throbber
Patent Application
`
`entitled
`
`BENDAMUSTINE PHARMACEUTICAL COMPOSITIONS
`
`Inventors;
`
`Jason Edward Brittain
`1580 Chiswick Ct.
`
`El Cajon, CA 92020
`
`Gary T. Elliott
`12433 Kingspine Ave.
`San Diego, CA 92131
`
`Joe Craig Franklin -
`11519 Kirby Place
`San Diego, CA92126
`
`CERTIFICATE OF MAILILNG BY “EXPRESS MAIL”
`“EXPRESS MAIL” Label Number: EU882120300US
`
`DATE OF DEPOSIT: Januag 14, 2005
`
`I hereby certify that this paper or fee is being deposited with the United States Postal Service “Express
`
`Mail-Post Office to Addressee” service under 37 C.F.R.§ 1.10 on the date indicated above and is addressed
`
`to : Mail Stop Provisional Patent Application, Commissioner for Patents, P.O. Box 1450, Alexandria, VA
`22313-1450.
`
`flmfw./Dd
`
`Robert W. Prince
`
`Number of Drawings: 6 (on 6 pages)
`Docket No.: O32PRV
`
`AGILA ET AL - EXHIBIT 1018 I
`0001
`
`0001
`
`AGILA ET AL - EXHIBIT 1018
`
`

`
`FIELD OF THE INVENTION
`
`The present invention pertains to the field of pharmaceutical compositions for the
`
`treatment of various disease states, especially neoplastic diseases and autoimmune
`
`diseases. Particularly, it relates to pharmaceutical formulations comprising nitrogen
`
`mustards, particularly the nitrogen mustard bendamustine, e.g., bendamustine HCI.
`
`BACKGROUND OF THE INVENTION
`
`The following description includes information that may be useful in understanding the
`
`present invention. It is not an admission that any such information is prior art, or
`
`relevant, to the presently claimed inventions, or that any publication specifically or
`
`implicitly referenced is prior art.
`
`Because of their high reactivity in aqueous solutions, nitrogen mustards are difficultito
`
`formulate as pharmaceuticals and are ofien supplied for administration in a lyophilized
`
`form that requires reconstitution, usually in water, by skilled hospital personal prior to
`
`administration. Once in aqueous solution, nitrogen mustards are subject to degradation
`
`by hydrolysis, thus, the reconstituted product should be administered to a patient as soon
`
`as possible after its reconstitution.
`
`Bendamustine, (4-{5-[Bis(2-chloroethy])amino]—1-methyl-2-benzimidazolyl} butyric
`
`acid, is an atypical structure with a benzimidazole ring, whose structure includes an
`
`active nitrogen mustard (see Formula. 1, which shows bendamustine hydrochloride).
`
`* CI/H
`I
`J/NWLiEN‘>—\_O>~OH -HCI
`'1
`
`Cl
`
`Formula I
`
`Bendamustine was initially synthesized in 1963 in the German Democratic Republic
`
`(GDR) and was available from 1971 to 1992 in that location under the name
`
`Cytostasan®. Since that time, it has been marketed in Germany under the tradename
`
`Ribomustin®. It has been widely used in Germany to treat chronic lymphocytic
`
`leukemia, Hodgkin’s disease, non-Hodgkin’s lymphoma, multiple myeloma, and breast
`
`cancer.
`
`0002
`
`0002
`
`

