throbber
Remington: The
`Science and Practice
`
`of Pharmacy
`
`Volume II
`
`AUROBINDO EX1018, 1
`
`AUROBINDO EX1018, 1
`
`

`

`1 9TH
`
`EDITION
`
`Remington:
`Practice of
`
`ALFONSO R GENNARO
`
`Chairman of the Editorial Board
`and Editor
`
`AUROBINDO EX1018, 2
`
`

`

`The Science and
`Pharmacy
`
`1995
`
`MACK PUBLISHING COMPANY
`
`Easton, Pennsylvania 18042
`
`AUROBINDO EX1018, 3
`
`AUROBINDO EX1018, 3
`
`

`

`Entered according to Act of Congress, in the year 1885 by Joseph P Remington,
`in the Office of the Librarian of Congress, at Washington DC
`
`Copyright 1889, 1894, 1905, 1907,1917, by Joseph P Remington
`
`Copyright 1926, 1936, by the Joseph P Remington Estate
`
`Copyright 1948, 1951, by The Philadelphia College of Pharmacy and Science
`
`Copyright 1956, 1960, 1965, 1970, 1975, 1980,1985, 1990, 1995, by The Philadelphia College of
`Pharmacy and Science
`
`All Rights Reserved
`
`Libraly of Congress Catalog Card No. 60—53334
`
`ISBN 0-912734-0443
`
`The use ofstruclni‘aljbrmnlasfram USAN and the USP Dictionary ofDrug Names is by
`permission ofThe USP Convention. The Convention is not responsiblefora'ny inaccuracy
`contained herein
`
`NOTICE—This terrt is not intended to represent, norshall it be interpreted to be, the equivalent
`ofor a substitutefor the official United States Pharmacopeia (USP) and/or the National
`Fornznlary (NF). In the event ofany (lifierence ordiscrepancy between. the current ofl‘icz‘al
`USP orNFstandards ofstrength, quality, purity, packaging and labelingfordrugs and
`representations oft/lent herein, the context and effect oft/2e oflicial compendia shall prevail.
`
`AUROBINDO EX1018, 4
`
`

