throbber
Drug Delivery Devices
`Issues in Drug Development
`
`Peter R. Byron
`
`Virginia Commonwealth University, Richmond, Virginia
`
`New techniques for delivery of drugs by inhalation are discussed
`in this article. Devices that promise to improve the efficiency of lung
`delivery are described along with some of the regulatory challenges
`faced by their development scientists. Although high delivery effi-
`ciencies are possible, such devices are expensive to develop and
`may only be feasible in the event that they are partnered with
`drugs whose therapeutic and economic value is truly enhanced by
`the effort invested in the process. Appropriate devices must also
`be selected after paying careful attention to the physicochemical
`and dosing demands associated with the drug substance to be.
`inhaled. Even newly launched commercial products display large
`variations in dose delivery to the lung, in spite of increased global
`efforts to regulate and ensure the uniformity of delivered doses
`and their aerosol size distributions; this because of variations in the
`inspiratory maneuvers used by patients and the lack of control
`exercised over these maneuvers by most new inhalers. Sophisti—
`cated electromechanical techniques are discussed as possible ways
`of overcoming some of the common difficulties associated with
`ensuring reproducibility of dose and drug delivery to the lung.
`
`Keywords: aerosol; drug; delivery devices; inhalers; lungs
`
`After a long period during which chlorofluorocarbon (CFC)-
`pressurized metered dose inhalers (MDIs) dominated the mar-
`ket, in the last 15 years several new inhalation devices have been
`introduced. Other novel devices remain “in the wings,” and these
`may yet be exploited by pharmaceutical companies for specific
`reasons (1). We are seeing increasing acceptance of the lungs
`as a route of systemic drug administration, as well as a number
`of new drugs offering novel therapeutic benefits for several acute
`and chronic lung disorders (2, 3). These developments represent
`significant opportunities for pharmaceutical companies, pro-
`vided they choose delivery systems that adequately “partner”
`each drug during its development. The regulatory hurdles facing
`inhaler developers have become more stringent as the global
`marketplace has extended the impact of the U.S.A.’s Food and
`Drug Administration; FDA is probably the strictest of the world’s
`regulatory authorities. Multidose inhalers must now be shown
`to deliver individual doses reproducibly throughout inhaler shelf—
`life, in temperatures and humidities that represent commonly
`experienced environmental conditions. Not only must doses be
`reproducible, but the particle or droplet size distributions from
`each inhaler must also be shown to be “stable” over the product’s
`lifetime, and the product proven to be manufactured reproduci-
`bly (3). This must be done to show that the clinical results that
`are presented to the regulators at the time of product submission
`are “representative” of the “to be marketed” inhaler. Some of
`the recent increased regulatory oversight has been stimulated
`by environmental demands to replace the CFC propellants in
`
`(Received in original form March 5, 2004; accepted in final form lune 76, 2004)
`Correspondence and requests for reprints should be addressed to Peter R. Byron,
`Ph.D., Professor and Chairman, Department of Pharmaceutics, Wrginia Common-
`wealth University, Richmond, VA 23298—0533. E-maiI: prbyron@vcu.edu
`Proc Am Thorac Soc Vol 1. pp 321-328, 2004
`DOI: 10.1513/pats.200403-023MS
`Internet address: www.atsjournals.org
`
`preSSUIiZed MDIs with non-ozone—depleting hydrofluoroalkanes
`(HFAs), at a time when many older drugs and inhalers were
`becoming subject to competition from the generic industry (1).
`Not surprisingly, there has been considerable commercial incen~
`tive, especially for innovator companies wishing to market new
`drugs, to attempt to launch such drugs in novel inhalers.
`From the drug point of View, inhalers contain mostly short-
`or long~acting topical bronchodilators (adrenergics as well as
`anticholinergics), antiinflammatory steroids, and antiallergics
`such as cromolyn sodiumr(3). We are also seeing proteins being
`delivered by this route for their loeal actions. Drugs like a—l-anti—
`trypsin and other antiproteases, genes, and oligonucleotides are
`all in clinical trial for local effects in the lung. Nebulized thNase
`was launched by Genentech some 5 years ago for treatment of
`patients with cystic fibrosis. Antiviral agents and vaccines are
`also under development for delivery as aerosols to enhance
`“local immunity” in different parts of the respiratory tract (2—6).
