throbber

`
`Downloaded from Downloaded from on May 5, 2017journal.pda.org on May 5, 2017journal.pda.org
`
`
`
`
`
`
`
`
`
`Solubility Principles and Practices for Parenteral Drug
`Dosage Form Development
`Stephanie Sweetana and Michael J. Akers
`
`PDA J Pharm Sci and Tech
`
`
`
`1996
`
`50,
`
` 330-342
`


`  
`

`

`
`
`Downloaded from on May 5, 2017journal.pda.org
`
`
`
`REVIEW ARTICLE
`
`Solubility Principles and Practices for Parenteral Drug Dosage
`Form Development
`
`STEPHANIE SWEETANA and MICHAEL J. AKERS*
`
`Pharmaceutical Sciences, Lilly Research Laboratories, Indianapolis, Indiana
`
`Introduction
`A common problem experienced in the early develop(cid:173)
`ment of drugs intended for parenteral, especially intrave(cid:173)
`nous, administration is the solubilization of a slightly
`soluble or water insoluble active ingredient. Drug solubi(cid:173)
`lization has been the subject of many scientific articles
`and textbooks (referenced throughout this article); yet
`despite this attention and available literature, product
`development scientists still encounter significant difficul(cid:173)
`ties in solving their solubility problems.
`Theories of solute solubilization are not easy to
`understand. Solubilization processes are amazingly com(cid:173)
`plex and require a fair amount of expertise in physical
`chemistry to interpret and apply current theoretical
`models. Much of the literature deals with solubilization
`theory and does not offer much practical help to the
`inexperienced scientist under a lot of pressure to find a
`solution to his/her solubility problem.
`This article intends to help the scientist in early drug
`formulation design for parenterally administered drug
`products by reviewing pertinent literature on solubiliza(cid:173)
`tion and reducing it to simple approaches one can use to
`solve solubility problems. The classical theories of solu(cid:173)
`bility, and how they relate to pharmaceutical systems of
`interest will be reviewed and practical applications
`discussed. Because of the common concerns regarding
`cosolvent toxicity and acceptability by medical and
`regulatory bodies, we also will treat this topic in some
`detail.
`
`I. Pertinent Theory of Solubilization of Drugs
`
`Solubility theories deal with conversion of a substance
`from one state to another, and the equilibrium phenom(cid:173)
`ena that are involved. Through pioneering work of
`Henry, Raoult and van't Hoff in the late 1800's, the
`properties of various solutions have been defined in
`theories. These early theories form the basis by which
`more complex systems, such as those encountered in the
`biological sciences, are compared and understood.
`No single theory can adequately explain solubility
`behavior of uncharged molecules in a variety of solvent
`systems. Each theory is suited for select combinations of
`
`Received June 29, 1995. Accepted for publication March 21, 1996.
`* Author to whom correspondence should be addressed: Lilly Corpo(cid:173)
`rate Center, Indianapolis, IN 46285.
`
`solutes and solvents where certain intermolecular forces
`are assumed to predominate, or conversely, be absent.
`The classical theories of solubility have been explained
`most simply in terms of intermolecular interactions.
`Ideal solution theory assumes solute-solute, solvent-
`solvent and solute-solvent interactions are completely
`uniform in strength and nature. An example of a
`solution behaving ideally is a non-polar solute in a
`non-polar solvent such as naphthalene in benzene.
`Regular solution theory evolved to account for the imbal(cid:173)
`ance of intermolecular interactions that often occur
`between dissimilar systems of a solute and solvent. The
`focus of this theory are systems of low polarity such as
`steroids in hydrocarbon solvents. Extended regular solu(cid:173)
`tion theory incorporated additional parameters such as
`dispersion, polar and hydrogen-bonding interactions
`into regular solution theory. Various approaches have
`been used to represent these molecular interactions,
`leading to a variety of models to predict and explain
`solubility behavior of polar solutes in polar systems, each
`with different approximations and assumptions (1–4).
`In most pharmaceutical systems, the routine applica(cid:173)
`tion of these models to predict solubility and simplify
`formulation development is complex. Most drugs of
`interest are ionizable, contain polar polyfunctional
`groups, and are capable of forming multiple hydrogen
`bonds. The majority of parenterally acceptable cosol¬
`vents—such as propylene glycol, polyethylene glycol,
`ethanol and water—are capable of self association
`through hydrogen bond formation. Such interactions
`may alter solvent structure and, as a result, influence
`solubility in an unpredictable manner (1). Examples of
`this phenomena are deviations from log-linear solubliza¬
`tion of nonpolar solutes in a polar cosolvent system (5).
`For the models to adequately describe solubility behav(cid:173)
`ior, proper weighting must be assigned to the relative
`importance of competing self-associations and strong
`intermolecular interactions. Currently this is being mod(cid:173)
`eled by various computer intensive group-contribution
`approaches, some of which allow for the mutual interac(cid:173)
`tions of various functional groups (1).
`In the biological sciences, many solutes of interest are
`capable of acting as acids or bases. In an ionizing media
`such as water, they may dissociate into ions which are
`usually highly water soluble. To what extent a molecule
`is ionized in an aqueous solution is largely dependent on
`its pKa and the pH of the media. The Henderson-
`
`330
`
`PDA Journal of Pharmaceutical Science & Technology
`
`