`
`Due to its degradation in aqueous solutions (like other nitrogen mustards), bendamustine
`
`is supplied as a lyophilized product. The current lyophilized formulation of
`
`bendamustine (Ribomustin®) contains bendamustine hydrochloride and mannitol in a
`
`sterile lyophilized form as a white powder for intravenous use following reconstitution.
`
`The finished lyophilisate is unstable when exposed to light. Therefore, the product is
`
`stored in brown or amber—colored glass bottles. The current lyophilized formulation of
`
`bendamustine contains degradation products that may occur during manufacturing of the
`
`drug substance and/or during the lyophilization process to make the finished drug
`
`product.
`
`Currently bendamustine is formulated as a lyophilized powder for injection with 100 mg
`
`of drug per 50 mL Vial or 25 mg of drug per 20 mL vial. The vials are opened and
`
`reconstituted as close to the time of patient administration as possible. The product is
`
`reconstituted with 40 mL (for the 100 mg presentation) or 10 mL (for the 25 mg
`
`presentation) of Sterile Water for Injection. The reconstituted product is further diluted
`
`into 500 mL, q.s., 0.9% Sodium Chloride for Injection. The route of administration is by
`
`intravenous infusion over 30 to 60 minutes.
`
`Following reconstitution with 40 mL Sterile Water for Injection, vials of bendamustine
`
`are stable for a period of 7 hours under room temperature storage or for 6 days upon
`
`storage at 2—8°C. The 500 mL admixture solution must be administered to the patient
`
`within 7 hours of vial reconstitution (assuming room temperature storage of the
`
`admixture).
`
`The reconstitution of the present bendamustine lyophilized powder is difficult. Reports
`
`from the clinic indicate that reconstitution can require at least fifteen minutes and may
`
`require as long as thirty minutes. Besides being burdensome and time-consuming for the
`
`healthcare professional responsible for reconstituting the product, the lengthy exposure of
`
`bendamustine to water during the reconstitution process increases the potential for loss of
`
`potency and impurity formation due to the hydrolysis of the product by water.
`
`0003
`
`0003
`
`

`
`Thus, a need exists for lyophilized formulations of bendamustine that are easier to
`
`reconstitute and which have a better impurity profile than the current lyophilate
`
`(lyophilized powder) fonnulations of bendamustine.
`
`German (GDR) Patent No. 34727 discloses a method of preparing (o-[5-bis-(B-
`
`chloroethyl)-amino-benzimidazolyl-(2)]-alkane carboxylic acids substituted in the 1-
`
`position.
`
`German (GDR) Patent No. 80967 discloses an injectable preparation of y-[1-methyl-5-
`
`bis-([3-chloroethyl)-amino-benzimaidazolyl-(2)]-butric acid hydrochloride.
`
`Gennan (GDR) Patent No. 159877 discloses a method for preparing 4-[1 —methyl-5—bis
`
`(2-chloroethyl) amino-benzimidazolyl-2)-butyric acid.
`
`German (GDR) Patent No. 159289 discloses an injectable solution of bendamustine.
`
`Ribomustin® bendamustine Product monograph (updated 1/2002)
`
`http://www.ribosepharm.de/pdf/ribomustin bendamustin/productmonographpdf provides
`
`information about Ribomustin® including product description.
`
`Ni et al. report that the nitrosourea SarCNU was more stable in pure tertiary butanol than
`
`in pure acetic acid, dimethyl sulfoxide, methylhydroxy, water or in TBA/water mixtures 0
`
`(Ni et al. (2001) Int]. J. Phamaceutics 226239-46).
`
`Lyophilized cyclophoshamide is known in the alt see e.g., US Patent Nos. 5,418,223;
`
`5,413,995; 5,268,368; 5,227,374; 5,130,305; 4,659,699; 4,537,883; and 5,066,647.
`
`The lyophilized nitrogen mustard Ifosfamide is disclosed in International Publication No.
`
`WO 2003/066027; US Pat. Nos. 6,613,927; 5,750,131; 5,972,912; 5,227,373; and
`
`5,204,335.
`
`Teagarden et al. disclose lyophilized formulations of prostaglandin E-1 made by
`
`dissolving PGE-1 in a solution of lactose and tertiary butyl alcohol (US Pat. No.
`
`5,770,230).
`
`0004
`
`0004
`
`

`
`SUMMARY OF THE INVENTION
`
`The present invention is directed to stable pharmaceutical compositions of nitrogen
`
`mustards, in particular lyophilized bendamustine and its use in treatment of various
`
`disease states, especially neoplastic diseases and autoimmune diseases.
`
`An embodiment of the invention is a pharmaceutical composition of bendamustine
`
`containing not more than about 0.5% to about 0.9% (area percent of bendamustine) HP1,
`
`as shown in Formula II, where the HP1 is the amount of HP] present at time zero after
`
`reconstitution of a lyophilized pharmaceutical composition of bendamustine as described
`
`herein.
`
`HO/\L
`N
`O
`Clf \(:[N:>_\_>_OH Formula 11.
`
`Another embodiment of the invention is a lyophilized preparation of bendamustine
`
`containing not more than about 0.1 % to about 0.3% bendamustine dimer as shown in
`
`Formula III at time zero after reconstitution.
`
`Ho’fi
`Ho’\/N
`
`HO
`
`0
`N
`7:
`I Ii>LN:*\_>‘°“
`
`\
`
`Formula III
`
`Yet another embodiment of the invention is a lyophilized preparation of bendamustine
`
`containing not more than about 0.5% to about 0.15% bendamustine ethylester, as shown
`
`in Formula IV at time zero after reconstitution
`
`C|
`
`%
`
`COOCH2CH3
`
`Formula IV.
`
`0005
`
`0005
`
`