`In the systemic drug area, only ergotamine has been traditionally
`delivered through the lung following its presentation as a me-
`tered dose (pressurized) inhaler (3). However, biotech compa-
`nies are attempting to deliver a number of hormones systemically
`via the lungs (e.g. calcitonin, hGH, PTH), and insulin is presently
`the subject of large Phase III trials by both Aventis—Pfizer and
`Novo~N0rdisk with their respective inhalation partners Nektar
`Therapeutics and Aradigrn Corporation (5, 7). In summary, the
`safety of the pulmonary route has gradually gained acceptance
`for macromolecular delivery (8) and there is a tremendous
`amount of interest and drug company research activity in this
`area. This will result in a large number of changes to inhaled
`drug therapy in the future.
`
`IMPROVING DRUG DELIVERY EFFICIENCY
`TO THE LUNG
`
`From the point of View of those wishing to pursue inhaled drug
`development, there are a number of frequent questions that are
`asked. These are presented, along with some rather general
`answers, in Table 1. It is clear from this Table that the choice
`of drug delivery technology to be pursued requires some unique
`expertise. “Efficient” dose delivery to the lung can be discussed
`by defining efficiency as the percentage of the dose depositing
`in the airways (DTL) relative to the delivered dose (dd) or, in
`some cases, the label claim on the inhaler (Table 1; Efficiency =
`100 - DTL/dd). It should come as no surprise to learn that high
`efficiencies are possible but, of course, the price of moving in
`that direction can be considerable. Motivating companies to
`accomplish high delivery efficiencies (in contrast to achieving
`reproducible but inefficient lung penetration) requires that some
`penalties are associated with less efficient delivery. In many
`cases, the lack of a penalty means that these “motivations” do
`not always exist. However, examples may be related to toxicol-
`ogy (e.g., oral corticosteroid deposition associated with candidia-
`sis and unnecessarily large total drug exposure), therapeutic
`need (e.g., insulin and other hormones require peripheral lung
`penetration to enable reasonable bioavailabilities to be achieved;
`3, 5), product misuse by patients (e.g., actual use of product
`encourages additional dosing and an increased incidence of ad-
`
`WATSON LABORATORIES, INC. , IPR2017-01622, Ex. 1030, P. 1 of 8
`
`WATSON LABORATORIES, INC. , IPR2017-01622, Ex. 1030, P. 1 of 8
`
`

`

`322
`
`PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL I
`
`2004
`
`TABLE 1. FAQS CONCERNING INHALER DEVICES WITH ANSWERS RELEVANT
`TO DEVELOPMENT SCIENTISTS
`
`Can N 100% of each "metered dose” be delivered to lung? No, all devices retain some of the (premetered) drug substance,
`and much of what is delivered is often deposited in the oropharynx.
`Can a large proportion of the "ex-mouthpiece dose” be delivered to lung? Yes, the ex-mouthpiece dose, or delivered dose‘,
`can be made to emphasize only the small "more respirable” particles.
`Can the close delivered to the lung (DTL’) be directed preferentially to the peripheral and/or the central ainivays? Yes, this
`is possible for normal individuals but it is not easily achieved for those with lung diseases.
`Are large payloads possible? Yes, values forDTL perinhalation S 4mg are possible. Above this value, multiple breaths or continuous
`inhalation (e.g. nebulizers) must be employed.
`Can “fragile molecules” be delivered intact? Yes, although process and formulation constraints are drug-specific.
`Are 2 2—year product shelf-lives possible? Yes, but formulation packaging and the use of solid-state drug in formulations are
`extremely important features in cases where reproducibility is a problem.
`Can the delivered dose, dd, and its emitted particle size distribution, psd, be made reproducible throughout inhaler lifetime?
`Yes. If this is not so, the product will not be accepted by regulators.
`Are all devices suitable for all drugs? No, the different device platforms are not generally applicable.
`Are some devices easier to develop? Yes, but present/y these are not the most efficient. High delivery efficiency platforms are
`more expensive to develop and launch.
`
`* Delivered dose, dd, is the dose leaving the inhaler mouthpiece when the inhaler is used according to the package insert.
`T The dose leaving the inhaler that deposits in the airways.
`
`verse events; 3, 4), or drug cost (e.g., drug substance costs consti-
`tute a large and prohibitive part of each inhaler’s value; 3).