`

`CHAPTER 94
`
`Sustained-Release Drug Delivery Systems
`
`Charles S L Chico, PhD
`Anda SR Pharmaceuticals, Inc
`
`Joseph R Robinson, PhD
`Professor of Pharmacy
`School of Pharmacy
`University of Wisconsin
`Madison, WI 50706
`
`The goal of any drug delivery system is to provide a therapeu—
`tic amount of drug to the proper site in the body to achieve
`promptly, and then maintain, the desired drug concentration.
`That is, the drug-delivery system should deliver drug at a rate
`dictated by the needs of the body over the period of treatment.
`This idealized objective points to the two aspects most impor-
`tant to drug delivery, namely, spatial placement and tempo-
`ral. delivery of a drug. Spatial placement relates to target-
`ing a drug to a specific organ or tissue, while temporal delivery
`refers to controlling the rate of drug delivery to the target
`tissue. An appropriately designed sustained—release drug de-
`livery system can be a major advance toward solving these two
`It is for this reason that the science and technol-
`ogy responsible for development of sustained-release pharma-
`ceuticals have been and continue to be the focus of a great
`deal of attention in both industrial and academic laboratories.
`There currently exist numerous products on the market formu-
`lated for both oral and parenteral routes of administration that
`claim sustained or controlled drug delivery. The bulk of
`research has been directed at oral dosage forms that satisfy
`the temporal aspect of drug delivery, but many of the newer
`approaches under investigation may allow for spatial place-
`This chapter will define and explain the nature
`of sustained-release drug therapy, briefly outline relevant
`physicochemical and biological properties of a drug that af-
`fect sustained-release performance and review the more com-
`mon types of oral and parenteral sustained-release dosage
`In addition, a brief discussion of some methods cur-
`rently being used to develop targeted delivery systems will be
`
`Conventional Drug Therapy
`
`To gain an appreciation for the value of sustained drug
`therapy it is useful to review some fundamental aspects of
`conventional drug delivery.‘ Consider single dosing of a
`hypothetical drug that follows a simple one-compartment phar-
`rnacokinetic model for disposition. Depending on the route
`of administration, a conventional dosage form of the drug, eg,
`a solution, suspension, capsule, tablet, etc, probably will pro-
`duce a drug blood level versus time profile similar to that
`shown in Fig 1. The term ”drug blood level” refers to the
`concentration of drug in blood or plasma, but the concentra-
`tion in any tissue could be plotted on the ordinate.
`It can be
`seen from this figure that administration of a drug by either
`intravenous injection or an extravascular route, eg, orally,
`intramuscularly or rectally, does not maintain drug blood
`levels within the therapeutic range for extended periods of
`time. The short duration of action is due to the inability of
`conventional dosage forms to control temporal delivery.
`If
`
`In this case the drug
`is shown in Fig 2 for the oral route.
`blood level reached and the time required to reach that level
`depend on the dose and the dosing interval. There are sev-
`eral potential problems inherent in multiple-dose therapy:
`1.
`If the dosing interval is not appropriate for the biological half-life of
`the drug, large “peaks" and “valleys” in the drug blood level may result.
`For example, drugs with short half-lives require frequent (losings to main-
`tain constant therapeutic levels.
`2. The drug blood level may not be within the therapeutic range at
`sufficiently early times, an important consideration for certain disease
`states.
`3. Patient noncompliance with the multiple—(losing regimen can result
`in failure of this approach.
`
`In many instances, potential problems associated with con-
`ventional drug therapy can be overcome. When this is the
`case, drugs given in conventional dosage forms by multiple-
`dosing can produce the desired drug blood level for extended
`periods of time. Frequently, however, these problems are
`significant enough to make drug therapy with conventional
`dosage forms less desirable than sustained-release drug
`therapy. This fact, coupled with the intrinsic inability of
`conventional dosage forms to achieve spatial placement, is a
`compelling motive for investigation of sustained-release drug
`delivery systems. There are numerous potential advantages
`of sustained-release drug therapy that will be discussed in the
`next section.
`
`Sustained-Release Drug Therapy
`
`As already mentioned, conventional dosage forms include
`solutions, suspensions, capsules, tablets, emulsions, aero-
`sols, foams, ointrnents and suppositories. For this discus-
`sion, these dosage forms can be considered to release their
`active ingredients into an absorption pool immediately.
`This is illustrated in the following simple kinetic scheme:
`1;
`.
`-
`k
`.
`It .
`Absorption
`Target
`I
`I
`Dosage
`a
`F01 m drug release P001
`absorption Al ea
`elimination
`The absorption pool represents a solution of the drug at the
`site of absorption, and the terms It}, It" and It}, are first-order
`rate constants for drug release, absorption and overall elimina-
`tion, respectively.
`Immediate release from a conventional
`dosage form implies that A7,. >>> A?“ or, alternatively, that
`absorption of drug across a biological membrane, such as the
`intestinal epithelium, is the rate-limiting step in delivery of the
`drug to its target area. For rronimrnediate-release dosage
`forms, It, <<< Ir", that is, release of drug from the dosage
`form is the rate-limiting step. This causes the above kinetic
`scheme to reduce to
`
`AUROBINDO EX1018, 5
`
`