`
`Still another embodiment of the invention is a lyophilized preparation of bendamustine
`
`containing not more than about 0.5 % to about 0.9% (area percent of bendamustine) HP1
`
`at the time of drug product release. An aspect of this embodiment is lyophilized
`
`preparations of bendamustine containing not more than about 0.5 % to about 0.9% (area
`
`percent of bendamustine) HP1 at the time of release of drug product where the
`
`lyophilized preparation is packaged in a vial or other pharmaceutically acceptable
`
`container.
`
`In yet another aspect of the invention, the lyophilized preparations of bendamustine are
`
`stable with respect to the amount of HP1 for at least about 6 months to about 36 months
`
`i or greater when stored at about 2° to about 30°. Preferred temperatures for storage are
`
`about 5° C and about room temperature.
`
`Another embodiment of the invention is a pharmaceutical dosage form that includes a
`
`pharmaceutical composition of bendamustine containing not more than about 0.5% to
`
`about 0.9% HP1 where the HP1 is the amount of HP1 present at time zero afier
`
`reconstitution of a lyophilized preparation of bendamustine of the present invention. In
`
`preferred aspects of the invention, the dosage form can be about 5 to about 500 mg of
`bendamustine, about 10 to about 300 mg of bendamustine, about 25 mg of bendamustine,
`
`about 100 mg of bendamustine, and about 200 mg of bendamustine.
`
`Yet another embodiment of the invention is a pharmaceutical dosage form that includes a
`
`lyophilized preparation of bendamustine containing not more than about 0.5% to about
`
`0.9% HP1. Preferred dosage forms can be about 5 to about 500 mg of bendamustine,
`
`about 10 to about 300 mg of bendamustine, about 25 mg of bendamustine, about 100 mg
`
`of bendamustine, and about 200 mg of bendamustine.
`
`In still another embodiment, the invention includes a pharmaceutical composition of
`
`bendamustine including bendamustine containing not more than about 0.5%‘to about
`
`0.9% HP1 (area percent of bendamustine) and a trace amount of one or more organic
`
`solvents, wherein said HP] is the amount of HP1 present at time zero after reconstitution
`
`of a lyophilized pharmaceutical composition of bendamustine as disclosed herein.
`
`In
`
`different aspects of this embodiment, the organic solvent is selected from one or more of
`
`tertiary butanol, n-propanol, n-butanol, isopropanol, ethanol, methanol, acetone, ethyl
`
`0006
`
`0006
`
`