`To achieve highly efficient drug delivery to lung requires that
`the aerosol creation process be controlled so that a fine droplet
`or particle cloud, containing a known drug concentration in a
`defined size distribution, is metered reproducibly into the pa—
`tient’s inhaled air stream. Provided a normal individual is inhal-
`ing slowly (o3 0.5 L/second), aerosol sizes, expressed as aero-
`dynamic diameters, should preferably be 3 to 7 pm for good
`tracheobronchial deposition, or between 1 and 3 pm for deposi-
`tion in the pulmonary regions (6). Breath holding is known to
`further enhance deposition in the pulmonary or alveolar regions
`(6). Unfortunately, although device designers can often accom~
`plish some of these requirements, patients rarely comply well
`with instruction leaflets given with inhalers. As a result, they
`often require regular counseling to reduce their use of inappro-
`priate inspiratory maneuvers, and in particular, often have diffi-
`culty coordinating their inspiration with the creation of the aero-
`sol cloud by the device (3, 4). For this reason, the reproducibility
`of DTL in vivo is often much worse than the apparent reproduc-
`ibility of the delivered dose assured the regulators during the
`drug development process (DTL and dd have large and small
`variance, respectively; Table 1). Furthermore, the presence of
`acute or chronic obstructive airways disease(s) in patients often
`precludes the delivery of a significant proportion of each dose
`to the lung periphery, irrespective of whether a device is selected
`which enables the predominant production of small aerosol par-
`ticles (1—3 pm) theoretically capable of penetrating the region
`in normal healthy volunteers (6).
`
`(9). There are two major types of hand—held nebulizer (3, 4).
`Air blast or “jet” nebulizers are cheapest and most likely to be
`used clinically, whereas continuous—output ultrasonic nebulizers
`have fallen out of favor recently for several reasons, including
`their inability to nebulize suspension formulations (10). Because
`the pharmaceutical vehicle used in nebulizer formulations is
`essentially aqueous, patients can use nebulizers for prolonged
`periods of time, to deliver quite large doses to the lung; tens of
`milligrams as opposed to micrograms are feasible in many cases.
`It is important to recognize that jet nebulizers can work quite
`well to create less than 5 pm, highly respirable, droplet aerosols
`for delivery of compounds that are either in solution or formu-
`lated as microfine—suspensions and placed in nebulizer reservoirs.
`There is thus no technologic reason why nebulizer therapy
`should not be used for effective drug delivery to the lung. There
`are, of course, economic reasons why they may be less than ideal
`if people are forced to use poorly designed, cheaply manufac~
`tured (variable) nebulizers repeatedly, as has been the case in
`some health centers. One problem then is economic; another
`may be the issue of convenience, because nebulizer systems are
`usually not very portable due to the need for a compressed gas
`supply.
`Most jet nebulizer designs force pressurized gas (usually air)
`from a nozzle (or jet), at high velocity past a liquid feed tube,
`so that the nebulizer solution is atomized at the capillary exit.
`The bulk of the aerosol mist (which may be traveling at up to
`sonic velocity) impacts against a baffle, drains back into the
`reservoir in the base, and recirculates. Only very small droplets
`(< N 5 pm, if the system is running correctly) escape the baffle
`and are available for inhalation. These droplets are produced
`at device-specific airflow rates and are inhaled along with “dilu-
`tion air” through a mouthpiece or mask arrangement. Clearly,
`Thus, the major factors that defeat inhaler design intentions are
`patient variables. Clearly, in the case of nebulizers used with
`some 50% of any aerosol produced by a nebulizer that is continu-
`sedentary patients, especially inhalers designed to deliver me-
`ously producing aerosol cannot be available during the patient’s
`exhalation because they are breathing tidally. Moreover, jet and
`tered doses, the inspiratory flow, tidal volume, and frequency
`baffle designs of different systems have hugely different efficien-
`of breathing are defined by the gas exchange needs of the patient
`cies, resulting in large differences between the times taken to
`and we may consider that some “control” exists over the patient’s
`administer a similar DTL, even from the same solution formula-
`average breathing pattern. If a nebulized aerosol is then pro-
`duced in a continuous fashion, patients may inhale and exhale
`tion. In all cases, aerosol generation quits before the entire drug
`its ouput for several minutes while seated. Unfortunately, the
`has left the reservoir. Most recently, several nebulizer manufac-
`way in which drugs are marketed as nebulizer solutions means
`turers (e.g., Pari, Aerogen) have designed systems to minimize
`that some of the potential “control” offered by this situation is
`‘drug losses during patient exhalation and minimize the times
`usually discarded by companies who market solutions for use
`taken to generate and deposit a given DTL (9). Thus, over the
`with undefined devices, using undefined operating conditions
`last 10 years these devices have become more efficient delivery
`WATSON LABORATORIES, INC. , IPR2017-01622, Ex. 1030, P. 2 of 8
`
`NEBULIZERS
`
`WATSON LABORATORIES, INC. , IPR2017-01622, Ex. 1030, P. 2 of 8
`
`

`

`i
`
`——‘.-::==:..
