throbber

`
` Parenteral Formulations of Small Molecules Therapeutics
`Part I
`Marketed in the United States (1999)
`Robert G. Strickley
`
`
`
` PDA J Pharm Sci and Tech
`
`1999
`
`53,
`
` 324-349
`
`MYLAN INST. EXHIBIT 1088 PAGE 1
`
`--
`

`

`REVIEW ARTICLE
`
`Parenteral Formulations of Small Molecules Therapeutics Marketed in
`the United States (1999)—Part I
`
`ROBERT G. STRICKLEY
`
`Axys Pharmaceuticals, Inc., South San Francisco, California
`
`Overview
`
`Introduction
`
`The chemical structure of a molecule determines the
`potential successful formulation approaches available to the
`parenteral scientist. However, there is no comprehensive
`listing of parenteral products with the chemical structure and
`formulation. A review of domestically marketed injectable
`product formulations of small molecule therapeutics is
`presented herein with the intent of compiling a comprehen-
`sive source of public information for the formulation
`scientist. The compilation lists the drug name, marketed
`name, chemical structure of the drug, marketed injectable
`formulation, preadministration preparation, route of admin-
`istration, company and the clinical indication (1–7).
`One purpose of this compilation is to assist the formula-
`tion scientist in being able to look at a drug’s chemical
`structure and then be able to determine possible formulation
`approaches. This compilation will also be useful for those
`interested in knowing what additives are currently used in
`injectable products and at what concentrations they are
`administered in practice. This compilation only focuses on
`marketed formulations and does not delve into the subject of
`preclinical or drug discovery formulations associated with
`early-stages pharmacokinetics or proof-of-concept pharma-
`codynamics, where the formulation scientist is not bound by
`regulatory constraints.
`There are a few published reviews on parenteral formula-
`tions (8) and in an excellent review article (9) Lilly
`scientists, Sweetana and Akers, discuss the various formula-
`tion approaches with detailed tables of examples. In a
`compendium of excipients for parenteral formulations (10)
`Genentech scientists, Powell, Nguyen and Baloian, list the
`acceptable excipients as well as their percent’s within the
`formulations, route of administration and pH. The compila-
`tion herein is an additional resource to the parenteral
`scientist by presenting the chemical structure and the
`formulation in a side-by-side fashion. An examination of
`this compilation reveals many examples of injectable formu-
`lation techniques to improve solubility or provide a sus-
`tained release. The next few sections highlight various
`formulation approaches with specific examples and tables,
`as well as general discussions of parenteral formulations.
`
`The word ‘‘parenteral’’ is Latin for ‘‘other than intestine,’’
`thus by definition the parenteral sciences not only includes
`injectable products but also transdermal, pulmonary, nasal,
`ophthalmic, and buccal routes of administration. However,
`in practice, parenteral usually refers to injectable products.
`Recently we have seen the commercialization of previously
`academic pursuits such as controlled-release formulations
`using microspheres, liposomes and polymeric gels, longer in
`vivo circulating times using PEGylated liposomes (also
`known as stealth liposomes) and PEGylated proteins, and
`new excipients such as cyclodextrin derivatives used as
`complexing agents for increasing water solubility of poorly
`soluble drugs. We have also seen the commercialization of
`injection devices such as prefilled syringes, dual chamber
`syringes containing solid drug and a liquid for reconstitu-
`tion, and will likely soon see needle-free injectors and
`pocket-size infusion pumps.
`
`Injectable Formulations
`
`Two key aspects of any successful injectable formulation
`are: 1) to achieve the required drug concentration, and 2) the
`drug must be chemically and physically stable in order to
`have a sufficient shelf-life, which is generally considered to
`be the time for 10% degradation. The ideal
`injectable
`formulation, from an in vivo tolerability point-of-view, is
`isotonic with physiological fluids and a neutral pH (i.e.,
`PBS: phosphate buffered saline, 0.01M sodium phosphate
`with 0.135M NaCl and 0.003M KCl, pH 7.4). However, in
`many instances the drug does not have sufficient water
`solubility at pH 7.4, and thus the formulation scientist must
`use a wide variety of solubilization techniques. If stability is
`insufficient to provide a two-year shelf-life, then the formu-
`lation scientist must either change the solution conditions to
`achieve both the solubility and stability requirements or
`develop a lyophilized product. This manuscript focuses on
`solubilization techniques for small molecules, and will not
`focus on stability or stabilization techniques.
`
`I. Solubilization Techniques
`
`1. pH Adjustment and Salts
`
`Editor’s Note: This review article on Injectable Products is being pub-
`lished in several parts. The next installment(s) will appear in subsequent
`issues of the Journal.
`Correspondence address: 180 Kimball Way, South San Francisco, CA
`94080.
`
`If the drug molecule is ionizable, then pH adjustment can
`be utilized to increase water solubility since the ionized
`molecular species has higher water solubility than its neutral
`species. Indeed, the most common solubilization technique
`is pH adjustment and weak acids are normally formulated at
`
`324
`
`PDA Journal of Pharmaceutical Science & Technology
`
`MYLAN INST. EXHIBIT 1088 PAGE 2
`
`

