366; Ho et al. (1997)
`
`Neurosurgery 4016 :1260-1268; Terzis et al. (1997) British J Cancer 75:1744-1752;
`
`Kilbourn et al. (1997) Disease-a-Month 43 :282-348; Frasci et al. (1997) J Clinical
`
`10
`
`Oncology 15:1409-1417; Sharma et al. (1997) International J Cancer 71:103-107;
`
`Georgiadis et al. (1997) Clinical Cancer Research 3 :449-454; Zhang et al. (1997) Cancer
`
`Chemother. Pharmacology40:81-86; EP 694303; WO 94/12031; and U.S. Patent Nos.
`
`5,733,888, 5,731,334, 5,719,265, 5,714,512, 5,703,117, 5,698,582, 5,696,153, 5,686,488,
`
`5,683,715, 5,681,846, 5,670,537, 5,665,761, 5,648,335, 5,648,090, 5,641,803, 5,633,228,
`
`15
`
`5,621,001, 5,616,608, 5,616,330, 5,614,549, 5,608,087, 5,604,202, 5,569,648, 5,583,153,
`
`5,580,899, 5,569,720, 5,565,478, 5,504,102, 5,496,846, 5,496,804, 5,478,860, 5,403,858.
`
`Numerous attempts have been made to produce aqueous solutions of hydrophobic
`
`drugs. For instance, formulations of cisplatin combined with dextran, polyglutamic acid,
`
`DNA, proteins, hyaluronic acid, etc. were compared. It was found that many of these
`
`20
`
`excipients were unacceptable as they bound the drug too tightly and did not release it on
`
`administration or did not bind enough drug to produce a pharmaceutically acceptable
`
`formulation. DNA was in the category of excipients which bound too tightly. Proteins,
`
`including serum albumin, were found to bind limited amounts of drug, only a portion of
`
`which was reversibly bound.
`
`25
`
`Albumins have been used as excipients as bulk stabilizers for a number of drug
`
`formulations, particularly biologicals such as interleukins and cytokines. Human serum
`
`albumin is a large component of interleukin-4 preparations. Meyer et al. (1994) Pharm.
`
`Res. 11: 1492-1495. Albumin has also been conjugated to drugs to increase uptake of the
`
`drug and derivatized albumins have been used to couple drugs and enhance uptake through
`
`30
`
`the blood-brain barrier. Sinn et al. (1990) Nucl. Med. Biol. 17:819-827; Pardridge et al.
`
`(1990) J Pharmacol. Exp. Ther. 255:893-899; Flume et al. (1989) Pharm. Acta Helv.
`
`64:351-352; and JP 61001622. WO 94/01090 describes broad formulations of hydrophilic
`
`7
`
`Actavis - IPR2017-01103, Ex. 1004,
`p. 9 of 162
`
`