`
`
`.M.__.¢.Imo-
`
`systems, but only if the physician insists that patients use the
`most modern technology. In general, if we choose reputable
`devices, define the amount of drug placed into the reservoir in
`a uniform volume (e.g., 3 ml), and ask patients to inhale and
`exhale through them until the system quits, then DTL should
`be somewhere between 8 and 14% of that which was initially
`instilled into the reservoir. The remainder is lost during exhala-
`tion or it is retained in the device. Aerogen, Omron, and Pari
`market hand-held ultrasonic (piezoelectric) devices that are
`smaller and less cumbersome than the compressor—driven sys-
`tems described above. These generate slightly larger aerosols
`and can accomplish similar values for DTL more quickly than
`air-blast systems. Although overall efficiencies are likely to con—
`tinue to improve for high-cost systems (perhaps approaching
`20—25% of the instilled dose), the overall efficiency of the present
`piezoelectric systems is frequently found to be similar to that
`for jet nebulizers (3, 4). Most recently, some device manufactur-
`ers have brought to market computer-controlled systems that
`monitor each patient’s breathing pattern and administer nebu-
`lizer output phased with inspiration, whence to enable control
`over DTL (11).
`From a clinical perspective, drugs in nebulizer solutions may
`be deposited in larger doses than those seen with many metered
`delivery devices. Also, patients who are seriously afflicted with
`obstructive lung conditions prefer to use nebulizer therapy, be-
`cause of the nebulizer’s generally more “respirable” output, its
`continuous generation, and its aqueous, often buffered, vehicle
`(12). However, assuming that the drug is an appropriate choice,
`nebulizer therapy can still fail to be effective because of poor
`nebulizer or compressor selection and an unsuitable choice of
`operating conditions. Notably, with more recent drug launches,
`Genentech’s “Pulmozyme” package insert is instructive. In ef-
`fect, this recommends the use of a small number of specific
`nebulizer systems with this product; only those that were found to
`be equivalent for in vitro/thNase delivery during Pulmozyme’s
`development are recommended in the package insert. This “co—
`marketing approach” is likely to occur more frequently for newly
`launched nebulizer drugs. Finally, it is important that nebulizer
`solutions should not be stored in nebulizer reservoirs; stored
`
`solutions may grow bacteria which can then be nebulized and
`inhaled. Devices should be washed and dried between uses, and
`dishwasher compatibility may thus be important.
`
`DOSE-METERING INHALER SYSTEMS
`
`Pressurized metered dose inhalers, dry powder inhalers, or other
`“bolus aerosol”-producing technologies are considered to be
`“controlled dose” or “dose-metering” systems because they are
`sold in forms in which they can only be used with a single drug
`and formulation. We should appreciate, however, that education
`and compliance continue to be issues that need to be addressed
`with all devices, if we hope to be able to control DTL (3, 4, 7).
`Metered aerosols are designed to be inhaled as boluses, unlike
`the homogeneous aerosols distributed throughout inhaled air by
`nebulizers. Bolus aerosols are known to deposit in different
`places, dependent upon when, in the inhalation, the “bolus” is
`released. In addition, many bolus pharmaceutical aerosols are
`unstable physically; that is, they may evaporate and/or have high
`tendencies to impact, decelerate, and/or sediment over short
`time periods.
`From a drug development perspective, there are numerous
`formulation issues that can impact the reproducibility of deliv-
`ered doses and aerosol size distributions from dose-metering in-
`haler systems. A thorough review of this area is beyond the scope
`of this article. However, the subject has been discussed elsewhere
`by the author and others from the point of view of pressurized
`
`metered dose inhalers (1, 13), powder inhalers (3, 14—18), and
`the physwal difficulties that are often found when attempting
`reproducible metering with extremely potent and unstable com—
`pounds, like formoterol (19).
