throbber
Journal of Liquid Chromatography & Related Technologies
`Copyright # Taylor & Francis Group, LLC
`ISSN 1082-6076 print/1520-572X online
`DOI: 10.1080/10826070701191151
`
`w
`
`, 30: 877–935, 2007
`
`Identification of Pharmaceutical Impurities
`
`Fenghe Qiu and Daniel L. Norwood
`Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, USA
`
`Abstract: Identification of pharmaceutical impurities is a critical analytical activity in
`the drug development process whose goal is to fully elucidate the chemical structures
`of unknown pharmaceutical impurities present in either drug substances or drug
`products above a particular threshold. Knowledge of the chemical structure of an
`impurity is essential to assess its toxicological implications and to gain an understand-
`ing of its formation mechanism. Knowledge of the formation mechanism is critical for
`improving the synthetic chemical processes and optimizing the drug formulation to
`reduce or eliminate the impurity. This article reviews the current regulatory require-
`ments for impurity identification and the chemical origins of various impurities, par-
`ticularly those derived from degradation of drugs. Strategies for identification of
`pharmaceutical impurities are discussed followed by an overview of the critical
`steps and the roles of various analytical techniques, such as HPLC-DAD, LC-MS,
`LC-NMR, GC-MS, and NMR, in pharmaceutical impurity identification, with an
`emphasis on applications of mass spectrometry based hyphenated techniques.
`Carefully selected examples are included to demonstrate key points in impurity
`formation and the appropriate application of various analytical techniques.
`
`Keywords: Pharmaceutical impurities, Hyphenated techniques, Impurity identification
`
`INTRODUCTION
`
`Impurity profiling and control is one of the most regulated areas in the pharma-
`ceutical industry.[1 – 7] According to ICH Q3A (R) “Impurities in the New
`Drug Substance.”[1] and ICH Q3B (R) “Impurities in the New Drug
`Product”,[2] a drug substance impurity is “any component of the new drug
`substance that
`is not
`the chemical entity defined as the new drug
`
`Address correspondence to Fenghe Qiu, Boehringer Ingelheim Pharmaceuticals Inc.,
`900 Ridgebury Rd., Ridgefield, CT 06877, USA. E-mail: fqiu@rdg.boehringer-
`ingelheim.com
`
`877
`
`IPR2020-00770
`United Therapeutics EX2021
`Page 1 of 59
`
`

`

`878
`
`F. Qiu and D. L. Norwood
`
`substance,” and a drug product impurity is “any component of the new drug
`product that is not the drug substance or an excipient in the drug product.”
`In a pharmaceutical product, an impurity is first and foremost a quality
`issue, since it could potentially compromise the efficacy of the drug
`product. Secondly, impurities also cause safety concerns.[9 – 12] Hence, any
`impurity present in a drug product must be fully understood, both qualitatively
`(chemically) and quantitatively, and qualified, if necessary, through toxico-
`logical assessment. From a chemical perspective, pharmaceutical impurities
`are inevitable because no chemical reaction has 100% selectivity and no
`chemical compound is “rock” stable. Nonetheless, it is possible to reduce
`impurities via synthetic process improvement and appropriate preformulation/
`formulation studies. Knowing the structure of an impurity is essential for
`allowing assessment of its toxicological implications and for understanding its
`formation mechanisms, which is critical knowledge for
`improving the
`synthetic chemical process and optimizing the formulation. Impurity identifi-
`cation is a specialized field in the pharmaceutical industry which requires
`specialized analytical facilities and expertise.[13 – 21] The goal of this review is
`to provide an overview of the current regulatory requirements on impurity
`identification, the chemical origins of various impurities, and the strategies
`and roles of various analytical techniques in pharmaceutical impurity identifi-
`cation, with an emphasis on mass spectrometry based hyphenated techniques.
`The examples presented in this review are used to illustrate key points in
`impurity formation chemistry and applications of various analytical techniques,
`particularly mass spectrometry based hyphenated techniques.
`
`REGULATORY REQUIREMENTS FOR PHARMACEUTICAL
`IMPURITY IDENTIFICATION
`
`ICH guidelines[1 – 3] classify impurities into three categories: organic impurities,
`inorganic impurities, and residual solvents. These impurities can be from a
`variety of sources, as given in Table 1. Organic impurities are derived from
`drug substance synthetic processes and degradation reactions in drug substances
`
`Impurity classification based on ICH guidelines Q3A(R),[1]
`Table 1.
`Q3B (R),[2] and Q3C[3]
`
`Organic impurities
`
`Inorganic impurities
`
`Residual solvents
`
`Starting materials
`Intermediates
`By-products
`Degradation products
`Reagents, ligands and catalysts
`Reagents, ligands and catalysts
`Heavy metals or other residual metals
`Inorganic salts
`Inorganic or organic liquids
`
`IPR2020-00770
`United Therapeutics EX2021
`Page 2 of 59
`
`