`

`
`
`Downloaded from on May 5, 2017journal.pda.org
`
`
`
`Hasselbalch equation is a mathematical expression of
`this relationship (3). In formulation development, con(cid:173)
`sideration of the amount of un-ionized drug in solution
`is helpful to avoid unexpected precipitation of this form.
`As the pH of a drug solution is changed, the amount of
`free acid or base may increase and eventually exceed the
`limited solubility of this form. It is possible to calculate
`the pH of precipitation and of maximum solubility, if the
`pKa of the molecule and the solubility of the un-ionized
`and ionized forms are known (3, 6). Generally, two pH
`units above or below the pHmax value establishes the
`desired pH for formulation. For drug molecules with
`multiple ionizable groups these equations are more
`complicated to apply and so experimentally generated
`solubility data are usually collected.
`Through our own experience, we find that theory gives
`us some direction with respect to experimental ap(cid:173)
`proaches, but we still need to rely on the empirical
`experimentation to screen for systems which offer the
`most promise in solubilizing water-insoluble drugs.
`
`II. Formulation Design
`
`Usually, the first approach used to increase the
`solubility of an insoluble drug in water is to form more
`water soluble salts. Berge and co-workers (7) wrote what
`is now a near classic review of salt form strategies
`acceptable for pharmaceuticals. If salt formation is not
`possible, e.g. too unstable, or does not render the
`molecule sufficiently water soluble, a series of formula(cid:173)
`tion approaches may be investigated. Table I summa(cid:173)
`rizes these general strategies. Often a useful approach to
`increase the aqueous solubility of an ionizable drug is
`pH adjustment. The next approach most frequently tried
`is the use of water-miscible cosolvents. Other ap(cid:173)
`proaches to be discussed briefly include the use of
`surface active agents and complexing agents. Develop(cid:173)
`ment of emulsified and colloidal drug delivery systems
`for intravenous administration are becoming more widely
`and successfully applied. They may confer to the en(cid:173)
`trapped or associated drug significantly different proper-
`
`TABLE I
`Summary of Parenteral Formulation Approaches
`
`Approach
`
`Examples
`
`pH adjustment
`
`pH 2 to 12
`
`Cosolvent
`
`Polyethylene glycol
`Propylene glycol
`Ethanol
`Dimethylacetamide
`
`Surface Active Agents
`
`Polysorbates
`Poloxamers
`Cremophor EL
`Lecithin
`Bile salts
`
`Complexing Agents
`
`Cyclodextrans
`Water-soluble vitamins
`
`Dispersed Systems
`
`Emulsions
`Liposomes
`Nanoparticles
`
`Important Formula
`Considerations
`
`Drug stability
`pH
`ions to buffer or adjust pH
`Drug precipitation upon infusion
`drug concentration
`use of buffer/buffer capacity
`infusion rate
`Formula irritation
`isotonicity
`infusion rate & duration
`drug vs vehicle
`drug precipitation
`
`Systemic toxicity
`total cosolvent administered
`Drug precipitation upon infusion
`drug concentration
`infusion rate
`Formula irritation
`isotonicity
`infusion rate & duration
`drug vs vehicle
`drug precipitation
`
`Hypersensitivity in animals
`Formula irritation
`isotonicity
`infusion rate & duration
`drug vs vehicle
`
`Purity of excipients and drugs
`Formula irritation
`isotonicity
`infusion rate & duration
`drug vs vehicle
`
`Sterility
`Particle size
`Pharmacokinetics
`Stability
`
`Useful Tests
`
`pH rate profile
`pH solubility profile
`Freezing point depression
`In vitro precipitation model
`In vivo phlebitis model
`In vitro cell lysis studies
`
`Mixture studies for maximum
`solubility
`In vitro precipitation model
`In vivo phlebitis model
`In vitro cell lysis studies
`
`In vivo phlebitis model
`In vitro cell lysis studies
`
`Phase solubility diagrams
`In vivo phlebitis model
`In vitro cell lysis studies
`
`Particle size
`
`Vol. 50, No. 5 / September–October 1996
`
`331
`
`