`
`Pressurized MDIs and Accessories
`
`MDIs remain the “gold standard” delivery system in many senses
`even though considerable efforts have had to be made to rede-
`velop acceptable valve and packaging systems, and to reformu—
`late products with HFA propellants (1). HFA-propelled MDIs
`are easily portable, tamper—proof, and multidose. They protect
`the remaining pressurized, liquefied product from oxidation,
`light, and water ingress while providing an inexpensive, mature
`technology with accurate liquid dose metering by volume (1, 3).
`Although reformulated systems have often chosen to mimic the
`inefficiency of the older CFC~propelled formulations, solution
`formulations are often feasible which can achieve values of DTL
`that approach half of the metered dose and exceed 50% of the
`delivered dose (20); this provided that close requirements are of
`the order of 100 ug or less. Moreover, MDIs are apparently
`easy to use, even though a considerable literature exists to show
`that in vivo values for DTL are variable, because of the difficul-
`ties that patients experience trying to coordinate inhalation with
`actuation of the MDI (4). The aerosol drug dose exits the MDI
`mouthpiece as a rapidly moving large droplet cloud. However,
`at about the distance of the back of the throat, the droplet
`diameter is reduced due to propellant evaporation, and a reason-
`able proportion of the “polydispersed” aerosol cloud is now
`small enough to penetrate the lung. It should not be surprising,
`therefore, that a proportion of each “metered dose” is lost in
`the actuator mouthpiece, and a further proportion is lost in the
`oropharynx due to inertial impaction of the “ballistic portion”
`of the spray. Spacers and reservoirs have come onto the market
`as compliance aids. These devices are shown diagrammatically
`in Figure 1; reservoirs can be differentiated from spacers because
`they contain a valve of some description, intended to retain the
`aerosol cloud created by the MDI, until the patient inhales.
`Spacers, on the other hand, contain no such value and simply
`distance the inhaler mouthpiece from the patient’s oropharynx.
`Both devices can reduce drug deposition in the back of the
`patient’s throat and enable extra time for evaporation to occur
`(4). Waste drug that would have been captured at the back of the
`patient’s throat can be partly retained in the spacer or reservoir.
`Notably, there is no reason why MDIs should not be developed
`with an attached spacer or reservoir for just this purpose, al—
`though many of the claims for the advantages of these devices
`fail to exemplify the problems known to be associated with
`multiple actuations into reservoirs, untoward delays between
`cloud generation and inhalation, and the effects of spacer and
`reservoir electrostatic charges; all of these effects dramatically
`reduce the DTL per dose from the MDI (13, 21). Notably, MDIs
`were rarely tested for efficacy or safety with such devices attached
`and their addition could therefore be construed by drug regula-
`tors as an example of product misuse (3, 4).
`Probably the greatest improvements likely to be associated
`with MDI usage in the future will involve the marketing of these
`devices as breath-actuated inhalers. There are many such new
`devices becoming available at present, although 3M has been
`marketing Autohaler (a breath~actuated CFC MDI) for a num—
`ber of years. Figure 1 and its legend describe an expensive option
`called Smartmist, developed by Aradigm Corporation and regis~
`tered (but presently not marketed) in the United States, with a
`number of excellent and sophisticated features (11). The device
`accepts and seals around regular pressurized MDIs and subjects
`these inhalers to microprocessor control. A built-in spirometer
`ensures that the patient receives the drug at the correct point
`
`WATSON LABORATORIES, INC. , IPR2017-01622, Ex. 1030, P. 3 of 8
`
`WATSON LABORATORIES, INC. , IPR2017-01622, Ex. 1030, P. 3 of 8
`
`

`

`324
`
`PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL I
`
`2004
`
`Lion
`
`Evapora
`, S
`
`imwnfl
`
`fitmme mm m if,
`
`WATSON LABORATORIES, INC. , IPR2017-01622, Ex. 1030, P. 4 of 8
`
`WATSON LABORATORIES, INC. , IPR2017-01622, Ex. 1030, P. 4 of 8
`
`
`

`

`‘3
`
`25
`
`These either measure the dose themselves (from a powder reser—
`voir) or they dispense and disperse individual doses which are pre-
`metered into blisters by the manufacturer. Turbohaler and Diskus,
`from AstraZeneca and GlaxoSmithKline, respectively, are rep-
`resentatives of the former and latter categories, although many
`other different designs are presently in development (17, 18, 23).
