throbber
REVIEW
`
`Particulate Matter in Injectable Drug Products
`
`STEPHEN E. LANGILLE, Ph.D.*
`
`Office of Pharmaceutical Science Center for Drug Evaluation and Research Food and Drug Administration 10903
`New Hampshire Ave, Bldg. 51, Rm. 4158 Silver Spring, MD 20993 ©PDA, Inc. 2013
`
`ABSTRACT: Clinicians have had concerns about particulate matter contamination of injectable drug products since
`the development of the earliest intravenous therapeutics. All parenteral products contain particulate matter, and
`particulate matter contamination still has the potential to cause harm to patients. With tens of millions of doses of
`injectable drug products administered in the United States each year, it is critical to understand the types and sources
`of particulate matter that contaminate injectable drug products, the possible effects of injected particulate matter on
`patients, and the current state of regulations and standards related to particulate matter in injectable drug products.
`Today, the goal of manufacturers, regulators, and standards-setting organizations should be to continue to minimize
`the risk of particle-induced sequelae, especially in high-risk patients, without trading unnecessary manufacturing
`burden for minimal safety gains.
`
`KEYWORDS: Injectable, Parenteral, Particulate matter, Pharmaceutical quality, Current good manufacturing practice
`(cGMP).
`
`LAY ABSTRACT: All injectable drug products are contaminated with some level of solid particulate matter, including,
`for example, fibers, dust, rubber, and silicone. These materials enter drug products primarily during the manufacturing
`process. The possible effects on patients of injectable drug products containing particulate matter depend on a number
`of factors. However, given the large number of patients receiving injectable drug products each year in the United
`States and the potential for particulate matter to cause harm to patients, it is critical to continue to minimize particulate
`matter contamination in injectable drug products. Manufacturing standards and regulations have helped improve
`manufacturing quality. Nevertheless, manufacturers, regulators, and standards-setting organizations must continue to
`work toward improving manufacturing quality and minimizing the risk of harm from particle contamination,
`especially in high-risk patients.
`
`Introduction
`
`One of the basic tenets of pharmaceutical quality is the
`manufacture of drug products that are free of micro-
`bial, chemical, and physical contaminants. Although
`microbial contamination of injectable drug products is
`fairly well understood, defined, and measureable, it
`remains difficult to achieve injectable drug products
`that are free of chemical and particulate matter con-
`
`* Corresponding Author: Stephen E. Langille, Ph.D.,
`Office of Pharmaceutical Science Center for Drug
`Evaluation and Research Food and Drug Administra-
`tion 10903 New Hampshire Ave, Bldg. 51, Rm. 4158
`Silver Spring, MD 20993. Telephone: 301-796-1557,
`e-mail: Stephen.Langille@fda.hhs.gov
`doi: 10.5731/pdajpst.2013.00922
`
`tamination. This is due, in part, to the nature of con-
`taminants, the current state of pharmaceutical manu-
`facturing, and the availability of extremely sensitive
`measuring techniques.
`
`Concerns about the clinical use of injectable drugs
`containing particulate matter can be traced to the
`earliest intravenous fluid therapies employed in the
`1830s. An Edinburgh physician named John Mackin-
`tosh, while developing methods of intravenous saline
`infusions to treat victims of a cholera outbreak, rec-
`ommended that the solutions be strained twice through
`leather rather than cotton or linen, which could allow
`“minute portions of flakey threads” to be injected into
`the patient (1). Although processing and filtration
`technologies for intravenous injections have evolved
`exponentially in the years since, concerns about the
`potential effects of injected particulate matter on pa-
`
`186
`
`PDA Journal of Pharmaceutical Science and Technology
`
`Eton Ex. 1108
`1 of 15
`
`