`

`pH . 5 (Table I), weak bases at pH , 7 (Table II).
`Zwitterionic molecules have multiple ionizable groups and
`can be either cationic, anionic or neutral (positive and
`negative charges cancel each other, for an overall net neutral
`molecule) and are usually formulated at a pH in which the
`drug is ionic (Table III). For example, both ciprofloxacin and
`sufentanil have a carboxylic acid and an amine, but are
`formulated as the cation at pH , 7. On the other hand, both
`ampicillin and cephapirin have a carboxylic acid and an
`amine or pyridine, but are formulated as the anion at pH . 5.
`
`The range in pH is quite broad and is between pH 2–12,
`and thus any molecule with a pKa between 3–11 can be
`potentially solubilized by pH adjustment. However, when
`using extremes in pH, care must be taken to minimize buffer
`capacity in order for the formulation to be in vivo compat-
`ible. When given intravenously, the formulation components
`are quickly diluted by the flow of blood and neutralized by
`the buffer capacity of blood. When given via intramuscular
`injection, the rate of dilution is reduced but rapid enough to
`still be able to inject in the range pH , 3–11. However,
`
`Vol. 53, No. 6 / November–December 1999
`
`325
`
`MYLAN INST. EXHIBIT 1088 PAGE 3
`
`

`

`when given subcutaneously the rate of dilution is reduced
`further with more potential for irritation at the injection site
`and thus the range is pH 3–6. For example, chlordiazepoxide
`is administered intravenously or intramuscularly and formu-
`lated at pH 3 with 20% propylene glycol and 4% TWEEN
`20. Phenytoin sodium is administered either intravenously
`or intramuscularly and formulated at pH 10–12 with 40%
`propylene glycol and 10% ethanol. Subcutaneous formula-
`
`tions are slightly acidic such as methadone at pH 3–6, and
`levorphanol at pH 4.3.
`Water-soluble salt forms (i.e., sodium salts of weak acids,
`or hydrochloride salts of weak bases) utilize the same
`principle of ionization, and are often the marketed form of
`the drug (Table IV). The most common cationic counterion
`is sodium which accounts for . 90% of the cations, and
`there are three meglumine salts, while only one salt each of
`
`326
`
`PDA Journal of Pharmaceutical Science & Technology
`
`MYLAN INST. EXHIBIT 1088 PAGE 4
`
`

`

`the cations potassium, tromethamine and calcium. There are
`many more anionic counterions and the most common is the
`hydrochloride salt followed by sulfate, mesylate, maleate
`and tartrate. When a salt is dissolved in non-buffered water,
`the resulting pH is generally ,2 pKa units away from the
`pKa, because protons are either added to (salt of a weak
`
`base) or taken away from water (salt of a weak acid). For
`example, gancyclovir is a weak acid with pKa2 5 9.4 and
`dissolving its sodium salt in water results in pH , 11.
`In order to maintain a desirable pH range, many formula-
`tions that utilize pH adjustment also use buffers to control
`pH (Table V). Buffers span the range of pH 2.5–11 and
`
`Vol. 53, No. 6 / November–December 1999
`
`327
`
`MYLAN INST. EXHIBIT 1088 PAGE 5
`
`

`

`include citrates, acetates, histidine, phosphate, tris(hydroxy-
`methyl)aminomethane, and carbonates. The buffer concen-
`tration must be high enough to maintain the desired pH, but
`must be balanced by in vivo tolerability considerations, and
`thus it is good practice to minimize buffer capacity of the
`administered formulation.
`
`2. Mixed Organic/Aqueous Formulations
`
`If pH adjustment alone is insufficient in achieving the
`desired solution concentration, then a combination of pH
`and organic solvent(s) is often employed. If the drug
`molecule is not ionizable then pH has no effect on solubility,
`
`328
`
`PDA Journal of Pharmaceutical Science & Technology
`
`MYLAN INST. EXHIBIT 1088 PAGE 6
`
`