`

`Identification of Pharmaceutical Impurities
`
`879
`
`Table 2.
`ICH reporting, identification and qualification threshold for organic impuri-
`ties in drug substance[1]
`
`Maximum
`daily dosea
`2 g/day
`
`.2 g/day
`
`Reporting
`threshold
`
`0.05%
`
`0.03%
`
`Identification
`threshold
`
`Qualification
`threshold
`
`0.1% or 1.0 mg TDIb,
`which ever is lower
`0.05%
`
`0.15% or 1.0 mg TDI,
`which ever is lower
`0.05%
`
`aThe amount of drug substance administered per day.
`bTDI: Total daily intake of the impurity.
`
`and drug products. Synthetic process related impurities can be derived from
`starting materials, intermediates, reagents, ligands, and catalysts used in the
`chemical synthesis, as well as by-products from the side-reactions of the
`chemical synthesis. Degradation products are derived from the chemical degra-
`dation of drug substances and drug products under storage or stress conditions.
`Control of the organic impurities in new drug substances is based on the
`Maximum Daily Dose and total daily intake (TDI) of the impurities. Table 2
`provides the ICH threshold for control of the organic impurities in new drug
`substances.[1] Depending on whether the Maximum Daily Dose higher or
`lower than 2 g, organic impurities in a new drug substance at (or greater than)
`0.05% or 0.1% require identification. Note that these thresholds do not apply
`to toxic impurities.[7,22,23] According to EMEA CHMP recommendations,[7]
`genotoxic impurities should be controlled based on compound-specific risk
`assessment. For an unstudied impurity (such risk assessment data do not
`exist), a threshold of toxicology concern (TTC) of 1.5 mg/day can be applied,
`with exceptions for aflatoxin-like, N-nitroso- and azoxy-compounds, which
`should be assessed based on compound-specific toxicity data.
`Control of organic impurities in new drug products are outlined in Table 3
`and Table 4.[2] Note that these thresholds are not the same as those for impu-
`rities in new drug substances given in Table 2. Based on the Maximum Daily
`Dose, the identification thresholds for organic impurities in new drug products
`are divided into 4 groups to give more consideration to low dose drug
`products. For most new drug products,
`the Maximum Daily Dose is
`between 10 mg – 2 g/day, therefore, any impurities at 0.2% or greater would
`have to be identified.
`
`Table 3. Reporting thresholds for impurities in new drug
`product[2]
`
`Maximum daily dose
`
`Reporting threshold
`
`1 g
`.1 g
`
`0.1%
`0.05%
`
`IPR2020-00770
`United Therapeutics EX2021
`Page 3 of 59
`
`