`

`tients continue, especially given the equally exponen-
`tial growth in the number of patients who could be
`affected.
`
`According to the American Hospital Association, U.S.
`hospitals admitted 37,479,709 patients in 2009 (2).
`Assuming an average intravenous solution administra-
`tion of 5 L per patient (3), nearly 190 million L of
`intravenous fluid are administered annually. Given
`these data, an accurate assessment is warranted of the
`factors causing particulate matter contamination of
`drug products, the patient risks associated with the
`administration of such contaminated drug products,
`and the current state of regulations and standards that
`provide the framework for achieving pharmaceutical
`quality.
`
`This article describes some of the sources of particu-
`late matter contamination in injectable drugs and the
`possible clinical effects that can result from such
`contamination. The article also reviews the develop-
`ment of standards and regulations to control contam-
`ination of injectable products and offers some prelim-
`inary next steps for manufacturers, regulators, and
`standards-setting organizations who are working to-
`gether to ensure patient safety.
`
`Classification and Sources of Particulate Matter
`
`Chapter ⬍788⬎ of the United States Pharmacopeia
`(USP), Particulate Matter in Injections (4), defines
`particulate matter as “mobile undissolved particles,
`other than gas bubbles, unintentionally present in the
`solutions”. Groves (5) divided injectable drug partic-
`ulate matter into two classes based on the source of the
`particulate matter: intrinsic particles, defined as those
`originally associated with the solution that were either
`not removed by filtration or precipitated out of the
`solution, and extrinsic particles, defined as those that
`enter the container or solution during manufacturing.
`USP Chapter ⬍1788⬎ Methods for the Determination
`of Particulate Matter in Injections and Ophthalmic
`Solutions (6) provides similar, but more specific, def-
`initions, classifying extrinsic particulate matter as “ad-
`ditive, foreign, unchanging, and not part of the formu-
`lation, package or assembly process”. It classifies
`intrinsic particulate matter as “associated with the
`package, formulation and/or assembly process and ca-
`pable of change upon aging”. USP Chapter ⬍1788⬎
`also notes that intrinsic particulate matter is not the
`same as inherent product characteristics such as the
`haze, coloration, or known populations of small par-
`
`ticles common to certain high-concentration protein
`formulations. This inherent particle category also in-
`cludes the normal particle size distribution of active
`pharmaceutical ingredients in suspensions and other
`common delivery forms (e.g., emulsions, lipids, etc.).
`Inherent particles or properties, when consistent and
`expected, may be completely acceptable.
`
`There are five general sources of particulate matter in
`injectable drug products: the environment, packaging
`materials, solution and formulation components, prod-
`uct packaging interactions, and process-generated par-
`ticles. Proper product development and appropriate
`manufacturing and packaging process design can suc-
`cessfully exclude particulate matter sourced from four
`of the five categories. The fifth category, particulate
`matter sourced from the environment, can be excluded
`only by use of highly controlled filling areas, rather
`than by an intimate understanding of the product,
`process, and container closure system. A list of poten-
`tial particle contaminants, their sources, and intrinsic/
`extrinsic natures as defined by USP Chapter ⬍1788⬎
`is presented in Table I.
`
`Note that certain types of particulate matter, includ-
`ing metal and glass, may be either
`intrinsic or
`extrinsic depending on the point at which they enter
`the container. For example, glass particles can enter
`the manufacturing process from the outside (extrin-
`sic, e.g., through the use of broken or poorly washed
`incoming vials) or come from inside the container
`through degradative change during product storage
`or from process-related glass breakage events (in-
`trinsic, e.g., lamellae, tunnel/oven, or during fill-
`ing). Likewise, metal particles can come from the
`containers, the manufacturing environment (extrin-
`sic, e.g., building materials), or the manufacturing
`process (intrinsic, e.g., blending equipment). Even
`particle levels that meet compendial or company
`target limits can be of concern. For example, so-
`called point-source contamination, which is the pre-
`domination of one particle type (7), may indicate
`the presence of process contribution or package
`instability that requires investigation and remedia-
`tion. An overall understanding of the product and
`processes and the establishment of methods that can
`control particulate matter contamination during de-
`velopment, manufacture, and packaging are essen-
`tial to be able to design systems capable of prevent-
`ing particulate matter
`contamination problems
`before they start (8, 9).
`
`Vol. 67, No. 3, May–June 2013
`
`187
`
`Eton Ex. 1108
`2 of 15
`
`