`
`acetate, dimethyl carbonate, acetonitrile, dichloromethane, methyl ethyl ketone, methyl
`
`isobutyl ketone, l-pentanol, methyl acetate, carbon tetrachloride, dimethyl sulfoxide,
`
`hexafluoroacetone, chlorobutanol, dimethyl sulfone, acetic acid, and cyclohexane.
`
`Preferred organic solvents include one or more of ethanol, methanol, propanol, butanol,
`
`isopropanol, and tertiary butanol. A more preferred organic solvent is tertiary butanol,
`
`also known as TBA, and t-butanol.
`
`The present invention involves a method for obtaining agency approval for a
`
`bendamustine product, the improvement which includes setting a release specification for
`
`bendamustine degradants at about 2.0 % to about 5.5 % (area percent bendamustine) or
`
`otherwise to achieve the pharmaceutical compositions described herein.
`
`Another embodiment is a method for obtaining agency approval for a bendamustine
`
`product, the improvement which includes setting a shelf-life specification for
`
`bendamustine degradants at about 5.0% to about 8.0% (area percent bendamustine)
`
`where the product is stored at about 2°C to about 30°C. Preferred temperatures for
`
`storage are about 5°C and about room temperature.
`
`Another embodiment of the invention is a process for manufacturing a lyophilized
`
`preparation of bendamustine which includes controlling for the concentration of
`
`bendamustine degradants in the final product, such that the concentration of
`
`bendamustine degradants is no more than about 5.5 % to about 2.0 % (area percent of
`
`bendamustine) at release or otherwise to achieve the pharmaceutical compositions
`
`described herein.
`
`The present invention discloses a process for manufacturing a lyophilized preparation of
`
`bendamustine which includes controlling for the concentration of bendamustine
`
`degradants in the final product, such that the concentration of bendamustine degradants is
`
`no more than about 8.0% to about 5.0% at the time of product expiration; wherein said
`
`product is stored at about 2°C to about 30°C.
`
`Another embodiment of the invention is a bendamustine pre-lyophilization solution or
`
`dispersion comprising one or more organic solvents where the solution or dispersions
`
`include at least one stabilizing concentration of an organic solvent which reduces the
`
`level of degradation of bendamustine so that the amount of HP1 produced during
`
`0007
`
`0007
`
`

`
`lyophilization from about 0 to 24 hours does not exceed about 0.5% to about 0.9% (area
`
`percent bendamustine). An aspect of this embodiment is the lyophilized powder
`
`produced from the pre-lyophilization solution or dispersion.
`
`Still another embodiment of the invention is a bendamustine pre-lyophilization solution
`
`or dispersion comprising one or more organic solvents where the solution or dispersions
`
`include at least one stabilizing concentration of an organic solvent which reduces the
`
`level of degradation of bendamustine so that the amount of bendamustine dimer produced
`
`during lyophilization from about 0 to 24 hours does not exceed about 0.5% to about 0.9%
`
`(area percent bendamustine). An aspect of this embodiment is the lyophilized powder
`
`produced from the pre-lyophilization solution or dispersion.
`
`The invention also discloses methods for preparing a bendamustine lyophilized
`
`preparation that includes dissolving bendamustine in a stabilizing concentration of an
`
`alcohol solvent of between about 5% to about 100% (v/v) alcohol; and lyophilizing the
`
`mixture wherein the bendamustine lyophilized preparation made from such methods
`
`contains not more than about 0.5% to about 0.9% (area percent of bendamustine) HP1 as
`
`shown in Formula II, wherein said HP1 is the amount of HP] present at time zero afier
`
`reconstitution of the lyophilized pharmaceutical composition of bendamustine. Other
`
`alcohol concentrations include about 5% to about 99.9%, about 3% to about 70%, about
`
`5% to about 60%, about 3% to about 50%, about 3% to about 40%, about 20% to about
`
`35%. Preferred concentrations of alcohol are from about 20% to about 30%. Preferred
`
`alcohols include one or more of methanol, ethanol, propanol, iso-propanol, butanol, and
`
`tertiary-butanol. A more preferred alcohol is tertiary-butanol. A preferred concentration
`
`of tertiary-butanol is about 20% to about 30%. An aspect of this embodiment is the
`
`addition of an excipient before lyophilization. A preferred excipient is mannitol.
`
`Preferred concentrations of bendamustine are from about 2 mg/mL to about 50 mg/mL.
`
`An aspect of this embodiment includes the lyophilized powder obtained from the
`
`methods of preparation disclosed herein.
`
`The invention also involves bendamustine formulations for lyophilization that include an
`
`excipient and a stabilizing concentration of an organic solvent. A preferred formulation
`
`includes bendamustine at a concentration of about 15 mg/mL, mannitol at a concentration
`
`0008
`
`0008
`
`