`

`
`
`
`
`
`
`DRUGBLOODLEVEL(°m°“"%ni)
`
`ExnavaECMor
`route
`
`
`TIME ('17!)
`
`Inoiiecfive
`Range
`
`
`
`
`
`DRUGBLOODLEVEL
`
`llIll,“UUUUUUUUU”I’llllflflflflllfllflflfllflflUZI
`
`
`
`
`TIME
`(hrl)
`
`Fig 1. Typical drug blood level versus time profiles for intravenous
`injections and an extravascular route of administration.
`
`Fig 3. Typical drug blood level versus time profiles for delayed-
`release drug delivery by a repeat-action dosage form.
`
`system must be directed primarily at altering the release rate
`by affecting the value of k)». The many ways in which this has
`been attempted will be discussed later in this chapter.
`Nonimmediate-release delivery systems may be divided con-
`veniently into four categories:
`1. Delayed release
`2. Sustained release
`a. Controlled release
`b. Prolonged release
`. Site-specific release
`Ll. Receptor release
`
`Delayed-release systems are those that use repetitive, inter-
`mittent closings of a drug from one or more immediate-release
`units incorporated into a single dosage form. Examples of
`delayed-release systems include repeat-action tablets and cap-
`sules, and enteric-coated tablets where timed release is
`achieved by a barrier coating. A delayed-release dosage form
`does not produce or maintain uniform drug blood levels within
`the therapeutic range, as shown in Fig 3, but, nonetheless, is
`more effective for patient compliance than conventional dos-
`age forms.
`Sustained-release systems include any drug delivery sys-
`tem that achieves slow release of drug over an extended
`period of time.
`If the systems can provide some control,
`whether this be of a temporal or spatial nature, or both, of
`drug release in the body, or in other words, the system is
`successful at maintaining constant drug levels in the target
`tissue or cells, it is considered a controlled—release system.
`If it is unsuccessful at this, but nevertheless prolongs therapeu-
`tic blood or tissue level of the drug for an extended period of
`time, it is considered a prolonged—release system. This is
`illustrated in Fig 4.
`Site-specific and receptor release refer to targeting of a
`drug directly to a certain biological location.
`In the case of
`site-specific release, the target is adjacent to, or in the dis—
`eased organ or tissue; for receptor release, the target is the
`particular receptor for a drug within an organ or tissue. Both
`of these systems satisfy the spatial aspect of drug delivery.
`
`nnmannaaanzamazanllanzzwuuuwzvluuuuv
`
`
`Tofic
`Range
`
`Thuapeuflc
`Range
`
`Inlilacfive
`Range
`
`llflflflflll'flflflfluylflfl
`
`
`
`
`
`
`
`DRUGeroooLEVEL(“”me
`
`
`TIME Mrfl
`
`Fig 2. Typical drug blood level versus time profile following oral
`multiple-dose therapy.
`
`Release Rate and Dose Considerati071.92
`
`Although it is not necessary or desirable to maintain a
`constant level of drug in the blood or target tissue for all
`therapeutic cases, this is the ideal goal of a sustained-release
`delivery system.
`In fact, in some cases optimum therapy is
`achieved by oscillating, rather than constant, drug levels.
`An example of this is antibiotic therapy, where the activity of
`the drug is required only during growth phases of the
`microorganism. A constant drug level will succeed at curing
`or controlling the condition, however, and this is true for most
`forms of therapy.
`The objective in designing a sustained-release system is to
`deliver drug at a rate necessary to achieve and maintain a
`constant drug blood level. This rate should be analogous to
`that achieved by continuous intravenous infusion where a
`drug is provided to the patient at a constant rate just equal to
`its rate of elimination. This implies that the rate of delivery
`must be independent of the amount of drug remaining in the
`dosage form and constant over time. That is, release from
`the dosage form should follow zero-order kinetics, as shown
`by
`
`k? = Rate In = Rate Out = kc ' Cd ' VII
`
`where It? is the zero—order rate constant for drug release
`(amount/time), Ice is the first-order rate constant for overall
`drug elimination (time’l), Cd is the desired drug level in the
`body (amount/volume) and Vd is the volume space in which
`the drug is distributed. The values of If,” Cd and Vd needed to
`calculate It? are obtained from appropriately designed single-
`dose pharmacokinetic studies. Equation 1 provides the
`method to calculate the zero-order release rate constant nec-
`essary to maintain a constant drug blood or tissue level for the
`simplest case where drug is eliminated by first-order kinetics.
`For many drugs, however, more complex elimination kinetics
`and other factors affecting their disposition are involved.
`This in turn affects the nature of the release kinetics necessary
`to maintain a constant drug blood level.
`recognize that while zero-order release may be desirable theo-
`
`I
`
`
`
`
`
`DRUGBLOODLEVEL(amoun/mL)
`
`
`
`TIME
`
`(hr!)
`
`Fig 4. Drug blood level versus time profiles showing the relation-
`ship between controlled-release (A), prolonged-release (B) and con-
`ventional—release (C) drug delivery.
`
`AUROBINDO EX1018, 6
`
`AUROBINDO EX1018, 6
`
`