`

`WO 00/06152
`
`PCT/US99/17179
`
`peptides and "sparingly water soluble" active compounds. Albumin is a cost-limiting
`
`component for use in drug stabilization. Thus, unless an unstable drug can be stabilized in
`
`some other fashion, albumin is not ideal as a bulk stabilizing agent. Further, native
`
`albumin is being phased out of use as it may contain infectious agents such as prions.
`
`5
`
`Replacement with recombinant albumin may result in an even more costly product.
`
`Therefore, in order to produce a commercially available, pharmaceutically acceptable
`
`albumin-bound drug, the drug must be bound reversibly to the albumin in a high molar
`
`ratio.
`
`The need remains for aqueous pharmaceutically acceptable formulations of
`
`10
`
`paclitaxel which are easy and inexpensive to prepare, produce fewer side effects, and in
`
`which the drug retains high water solubility and activity.
`
`SUMMARY AND OBJECTS OF THE INVENTION
`
`In one embodiment, the invention provides an optically clear, pharmaceutically
`
`15
`
`acceptable aqueous composition comprising paclitaxel or a derivative thereof, serum
`
`albumin or a fragment thereof, and a pharmaceutically acceptable vehicle. In various
`
`embodiments, the composition comprises no more than 10% organic solvent, and has a pH
`
`of about 3.0 to about 4.8 (the pl of albumin). In various embodiments, the composition
`
`comprises about 1 to about 10%, about 2 to about 8%, or about 4 to about 6% v/v
`
`20
`
`(volume/volume) organic solvent. In a preferred embodiment, the composition is essential
`
`free of organic solvent. The organic solvent is preferably an alcohol, most preferably
`
`ethanol. In various embodiments, the pH is about 3.0 to about 4.8, about 4.0 or less, about
`
`3.0 to about 4.0, or about 3.4 to about 3.8. In various embodiments, the ratio of paclitaxel
`
`or derivative thereof to albumin is at least about 1 :5, at least about 1 :4, at least about 1 :2, at
`
`25
`
`least about 1: 1, or at least about 2: 1. In various embodiments, the serum albumin is
`
`defatted, undefatted or a mixture of defatted and undefatted forms. In various
`
`embodiments, the serum albumin is mammalian, preferably human. In various
`
`embodiments, the serum albumin is at least about 50%, at least about 60%, at least about
`
`70%, at least about 80%, or at least about 90% monomeric. In various embodiments, at
`
`30
`
`least about 70%, at least about 80%, at least about 85%, or at least about 90% of the
`
`paclitaxel or derivative thereof is bound to albumin. In another embodiment, the
`
`composition is lyophilized. In another embodiment, the composition is reconstituted from a
`
`8
`
`Actavis - IPR2017-01103, Ex. 1004,
`p. 10 of 162
`
`

`

`WO 00/06152
`
`PCT/US99/17179
`
`lyophilized formulation. In various embodiments, the concentration of paclitaxel is greater
`
`than about 25 µg/ml, greater than about 50 µg/ml, greater than about 100 µg/ml, greater
`
`than about 200 µg/ml, greater than about 300 µg/ml, greater than about 400 µg/ml, or
`
`greater than about 500 µg/ml. In another embodiment, the composition is coated onto an
`
`5
`
`implantable device such as a stent or wrap. In some embodiments, the device is catheter(cid:173)
`
`based and/or used in conjunction with surgery. In some embodiments, the coating prevents
`
`restenosis, local tumor growth or tissue over-growth and/or chronic inflammation.
`
`The composition is characterized by having optical clarity for a length of time
`
`sufficient to administer to a patient or to process further (e.g., subject to drying). In another
`
`10
`
`embodiment, the composition is optionally dried and stored as a dried "storage-stable"
`
`composition. The dried preparation of the composition is resolubilized prior to
`
`administration. In a preferred embodiment, the drying process is lyophilization. In one
`
`embodiment, the composition prior to drying comprises Mcllvaine buffer. In another
`
`embodiment, the lyophilized preparation of the composition is optionally reconstituted with
`
`15
`
`a physiologically acceptable vehicle, such as Mcllvaine buffer, water, a sugar solution such
`
`as dextrose or glucose, or certain saline solutions, including dilutions of saline. The
`
`reconstituted compositions can be essentially free of solvent, which can be removed in the
`
`lyophilization step. The resolubilized composition can be 2-10 times more concentrated
`
`than the original pharmaceutically acceptable composition, depending on the concentration
`
`20
`
`of paclitaxel in the pre-lyophilized composition. Thus, the invention encompasses a
`
`resolubilized composition which is optically clear for at least 8 hours after reconstitution.
`
`The composition comprises less than 10% organic solvent and has a pH of about 3 .0 to
`
`about 4.8 upon reconstitution, at least about 70% of the paclitaxel introduced into the
`
`composition is bound to the serum albumin, and the paclitaxel concentration in the
`
`25
`
`composition is at least 50 µg/ml. The invention further encompasses methods of
`
`administration of the reconstituted composition wherein a therapeutically effective amount
`
`of paclitaxel can be administered as a 1 to 3 hour (or greater) injection or as a bolus.
`
`In another embodiment, the invention encompasses a method of treatment,
`
`comprising administering to a patient a therapeutically effective amount of an optically
`
`30
`
`clear, pharmaceutically acceptable aqueous composition comprising paclitaxel or a
`
`derivative thereof, serum albumin and a pharmaceutically acceptable vehicle, as described
`
`above. The indication to be treated with the composition can include any indication known
`
`9
`
`Actavis - IPR2017-01103, Ex. 1004,
`p. 11 of 162
`
`