`
`of about 25.5 mg/mL, tertiary-butyl alcohol at a concentration of about 30% (v/v) and
`
`water. Included in this embodiment of the invention are the lyophilized preparations
`
`made from such bendamustine formulations.
`
`Included in the inventions are methods of treating a medical condition in a patient that
`
`involve administering a therapeutically effective amount of a pharmaceutical composition
`
`of the invention where the condition is amenable to treatment with said pharmaceutical
`
`composition. Some conditions amenable to treatment with the compositons of the
`
`invention include chronic lymphocytic leukemia (CLL), Hodgkin’s disease, non-
`
`Hodgkin’s lymphoma (NHL), multiple myeloma (MM), breast cancer, small cell lung
`cancer, hyperproliferative disorders, and an autoimmune disease. Preferred conditions
`
`include NHL, CLL, brest cancer, and MM. Preferred autoimmune diseases include
`
`rheumatoid arthritis, multiple sclerosis or lupus.
`
`Also included in the invention are methods of treating in which the pharmaceutical
`
`compositions of the invention are in combination with one or more anti-neoplastic agents
`
`where the antineoplastic agent is given prior, concurrently, or subsequent to the
`
`administration of the pharmaceutical composition of the invention. Preferred
`antineoplastic agents are antibodies specific for CD20.
`
`Another embodiment of the invention is a lyophilization cycle for producing lyophilized
`
`bendamustine preparations of the invention. A preferred lyophilization cycle includes a)
`
`freezing to about -50°C over about 8 hours; b) holding at -50°C for about 4 hours; c)
`
`ramping to -25°C over about 3 hours; d) holding at -10°C for 30 hours; e) ramping to
`
`between about 25°C and about 40°C or higher for about 3 hours; t) holding between
`
`about 25°C and about 40°C for about 25 hours; g) ramping to about 20°C in 1 hour; h)
`
`unloading at about 20°C, at a pressure of 13.5 psi in a pharrnaceutically acceptable
`
`container that is hermetically sealed; wherein the pressure is about 150 microns
`
`throughout primary drying and 50 microns throughout secondary drying. An aspect of
`
`this cycle involves step (e) which is ramped to about 30-35°C for 3 hours and then
`
`ramped to 40°C for 5 hours. Another aspect of this embodiment is the lyophilized
`
`powered prepared from such lyophilization cycles.
`
`0009
`
`0009
`
`

`
`The invention also encompasses a pharmaceutical dosage form of bendamustine
`
`containing not more than about 0.5 % to about 0.9% HP1 (area percent of bendamustine)
`
`wherein said dosage form comprises a vial or other pharmaceutically acceptable
`
`container, wherein said HP1 is the amount of HP1 present pre-reconstitution or at time
`
`zero after reconstitution of said dosage form. Preferred concentrations of bendamustine
`
`include about 120 to about 500 mg/container, about 100 mg/container, about 5 mg to about
`
`2 g/container and about 170 mg/container.
`
`Another aspect of the invention is a lyophilized preparation of bendamustine that can be
`
`reconstituted in water at room temperature in under about 10 minutes to about 5 minutes.
`
`Another embodiment of the invention is a method for reducing the formation of
`
`bendamustine degradants in a lyophilized preparation by preparing a pre-lyophilization
`
`solution or dispersion of one or more organic solvents, wherein said solution comprises at
`least one stabilizing concentration of an organic solvent which reduces the level of
`
`degradation of bendamustine so that the amount of HP] produced during lyophilization
`
`from about 1 to 24 hours does not exceed about 0.5% to about 0.9% (area percent
`
`bendamustine) and said pre-lyophilization solution or dispersion is prepared using one or
`
`more means to reduce free radical production. Such means can include one or more of
`
`the following: 1) making the pre-lyophilization solution or dispersion at temperatures
`
`below about 40°C; 2) adding an antioxidant such as an excipient that has antioxidant
`
`properties, including mannitol; 3) preparing the pre-lyophilization or dispersion under
`
`gas; preparing the pre-lyophilization solution or dispersion under conditions devoid of
`
`ultraviolet light; and 4) the pre-lyophilization solution or dispersion in a container that
`
`blocks or reduces ultraviolet light.
`
`The present invention also includes pre-lyophilized pharmaceutical compositions of
`
`bendamustine. A preferred pre-lyophilized composition includes bendamustine HC1
`
`about 15 mg/mL, mannitol about 25.5 mg/mL, about 30% (v/v) tertiary-butyl alcohol,
`
`and water.
`
`These and other embodiments of the invention are described hereinbelow or are evident
`
`to persons of ordinary skill in the art based on the following disclosures.
`
`10
`
`0010
`
`0010
`
`