`

`
`
`Downloaded from on May 5, 2017journal.pda.org
`
`
`
`ties from the free form, providing the opportunity to
`prolong drug presence in the bloodstream or to alter
`disposition in the body. "Heroic" methods, reported in
`the literature for various cancer drugs, will also be
`reviewed although these methods use types and amounts
`of excipients that probably would not commonly be
`considered approvable for intravenous administration.
`The basis for reliable formulation development is
`accurate determination of solubility. Traditional method­
`ology is the "equilibrium method" (8) where excess drug
`is added to the solvent system, and some means of
`agitation
`is employed under constant
`temperature.
`Samples are withdrawn, filtered, and analyzed for drug
`concentration over a period of time and equilibration is
`demonstrated by uniformity of the data over the time
`interval. For sparingly soluble drugs where equilibria are
`slow, accurate determinations of solubility may be diffi­
`cult. Useful techniques in these instances include using
`highly specific analytical methods to detect parent com­
`pounds, minimizing the amount of excess solid added,
`and assuring sufficient equilibration time (1). Solid state
`factors and batch-to-batch variation (different poly­
`morphs, hydration state, crystallinity, crystal homogene­
`ity, and impurities) may affect reproducibility of drug
`solubility determinations.
`
`A. pH Adjustment
`
`Current FDA approved marketed parenteral prod­
`ucts range in pH from 2 to 11. A comprehensive listing of
`these products may be found in Table II. For biocompat-
`ability reasons, formulation of injectables within the pH
`ranges of 4 to 8 is most common. However, to achieve
`sufficient drug solubility, a pH outside this range may be
`necessary.
`The pH at which a product is formulated is usually
`determined from the pH solubility and pH rate profiles
`of the drug (9). A recent example of their application to
`aid parenteral formulation development is CI-988, a
`cholecystokinin-B receptor antagonist (10).
`Additional formulation variables to be considered are
`the necessity of a buffer, buffer capacity, and drug
`concentration. These can influence supersaturated drug
`concentrations in the bloodstream, a condition that may
`lead to in vivo drug precipitation. The blood is very
`efficient at pH neutralization and normally maintains a
`narrow pH range of 7.38 to 7.42. For example, a low
`incidence of phlebitis was observed in the rabbit ear vein
`model when solutions over the pH range of 3 to 11, with
`buffer concentrations of approximately 0.3 M, were
`administered in a single small volume (1 mL) bolus dose
`(11). Simple screening tests consisting of a computa­
`tional model where drug solubility is plotted as a
`function of dilution, and in vitro dilution experiments
`were shown to be effective tools in evaluating the ability
`of the pH-solubilized drug to remain in solution dilution
`(12, 13). Davio et al. (14) showed that in vivo precipita­
`tion of the pH-solubilized drug ditekiren was dependent
`upon drug concentration and infusion rate. Low concen­
`tration drug solutions, which are rapidly diluted below
`
`saturation solubility, and rapid infusions were preferred
`to minimize precipitation.
`The most commonly used buffer components in paren­
`teral products and their pKa's are; citric acid (3.13, 4.76,
`6.40), acetic acid (4.76) and phosphoric acid (2.15, 7.20,
`12.33). When buffers are employed, the stability of the
`molecule must also be considered, since it may be
`influenced by the ions in solution (9). Examples of buffer
`catalyzed solution degradation include famotidine, a
`histamine H2 receptor inhibitor (15) and loracarbef, a
`zwitterionic cephalosporin (16).
`
`B. Use of Cosolvents
`In recent years, surveys of FDA-approved parenteral
`products (17–19) show five water-miscible cosolvents—
`glycerin, ethanol, propylene glycol, polyethylene glycol,
`and N,N,-dimethylacetamide—as components of sterile
`formulations (Table III and IV). Cosolvents are em­
`ployed in approximately 10% of FDA approved paren­
`teral products. They are useful because they may often
`provide exponential increases in solubility (20) and also
`allow exclusion of water for compounds susceptible to
`hydrolysis.
`Investigation of the solubilizing potential of various
`cosolvents may be approached empirically by determin­
`ing the compounds solubility in cosolvent compositions
`similar to marketed products (21–23), or by one of
`several systematic approaches, such as log-linear solubil­
`ity relationships or statistical experimental design.
`In the study of log-linear solubility relationships,
`Yalkowsky and Roseman (20) investigated a range of
`solutes in binary cosolvent mixtures of ethanol, propyl­
`ene glycol, and glycerin in water and discussed the
`closeness of fit of apparent solubility to a log-linear
`solubility equation. Briefly, this technique involves experi­
`mentally determining the solubility of a compound in
`increasing percentages of a cosolvent and generating a
`semi-logarithmic plot of the apparent solubility of the
`drug as a function of the volume-fraction of the cosol­
`vent. Using the slope and the solubility of the compound
`in pure water, an equation may be written to describe
`the solubility in a binary system.
`Assuming that the log-linear increases in solubility of
`individual cosolvents are additive, equations may also be
`written for ternary and quaternary mixed cosolvent
`systems (24). Mathematically, these relationships are
`described by the following equations:
`
`Binary cosolvent system
`
`log Cx = log Cw. + αxfx
`Ternary cosolvent system
`
`log Cx = log Cw. + αxfx + αafa
`Quaternary cosolvent system
`
`log Cx = log Cw + αxfx + αafa + αbfb
`
`where Cw is the drug solubility in water; α's are the
`slopes of the semi logarithmic plots; Cx is the drug
`solubility; f is the volume fraction of the cosolvent; and
`the subscripts a, b, x denote the cosolvents A, B, and X
`
`332
`
`PDA Journal of Pharmaceutical Science & Technology
`
`