`

`CHAPTER 94
`
`retically, nonzero-order release may be equivalent clinically to
`constant release in many cases. Aside from the extent of
`intra- and intersubject variation is the Observation that, for
`many drugs, modest changes in drug tissue levels do not result
`in an improvement in clinical performance. Thus, a noncon-
`stant drug level may be indistinguishable clinically from a
`constant drrrg level.
`To achieve a therapeutic level promptly and sustain the
`level for a given period of time, the dosage form generally
`consists of two parts:
`an initial priming dose, D,, that re-
`leases drug immediately and a maintenance or sustaining
`dose, D,,,. The total dose, W, thus required for the system is
`
`W=Di+Dm
`
`(2)
`
`For a system where the maintenance dose releases drug by a
`zero-order process for a specified period of time, the total
`
`Table 1—Potential Advantages of Sustained Drug Therapy
`1. Avoid patient compliance problems
`‘2. Employ less total drug
`a. Miuiruize or eliminate local side effects
`b. Minimize or eliminate systemic side effects
`c. Obtain less potentiation or redrrction in drug activity with
`chronic use
`d. Minimize drug accumulation with chronic dosing
`Improve efficiency in treatment
`a. Cure or control condition more promptly
`b. Improve control of condition, ie, redrrce fluctuation in drug
`level
`c. Improve bioavailability of some drugs
`(1. Make use of special effects, eg, sustained-release aspirin for
`morning relief of arthritis by (losing before bedtime
`4. Economy
`
`3.
`
`W = D, + kETd
`
`(3)
`
`where It? is the zero-order rate constant for drug release and
`T,, is the total time desired for sustained release from one
`If the maintenance dose begins the release of drug at
`the time of dosing (t = 0), it will add to that which is provided
`by the initial dose, thus increasing the initial drug level.
`In
`this case a correction factor is needed to account for the added
`drrrg from the maintenance (lose:
`W = D,- + kgTd — kIQT)1)
`
`(4)
`
`The correction factor, k?T,, is the amount of drug provided
`during the period from t = to the time of the peak drug level,
`T,,. No correction factor is needed if the dosage form is
`constructed in such a fashion that the maintenance dose does
`not begin to release drug until time T1,.
`It already has been mentioned that a perfectly invariant
`drug blood or tissue level versus time profile is the ideal goal
`of a sustained-release system. The way to achieve this, in the
`simplest case, is by use of a maintenance dose that releases its
`drug by zero-order kinetics. However, satisfactory approxi-
`mations of a constant drug level can be obtained by suitable
`combinations of the initial dose and a maintenance dose that
`releases its drug by a first-order process.
`The total dose for
`
`W = Di + (k’P Crl/kr) V11
`
`(5)
`
`where k7,. is the first-order rate constant for drug release
`(time—1), and [cm 0,, and Vd are as defined previously.
`If the
`maintenance dose begins releasing drug att = 0, a correction
`factor is required just as it was in the zero-order case. The
`correct expression in this case is
`
`(6)
`W = Di + (chd/k’r)Vd _ Dmk’e T1)
`In order to maintain drug blood levels within the therapeutic
`range over the entire time course of therapy, most sustained-
`release drug delivery systems are, like conventional dosage
`forms, administered as multiple rather than single doses.
`For an ideal sustained-release system that releases drug by
`zero-order kinetics, the multiple dosing regimen is analogous
`to that used for a constant intravenous infusion, as discussed
`in Chapter 42. For those sustained-release systems having
`release kinetics other than zero-order, the multiple dosing
`regimen is more complex and its analysis is beyond the scope
`of this chapter; Welling and Dobrinska3 provide more detailed
`
`Potential Advantages of Sustained Drug Therapy
`
`drug therapy. Minimizing or eliminating patient compliance
`problems is an obvious advantage of sustained-release therapy.
`Because of the nature of its release kinetics, a sustained-
`release system should be able to use less total drug over the
`time course of therapy than a conventional preparation. The
`advantages of this are a decrease or elimination of both local
`and systemic side effects, less potentiation or reduction in
`drug activity with chronic use and minimization of drug accu-
`mulation in body tissues with chronic dosing.
`Unquestionably the most important reason for sustained-
`drug therapy is improved efficiency in treatment, ie, optimized
`therapy. The result of obtaining constant drug blood levels
`from a sustained-release system is to achieve promptly the
`desired effect and maintain it for an extended period of time.
`Reduction or elimination of fluctuations in the drug blood
`level allows better disease state management.
`In addition,
`the method by which sustained release is achieved can im-
`prove the bioavailability of some drugs. For example, drugs
`susceptible to enzymatic inactivation or bacterial decomposi-
`tion can be protected by encapsulation in polymer systems
`suitable for sustained release. For drugs that have a “spe-
`cific window" for absorption, increased bioavailability can be
`attained by localizing the sustained-release delivery system in
`certain regions of the gastrointestinal tract.
`Improved effi-
`ciency in treatment also can take the form of a special thera-
`peutic effect not possible with a conventional dosage form
`(see Table 1).
`The last potential advantage listed in Table 1, that of
`economy, can be examined from two points of view.
`Although the initial unit cost of most sustained-drug delivery
`systems usually is greater than that of conventional dosage
`forms because of the special nature of these products, the
`average cost of treatment over an extended time period may
`be less. Economy also may result from a decrease in nursing
`time/ hospitalization, less lost work time, etc.
`
`Drug Properties Relevant to Sustained-Release
`Formulation
`
`The design of sustained-release delivery systems is subject
`to several variables of considerable importance. Among these
`are the route of drug delivery, the type of delivery system, the
`disease being treated, the patient, the length of therapy and
`the properties of the drug. Each of these variables are inter-
`related and this imposes certain constraints upon choices for
`the route of delivery, the design of the delivery system and the
`length of therapy. Of particular interest to the scientist de—
`signing the system are the constraints imposed by the proper-
`ties of the drug.
`It is these properties that have the greatest
`
`AUROBINDO EX1018, 7
`
`