`

`TABLE I
`Types and Sources of Injectable Particulate Matter
`
`Source
`
`Environment (including
`personnel)
`
`Packaging material
`
`Solution and formulation
`components
`
`Product–package
`interactions
`
`Process-generated
`particulate matter
`
`Particulate Material
`
`Dust
`Fibers
`Biologics—insect parts, microorganisms, pollens
`Fibers of anthropogenic origin
`Hair
`Skin
`Paint/coating chips
`Rust
`Metal (non-product contact types)
`Minerals
`Polymers (unknown source)
`Glass (e.g., carry over from components)
`Extraneous Material (e.g., carry over from
`rubber stopper components)
`Rubber
`Glass
`Polymers
`Silicone
`Precipitates
`Oligomers
`Degradants
`Agglomerates
`Undissolved material
`Glass lamellae
`Silica
`Rubber
`Plastic
`Metal (e.g., stainless steel from processing
`equipment)
`Filter and Consumables fibers
`Glass (from breakage events)
`
`Intrinsic/Extrinsic
`
`Extrinsic
`
`Intrinsic
`
`Intrinsic
`
`Intrinsic
`
`Intrinsic
`
`Clinical Effects of Injected Particulate Matter
`
`Route of Administration
`
`Many clinical effects have been documented in sub-
`jects who have received injections containing partic-
`ulate matter contamination. Examples include phlebi-
`tis (3, 10 –13), pulmonary emboli (14 –16), pulmonary
`granulomas (3, 11, 17), immune system dysfunction
`(3, 18), pulmonary dysfunction (13, 15), infarction
`(15, 19), and death (14, 20 –22). The patient risk
`associated with the injection of drugs containing par-
`ticulate matter depends on a number of factors, includ-
`ing the route of administration used, the particle size
`and shape, the number of particles injected, the parti-
`cle composition, and the patient population.
`
`The route of pharmaceutical product administration
`can influence the deposition of the injected particles,
`the total particle load administered to the patient, and
`the overall risk to the patient. Immunologically inert
`particles, such as glass or cellulosic fibers, delivered
`via intramuscular and subcutaneous routes have re-
`ceived little attention with regard to their potential for
`causing adverse events due to the fact that the deliv-
`ered volumes (and the overall particle load) are rela-
`tively small, the risk of a systemic reaction is low, and
`the ability of these particles to migrate far from the
`injection site is negligible (23). However, vascular
`
`188
`
`PDA Journal of Pharmaceutical Science and Technology
`
`Eton Ex. 1108
`3 of 15
`
`

`

`injections make possible the delivery of greater vol-
`umes of fluids and the broader dissemination and
`deposition of particulate matter throughout the body.
`
`administration should be considered during product
`development when assessing the critical quality attri-
`butes for a given product (26).
`
`Because the size of veins increases in the direction of
`blood flow, most particles injected intravenously will
`travel through the venous system to the heart on their
`way to the lungs via the pulmonary artery. The diam-
`eter of capillaries is approximately 6 – 8 um. As a
`result, most particles larger than 6 – 8 um will remain
`in the pulmonary capillaries, with smaller particles
`passing through the lungs and depositing in organs
`such as the liver and spleen, where they are processed
`by phagocytic cells of the reticuloendothelial system
`(16). Phagocytic overload of the reticuloendothelial
`system by large numbers of particles has the potential
`to block the system and lead to secondary infections in
`a debilitated host (3). There is little information in the
`literature regarding the ability of the immune system
`to clear relatively large (⬎10 um) inorganic particles
`(e.g., rubber, glass, and metal) lodged in organs such
`as the lung or what effect, if any, the accumulation of
`such particles in vital organs may have over time.
`
`Because arteries decrease in size with the direction of
`blood flow, the inadvertent administration of intra-