`

`
`
`Downloaded from on May 5, 2017journal.pda.org
`
`
`
`TABLE II
`Examples of Marketed Parenteral Products with Solution pH Outside Range of 4 to 8 (18, 19)
`
`pH
`Adjustment
`
`Generic
`Name
`
`Trade Name
`
`Marketed
`Form
`
`Routes
`
`Lactic acid, NaOH
`Benzenesulfonic acid
`
`Amrinone Lactate
`Atracurium Besylate
`
`Inocor (Sanofi Winthrop)
`Tracrium (Burroughs
`Wellcome)
`Librium (Roche)
`Emete-Con (Roerig)
`Cipro I.V. (Miles)
`DTIC-Dome (Miles)
`Intropin (DuPont)
`Cardizem (Marion Merrell
`Dow)
`Vibramycin IV (Roerig,
`Elkins-Sinn)
`Inapsine (Janssen)
`Ergotrate Maleate (Lilly)
`
`Solution
`Solution
`
`Powder
`Powder
`Concentrate
`Powder
`Solution
`Solution
`
`IB, IF
`IB, IF
`
`IB
`IM, IF
`IF
`IB, IF
`IF
`IF, IB
`
`Powder
`
`IF
`
`Solution
`Solution
`
`IM, IF, IB
`IM, IB
`
`Innovar (Janssen)
`
`Solution
`
`IM, IB, IF
`
`Robinul (Robins)
`Haldol (McNeil)
`Normodyne (Schering)
`Trandate (Glaxo)
`Aldomet Ester HCl(Merck)
`Methergine (Sandoz)
`
`Versed (Roche)
`Primacor (Sanofi Winthrop)
`Minocin (Lederle)
`Nubain (DuPont)
`Narcan (DuPont)
`Zofran (Cerenex)
`Pitocin (Parke-Davis)
`Papaverine HCl (Lilly)
`Pyridoxine HCl (Steris)
`Priscoline HCl (Ciba)
`
`Solution
`Solution
`Solution
`
`Solution
`Solution
`
`Solution
`Solution
`Powder
`Solution
`Solution
`Solution
`Solution
`Solution
`Solution
`Solution
`
`IM, IB
`IM
`IB, IF
`
`IF
`IM, F
`
`IM, IF
`IF
`IF
`IM, IB
`IM, IB, IF
`IF
`IF
`IB, IF
`IM, IB
`IB, IM
`
`Diamox (Lederle)
`Zovirax (Burroughs
`Wellcome)
`Aminophylline (Abbott,
`Elkins-Sinn, American
`Regent)
`Amytal Na (Lilly)
`Imuran (Burroughs
`Wellcome)
`Polycillin-N (Apothecon)
`Totacillin-N (Beecham)
`Omnipen-N (Wyeth)
`Celestone Phosphate
`(Schering)
`Sodium Diuril (Merck)
`Hyperstat (Schering)
`Stilphostrol (Miles)
`
`Powder
`Powder
`
`IM, IB, IF
`IF
`
`Solution
`
`IB, IF
`
`Powder
`Powder
`
`Powder
`
`IM, IF
`IB, IF
`
`IM, IB, IF
`
`Solution
`
`IB, IM
`
`Powder