`In general, the premetered drug-in-blister approach is the easier
`type to develop because the reproducibility of the metered dose
`can be assured during the drug formulation packaging and device
`assembly processes. Provided then, that the device and formula-
`tion design is such that (1 ) the blisters empty well and (2) formula-
`tion and drug adhesion to the device is minimal, the delivered dose
`can usually be shown to the regulators to be reproducible (17, 18).
`With a device like Turbohaler, however, whose operating princi-
`ples are well known (23) and will not be repeated here, the
`powder metering system delivers doses in vitro that are much
`more variable than those derived from premetered blister packs.
`This intrinsic variability, which increases after storage and/or
`transport, must therefore be shown in the clinic (for each sepa-
`rate drug formulation) to have no therapeutic, or toxicologic
`consequences. The effort is complicated further if we observe
`that even with quite sophisticated powder inhaler devices, the
`mode of use and ambient environmental conditions can often
`
`define drug doses reaching the patient’s lungs (15 , 16). The advice
`to “keep your powder dry” continues to hold, and in many of
`these devices, designers and manufacturers have gone to some
`lengths to reduce the likelihood of water vapor ingress creating
`problems. With Turbohaler and others, desiccant is included
`(23) and the overcap should be kept firmly in place when the
`inhaler is not being used. Exhalation into the device, which
`should never be practiced, is also often addressed in patient
`information leaflets.
`
`The systems discussed so far are known as “passive” DPIs,
`because of their reliance upon “patient power” for the purposes
`of drug aerosolization. These all deliver variable doses and show
`delivery efficiencies to the lung which depend on the inspiratory
`effort the patient expends during inhaler use (inhale faster
`through these and DTL, even the delivered dose in some in-
`stances, increases). Ironically, however, clinical studies with Tur-
`bohaler (which shows high in vitro variability; 15) show that its
`in vivo dosing variance is smaller than that of the MDI (24),
`which suffers from high in viva dosing variance, due to coordina-
`tion problems at the patient interface. In vitro, the delivered
`dose can also often be shown to be dependent on a term known
`as “acceleration” or the rate at which a given airflow through
`the inhaler is approached during testing (17). We have illustrated
`
`Byron: Drug Delivery Devices
`
`in the inspiration, (e.g., shortly after they commence inhalation
`from residual volume). It also ensures that they are inhaling
`slowly as fast inhalation will require repeat attempts; an indicator
`light will inform the patient if the dose is inhaled (subsequently)
`at the correct rate. Because the device’s memory can be down-
`loaded, the health professional can check whether or not people
`are complying with recommendations. This type of technology,
`therefore, is clearly one way of removing some of the control
`from the patient, and thus ensuring improved benefits by virtue
`of reducing the variance of in vivo DTL (4, 22); at the very least,
`it should enable improvements to be made in the analysis of
`results during and following ambulatory clinical trials.
`
`Dry Powder Inhalers
`
`The present popularity of novel dry powder inhaler (DPI) devel-
`opment commenced at the time that CFC replacement became
`an issue. These devices are now much more sophisticated than
`used to be the case when only single-dose, capsule—loading sys-
`tems existed (e.g., Spinhaler and Rotahaler from Aventis and
`GlaxoSmithKline, respectively). From a user perspective, the
`number of different designs that are marketed or in development
`will itself create problems for physicians and patient educators.
`We already have difficulty teaching patients to use MDIs; imag-
`ine the increasing complexity as the variety of inhaler options
`increases further. lmportantly, from the point of view of DTL
`variance, most DPIs only deliver drugs when the patient inhales
`through them. As a consequence, the issue of “coordination”
`between actuation and inhaling disappears. However, because
`small volume powder metering is never as precise as the measure-
`ment of liquids, and because DPIs are generally less robust than
`MDls, there are many alternative (and often inhaler—specific)
`ways in which patients can misuse these inhalers. As a result,
`patients may fail to receive therapy for a variety of reasons
`(e.g., exhalation into the device, loading the device in incorrect
`orientations and inappropriate storage conditions may affect
`different DPls in different ways) (4, 15, 16).