`

`but solubility enhancement can often be accomplished by a
`combination of aqueous and organic solvents (i.e., a cosol-
`vent). The currently used organic solvents used in mixed
`organic/aqueous formulations are propylene glycol, ethanol,
`polyethylene glycol 300 or 400, cremophor EL, TWEEN 80,
`sorbitol, glycerin and dimethylacetamide (DMA) (Table VI).
`As with any formulation additive, the concentration that is
`administered should be minimized to avoid any in vivo
`complications such as local irritation or precipitation at the
`injection site. Many cosolvent formulations are marketed
`using rather high concentrations of organic solvent, and are
`usually but not always diluted prior to injection. For
`example, propylene glycol
`is 50% of the fenoldopam
`marketed formulation, but is diluted to ,1% for IV infusion.
`However, propylene glycol is ,70% of the oxytetracycline
`marketed formulation and is injected intramuscularly with-
`out dilution.
`Similar to formulations using pH adjustment, of the three
`main routes of administration (i.e., intravenous, intramuscu-
`lar and subcutaneous), the subcutaneous route has the most
`constraints when using cosolvent due to the reduced volume
`flow away from the injection site compared to intravenous
`and intramuscular. As a result, only three cosolvent products
`are administered subcutaneously and the amount of organic
`solvent is limited to ethanol 6% (dihydroergotamine), glyc-
`erin 32% (epinephrine), and propylene glycol 10% (hydrala-
`zine). Whereas, the intravenous bolus route can use ethanol
`up to 20% (paricalcitrol), PEG 300 up to 50% (methocarba-
`mil), and propylene glycol up to 68% (phenobarbitol). The
`intramuscular route has similar in vivo constraints to the
`intravenous route, but can tolerate even more organic
`solvent (see section I.3, Totally Organic Solution Formula-
`tions).
`Surfactant formulations seem to be on the increase with
`excipients Cremophor EL and TWEEN 80 leading the way.
`These formulations, in general, are supersaturated upon
`dilution and must be used soon after dilution into IV
`compatible fluids. For example, cremophor EL is 11% of the
`miconazole marketed formulation, but is diluted to 1% for
`IV infusion. Also, TWEEN 80 is 10% of the amiodarone
`marketed formulation, but is diluted to 0.4% for IV infusion.
`However, cremophor EL at 10% or TWEEN 80 at 25% can
`be administered by IV infusion (see section I.3).
`
`3. Totally Organic Solution Formulations
`
`Molecules that are non-ionizable (have pKa , 2, or
`pKa . 11) and non-polar are water insoluble with no effect
`of pH on solubility, and thus are the most challenging for the
`formulation scientist. These water-insoluble molecules can
`be formulated in 100% organic solvent, which is then
`usually but not always diluted prior to administration (Table
`VII). For example, busulfan is marketed in 33% dimethyl-
`acetamide and 67% PEG 400, but is diluted 10-fold prior to
`IV infusion. The lorazepam marketed formulation is 80%
`propylene glycol, 18% ethanol and 2% benzyl alcohol, but is
`diluted 2-fold for IV bolus injection, but not diluted for
`intramuscular injection. Paclitaxel is marketed with 51%
`cremophor EL and 49% ethanol, but is diluted 5- to 20-fold
`for IV infusion. Docetaxel is marketed in 100% TWEEN 80,
`but is diluted to 25% for IV infusion.