`arterially injected particles that are too large to pass
`through arterioles and capillaries may cause occlu-
`sions that could affect blood flow to tissues down-
`stream of the injection site. The physiological effects
`of any such occlusion will depend upon the size of the
`particle and the collateral circulation available to the
`affected area (23). Ironically, smaller particles capable
`of blocking terminal arterial vessels—and causing in-
`farctions—may be more detrimental than larger parti-
`cles capable of arteriole occlusion due to the reduced
`collateral blood supply available to the affected tissue
`(24). The inadvertent intravascular injection of corti-
`costeroid formulations containing particles has been
`linked to adverse central nervous system sequelae in
`humans not observed with non-particulate steroid for-
`mulations (24). A study involving pigs injected in the
`vertebral artery with particulate- or non-particulate-
`based steroids yielded similar results, with pigs receiv-
`ing the particulate-containing steroids displaying brain
`stem edema and significant tissue damage (25).
`
`Other routes of administration, such as the intrathecal,
`epidural,
`intraocular, and intracranial
`routes, may
`carry different risks due to the direct delivery of the
`particulate matter to specific areas of the body. The
`risks of particulate matter delivered via these routes of
`
`Size and Shape
`
`The size and shape of an injected particle can affect
`both its deposition within the body and its clinical
`effects on the subject. Rabbits injected with radiola-
`beled polystyrene particles of different sizes showed
`rapid deposition of 15.8 um particles in the lungs
`while 1.27 um particles were deposited mainly in the
`liver (16). Similar results were obtained when dogs
`were injected intravenously with radiolabeled micro-
`spheres of 3, 5, 7, and 12 um in diameter. The 7 and
`12 um particles were deposited primarily in the lungs,
`while the 3 and 5 um particles migrated mainly to the
`spleen and liver. As expected, clearance from the
`bloodstream was size-dependent, with the larger par-
`ticles clearing first (27). Rabbits injected with 5 um
`diethylaminoethyl (DEAE) cellulose fibers demon-
`strated deposition primarily in the lungs, but also in
`the liver and kidneys (16). Rabbits injected intrave-
`nously with 30 um DEAE cellulose fibers died within
`4 minutes of administration due to an acute toxic
`response (tachycardia, dyspnoea, dystaxia) caused by
`pulmonary emboli (16). In contrast, 40 to 60 um
`DEAE cellulose microspheres, although entrapped by
`the lung, caused no adverse reactions and each of the
`rabbits injected survived until the completion of the
`study (16). These studies suggest that the shape of a
`particle may be just as important as its size when
`determining its potential for harm. Certainly the total
`particle load must be considered as well.
`
`Due to the obvious challenges associated with con-
`trolled clinical studies to investigate the effects of
`injected particles in humans, little is known about the
`risk to diverse patient populations posed by particles
`of various sizes, shapes, and composition injected via
`different routes of administration. Adverse event re-
`ports and autopsy results are the only sources of
`information about the effects of larger particles on
`patient populations. Visible particulate matter com-
`posed of calcium salt precipitates in drug admixtures
`has caused a number of serious clinical events (21). In
`1994, two young female patients undergoing treatment
`for pelvic infections died of pulmonary emboli follow-
`ing intravenous administration of total nutrient admix-
`tures containing FreAmine III as an amino acid source
`(14, 20). Analysis of the precipitate isolated from the
`admixtures administered to each patient revealed the
`
`Vol. 67, No. 3, May–June 2013
`
`189
`
`Eton Ex. 1108
`4 of 15
`
`