`

`Physicochenrical. Properties
`
`Aqueous Solubility and pKa—It is well known that in
`order for a drug to be absorbed it first must dissolve in the
`aqueous phase surrounding the site of administration and
`then partition into the absorbing membrane. Two of the
`most important physicochemical properties of a drug that
`influence its absorptive behavior are its aqueous solubility
`and,
`if it is a weak acid or base (as are most drugs),
`its
`pKa. These properties play an influential role in performance
`of nonsustained-release products; their role is even greater in
`sustained-release systems.
`The aqueous solubility of a drug influences its dissolution
`rate, which in turn establishes its concentration in solution
`and hence the driving force for diffusion across membranes.
`Dissolution rate is related to aqueous solubility as shown by
`the Noyes-Whitney equation which, under sink conditions, is
`
`dC/dt = kDAc,
`
`(7)
`
`where dC/dt is the dissolution rate, kl, is the dissolution rate
`constant,A is the total surface area of the drug particles and C,
`is the aqueous saturation solubility of the drug. Tire dissolu—
`tion rate is constant only if surface area, A, remains constant,
`but the important point to note is that the initial rate is propor-
`tional directly to aqueous solubility Cs. Therefore, the aque-
`ous solubility of a drug can be used as a first approximation of
`its dissolution rate. Drugs with low aqueous solubility have
`low dissolution rates and usually suffer oral bioavailability
`problems.
`It will be recalled from Chapter 16 that the aqueous solubil-
`ity of weak acids and bases is governed by the pKa of the
`compound and the pH of the medium. Fora weak acid
`
`u’
`S, = 50(1 + K ’[H+]) = 80(1 + 10PH‘I’K")
`
`(8)
`
`where S, is the total solubility (both the ionized and unionized
`forms) of the weak acid, SO is the solubility of the unionized
`form, K“ is the acid dissociation constant and [H+] is the
`hydrogen ion concentration of the medium. Equation 8 pre-
`dicts that the total solubility, 8,, of a weak acid with a given pKa
`can be affected by the pH of the medium.
`Similarly, for a
`weak base
`
`5; = 30(1 + [H+]/K(r) = 50(1 + IOPKFPH)
`
`(9)
`
`where S, is the total solubility (both the conjugate acid and
`free-base forms) of the weak base, So is the solubility of the
`free-base form and K" is the acid dissociation constant of the
`conjugate acid. Analogous to Eq 8, Eq 9 predicts that the
`total solubility, 5,, of a weak base whose conjugate acid has a
`given pKa can be affected by the pH of the medium.
`Considering the pH-partition hypothesis, the importance of
`Eqs 8 and 9 relative to drug absorption is evident. The pH-
`partition hypothesis simply states that the Lin-ionized form of
`a drug will be absorbed preferentially, in a passive manner,
`through membranes. Since weakly acidic drrrgs will exist in
`the stomach (pH = l to 2) primarily in the un-ionized form,
`their absorption will be favored from this acidic environment.
`On the other hand, weakly basic drugs will exist primarily in
`the ionized form (conjugate acid) at the same site, and their
`absorption will be poor.
`In the upper portion of the small
`intestine, the pH is more alkaline (pH = 5 to 7) and the reverse
`will be expected for weak acids and bases. The ratio of Eq 8
`or 9 written for either the pH of the gastric or intestinal fluid
`and the pH of blood is indicative of the driving force for
`absorption based on pH gradient. For example, consider the
`ratio of the total solubility of the weak acid aspirin in the blood
`
`from the stomach. The same calculation for intestinal pH
`(about 7) yields a ratio close to 1, implying a less-favorable
`driving force for absorption at that location.
`release of an ionizable drug from a sustained-release system