`
`4. Cyclodextrins
`
`Some molecules can be solubilized by forming an inclu-
`sion complex with a cyclodextrin. Cyclodextrins have a
`hydrophilic exterior and a hydrophobic interior core of
`specific dimensions, and thus molecules with a non-polar,
`aromatic or heterocyclic ring can potentially fit inside the
`core. Increased water solubility through molecular complex-
`ation with cyclodextrins has advantages over the cosolvent
`approach since upon dilution a 1:1 complex between cyclo-
`dextrin and drug will not precipitate, but a drug dissolved in
`a cosolvent often precipitates upon dilution. Two cyclodex-
`trins have been accepted for human injectable use with the
`approval of alprostidol alfadex and itraconazole. Alprostidol
`alfadex is marketed as a lyophilized powder with a-cyclodex-
`trin and is administered intracavernosally. Itraconazole was
`approved in April 1999 as a solution with 40% hydroxypro-
`pyl-b-cyclodextrin and is administered by intravenous infu-
`sion after a 2-fold dilution with saline (6). The next
`cyclodextrin likely to be approved is sulfobutylether-b-
`cyclodextrin, which is in the clinical formulation of ziprasi-
`done for intramuscular injection (11).
`
`5. Emulsions
`
`Oil-soluble molecules are generally neutral uncharged
`and non-polar molecules, but can be formulated for intrave-
`nous administration by the use an oil-in-water emulsion.
`Emulsions can solubilize oil-soluble drugs since the drug
`partitions into the oil phase. A typical emulsion is composed
`of water with 10–20% soybean and/or safflower oil, 2%
`glycerol, 1% egg lecithin and pH 7–8, and is injected by
`either IV bolus or IV infusion. The only marketed emulsion
`formulation is propofol, which is in a typical emulsion
`composed of 10% soybean oil containing 10 mg/mL drug.
`The total parenteral nutrition (TPN) formulations are the
`lipid emulsions Intralipid and Liposyn, which are adminis-
`tered by intravenous infusion as nutritional supplements.
`
`6. Prodrugs
`
`Molecules which contain an alcohol, phenol, carboxylic
`acid, amine, hydantoin functional group can potentially be
`derivatized as a prodrug. Once the prodrug is administered
`in vivo, the promoiety is hydrolyzed by either esterases or
`phosphatases releasing the parent drug. Although prodrugs
`are normally associated with orally administered products
`for better oral bioavailability, many parenteral products are
`prodrugs (Table VIII).
`The versatility of the prodrug approach is demonstrated
`with prodrugs that in design either increase or decrease
`water solubility. A water-soluble prodrug has an electroni-
`cally charged promoiety, while a water insoluble prodrug
`has been derivatized to be a neutral molecule (see section
`II.7b). Recently, a few water-soluble phosphate ester pro-
`drugs have been developed and marketed in order to replace
`the original formulations that contain high concentrations of
`organic solvent. The phenol-containing etoposide (etoposide
`phosphate) is derivatized as a water-soluble phosphate ester.
`Water-soluble phosphate esters are also prodrugs for alcohol-
`containing betamethasone, clindamycin, dexamethasone,
`fludarabine, hydrocortisone, and prednisolone. The hydan-
`
`Vol. 53, No. 6 / November–December 1999
`
`329
`
`MYLAN INST. EXHIBIT 1088 PAGE 7
`
`