`Solution
`Solution
`
`Solution
`Solution
`Solution
`
`Powder
`Powder
`
`Powder
`
`IB, IF
`IB
`IF
`
`IB, IF
`IB
`IM, IB, IF
`
`IF
`IM, IB, IF
`
`IB, IF
`
`333
`
`pH
`(constituted)
`
`p H <4
`3.2–4
`3.25–3.65
`
`3
`3–4
`3.3–3.9
`3–4
`2.5–4.5
`3.7–4.1
`
`1.8–3.3
`
`3–3.8
`2.7–3.5
`
`3.2–3.8
`
`2–3
`3–3.6
`3–4
`
`3–4.2
`2.7–.5
`
`3
`3.2–4
`2–2.8
`3.5
`3–4
`3.3–4
`2.5–4.5
`3–4
`2–3.8
`3–4
`
`pH>8
`9.2
`10.5–11.6
`
`8.6–9
`
`9.6–10.4
`9.6
`
`8-10
`
`8.5
`
`9.2–10
`11.6
`9–10.5
`
`9.2
`8–11
`8–9.3
`
`11
`8.1
`
`Lactic acid, HC1
`Citric acid
`NaOH, HC1
`Citric acid, Na citrate
`
`Lactic acid
`Lactic acid, ethyl
`lactate
`Lactic acid
`
`NaOH/HC1
`Lactic acid
`
`NaOH, citric acid
`Tartaric acid
`
`NaOH, HCl
`
`Na citrate, citric acid
`HCl
`Citric acid, Na citrate
`Acetic acid
`NaOH
`
`Tartaric acid, Na
`citrate
`
`HCl/NaOH
`
`NaOH
`
`Na2HPO4, NaOH
`
`NaOH
`NaOH
`
`NaOH
`NaOH
`NaOH
`
`Chlordiazepoxide HCl
`Benzquinamide HCl
`Ciprofloxacin
`Dacarbazine
`Dopamine HCl
`Diltiazem HCl
`
`Doxycycline Hyclate
`
`Droperidol
`Ergonovine Maleate
`
`Fentanyl Citrate and
`Droperidol
`Glycopyrrolate
`Haloperidol Lactate
`Labetalol HCl
`
`Methyldopate HCl
`Methylergonovine
`Maleate
`Midazolam HCl
`Milrinone Lactate
`Minocycline HCl
`Nalbuphine HCl
`Naloxone HCl
`Ondansetron HCl
`Oxytocin
`Papaverine HCl
`Pyridoxine HCl
`Tolazoline HCl
`
`Acetazolamide Na
`Acyclovir Na
`
`Aminophylline
`
`Amobarbital Na
`Azathioprine Na
`
`Ampicillin Na
`
`Betamethasone Na
`PO4
`Chlorothiazide Na
`Diazoxide
`Diethylstilbestrol
`Diphosphate
`Fluorouracil
`Folic acid
`Lasix
`
`Ganciclovir Na
`Leucovorin Ca
`
`Vol.50, No. 5 / September–October 1996
`
`Fluorouracil (Roche)
`Folvite (Lederle)
`Furosemide
`(Hoechst-Roussel)
`Cytovene (Syntex)
`Wellcovorin (Immunex,
`Burroughs Wellcome)
`Methohexital Na
`Na carbonate
`9.5–10.5
`Brevital Na (Lilly)
`IM = intramuscular, IF = intravenous infusion, IB = intravenous direct injection.
`
`