`DPI formulations may either be drug mixed with a large
`particle size excipient (or diluent, e.g. lactose), to aid with powder
`flow, or it may consist of drug alone (3, 4). In all cases, the
`powder formulations are cleverly processed using proprietary
`techniques that are responsible for the DPI in question having
`apparently reproducible properties when tested in vitro under
`known, but constant, flow versus time profiles (e.g. for delivered
`dose and size distribution). From the development perspective,
`modern “passive” multidose DPIs (in which the patient provides
`the energy for powder dispersion) fall into two main categories.
`
`Figure 1. Photographs and diagrams of some of the inhalers described in the text. (a) Diagrammatic representation of MD! actuation into reservoir
`device prior to inhalation of the aerosol cloud from the reservoir. (b) Aradigm Corporation’s Smartmist "breath actuator” designed to house an
`existing MDl product and enable microprocessor control of its operation. Ensures actuation at “correct” point of inspiration, an appropriate
`inhalation rate and downloading to a PC for compliance checks (11). (c) Nektar’s Pulmonary Delivery System presently in Phase 11! trials with
`insulin as Pfizer-Aventis’ Exubera product. The device expels a puff of compressed air through a "transjector" inserted into a prepackaged blister
`containing spray dried insulin with proprietary excipients. The air puff creates an aerosol cloud for the patient to inhale through a mouthpiece
`from the integral reservoir atop the device. (d) Boehringer lngelheim’s Respimat "Soft mist inhaler.” A disposable, liquid self—metering device for
`small-volume (15 iii) aqueous and semi-aqueous solutions; lower part of diagram shows the conjunction of high pressure conduits leading to two
`opposed 10pm jets that create the fine mist as the spray streams converge; the company claim that the long duration (> 1 second) of the trigger-
`actuated aerosol cloud will enable "greater coordination and lung delivery” (24). (e) Diagram of Aradigm’s AERX operating principles; premetered
`drug solutions, in sterile blister-packs are pumped through an array of single—use, laser-drilled jets within the drug storage blister; results in efficient
`aerosolization and employs breath actuation and inspiration control, as well as patient education features. Device is expensive but has high delivery
`efficiency, humidity and temperature independence, and feedback on device usage during clinical trials (1 8, 19). (f) Picture of Chrysalis TechnologieS’
`prototype offering liquid metering, vaporization and condensation with claimed "pharmaceutical quality" for some drugs. Liquid is pumped and
`simultaneously subjected to microprocessor-controlled heating, during passage through a "capillary aerosol generator”; device can produce different
`aerosol droplet sizes commencing with MMADs in the submicron range (21, 22).
`
` WATSON LABORATORIES, INC. , IPR2017-01622, Ex. 1030, P. 5 of 8
`
`WATSON LABORATORIES, INC. , IPR2017-01622, Ex. 1030, P. 5 of 8
`
`

`

`326
`
`PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL I
`
`2004
`
`the dependency of the fine particle dose on flowrate for marketed
`cromolyn sodium capsules in Spinhaler (UK product; lactose-
`free formulations) by using a constant throughput of 4 L of
`ambient room air at different flow rates. Whereas the delivered
`
`lations and are designed to minimize the effect that the patient
`has on the particle size distribution of the aerosolized drug dose.
`
`Novel Solution Metering Inhaler Devices
`
`Figures 1d, 1e, and 1f show some of the new solution metering
`devices that are currently in development for use with inhaled
`drugs. Several other companies are active in the area (Aerogen,
`Aradigm, Batelle Pharma, Chrysalis Technologies, Boehringer-
`Ingelheim, and Sheffield Pharmaceuticals; 26—34). All are devel—
`oping “propellant—free” inhalers that offer the advantage of liq—
`uid metering (for dosing precision) and an energy source to
`enable patient-independent, reproducible “active” aerosol pro-
`duction. Some devices are mechanical while others are electro-
`
`dose changed minimally (20 i 3 mg), the fine particle dose
`(dose < 5 pm aerodynamic diameter) differed by more than an
`order of magnitude in the range 30—100 L/minute (15, 16). Not
`surprisingly, all these dependencies are functions of both the
`inhaler and the formulation. For example, Turbohaler (With
`terbutaline sulfate) shows a strong dependence on flowrate,
`whereas Rotahaler and Diskhaler (albuterol sulfate) exhibit
`much less effect (14). To some extent this is predictable because
`the latter devices are rather inefficient and their designs ensure
`mechanical. The latter appear to have some added challenges
`only that they empty fairly well, not that they disperse powder
`from a development perspective, because of the need to demon-
`e

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