`

`presence of calcium and phosphorous salts matching
`those found in the pulmonary microvasculature of the
`autopsy specimens. Co-administration of the antibiotic
`ceftriaxone and calcium-containing intravenous solu-
`tions to neonates resulted in eight adverse event re-
`ports and seven deaths. One patient experienced car-
`diopulmonary arrest after a white precipitate in the
`patient’s intravenous tubing was pushed into the infant
`in an effort to clear the tubing (28). Pulmonary emboli
`were reported in multiple cases, and autopsies re-
`vealed the presence of white crystalline precipitates in
`the lungs, heart, kidney, and liver (21, 28). Both the
`ceftriaxone and FreAmine III incidents resulted in the
`issuance of U.S. Food and Drug Administration (FDA)
`drug safety warnings regarding the potential for cal-
`cium precipitation in these drug products (28, 29).
`Cant et al. (22) reported the case of a premature
`neonate who was treated with an umbilical artery
`catheter shortly after birth. Injections were made into
`the catheter using polypropylene syringes. The cathe-
`ter was removed on day 4, but the patient soon devel-
`oped abdominal distension and died at 52 days of age.
`An autopsy revealed acute infarction of the small
`bowel and the presence of polypropylene fragments of
`50 to 200 um in size. Although this may be the only
`documented case of a fatality resulting from injection
`of material derived from a pharmaceutical container
`closure system, the case underscores the vulnerability
`of neonates to sequelae resulting from the infusion of
`particles and suggests that the intra-arterial route of
`administration may carry additional risks.
`
`Number
`
`Estimates are that patients in intensive care receive
`more than a million injected particles ⬎2 microns in
`size daily (18, 30, 31). One method for controlling the
`particle load administered to critically ill patients has
`been through the use of final filters. A controlled
`clinical study of 88 infants receiving either filtered or
`unfiltered infusions via a central line revealed signif-
`icant reductions in the incidence of complications such
`as thrombi and necrotizing enterocolitis (32). Studies
`on adult patients using 0.22 and 0.45 um intravenous
`in-line filters seem to indicate that the use of in-line
`filters reduced the incidence and time of onset of
`particle-induced phlebitis (3). In vitro studies also
`showed that human macrophages and epithelial cells
`displayed decreased cytokine production following
`exposure to silicone particles mimicking those ob-
`tained from intravenous line filters obtained from pe-
`diatric intensive care units (18). However, the use of
`
`final filters may present other problems, such as the
`possibility of drug product reaction with or absorption
`by the filter material or impaired fluid flow through the
`filter. Opinions vary regarding the economic benefit of
`in-line filtration to remove microorganisms and par-
`ticulate matter during drug product infusion (32–36).
`Nevertheless, a review of clinical case reports involv-
`ing calcium phosphate precipitation in intravenous
`admixtures revealed that the use of in-line filtration
`made the difference between non-fatal and fatal cases
`(37). Thus, the use of in-line filtration for extempora-
`neously prepared, multi-component
`intravenous ad-
`mixtures may be prudent.
`
`Composition
`
`Barber (23) provides an excellent review of several
`pre-1980 animal studies involving different types of
`particulate matter (filter paper, glass, rubber, hair,
`polystyrene, plastic, and insoluble drug residues) in
`various animal models (rabbits, dogs,
`rats, mice,
`guinea pigs, and hedgehogs). The clinical effects seen
`in these studies range from relatively minor tissue
`damage associated with the administration of silicone
`and polystyrene particles to rabbits and dogs, to more
`serious reactions such as local inflammation, the for-
`mation of pulmonary granulomas, and death in rabbits,
`dogs, and rats injected with plastics, ground filter
`paper, or large numbers of polystyrene particles ⬎40
`um in size.
`
`One of the most common contaminants of injectable
`drug products is glass derived from the manufacturing
`process, reaction of the drug with the container closure
`system, or that produced by opening glass ampoules
`(36, 38, 39, 40). Recent glass delamination issues
`involving multiple drug products have increased con-
`cern about the risk posed by glass particles and interest
`in developing methods to control
`the formation of
`glass lamellae over the product shelf life (40, 41).
`Sequelae attributed directly to glass particles include
`phlebitis (3), pulmonary granulomas (31), systemic
`inflammatory response syndrome (18), and adult re-
`spiratory distress syndrome (34). Studies have also
`suggested that glass particle–induced sequelae may
`require considerable time to develop and, as a conse-
`quence, may often be overlooked (38, 39, 42).
`
`Another common pharmaceutical contaminant is metal
`particles (43, 44). Although the most common source
`of metal particles is processing equipment, they have
`also been found to contaminate the raw materials used
`
`190
`
`PDA Journal of Pharmaceutical Science and Technology
`
`Eton Ex. 1108
`5 of 15
`
`