`

`
`
`Downloaded from on May 5, 2017journal.pda.org
`
`
`
`TABLE III
`Cosolvent Concentrations in Some Currently Marketed Parenterals (18, 19)
`
`Cosolvent in
`Marketed Vehicle
`
`Generic
`Name
`
`Ethanol 100%
`
`Carmustine
`
`Trade Name
`
`Marketed
`Form
`
`BiCNU (Bristol-Myers
`Oncology)
`
`Drug
`+ Diluent
`
`Routes
`
`Administration
`
`Appx.
`Vehicle
`per Dose
`
`IF
`
`Dilute 1:10
`
`3 ml
`
`Propylene glycol 40%
`Ethyl alcohol 10%
`
`Propylene Glycol 40%
`Alcohol 10%
`
`Benzyl alcohol 5%
`Propylene glycol 50%
`
`Propylene glycol 25%
`Ethanol 25%
`
`Propylene glycol
`10.36%
`
`Ethanol 10%
`
`Diazepam
`
`Valium (Roche)
`
`Solution
`
`IM, IB
`
`Direct injection
`
`0.5–4 ml
`
`Digoxin
`
`Dimenhydrinate
`
`Lanoxin (Burroughs
`Wellcome)
`
`Dimenhydrinate
`(Steris)
`
`Solution
`
`IB
`
`Direct injection
`
`1–3 ml
`
`Solution
`
`IM, IF
`
`Dilute 1:10
`
`1 ml
`
`Esmolol HCl
`
`Brevibloc (DuPont)
`
`Concentrate
`
`IF
`
`Dilute 1:25
`
`1–10 ml
`
`Hydralazine HCl
`
`Apresoline HCl (Ciba)
`
`Solution
`
`IM, IB
`
`Direct injection
`
`0.5–1 ml
`
`Ketorolac
`Tromethamine
`
`Lorazepam
`
`Toradol (Syntex)
`
`Solution
`
`IM only
`
`Direct Injection IM
`
`1 ml
`
`Solution
`
`IM, IB
`
`PEG 400 0.18 ml/ml
`Benzyl alcohol 2%
`Propylene glycol
`
`Povidone 20 mg
`Diluent (10 ml)
`Propylene glycol 6 ml
`Ethanol 0.52 mL
`
`Ethanol 30%
`Propylene glycol 30%
`
`Propylene glycol 40%
`Alcohol 10%
`
`Alcohol 10%
`Propylene glycol 67.8%
`
`Propylene glycol 40%
`Alcohol 10%
`
`Ativan (Wyeth-
`Ayerst)
`
`IM Direct Injection
`Dilute 1:1 IV
`
`1 ml
`
`Melphalan HC1
`
`Alkeran (Burroughs
`Wellcome)
`
`Drug
`+ Diluent
`
`IF
`
`Dilute constitute
`> 1:10
`
`10 ml
`
`Nitroglycerin
`
`Tridil (DuPont)
`
`Concentrate
`
`IF
`
`Dilute 1:100
`
`2.5–10 ml
`
`Pentobarbital Na
`
`Nembutal (Abbott)
`
`Solution
`
`IM, IB
`
`Slow direct injection
`
`2 ml
`
`Phenobarbital Na
`
`Luminal Na (Sanofi
`Winthrop)
`
`Solution
`
`IM. IB
`
`Direct injection
`
`1 ml
`
`Phenytoin Na
`
`Dilantin (Parke Davis)
`
`Solution
`
`IM, IB
`
`Direct injection
`
`3–5 ml
`
`Polyethylene glycol 50%
`
`Secobarbital Na
`
`Propylene glycol 40%
`Ethanol 10%
`
`Trimethoprim-Sul(cid:173)
`famethoxazole
`
`Secobarbital Na
`(Wyeth-Ayerst)
`
`Septra (Burroughs
`Wellcome)
`Bactrim (Roche)
`
`Solution
`
`IM, IB, IF
`
`Direct injection
`
`1–5 ml
`
`Concentrate
`
`IF
`
`Dilute 1:25
`
`5–10 ml
`
`Amsacrine
`
`N,N-Dimethylacet-
`Drug
`Amsidine Concen(cid:173)
`amide 100%
`+ Diluent
`trate (Parke-Davis)"
`IM = intramuscular. IF = intravenous infusion, IB = intravenous direct injection.
`a Drug available outside the United States.
`
`IF
`
`Dilute 1:500
`
`1.5 ml
`
`respectively. In its simplest form, determining the drug
`solubility in water and pure cosolvents would allow
`estimation of the amount and type of cosolvent required
`to attain a desired solubility. In most cases however,
`deviations from log-linear increases of solubility occur in
`aqueous cosolvent mixtures as indicated by curvature in
`the solubility plots. The deviations are attributed to
`solvent-solvent interactions (5, 25).
`For first approximations of solubility however, this
`approach has been shown to be useful (26–27). Chien
`(28) used this technique and polarity indexes of cosol(cid:173)
`vents to calculate the polarity of a solution that pro(cid:173)
`duced the greatest solubility of the drug metronidazole.
`
`Aqueous/cosolvent ratios of corresponding polarity could
`then be calculated for other cosolvent systems to provide
`qualitative identification of solubility maximums (29,
`30). Polarity indexes of common water miscible cosol(cid:173)
`vents have been tabulated and discussed by Rubino and
`Yalkowsky. These indexes reflect the cohesive proper(cid:173)
`ties of the solvent (solubility parameter and interfacial
`tension), hydrogen bonding ability (proton donor and
`acceptor density), and polarity (dielectric constant).
`Another solubility determination approach particu(cid:173)
`larly helpful for complex mixtures is a statistical experi(cid:173)
`mental design (31). Identifying the optimum combina(cid:173)
`tion of cosolvents for solubilization may reduce the net
`
`334
`
`PDA Journal of Pharmaceutical Science & Technology
`
`