`should be ”programmed” in accordance with the variation in
`pH of the different segments of the gastrointestinal (GI) tract
`so that the amount of preferentially absorbed species, and
`thus the plasma level of drug, will be approximately constant
`throughout the time course of drug action.
`In general, extremes in the aqueous solubility of a drug are
`undesirable for formulation into a sustained-release product.
`A drug with very low solubility and a slow dissolution rate will
`exhibit dissolution-limited absorption and yield an inherently
`sustained blood level.
`In most instances, formulation of such
`a drug into a sustained-release system is redundant. Even if
`a poorly soluble drug was considered as a candidate for formu-
`lation into a sustained-release system, a restraint would be
`placed upon the type of delivery system which could be used.
`For example, any system relying upon diffusion of drug
`through a polymer as the rate-limiting step in release would be
`unsuitable for a poorly soluble drug, since the driving force for
`diffusion is the concentration of drug in the polymer or solu-
`tion and this concentration would be low. For a drug with
`very high solubility and a rapid dissolution rate, it often is
`quite difficult to decrease its dissolution rate and slow its
`absorption. Preparing a slightly soluble form of a drug with
`normally high solubility is, however, one possible method for
`preparing sustained-release dosage forms. This will be elabo-
`rated upon elsewhere in this chapter.
`Partition Coefficient—Between the time that a drug is
`administered and the time it is eliminated from the body, it
`must diffuse through a variety of biological membranes which
`act primarily as lipid-like barriers. A major criterion in evalu-
`ation of the ability of a drrrg to penetrate these lipid mem-
`branes is its apparent oil/water partition coefficient, defined
`as
`
`K: CO/C",
`where CO is the equilibrium concentration of all forms of the
`drug, eg, ionized and un-ionized, in an organic phase at equilib-
`rium, and Cu. is the equilibrium concentration of all forms in an
`aqueous phase. A frequently used solvent for the organic
`phase is 1-octanol. Although not always valid, an approxima-
`tion to the value of K may be obtained by the ratio of the
`solubility of the drug in l-octanol to that in water.
`drugs with extremely large values of K are very oil-soluble and
`will partition into membranes quite readily. The relationship
`between tissue permeation and partition coefficient for the
`drug generally is known as the H(msch correlation, discussed
`in Chapter 28.
`In general, it describes a parabolic relation-
`ship between the logarithm of the activity of a drug or its
`ability to be absorbed and the logarithm of its partition coeffi—
`cient for a series of drugs as shown in Fig 5. The explanation
`for this relationship is that the activity of a drug is a function of
`its ability to cross membranes and interact with the receptor;
`as a first approximation, the more effectively a drug crosses
`membranes, the greater its activity. There is also an opti-
`mum partition coefficient for a drug at which it most effec-
`tively permeates membranes and thus shows greatest activity.
`Values of the partition coefficient below this optimum result in
`decreased lipid solubility, and the drug will remain localized in
`the first aqueous phase it contacts. Values larger than the
`optimum result in poorer aqueous solubility, but enhanced
`lipid solubility and the drug will not partition out of the lipid
`membrane once it gets in. The value of K at which optimum
`activity is observed is approximately 1000/1 in l-octanol/
`water. Drugs with a partition coefficient that is higher or
`
`AUROBINDO EX1018, 8
`
`AUROBINDO EX1018, 8
`
`