`
`BRIEF DESCRIPTION OF THE DRAWINGS
`
`Fig. 1 shows the solubility of bendamustine at various temperatures for two different
`
`solutions of bendamustine in tertiary butanol.
`
`Fig. 2 shows the purity results of an HPLC analysis after incubating bendamustine in
`
`various alcohols for 24 hours at 5°C. Results are presented as the area percent of the
`
`bendamustine peak.
`
`Fig. 3 shows HP1 (Formula II) formation after 24 hours in various alcohol/water co-
`
`solvents at 5°C
`
`Fig 4 shows dimer (Formula III) formation after 24 hours in various alcohol/water co-
`
`solvents at 5°C
`
`Fig. 5- shows a lyophilization cycle for bendamustine using a TBA/water cosolvent.
`
`Fig. 6 shows a chromatogram for Ribomustin® using HPLC method No.-1.
`
`DETAILED DESCRIPTION OF THE INVENTION
`
`Definitions:
`
`As used herein, the terms “formulate” refers to the preparation of a drug, e.g.,
`
`bendamustine, in a form suitable for administration to a mammalian patient, preferably a
`
`human. Thus, "formulation" can include the addition of pharmaceutically acceptable
`
`excipients, diluents, or carriers.
`
`Asused herein, the term “lyophilized powder” or “lyophilized preparation” refers to any
`
`solid material obtained by lyophilization, i.e., freeze-drying of an aqueous solution. The
`
`aqueous solution may contain a non-aqueous solvent and/or a solution composed of an
`
`aqueous and non-aqueous solvent(s).
`
`By “stable pharmaceutical composition” is meant any phannaceutical composition
`
`having sufficient stability to have utility as a pharmaceutical product. Preferably, a stable
`
`11
`
`0011
`
`0011
`
`

`
`pharmaceutical composition has sufficient stability to allow storage at a convenient
`
`temperature, preferably between -20°C and 40°C, more preferably about 2°C to about
`
`30°C, for a reasonable period of time, e.g., the shelf-life of the product which can be as
`
`short as one month but is typically six months or longer, more preferably one year or
`
`longer even more preferably twenty-four months or longer, and even more preferably
`
`thirty-six months or longer. The shelf-life or expiration can be that amount of time where
`
`the active ingredient degrades to a point below 90% purity. For purposes of the present
`
`invention stable pharmaceutical composition includes reference to pharmaceutical
`
`compositions with specific ranges of impurities asdescribed herein. Preferably, a stable
`
`pharmaceutical composition is one which has minimal degradation of the active
`
`ingredient, e.g., it retains at least about 85 % of undegraded active, preferably at least
`
`about 90 %, and more preferably at least about 95%, after storage at 2-30°C for a 2-3 year
`
`period of time.
`
`By “degraded” is meant that the active has undergone a change in chemical structure.
`
`The term "therapeutically effective amount" as used herein refers to that amount of the
`
`compound being administered that will relieve to some extent one or more of the
`
`symptoms of the disorder being treated. In reference to the treatment of neoplasms, a
`
`therapeutically effective amount refers to that amount which has the effect of (1)
`
`reducing the size of the tumor, (2) inhibiting (that is, slowing to some extent, preferably
`
`stopping) tumor metastasis, (3) inhibiting to some extent (that is, slowing to some extent,
`
`preferably stopping) tumor growth, and/or, (4) relieving to some extent (or, preferably,
`
`eliminating) one or more symptoms associated with the cancer. Therapeutically effective
`
`amount can also mean preventing the disease from occurring in an animal that may be
`
`predisposed to the disease but does not yet experience or exhibit symptoms of the disease
`
`(prophylactic treatment). Further, therapeutically effective amount can be that amount
`
`that increases the life expectancy of a patient afflicted with a terminal disorder. Typical
`
`therapeutically effective doses for bendamustine for the treatment of non-Hodgkin’s
`
`lymphoma can be from about 60-120 mg/m2 given as a single dose on two consecutive
`
`days. The cycle can be repeated about every three to four weeks. For the treatment of
`
`chronic lymphocytic leukemia (CLL) bendamustine can be given at about 80-100 mg/m2
`
`on days 1 and 2. The cycle can be repeated afier about 4 weeks. For the treatment of
`
`12
`
`0012
`
`0012
`
`