`

`toin-containing phenytoin prodrug (fosphenytoin) is deriva-
`tized in a unique fashion as a water-soluble hydroxymethyl
`phosphate ester, which after in vivo enzymatic phosphate
`ester cleavage, the resulting hydroxymethyl intermediate
`quickly dissociates to phenytoin and formaldehyde (12).
`Other water solubilizing prodrug approaches are a succinate
`ester of the alcohol methylprednisolone, and a piperidine
`carbamate in irinotecan a prodrug for a phenol drug.
`Prodrugs can also be used for stability reasons. For
`example, alatrofloxacin is the alanine-alanine dipeptide
`prodrug for the primary amine trovafloxacin which is
`
`unstable in solution. The prodrug alatrofloxacin is marketed
`as a solution at pH 3.4–4.3.
`
`II. Sustained-Release Techniques
`
`The research in controlled release during the 1970s has in
`the 1990s become a commercial realization with the ap-
`proval of liposomal, polymeric microsphere and polymeric
`gel formulations. However, traditional approaches are still in
`use such as suspensions, prodrugs and oil depots.
`
`330
`
`PDA Journal of Pharmaceutical Science & Technology
`
`MYLAN INST. EXHIBIT 1088 PAGE 8
`
`

`

`7a. Suspension Formulations
`
`Suspension formulations provide a sustained-release de-
`pot at the injection site that releases prodrug by dissolution.
`Suspensions used for sustained delivery are composed of a
`drug dispersion in either an aqueous or oil-based suspension
`(Table IX).
`Almost all suspensions are administered intramuscularly,
`intralesionally or intra-articularly. The only subcutaneously
`administered suspension of a small molecule (many proteins
`are administered subcutaneous, e.g., human insulin) is
`epinephrine, which is administered every 6 hours and is
`formulated in 32% glycerin providing both rapid (drug in
`solution) and sustained activity (crystalline drug in suspen-
`sion). The only sesame oil suspension is the anti-rheumatic
`
`aurothioglucose, which is administered intramuscularly ev-
`ery 1–4 weeks.
`
`7b. Prodrugs in Suspension Formulations
`
`Most of the other suspension formulations are aqueous-
`based and contain water-insoluble prodrugs which are
`lipophilic esters of alcohols. For example, hydrocortisone
`acetate and dexamethasone acetate are acetate esters of their
`alcohol-containing parent drug, and are administered intra-
`muscularly, intralesionally or intra-articularly once every
`1–3 weeks. The contraceptive medroxyprogesterone acetate
`is administered intramuscularly once every 13 weeks. Aque-
`ous-based suspensions typically contain TWEEN 80 at
`,0.75–4 mg/mL (0.4%) along with a suspending agent such
`
`Vol. 53, No. 6 / November–December 1999
`
`331
`
`MYLAN INST. EXHIBIT 1088 PAGE 9
`
`

`

`332
`
`PDA Journal of Pharmaceutical Science & Technology
`
`MYLAN INST. EXHIBIT 1088 PAGE 10
`
`

`

`Vol. 53, No. 6 / November–December 1999
`
`333
`
`MYLAN INST. EXHIBIT 1088 PAGE 11
`
`

`

`334
`
`PDA Journal of Pharmaceutical Science & Technology
`
`MYLAN INST. EXHIBIT 1088 PAGE 12
`
`

`

`Vol. 53, No. 6 / November–December 1999
`
`335
`
`MYLAN INST. EXHIBIT 1088 PAGE 13
`
`

`

`336
`
`PDA Journal of Pharmaceutical Science & Technology
`
`MYLAN INST. EXHIBIT 1088 PAGE 14
`
`

`

`Vol. 53, No. 6 / November–December 1999
`
`337
`
`MYLAN INST. EXHIBIT 1088 PAGE 15
`
`

`

`338
`
`PDA Journal of Pharmaceutical Science & Technology
`
`MYLAN INST. EXHIBIT 1088 PAGE 16
`
`

`

`Vol. 53, No. 6 / November–December 1999
`
`339
`
`MYLAN INST. EXHIBIT 1088 PAGE 17
`
`

`

`340
`
`PDA Journal of Pharmaceutical Science & Technology
`
`MYLAN INST. EXHIBIT 1088 PAGE 18
`
`