`

`CHAPTER 94
`
`
`log K
`
`Fig 5. Typical relationship between drug activity and partition coef-
`ficient, K, generally known as the Hansch correlation.
`
`lower than the optimum are, in general, poorer candidates for
`formulation into sustained-release dosage forms.
`Drug Stability—Of importance for oral dosage forms is
`the loss of drug through acid hydrolysis and/01‘ metabolism in
`Since a drug in the solid state undergoes
`degradation at a much slower rate than a drug in suspension or
`solution, it would seem possible to improve significantly the
`relative bioavailability of a drug, which is unstable in the GI
`tract, by placing it in a slowly available sustained—release
`form. For those drugs that are unstable in the stomach, the
`most appropriate sustaining unit would be one that releases
`its contents only in the intestine. The reverse is the case for
`those drugs that are unstable in the environment of the intes-
`tine; the most appropriate sustaining unit in this case would be
`one that releases its contents only in the stomach. However,
`most sustained-release systems currently in use release their
`contents over the entire length of the GI tract. Thus, drugs
`with significant stability problems in any particular area of the
`GI tract are less suitable for formulation into sustained-
`release systems that deliver their contents uniformly over the
`length of the GI tract. Delivery systems that remain localized
`in a certain area of the GI tract eg, bioadhesive drug delivery
`system, and act as reservoirs for drug release are much more
`advantageous for drugs that not only suffer from stability
`problems but have other bioavailability problems as well.
`Development of this type of system is still in its infancy.
`The presence of metabolizing enzymes at the site of absorp-
`tion is not necessarily a negative factor in sustained—release
`Indeed, the prodr'ug approach to drug delivery
`takes advantage of the presence of these enzymes to regener-
`ate the parent molecule of an inactive drug derivative. This
`will be amplified upon below and in Chapter 28.
`
`Protein Binding
`
`Chapters 14 and 43 described the occurrence of drug bind
`ing to plasma proteins (eg, albumin) and the resulting reten-
`tion of drug in the vascular space. Distribution of the drug
`into the extravascular space is governed by the equilibrium
`process of dissociation of the drug from the protein. The
`drug—protein complex can serve therefore as a reservoir in the
`vascular space for sustained drug release to extravascular
`tissues, but only for those drugs that exhibit a high degree of
`binding. Thus, the protein binding characteristics of a drug
`can play a significant role in its therapeutic effect, regardless
`of the type of dosage form. Extensive binding to plasma
`proteins will be evidenced by a long half-life of elimination for
`the drug, and such drugs generally do not require a sustained-
`release dosage form. However, drugs that exhibit a high
`degree of binding to plasma proteins also might bind to bio-
`polymers in the GI tract, which could have an influence on
`sustained-drug delivery.
`
`Some drugs that exhibit greater than 95% binding at therapeu-
`tic levels are amitriptyline, bishydroxycoumarin, diazepam,
`diazoxide, dicumarol and novobiocin.
`Molecular Size and Difi'usivity—As previously discussed,