`

`880
`
`F. Qiu and D. L. Norwood
`
`Table 4. Identification thresholds for organic impurities in new drug
`product[2]
`
`Maximum daily dose
`
`Identification threshold
`
`,1 mg
`1 – 10 mg
`.10 mg22 g
`.2 g
`
`1.0% or 5mg TDI, which ever is lower
`0.5% or 20 mg TDI, which ever is lower
`0.2% or 2 mg TDI, which ever is lower
`0.1%
`
`Leachables are a separate class of drug product impurities, however,
`control of leachbales is not covered by ICH guidance.[2] Recently, the PQRI
`(Product Quality Research Institute) Leachables and Extractables Working
`Group proposed safety and qualification thresholds for leachables in OINDP
`(Orally Inhaled and Nasal Drug Products) to the regulatory authorities.[8]
`The proposed Safety concern threshold (SCT) is 0.15 mg/day, and the Quali-
`fication Threshold (QT) is 5 mg/day for an individual leachable in an OINDP.
`Note that proposed safety thresholds only apply to OINDP and not other drug
`product type (e.g., injectables, ophthalmic, etc.)
`Inorganic impurities are, in most cases, introduced from the synthetic
`process of the drug substance (e.g., catalyst), or as impurities present in exci-
`pients. Analysis and control of inorganic impurities usually follows pharma-
`copoeial monographs or other appropriate procedures and will not be
`discussed further in this review.
`Residual solvents are defined as organic volatile chemicals that are used
`or produced in the manufacture of drug substances or excipients, or in the
`preparation of drug products.[3] Residual solvents are divided by a risk assess-
`ment approach into three classes.
`Class 1 solvents are known human carcinogens, strongly suspected human
`carcinogens, and environmental hazards; therefore, these solvents should be
`avoided in the production of drug substance, excipients, or drug products,
`unless their use can be strongly justified in a risk-benefit assessment. If unavoid-
`able, the level of an individual Class 1 residual solvent should be strictly
`controlled below the concentration limits (for example the limit for benzene is
`2 ppm).[3] Class 2 solvents are non-genotoxic animal carcinogens or possible
`causative agents of other irreversible toxicity such as neurotoxicity or teratogeni-
`city. Class 2 solvents are controlled according to the PDEs (Permitted Daily
`Exposure) and Maximum Daily Dose (Option 1 and Option 2). ICH Q3C[3]
`provides PDEs of all Class 2 solvents. Class 3 solvents are solvents with low
`toxic potential to man. It is recommended that amounts of these residual
`solvents of 50 mg per day or less would be acceptable without justification.
`For solvents for which no adequate toxicological data are available, man-
`ufacturers should supply justification for residual levels of these solvents in
`pharmaceutical products.
`
`IPR2020-00770
`United Therapeutics EX2021
`Page 4 of 59
`
`

`

`Identification of Pharmaceutical Impurities
`
`881
`
`ORIGINS OF PHARMACEUTICAL IMPURITIES
`
`Organic impurities can be originated from a variety of sources in both drug
`substances and drug products. Generally, the origins of impurities can be
`categorized into the following sources: synthetic process of
`the drug
`substance, degradation of the drug substance, container/closer and packing
`materials, and extraneous contaminants.
`
`Impurities Originating from Drug Substance Synthetic Processes
`
`Most small molecule drug substances are chemically synthesized. Chemical
`entities, other than the drug substance, that are involved or produced in the
`synthetic process can be carried over to the final drug substance as trace
`level impurities. These chemical entities include raw materials, intermediates,
`solvents, chemical reagents, catalysts, by-products, impurities present in the
`starting materials, and chemical entities formed from those starting material
`impurities (particularly those involved in the last steps of the synthesis).
`These impurities are usually referred to as process impurities. The goal of
`process impurity identification is to determine the structures and origins of
`these impurities. This knowledge is critical for improving the synthetic
`chemical process, in order to eliminate or minimize process impurities.
`
`Process Impurities Originating from Starting Materials and
`Intermediates
`
`Starting materials and intermediates are the chemical building blocks used to
`construct the final form of a drug substance molecule. Unreacted starting
`materials and intermediates, particularly those involved in the last a few
`steps of the synthesis, can potentially survive the synthetic and purification
`process and appear in the final product as impurities.[23] For example, in the
`synthesis of tipranavir drug substance, the “aniline” is the intermediate in
`the last step of the synthesis.[24] Because the similarity between the structures
`of the “aniline” and the final product, it is difficult to totally eliminate it in the
`subsequent purification step. Consequently, it appears in the drug substance at
`around 0.1%.
`
`Process Impurities Originating from Impurities in the Starting Materials
`
`Impurities present in the staring materials[25 – 27] could follow the same
`reaction pathways as the starting material itself, and the reaction products
`could carry over to the final product as process impurities. Knowledge of
`the impurities in starting materials helps to identify related impurities in
`the final product, and to understand the formation mechanisms of these
`related process impurities. An often encountered scenario involves starting
`
`IPR2020-00770
`United Therapeutics EX2021
`Page 5 of 59
`
`