`

`as sodium carboxymethylcellulose at ,5 mg/mL (i.e.,
`dexamethasone acetate), PEG 3350 at 30 mg/mL (i.e.,
`methylprednisolone acetate) or sorbitol at 50% (i.e., triam-
`cinolone hexacetonide).
`
`8. Depots
`
`Sesame oil formulations of oil-soluble drugs provide a
`sustained-release depot at the injection site that releases drug
`by diffusion-like uptake of oil. For example, the prodrugs
`haloperidol deconate and testosterone enanthate are formu-
`lated in 100% sesame oil and administered intramuscularly
`once a month.
`
`9. Liposomes
`
`An exciting new era of the parenteral sciences began with
`the approval of liposomal products. A liposome is a lipid
`bilayer and an aqueous-based multilayered spherical drug
`delivery system where the drug is encapsulated inside the
`liposome, and is released as the liposome is eroded in vivo. A
`typical liposome formulation contains water with lipid at ,5
`mg/mL, an isotonicifier, a pH 5–8 buffer, and with or without
`cholesterol. These liposomes are injected either by IV
`infusion or intrathecally. Table X lists the six currently
`available liposomal products of the four drugs amphotericin
`B (3 liposome formulations), cytarabine, daunorubicin and
`doxorubicin. The amphotericin B liposomal products are
`administered by IV infusion and have an in vivo elimination
`half-life of 40–150 hours. The daunorubicin liposomal
`formulation has an in vivo half-life of 4.4 hours compared to
`0.8 hours for the conventional formulation (1, pg. 1970). The
`cytarabine liposomal formulation, Depocyt, is administered
`intrathecally once every 2 weeks, while the conventional
`formulation is given twice per week.
`To further increase the in vivo circulating times, lipo-
`somes can be covalently derivatized with polyethylenegly-
`col to produce PEGylated or stealth liposomes. The only
`commercially available PEGylated liposome is doxorubicin
`in Doxil and is administered by IV infusion and has a
`half-life of 50–55 hours (1, pg. 2985). The proteins adeno-
`sine deaminase (Adagen) and asparginase (Oncaspar) are
`also available as a PEGylated derivative.
`
`10. Polymeric Microspheres
`
`The era of controlled release using polymeric micro-
`spheres began with the approval of the peptide leuprolide as
`lupron depot. The drug is incorporated into a biocompatible
`polymer and transformed into lyophilized microspheres
`during the manufacturing process. The reconstituted micro-
`spheres are injected intramuscularly and slowly erode in
`vivo, releasing the drug. In the marketed formulation, leupro-
`lide is in lyophilized microspheres with DL-lactic/glycolic
`acid copolymer (PLGA), gelatin and mannitol, which is then
`reconstituted prior to administration to a suspension using an
`aqueous solution of sodium carboxymethylcellulose, TWEEN
`80 and mannitol. The microspheres provide a depot of drug
`and are administered once every 1–4 months, depending on
`the dose (3.75 mg/l month, 30 mg/4 months). One of the
`leuprolide formulations uses a dual chamber syringe for ease
`of reconstitution and administration.
`
`A polymeric PLGA microsphere formulation of human
`growth hormone (Nutropin Depot) finished Phase III clinical
`trials in 1999 (13). In this formulation, human growth
`hormone is made into an insoluble complex with zinc, and
`encapsulated into PLGA microspheres in a non-aqueous
`cryogenic process (14). The resulting free-flowing powder is
`reconstituted to a suspension prior to subcutaneous or
`intramuscular administration.
`
`11. Polymeric Gels
`
`Polymeric gels provide a depot of drug that is released
`over 1–4 weeks. The era of controlled release using poly-
`meric gels began with the approval of doxycycline hyclate
`which is available as a 7-day controlled-release system that
`is a solution upon subgingival administration, but solidifies
`upon contact with the crevicular fluid. This product
`is
`marketed as Atridoxt in a Atrigel Delivery System which is
`a two-syringe set-up where syringe A contains the polymer
`poly(DL-lactide) dissolved in N-methyl-2-pyrrolidone, and
`syringe B contains solid doxycycline. Upon coupling the
`two syringes, the liquid in syringe A is injected into syringe
`B and repeatedly mixed to complete dissolution, and then
`the yellow viscous liquid is administered subgingivally.
`Local delivery directly into tumors of the anti-tumor
`cancer drugs fluorouracil and cisplatin, as well a subcutane-
`ous injection of leuprolide are in clinical trials using a
`polymeric gel formulation.
`
`III. Containers/Vials
`
`Most injectable products are still marketed in traditional
`vials, ampules and infusion bags. However, there is in-
`creased use of more convenient containers such as prefilled
`syringes, dual chamber syringes and pen-type injectors.
`Prefilled syringes are especially useful in emergency situa-
`tions such as in the use of the antithrombotics dalteparin,
`danaparoid and enoxaparin; the analgesics morphine, hydro-
`morphone, fentanyl, lidocaine and sumatripan; the sedatives
`lorazepam and propofol; and the antihypertensive labetalol.
`Dual chamber syringes are used to avoid the usual manipula-
`tions involved in reconstitution of a lyophilized powder, and
`one syringe contains the solid drug while the second syringe
`contains the liquid diluent, which are mixed prior to
`administration. Products that use a dual chamber set-up
`include diltiazem, doxycycline and leuprolide. Pen-type
`injectors such as NovoPent with insulin involve a 1–3 mL
`cartridge that goes into the pen-like delivery device, and the
`epinephrine autoinjector for intramuscular self-administra-
`tion.
`
`IV. Future
`
`The future is promising for the formulation sciences, in
`general, and also for the parenteral formulation sciences.
`New parenteral achievements will likely include targeted
`delivery, more sophisticated controlled delivery, novel for-
`mulations and new excipients, which may utilize new
`technologies and be marketed in new devices. Biotechnol-
`ogy proteins and antibodies will likely continue to be at the
`forefronts of the parenteral sciences. The new and exciting
`field of gene therapy will likely rely on injectable and
`
`Vol. 53, No. 6 / November–December 1999
`
`341
`
`MYLAN INST. EXHIBIT 1088 PAGE 19
`
`