`a drug must diffuse through a variety of biological membranes
`during its time course in the body.
`In addition to diffusion
`through these biological membranes, drugs in many sustained-
`release systems nurst diffuse through a rate-controlling mem-
`brane or matrix. The ability of a drug to diffuse through
`membranes, its so called diffusivity (diffusion coefficient), is a
`function of its molecular size (or molecular weight). An im-
`portant influence upon the value of the diffusivity, D, in poly-
`mers is the molecular size (or molecular weight) of the diffus-
`ing species.
`In most polymers, it is possible to relate log D
`empirically to some function of molecular size, as shown in Eq
`12:“
`
`logD = —.5‘,, log 0 + k, = —5M logM + kt,”
`
`(12)
`
`where r) is molecular volume, M is molecular weight and 5,, SM,
`It, and k,,, are constants.
`The value of D thus is related to the
`size and shape of the cavities as well as size and shape of
`drugs. Generally, values of the diffusion coefficient for inter-
`mediate-molecular-weight drugs, ie, 150 to 400, through flex-
`ible polymers range from 10’“ to 10‘9 cmZ/sec, with values
`on the order of 10’8 being most common.5 A value of ap-
`proximately 10‘6 is typical for these drugs through water as
`the medium.
`It is of interest to note that the value of D for
`one gas in another is on the order of 10’1 c1112/ sec, and for one
`liquid through another, 10“3 cmz/sec.
`F‘or drugs with a mo-
`lecular weight greater than 500, the diffusion coefficients in
`many polymers frequently are so small that they are difficult to
`quantify, ie, less than 10‘12 cmZ/sec. Thus, high-molecular-
`weight drugs and/ or polymeric drugs should be expected to
`display very slow-release kinetics in sustained-release devices
`using diffusion through polymeric membranes or matrices as
`the releasing mechanism.
`
`Biological. Properties
`
`Absorption—The rate, extent and uniformity of absorp-
`tion of a drug are important factors when considering its
`formulation into a sustained-release system. Since the rate-
`lirniting step in drug delivery from a sustained-release system
`is its release from a dosage form, rather than absorption, a
`rapid rate of absorption of the drug relative to its release is
`essential if the system is to be successful. As stated previ-
`ously in discussing terminology, k, <<< It... This becomes
`most critical in the case of oral administration. Assuming
`that the transit time of a drug through the absorptive area of
`the GI tract is between 9 and 12 hours, the maximum absorp-
`tion half-life should be 3 to 4 hours.6 This corresponds to a
`minimum absorption rate constant It}, of 0.17 hi“1 to 0.28 hr"
`necessary for about 80 to 95% absorption over a 9- to 12-hour
`transit time. For a drug with a very rapid rate of absorption
`(ie, k" >> 0.23 hr"), the above discussion implies that a
`first-order release-rate constant k,- less than 0.17 hr—l is likely
`to result in unacceptably poor bioavailability in many patients.
`Therefore, slowly absorbed drugs will be difficult to formulate
`into sustained-release systems where the criterion that k, <<<
`It}, must be met.
`The extent and uniformity of the absorption of a drug, as
`reflected by its bioavailability and the fraction of the total dose
`absorbed, may be quite low for a v

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