`

`in drug product formulation (43). Lead and chromium
`are considered among the most dangerous metal con-
`taminants, but serious adverse events related to alu-
`minum ingestion have also been well documented
`(43– 46). Aluminum toxicity in premature infants has
`been linked to total parenteral nutrition admixtures
`(45, 46) and contributed to the issuance of FDA reg-
`ulations regarding the aluminum content of drug prod-
`ucts used for total parenteral nutrition (47). By far, the
`most common and expected type of metal particles
`found in liquid injectables is stainless steel (44). Re-
`cent drug product recalls due to the presence of stain-
`less steel particles in lipid emulsions requiring high
`sheer force manufacturing processes have necessitated
`the development of modified manufacturing processes
`and visual inspection methods to detect potentially
`harmful levels of metallic particles (48).
`
`In the class of inherent particles, proteinaceous par-
`ticulate matter poses a unique risk to the patient be-
`cause of the unintended host immune responses it may
`elicit (49). Host responses resulting in antibody-me-
`diated neutralization of the protein’s active site or the
`production of drug-induced antibodies to the therapeu-
`tic version of an endogenous protein have the potential
`to cause catastrophic cross-reaction and neutralization
`of the endogenous protein (49, 50). Although the
`precise characteristics of proteinaceous particles asso-
`ciated with immune response elicitation are not well
`understood (50, 51), it is believed that protein aggre-
`gation and the formation of repetitive arrays of anti-
`gens are contributing factors in the elicitation of an
`immune response (49).
`
`The causative factors of protein aggregation are also
`poorly understood. However, the complex manufac-
`turing and purification methods used in the processing
`and handling of therapeutic proteins can influence a
`number of different product characteristics, including
`particle size (49, 50, 52). One potential cause of
`protein aggregation is the presence of extrinsic partic-
`ulate matter that may serve as nucleation sites for the
`formation of larger particles (52). The overall net
`charge of the protein solution has also been shown to
`influence the degree of protein aggregation and the
`type of protein aggregates formed in a given solution.
`Studies have shown that solubilized proteins held un-
`der conditions near their isoelectric point may be
`prone to aggregation into spherical particles due to
`exposure of hydrophobic residues to the solvent
`caused by the solution’s low net charge (52, 53).
`Alternatively, proteins held under conditions of high
`
`net charge tend to organize into amyloid fibrils due to
`electrostatic repulsion and slow aggregation (53). The
`potential for aggregation and the monitoring of sub-
`visible protein particles in the 0.1 to 10 um size range
`should be considered during biologic product devel-
`opment and surveillance programs (49).
`
`Finally, it is important to distinguish between hard and
`soft particles. A hard particle is a rigid structure that
`often has a non-spherical, irregular shape. Such particles
`are inflexible and, if large enough (⬎5 ␮m), are more
`likely to produce mechanical obstruction and vascular
`emboli. All of the particles described above would be
`considered hard particles capable of inducing embolic
`phenomena upon intravascular (i.e., intravenous, arterial)
`infusion. In contrast, a soft particle is a flexible, or
`deformable, structure that is spherical, such as an emul-
`sified oil droplet. As with hard particles, soft particles
`may also produce embolic phenomena upon intravascu-
`lar infusion, but due to their deformable characteristics,
`the embolism is incomplete, that is, when large hard
`particles flow towards a vessel with a smaller diameter,
`the occlusion cannot be overcome by compensatory in-
`creases in venous and/or arterial pressures. When the
`same-sized soft particles flow towards the same small
`vessel, the occlusion can be overcome due to the malle-
`ability of the lipid droplet. As the occlusion clears,
`however, the large-diameter droplets can accumulate in
`downstream vital organs, such as the liver, producing a
`systemic inflammatory response (54, 55).
`
`Patient Population
`
`The patient populations that may be most at risk for
`particulate matter–related sequelae include patients with
`existing tissue damage, critically ill patients, and neo-
`nates (3, 15, 18, 31, 38, 55). Two recent animal studies
`investigated the effects of injected particles in the pres-
`ence of pre-existing tissue damage. Schaefer et al. (15)
`demonstrated that the injection of particles from two
`generic antibiotics containing particulate matter levels 4
`to 50 times higher than those found in the innovator drug
`resulted in a nearly 50% loss of damaged capillary net-
`works in the ischemic muscle tissue of hamsters as
`compared to muscle treated with the innovator drug
`product. Passing the generic formulations through a
`0.2 um filter prior to administration eliminated the dam-
`aging effect of the generic antibiotics (15). Lehr et al.
`(56) conducted a similar study comparing the clinical
`effects of particles from the innovator and generic man-
`ufacturers of cefotaxime. Although the capillary perfu-
`sion of healthy muscle was not affected by the intrave-
`
`Vol. 67, No. 3, May–June 2013
`
`191
`
`Eton Ex. 1108
`6 of 15
`
`