`

`342
`
`PDA Journal of Pharmaceutical Science & Technology
`
`MYLAN INST. EXHIBIT 1088 PAGE 20
`
`

`

`Vol. 53, No. 6 / November–December 1999
`
`343
`
`MYLAN INST. EXHIBIT 1088 PAGE 21
`
`

`

`344
`
`PDA Journal of Pharmaceutical Science & Technology
`
`MYLAN INST. EXHIBIT 1088 PAGE 22
`
`

`

`Vol. 53, No. 6 / November–December 1999
`
`345
`
`MYLAN INST. EXHIBIT 1088 PAGE 23
`
`

`

`346
`
`PDA Journal of Pharmaceutical Science & Technology
`
`MYLAN INST. EXHIBIT 1088 PAGE 24
`
`

`

`Vol. 53, No. 6 / November–December 1999
`
`347
`
`MYLAN INST. EXHIBIT 1088 PAGE 25
`
`

`

`solution formulations for delivery of antisense oligonucleo-
`tides (15, 16), such as with the anti-sense ophthalmic
`product fomiversen (Vitravene). In general, formulation
`approaches along with drug design will be the means to
`achieve optimal drug delivery based upon therapeutic needs.
`New approaches could include nanoparticles (17), submi-
`cron solid particles coated with either natural or semisyn-
`thetic phospholipids (18), mixed-micelles, microemulsions
`for injection (19), and soluble self-assembled block copoly-
`mers to either solubilize drug in a micelle-like structure
`[PEO-b-PAA-DOX, poly(ethylene oxide)-block-poly(aspar-
`tic acid)-doxorubicin] or covalently bind drug (20). ‘‘Smart’’
`controlled-release systems that deliver drug when needed
`could be the next generation in controlled release, including
`pulsatile delivery to mimic human circadian rhythms or
`normal hormone production. The release of drug could be
`triggered by timed events or more sophisticated means, such
`as a chemical stimulus, photosensors, blood pressure sen-
`sors, or some type of biofeedback mechanism. New excipi-
`ents will
`likely be approved, such as sulfobutyl ether
`b-cyclodextrin, tetraglycol, triglyme, transcutol, 2-pyrrol-
`idone (Soluphort P), glycerol formal, Solutol HS-15, and
`poloxamers which will expand the number of formulation
`additives available to the formulation scientist.
`Devices such as needle-free injectors (already in use with
`vaccines) for both solutions and solids (21) could revolution-
`ize the manner in which injectable drugs are administered.
`The increased emphasis on home health care will likely
`result in home infusion devices and set-ups such as battery
`operated and/or pocket-sized infusion pumps. We are likely
`to continue to see more applications of convenient injection
`devices, prefilled syringes, dual chamber devices and ready-
`to-use solutions.
`
`Advanced technologies will likely be used in commercial
`production of future parenteral products; for example, the
`use of nanoparticles for injection of water-insoluble drugs.
`Supercritical fluid processing to form spherical micropar-
`ticles (22) and perhaps a designed distribution of particle
`size has tremendous potential in future formulations and
`pharmaceutical manufacturing.
`Combinations of novel formulations and novel delivery
`systems that are in active research (23) will certainly be
`developed. One can imagine the many combinations of
`needle-free injection of solutions or solids, controlled-
`release systems, ‘‘stealth’’ carriers, targeted delivery, vac-
`cines, gene therapy, antibodies and specially designed small
`molecules. Yes, as the parenteral sciences continue to
`mature, future products will be science fiction come true!
`
`Notes on the Compilation
`
`A few comments on the compilation are in order to help
`the reader understand the table format, chemical structures,
`some occasional additional information, highlighted por-
`tions, and abbreviations.
`
`1) The order of lines within the formulation box is:
`a) Solution or lyophilized powder
`b) Drug concentration or amount (i.e., mg/mL, mg,
`units/mL, etc.)
`c) Excipients and concentration or amount (i.e.,
`mg/mL, %, mg, etc.)
`—organic solvent(s)
`—suspending agent(s)
`—bulking agent(s)
`—isotonicifier(s)
`
`348
`
`PDA Journal of Pharmaceutical Science & Technology
`
`MYLAN INST. EXHIBIT 1088 PAGE 26
`
`