`
`Hodgkin’s disease (stages II-IV), bendamustine can be given in the “DBVBe regimen”
`
`with daunorubicin 25 mg/m2 on days 1 and 15, bleomycin 10 mg/m2 on days 1 and 15,
`
`vincristine 1.4 mg/m2 on days 1 and 15, and bendamustine 50 mg/m2 on days 1-5 with
`
`repetition of the cycle about every 4 weeks. For breast cancer, bendamustine (120
`
`mg/m2) on days 1 and 8 can be given in combination with methotrexate 40 mg/m2 on
`
`days 1 and 8, and 5-fluorouracil 600 mg/m2 on days 1 and 8 with repetition of the cycle
`
`about every 4 weeks. Asa second-line of therapy for breast cancer, bendamustine can be
`
`given at about 100-150 mg/m2 on days 1 and 2 with repetition of the cycle about every 4
`
`weeks.
`
`As used herein "neoplastic" refers to a neoplasm, which is an abnormal growth, such
`
`growth occurring because of a proliferation of cells not subject to the usual limitations of
`
`growth. As used herein, "anti-neoplastic agent" is any compound, composition,
`
`admixture, co-mixture, or blend which inhibits, eliminates, retards, or reverses the
`
`neoplastic phenotype of a cell.
`
`As used herein "hyperproliferation" is the overproduction of cells in response to a
`
`particular growth factor. "Hyperproliferative disorders" are diseases in which the cells
`
`overproduce in response to a particular growth factor. Examples of such
`
`"hyperproliferative disorders" include diabetic retinopathy, psoriasis, endometriosis,
`
`cancer, macular degenerative disorders and benign growth disorders such as prostate
`
`enlargement.
`
`As used herein, the term “vial” refers to any walled container, whether rigid or flexible.
`
`"Controlling" as used herein means putting process controls in place to facilitate
`
`achievement of the thing being controlled. For example, in a given case, "controlling"
`
`can mean testing samples of each lot or a number of lots regularly or randomly; setting
`
`the concentration of degradants as a release specification; selecting process conditions,
`
`e.g., use of alcohols and/or other organic solvents in the pre-lyophilization solution or
`
`dispersion, so as to assure that the concentration of degradants of the active ingredient is
`
`not unacceptably high; etc. Controlling for degradants by setting release specifications .
`
`for the amount of degradants can be used to facilitate regulatory approval of a
`
`13
`
`0013
`
`0013
`
`

`
`pharmaceutical product by a regulatory agency, such as the U.S. Food and Drug
`
`Administration and similar agencies in other countries or regions ("agency").
`
`The term "pharmaceutically acceptable” as used herein means that the thing that is
`
`pharmaceutically acceptable, e.g., components, including containers, of a pharmaceutical
`
`composition, does not cause unacceptable loss of pharmacological activity or
`
`unacceptable adverse side effects. Examples of pharmaceutically acceptable components
`
`are provided in The United States Pharmacopeia (USP), The National Formulary (NF),
`
`adopted at the United States Pharmacopeial Convention, held in Rockville, Md. in 1990
`
`and FDA Inactive Ingredient Guide 1990, 1996 issued by the U.S. Food and Drug
`
`Administration (both are hereby incorporated by reference herein, including any
`
`drawings). Other grades of solutions or components that meet necessary limits and/or
`
`specifications that are outside of the USP/NF may also be used.
`
`The term “pharmaceutical composition” as used herein shall mean a composition that is
`
`made under conditions such that it is suitable for administration to humans, e.g., it is
`
`made under GMP conditions and contains pharmaceutically acceptable excipients, e.g.,
`
`without limitation, stabilizers, bulking agents, buffers,_ carriers, diluents, vehicles,
`
`solubilizers, and binders. As used herein pharmaceutical composition includes but is not
`
`limited to a pre-lyophilization solution or dispersion as well as a liquid form ready for
`
`injection or infusion.
`
`A “pharmaceutical dosage form” as used herein means the pharmaceutical compositions
`
`disclosed herein being in a container and in an amount suitable for reconstitution and
`
`administration of one or more doses, typically about 1-2, 1-3, 1-4, 1-5, 1-6, 1-10, or about
`
`1-20 doses. A “pharmaceutical dosage form” -as used herein also means a lyophilized
`
`pharmaceutical composition disclosed herein in a container and in an amount suitable for
`
`reconstitution and delivery of one or more doses, typically about 1-2, 1-3, 1-4, 1-5, 1-6,
`
`1-10, or about 1-20 doses. The pharmaceutical dosage form can comprise a vial or
`
`syringe or other suitable pharmaceutically acceptable container. The pharmaceutical
`
`dosage form suitable for injection or infusion use can include sterile aqueous solutions or
`
`dispersions or sterile powders comprising an active ingredient which are adapted for the
`
`extemporaneous preparation of sterile injectable or infusible solutions or dispersions. In
`
`14
`
`0014
`
`0014
`
`