`

`nous injection of the concentrated antibiotic particles,
`post-ischemic muscle tissue demonstrated reduced cap-
`illary perfusion following intravenous treatment with the
`concentrated particle solutions. Histological sections re-
`vealed that particulate matter caused mechanical disrup-
`tion of the circulation to striated muscle. These findings
`suggest that injected particles could be more detrimental
`to patents with existing tissue damage, such as in the case
`of trauma, surgery, or sepsis (13).
`
`The heavy particle loads incurred by critical care
`patients due to the sheer volume of administered in-
`travenous solutions have been linked to adult respira-
`tory distress syndrome (34), systemic inflammatory
`response syndrome (SIRS) (57), and immune system
`dysfunction (3). Walpot et al. (34) used energy-dis-
`persive x-ray analysis to show that critical care pa-
`tients may be more susceptible to particulate matter
`deposition in pulmonary tissue than healthy subjects.
`Additional studies have suggested that in-line filtra-
`tion may benefit critically ill infants by offering pro-
`tection against SIRS and other glass particle–induced
`adverse events (38, 57).
`
`The potential effect of heavy particle loads on neo-
`nates was also reported by Puntis et al. (31), who
`compared the necropsy results of 32 infants who died
`of sudden infant death syndrome with those of 41
`infants who died following total parenteral nutrition
`therapy. Two of
`the 41 parenterally fed patients
`showed widespread pulmonary granulomas containing
`material such as glass fragments and cotton fibers. No
`such granulomas were identified in the patients who
`expired due to sudden infant death syndrome. Al-
`though the capillary diameter of neonates is the same
`as those of adults, the overall number of blood vessels
`and the diameter of the major blood vessels are
`smaller in children as compared to adults, a factor that
`could accentuate the effects of injected particles rela-
`tive to the effects seen in adult patients.
`
`Relevant Regulations and Standards
`
`Over the years, a number of statutes and regulations have
`been enacted and implemented intended to control par-
`ticulate matter contamination of injectable drug products.
`Nevertheless, federal regulations pertaining to current
`good manufacturing practice (cGMP) do not specifically
`address the subject of particulate matter, but do contain
`several passages applicable to particulate matter contam-
`ination. With regard to the effect
`that
`the container
`closure system might have on particulate matter in a
`
`product, regulations at 21 CFR 211.94(a) state that “drug
`product containers and closures shall not be reactive,
`additive or absorptive so as to alter the safety, identity,
`strength, quality, or purity of the drug beyond the official
`or established requirements”. Regulations at 21 CFR
`211.165(a) and (f) state that “for each batch of drug
`product there shall be appropriate laboratory determina-
`tion of satisfactory conformance to final specifications
`for the drug product” and “drug products failing to meet
`established standards or specifications . . . shall be re-
`jected”. These regulations apply to the visible and sub-
`visible particulate matter specifications cited in drug
`product applications (see Table II).
`
`Section 501 of the Federal Food, Drug, and Cosmetic
`Act (FD&C Act) states that a drug or device will be
`considered adulterated (1) “if it has been prepared,
`packed, or held under insanitary conditions whereby it
`may have been contaminated with filth, or whereby it
`may have been rendered injurious to health”; (2)
`“if . . . the facilities or controls used for its manufac-
`ture, processing, packaging, or holding do not conform
`or are not operated or administered in conformity with
`current good manufacturing practice to assure that the
`drug . . . meets the quality and purity characteristics,
`which it purports or is represented to possess”; or (3)
`“if it purports to be or is represented as a drug the
`name of which is recognized in an official compen-
`dium, and its strength differs from, or its quality or
`purity falls below,
`the standards set forth in such
`compendium”. Although a manufacturer’s level of
`compliance with Section 501 of the FD&C Act may be
`subject to interpretation, the emphasis on cGMP and
`compendial standards is clear.
`
`The first compendial standard for visible particles in
`drugs for use in the United States came in 1936 when
`the National Formulary VI stated that injectable so-
`lutions were to be “substantially free from precipitate,
`cloudiness or turbidity, specs or flecks, fibers or cotton
`hairs, or any undissolved material” (58). In 1942, the
`USP and the American Pharmaceutical Association
`(publisher of the National Formulary at
`that
`time)
`stated that aqueous injections should be “substantially
`free” of particles discernable with the naked eye. This
`definition eventually evolved into those currently used
`by the three major pharmacopeia stating that injectable
`drug products should be “essentially free” (USP),
`“practically free” (European Pharmacopeia) or free of
`“readily detectable” (Japanese Pharmacopeia) visible
`particles (58).
`
`192
`
`PDA Journal of Pharmaceutical Science and Technology
`
`Eton Ex. 1108
`7 of 15
`
`

`

`matter”
`particulate
`visible
`changein
`
`Criteria—“Clear”
`2.9.20Acceptance
`
`container
`
`“Nosignificant
`
`EuropeanPharmacopeia.
`
`2.3⫾2.3
`
`153⫾33.8
`
`⬍788⬎
`
`6000/600per
`
`N/A
`
`Nospecification
`
`6.8⫾11.7
`
`423⫾157.9
`
`⬍788⬎
`
`provided
`
`visible.”
`particleswhichare
`containoneormore
`Solutionmustnot
`
`“Pass”
`
`“Complies”
`
`“Solutionmustbeclear.
`Particles”USP⬍1⬎
`
`“FreeofVisible
`
`1⫾1.4
`
`0⫾0
`
`121.5⫾92.6
`
`133.3⫾57.7
`
`provided
`
`N/A
`
`N/A
`
`Nospecification
`
`11

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