`

`—preservative(s)
`—buffer
`d) pH
`2) Some drugs have the pKa listed, but this is not compre-
`hensive and is added for informative purposes.
`3) The chemical structures are drawn in most instances as
`the neutral species even though the market product
`may be a salt form.
`4) In the drug name, the counter ion is in lower case, but a
`covalently bound prodrug moiety is capitalized.
`5) Some entries were not found in the 1999 PDR at all or
`not as injectables, but were found in other references.
`In these cases ‘‘(Not in 1999 PDR)’’ is added under the
`marketed name.
`6) Various portions of some entries are highlighted in
`bold typeface, in order to help the reader clearly notice
`key formulation aspect(s).
`7) Some drugs are marketed in multiple formulations,
`and in these cases the formulations are numbered.
`8) There are some peptide entries to highlight new formu-
`lation approaches.
`9) Abbreviations used herein (Table XI).
`
`References
`
`1. Physician’s Desk Reference, 53rd ed., Medical Economics Company,
`Inc., Montvale, NJ, 1999.
`2. Lawrence A. Trissel, Handbook on Injectable Drugs, 10th ed., Ameri-
`can Society of Health-System Pharmacists, Inc., Bethesda, MD, 1998.
`3. Lynn D. Phillips and Merrily A. Huhn. Manual of IV Medications, 2nd
`ed., Lippincott-Raven Publishers, Philadelphia, PA, 1999.
`4. D. A. Hussar, ‘‘New drugs of 1998,’’ Journal of the American Pharma-
`ceutical Association, 39, 151 (1999).
`5. D. A. Hussar, ‘‘New drugs of 1997,’’ Journal of the American Pharma-
`ceutical Association, 38, 155 (1998).
`6. Food and Drug Administration internet website, http://www.fda.gov/,
`See human drugs, then what’s new, then new and generics approvals.
`7. A. Depalma, Managing editor, http://pharmaceuticalonline.com/, Weekly
`update on applications and approvals.
`
`8. Y-C J. Wang and R. R. Kowal, ‘‘Review of excipients and pHs for
`parenteral products used in the United States,’’ J. Parent. Sci. Technol.,
`34, 452 (1980).
`9. S. Sweetana and M. J. Akers, ‘‘Solubility principles and practices for
`parenteral dosage form development,’’ PDA J. Parent. Sci. Technol.,
`50, 330 (1996).
`10. M. F. Powell, T. Nguyen, and L. Baloian, ‘‘Compendium of excipients
`for parenteral formulations,’’ PDA J. Parent. Sci. Technol., 52, 238
`(1998).
`11. Y. Kim, D. A. Oksanen, W. Massrfski, J. F. Blake, E. M. Duffy, and B.
`Chrunyx, ‘‘Inclusion complexation of ziprasidone mesylate with b-cy-
`clodextrin sulfobutyl ether,’’ Journal of Pharmaceutical Sciences, 87,
`1560 (1998).
`12. V. J. Stella, ‘‘A case for prodrugs: Fosphenytoin,’’ Adv. Drug Del. Rev.,
`19, 331 (1996).
`13. A. Depalma, ‘‘Alkermers and genentech expand collaboration for
`nutropin depot,’’ feature article, April 19, 1999, http://pharmaceuticalon-
`line.com
`14. O. L. Johnson, W. Jaworowicz, J. L. Cleland, L. Bailey, M. Charnis, E.
`Duenas, C. Wu, D. Shepard, S. Magil, T. Last, A. J. S. Jones, and S. D.
`Putney, ‘‘The stabilization and encapsulation of human growth hor-
`mone into biodegradable microspheres,’’ Pharmaceutical Research,
`14, 730 (1997).
`15. David A. Putnam, ‘‘Antisense strategies and therapeutic applications.’’
`Am. J. Health-Syst. Pharm., 53, 151 (1996).
`16. R. L. Juliano, S. Alahari, R. Kole, and M. Cho, ‘‘Antisense pharmaco-
`dynamics: Critical issues in the transport and delivery of antisense
`oligonucleotides,’’ Pharmaceutical Research, 16, 494 (1999).
`17. G. Caponetti, J. S. Hrkach, B. Kriwet, M. Poh, N. Lotan, P. Colombo,
`and R. Langer, ‘‘Microparticles of novel branched copolymers of lactic
`acid and amino acids: Preparation and characterization,’’ Journal of
`Pharmaceutical Sciences, 88, 136 (1999).
`18. S. N. Pace, G. W. Pace, I. Parikh, and A. K. Mishra, ‘‘Novel

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