`

`WO 00/06152
`
`PCT /US99/17179
`
`in the art to be treatable with paclitaxel, including, but not limited to, cancer. Preferably,
`
`the cancer affects cells of the bladder, blood, bone, brain, breast, cervix, colon, epithelium,
`
`digestive tract, head/neck, kidneys, liver, lung, mouth, ovaries, pancreas, prostate gland,
`
`skin, stomach, testicles, or tongue. The indication can also include, but is not limited to,
`
`5
`
`paclitaxel-treatable indications such as Alzheimer's disease, kidney disease, peripheral
`
`neuropathy, psoriasis, restenosis, rheumatoid arthritis, systemic lupus erythematosus,
`
`surgical adhesions, or tissue overgrowth after surgery. Preferably, the patient is a mammal.
`
`More preferably, the mammal is a human.
`
`The composition and methods of use thereof can optionally further comprise an
`
`10
`
`additional biologically active ingredient, including but not limited to those known to
`
`function synergistically with paclitaxel. In various embodiments, the additional agent
`
`includes, but is not limited to, G-CSF (granulocyte colony-stimulating factor), GM-CSF
`
`(granulocyte macrophage colony-stimulating factor), IL-4 (interleukin 4), IGF-I, analide
`
`derivatives, antiarthritics (e.g., an angiogenesis inhibitor), antibodies specific to cancer
`
`15
`
`cells, antineoplastics (e.g., carboplatin, cyclophosphamide, estramustine phosphate, and
`
`etoposide ), doxorubicin, immunosuppressants (e.g., cisplatin and cyclophosphamide ),
`
`steroidal and non-steroidal hormone (e.g., cortisone), transduction inhibitors, and vitamins
`
`(e.g., vitamin C). The composition can further comprise low concentrations of excipients
`
`such as polyethylene glycol, detergents, organic solvents, or organic or inorganic acids.
`
`20
`
`In another embodiment, the invention encompasses a method of making an optically
`
`clear, pharmaceutically acceptable aqueous composition comprising paclitaxel or a
`
`derivative thereof, serum albumin and a pharmaceutically acceptable vehicle, as described
`
`above, comprising the steps of preparing a solution of the paclitaxel or a derivative thereof,
`
`preparing a solution of serum albumin, and slowly combining the solutions. Due to stable
`
`25
`
`binding of Ptx to serum albumin, the rate of addition of the Ptx solution to the albumin
`
`solution can be decreased to assist in more optimal loading of Ptx onto albumin. The
`
`paclitaxel solution can, for example, be added dropwise at a controlled rate; this rate can
`
`be, for example, at about 0.1 to 10 ml/min, e.g., 1 ml/min or slower, and the drop size can
`
`be 8 to 20 µ1. In various embodiments, the ratio of paclitaxel or derivative thereof to
`
`30
`
`albumin is at least about 1: 1 or at least about 2: 1, and the solutions are combined at a
`
`temperature below room temperature, about 2°C to 8°C, or about 4°C. In various
`
`embodiments, the ratio of paclitaxel or derivative thereof to albumin is at least about 1: 5, at
`
`10
`
`Actavis - IPR2017-01103, Ex. 1004,
`p. 12 of 162
`
`

`

`WO 00/06152
`
`PCT/US99/17179
`
`least about 1 :4, at least about 1 :2, at least about 1: 1, or at least about 2: 1. It is anticipated
`
`that ratios of 3: 1 and possibly even 4: 1 can be achieved according to the invention
`
`described herein, by controlling the rate of addition of the paclitaxel to the albumin solution
`
`to a degree that does not interfere with continued stability during processing.
`
`5
`
`Preferably the paclitaxel is "optimally concentrated." This term means that the
`
`paclitaxel concentration in the composition allows a solvent concentration of 1 - 10% v/v.
`
`The molar ratio of paclitaxel:albumin and the final concentration of paclitaxel in the
`
`albumin solution are optimized, such that the paclitaxel remains in solution for a length of
`
`time practical for administration or lyophilization/reconstitution. We have found that the
`
`

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