`

`
`
`Downloaded from on May 5, 2017journal.pda.org
`
`
`
`TABLE IV
`Surfactant Concentrations in Some Currently Marketed Parenterals (18, 19)
`
`Solubilizer in
`Marketed Vehicle
`
`NN-dimethylacetamide 60
`mg/ml
`Cremophor EL 500
`mg/ml
`Dehydrated alcohol
`42.7%
`
`Polysorbate 80 20 mg/ml
`Propylene glycol 20.7
`mg/ml
`
`Cremophor EL 527
`mg/ml
`Ethanol 49.7%
`Propylene glycol 30%
`Polysorbate 80 1.6%
`Polysorbate 20 0.028%
`
`Polysorbate 80 4%
`Propylene glycol 20%
`
`Cremophor EL 650
`mg/ml
`Alcohol 32.9%
`
`Polyethylene glycol 300
`650 mg/ml
`Ethyl alcohol 30.5% v/v
`Polysorbate 80 8%
`Polyoxyethylated fatty
`acid 7.0%
`
`Generic Name
`
`Trade Name
`
`Routes
`
`Administration
`
`Appx.
`Vehicle
`per Dose
`
`Teniposide
`
`Vumon (Bristol-Myers
`Squibb)
`
`IF
`
`Dilute 1:10 or 1:100
`
`5–9 ml
`
`Phytonadione
`
`Konakion (Roche)
`
`IM only
`
`Direct injection IM
`
`1–2.5 ml
`
`Paclitaxel
`
`Taxol (Bristol-Myers
`Squibb)
`
`Multivitamins
`
`M.V.I.-12 (Astra)
`
`IF
`
`IF
`
`Dilute 1:5 or 1:20
`
`20 ml
`
`Dilute 1:100 or 1:500
`
`5 ml
`
`Chlordiazepoxide HCl
`
`Librium (Roche)
`
`IM only
`
`Direct injection IM
`
`2 ml
`
`Cyclosporine
`
`Sandimmune (Sandoz)
`
`IF
`
`Dilute 1:20–1:100
`
`5 ml
`
`Etoposide
`
`VePesid (Bristol-Myers
`Oncology)
`
`IF
`
`Dilute 1:100
`
`5 ml
`
`Phytonadione
`
`AquaMEPHYTON
`(Merck)
`
`IM, IB
`
`Direct inject IM,
`preferred
`
`1–2.5 ml
`
`PEG40 castor oil 0.115
`ml/ml
`
`Miconazole
`
`Monistat i.v. (Janssen)
`
`IF
`
`Dilute 1:10
`
`20 ml
`
`Direct injection IM
`
`1–2 ml
`
`Direct infusion
`
`20–100 ml
`
`Polysorbate 80 12%
`
`Vitamin A
`
`Polysorbate 80 0.008%
`
`Alteplase
`
`Aquasol A Parenteral
`(Astra)
`
`Activase (Genentech)
`
`Na desoxycholate 0.41%
`
`Amphotericin B
`
`Fungizone (Apothecon)
`
`IM
`
`IF
`
`IF
`
`Polysorbate 20 0.40%
`
`Calcitriol
`
`Polysorbate 80 0.04%
`
`Cefazolin Na
`
`Polysorbate 80 0.004%
`
`Filgrastim
`
`Sodium dodecyl sulfate
`0.18 mg/ml
`
`Aldesleukin
`
`Calcijex (Abbott)
`
`Kefzol (Lilly)
`Ancef (SmithKline
`Beecham)
`
`Neupogen (Amgen)
`
`Proleukin (Cetus
`Oncology)
`
`Polysorbate 80 10%
`
`Cordarone X IV (Sanofi
`Winthrop)a
`IM = intramuscular, IF = intravenous infusion, IB = intravenous direct injection.
`a Drug available outside the United States.
`
`Amiodarone HCl
`
`Dilute 1:50
`
`IB
`IM, IF, IB
`
`Direct injection
`
`Direct injection
`
`3–20 ml
`
`0.5–1 ml
`
`IB
`
`IF
`
`IF
`
`Direct injection
`
`0.25–3 ml
`
`Dilute 1:42
`
`Dilute 1:50
`
`1.2 ml
`
`3–7 ml
`
`amount of cosolvent in the formula (26). They also
`facilitate the study of systems characterized by non(cid:173)
`linear increases in solubility. Optimization techniques in
`pharmaceutical formulation have recently been re(cid:173)
`viewed (32). An example of their use is a simplex search
`for solvent blends producing maximum drug solubility
`(2).
`Acceptable levels of cosolvent in parenteral formula(cid:173)
`tions are not easily defined. A review of currently
`
`marketed parenteral products shows that percentages
`range from 10 to 100% (Table III and IV). Appropriate
`product amounts are often a matter of considering a
`diverse set of factors such as; 1) administration condi(cid:173)
`tions, 2) total dose, 3) target population and 4) duration
`of therapy. Toxicity and adverse clinical effects of
`common cosolvents are summarized (33–34). Recent
`safety assessment reviews of propylene glycol (35) poly(cid:173)
`ethylene glycol (36) and glycerol (37) have been pub-
`
`Vol. 50, No. 5 / September–October 1996
`
`335
`
`