`
`all cases, the ultimate dosage form should be sterile, fluid and stable under the conditions
`
`of manufacture and storage. The liquid carrier or vehicle can be a solvent or liquid
`
`dispersion medium comprising, for example, water, ethanol, a polyol such as glycerol,
`
`propylene glycol, or liquid polyethylene glycols and the like, vegetable oils, nontoxic
`
`glyceryl esters, and suitable mixtures thereof. The prevention of the growth of
`microorganisms can be accomplished by various antibacterial and antifungal agents, for
`
`example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
`
`As used herein, the term "excipient" means the substances used to formulate active
`
`pharmaceutical ingredients (API) into pharmaceutical formulations; in a preferred
`
`embodiment, an excipient does not lower or interfere with the primary therapeutic effect
`
`of the API. Preferably, an excipient is therapeutically inert. The term "excipient"
`
`encompasses carriers, diluents, vehicles, solubilizers, stabilize_rs, bulking agents, and
`
`binders. Excipients can also be those substances present in a pharmaceutical formulation
`
`as an indirect or unintended result of the manufacturing process. Preferably, excipients
`
`are approved for or considered to be safe for human and animal administration, i.e.,
`
`GRAS substances (generally regarded as safe). GRAS substances are listed by the Food
`
`and Drug administration in the Code of Federal Regulations (CFR) at 21 CFR § 182 and
`
`21 CFR § 184, incorporated herein by reference. Preferred excipents include, but are not
`
`limited to, hexitols, including mannitol and the like.
`
`As used herein “a stabilizing concentration of an organic solvent” or “a stabilizing
`
`concentration of an alcohol” means that amount of an organic solvent or alcohol that
`
`reduces the level of degradation of bendamustine to achieve a specified level of
`
`degradants in the final drug product. For example, with respect to the degradant HP1, a
`
`stabilizing concentration of an organic solvent is that amount which results in an HP1
`
`concentration (area percent of bendamustine) of less than about 0.9 %, preferably less
`
`than 0.8 %, more preferably less than 0.7 % and even more preferably less than 0.6 %.
`
`With respect to the overall degradant concentration of the final drug product, a stabilizing
`
`concentration of an organic solvent is that amount that results in a total degradant
`
`concentration (at the time of drug product release) of less than about 8% (area percent
`
`bendamustine), preferably less than about 7 %, more preferably less than about 6%, and
`
`even more preferably less than about 5.5 %. By "area percent of bendamustine" is meant
`
`15
`
`0015
`
`0015
`
`

`
`the amount of a specified degradant, e.g., HPI , relative to the amount of bendamustine as
`
`determined, e.g., by HPLC.
`
`The term “organic solvent” means an organic material, usually a liquid, capable of
`
`dissolving other substances.
`
`As used herein, “trace amount of an organic solvent” means an amount of solvent that is
`
`equal to or below recommended levels for pharmaceutical products, for example, as
`
`recommended by ICH guidelines (International Conferences on Harmonization,
`
`Impurities-- Guidelines for Residual Solvents. Q3C. Federal Register.
`
`l997;62(247):67377). The lower limit is the lowest amount that can be detected.
`
`The term “at release” means the drug product has met the release specifications and can
`
`be used for its intended pharmaceutical purpose.
`
`A. General
`
`The invention provides stable, pharmaceutically acceptable compositions prepared from
`
`bendamustine. In particular, the invention provides formulations for the lyophilization of
`
`bendamustine HCl. The lyophilizedpowder obtained from such formulations is more
`
`easily reconstituted than the presently available lyophilized powder of bendamustine.
`
`F

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