`

`
`
`Downloaded from on May 5, 2017journal.pda.org
`
`
`
`TABLE V
`Some Currently Marketed Parenterals Utilizing Complexing
`Agents, Mixed Micelles, or Lipid Systems
`
`Solubilizer
`System
`
`Generic
`Name
`
`Trade Name
`
`investigation (41). Detailed reviews of micelle struc(cid:173)
`tures, characterization techniques, and pharmaceutical
`applications have been published (42, 43).
`The toxicity of surfactants reported in the literature
`prior to 1983 are summarized by Attwood and Florence
`(43). Reviews on the pharmacology of polysorbate 80
`(44) and the incidence of clinical side effects of Cremo(cid:173)
`phor EL® (42) have been published. Children and
`newborns may be particularly sensitive to these agents
`and administration to this population is discussed (46).
`
`D. Use of Complexing Agents
`
`Complexation of water insoluble drugs usually in(cid:173)
`volves the incorporation of the drug within the inner
`core of the complexing agent so that the outer hydro-
`philic groups of the complexing agent interacts with
`water rendering the complex soluble.
`An example of successful application of this technol(cid:173)
`ogy is Amphocil®, a lipid complex formed between
`amphotericin B and sodium cholesteryl sulfate, a natu(cid:173)
`rally occurring cholesterol metabolite (47). In solution,
`the complex is postulated to be a stable disc-like
`structure that remains intact in the systemic circulation.
`Comparative studies in animals with micelle solubilized
`amphotericin B (Fungizone®) have shown a significant
`reduction in systemic toxicity as a result of altered
`systemic distribution and elimination characteristics (48).
`Naturally occurring cyclodextrins, particularly ß-cyclo¬
`dextrin, are able to complex water insoluble drugs and
`render them soluble in water. However, ß-cyclodextrin
`have been associated with renal toxicity upon parenteral
`administration. The toxicity has been attributed the low
`aqueous solubility of ß-cyclodextrin and precipitation in
`the kidney. Newer cyclodextrins are chemically modified
`to improve water solubility and increase their usefulness
`(49), Brewster et al, (50) have described the preparation
`and successful use of chemically modified cyclodextrins
`such as 2-hydroxypropyl-ß-cyclodextrin in solubilizing
`and even stabilizing various proteins and peptides.
`An example where the drug was not incorporated
`within some kind of matrix, but combined with an
`additive to obtain basically a soluble salt complex
`involved ascorbic acid (51). Similarly, tromethamine has
`been reported to solubilize zomepirac, an anionic drug,
`by micelle (association colloid) formation (52). The
`aqueous solubility of metronidazole was reported to be
`enhanced by the water soluble vitamins nicotinamide,
`ascorbic acid or pyridoxine HCl (32). A cage-like struc(cid:173)
`ture formed by the vitamins around molecules of metro(cid:173)
`nidazole was postulated.
`
`E. Emulsion Systems
`
`If a molecule has sufficient lipid solubility, emulsions
`may be employed. Typical emulsions contain triglyceride-
`rich vegetable oils and lecithin and may also contain
`nonionic surface active agents as emulsifying agents.
`Insoluble drugs may be incorporated into commercial
`fat emulsions or through emulsification of the oil-
`solubilized drug. The former is usually not successful
`
`Complexing Agents
`Hydrolyze
`gelatin 0.7%
`Ethylenediamine
`
`Corticotropin
`
`Aminophylline
`
`Amphotericin B
`
`Amphotericin B
`
`Diazepam
`
`Vitamin K
`
`DMPG and
`DMPC lipid
`complex
`Na cholesteryl
`sulfate, col(cid:173)
`loidal disper(cid:173)
`sion
`Mixed Micelles
`Glycocholic acid-
`lecithin
`Glycocholic acid-
`lecithin
`Emulsions or Liposomes
`Lipid emulsion
`Diazepam
`
`Athcar (Rhone-
`Poulenc Rorer)
`Aminophylline
`(Abbott, Elkins-
`Sinn, American
`Regent)
`Alelcet (The Lipo(cid:173)
`some Co.)a
`
`Amphocil (Lipo(cid:173)
`some Tech(cid:173)
`nology)
`
`Valium MM
`(Roche)a
`Konakion/120
`(Roche)a
`
`Lipid emulsion
`Lipid emulsion
`
`Propofol
`Perfluorodecalin
`
`Liposome
`
`Amphotericin B
`
`Dizac (Ohmeda)
`Diazemuls
`(Dumex)a
`Diprivan (Zeneca)
`Fluosol-DA (Alpha
`Therapeutics)
`AmBisome
`(Vestar)a
`IM = intramuscular, IF = intravenous infusion. IB = intravenous
`direct injection.
`a Drug available outside the United States.
`
`lished. Local toxicity of several cosolvent vehicles in
`animals is summarized in Table VI.
`
`C. Use of Surface Active Agents
`
`Surface active agents are usually incorporated into
`parenterals to provide one of several desirable proper(cid:173)
`ties; 1) increase drug solubility through micellization, 2)
`to prevent drug precipitation upon dilution (38), 3)
`improve the stability of a drug in solution by incorpora(cid:173)
`tion of the drug into a micellar structure (39), and 4) in
`protein formulations, prevent aggregation due to liquid/
`air or liquid/solid interfacial interactions.
`Table IV provides examples of FDA-approved paren(cid:173)
`teral products containing surface active agents. While
`many different types of surfactants exist (40), only an
`extreme few have precedence for use in parenteral
`products. For example, for stabilization of proteins
`against problems of aggregation, only polyoxyethylene
`sorbitan monooleate (polysorbate 80) is an FDA-
`approved surfactant (18). Other surfactants which have
`been used in parenteral products are poloxamer 188
`(polyoxyethylene-polyoxypropylene copolymer), polysor(cid:173)
`bate 20 and 40 (polyoxyethylene-polyoxypropylene (poly(cid:173)
`oxyethylene sorbitan monofatty acid esters), Cremophor
`EL® and Emulphor EL 719® (polyethoxylated fatty acid
`esters and oils). Which surfactant is most effective as a
`solubilizer or stabilizer is often a matter of empirical
`
`336
`
`PDA Journal of Pharmaceutical Science & Technology
`
`

`

`
`
`Downloaded from on May 5, 2017journal.pda.org
`
`
`
`because drugs influence the stability of these commer(cid:173)
`cial emulsions (53).
`Emulsion formulas have shown advantages over high
`cosolvent levels by reducing local venous irritation (54).
`While emulsion

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