`

`882
`
`F. Qiu and D. L. Norwood
`
`materials containing structural analogs, positional isomers, etc., as trace
`level impurities. These impurities are difficult to separate and remove from
`the starting material during its manufacturing process. When the starting
`material is used for synthesis of a drug substance, these impurities will
`likely form analogs or isomers of the desired drug substance. Some real
`world examples of process impurities derived from impurities in starting
`materials were reported by Go¨ro¨g et al.[28 – 30] One such example is the
`presence of a 4-trifluoromethyl positional isomer in 3-trifluoromethyl-a-ethyl-
`benzhydrol (flumecinol), due to the presence of 4-trifluoromethylbenzene
`impurity in the starting material, 3-trifluoromethylbenzene. A second
`example involves a 2-methyl analogue present as a trace impurity in tolperi-
`sone, due to the presence of 2-methylpropiophenone in the starting material,
`4-methylpropiophenone.
`
`Process Impurities Originating from Chemical Reagents, Ligands and
`Catalysts
`
`Chemical reagents, ligands, and catalysts used in the synthesis of a drug
`substance can be carried over to the final products as trace level impurities.
`For example, carbonic acid chloromethyl
`tetrahydro-pyran-4-yl ester
`(CCMTHP), which is used as an alkylating agent in the synthesis of a ß-
`lactam drug substance, was observed in the final product as an impurity.[31]
`Many chemical reactions are promoted by metal based catalysts. For
`instance, a Ziegler-Natta catalyst contains
`titanium, Grubb’s catalyst
`contains ruthenium, and Adam’s catalyst contains platinum, just to name a
`few. It is expected that these heavy metals will appear in drug substances at
`trace levels when such catalysts are employed. Most heavy metals have
`safety concerns; therefore, specifications and analytical methods should be
`in place to monitor the heavy metals involved in the process.[32,33] Garrett
`et al.[34] recently reviewed the practices for meeting palladium specifications
`in drug substances. In some cases, reagents or catalysts may react with
`intermediates or final products to form by-products. Go¨ro¨g et al.[35] reported
`that pyridine, a catalyst used in the course of synthesis of mazipredone,
`reacts with an intermediate to form a pyridinium impurity. Figure 1 shows
`a process where t-BuOK (potassium t-butyloxide) is used to extract a
`proton from an intermediate and promote the coupling of two intermediates
`to form a drug substance. During this process, t-butyloxyl also attacks
`the cyclopentanol ester moiety of one intermediate to form an impurity
`with a molecular weight of M-12 (M being molecular weight of the
`intermediate).
`Another example of reagent related impurities is the observation of
`elemental sulfur (S8) in a batch of bulk drug. In this case, the last step of
`the synthesis involves Na2S2O3 as a reducing reagent
`to scavenge the
`residual oxidant. When Na2S2O3 is used in excessive amount, elemental
`
`IPR2020-00770
`United Therapeutics EX2021
`Page 6 of 59
`
`

`

`Identification of Pharmaceutical Impurities
`
`883
`
`Figure 1. Formation of a process impurity (M-12) due to the side reaction between an
`intermediate and a reagent.
`
`sulfur is formed, according to the proposed mechanism:
`
`þ 1=8S8 ðsolidÞ
`
` 3
`
`
`
`þ Hþ!HSO
`
`S2O2
`
`3
`
`Elemental sulfur could not be eliminated by the subsequent cleaning steps; as
`a consequence, it appeared in the final product as process impurities.
`
`Process Impurities Originating from By-Products of the Synthesis
`
`The selectivity of a chemical reaction is rarely 100%, and side-reactions are
`common during the synthesis of drug substances. By-products from the side-
`reactions are among the most common process impurities in drugs. By-pro-
`ducts can be formed through a variety of side reactions, such as incomplete
`reaction, overreaction,
`isomerization, dimerization,
`rearrangement, or
`unwanted reactions between starting materials or
`intermediates with
`chemical reagents (e.g., as shown in Figure 1) or catalysts.[35] There are
`numerous case studies in identification and control of by-product related
`process impurities in the literature.[35 – 43] The following three examples
`demonstrate typical formation pathways of by-products of drug substances.
`Proudfoot et al.[43] reported that isomeric by-products are formed in the
`course of the synthesis of nevirapine ring systems. The mechanism of
`formation of these isomeric impurities is through a so called Smiles
`rearrangement. Horvath et al.[44,45] reported a case where an over-reaction
`product, an epimeric impurity and a dimeric impurity were formed
`17a-ethinyl-17-hydroxy
`during
`the
`synthesis
`of
`steroids. During
`preparation of LY297802 tartrate, Olsen and Baertschi[46] observed two
`by-products in the step from intermediate I to intermediate II due to
`elimination of HCl and hydroxylation, as shown in Figure 2. These
`by-products were able to undergo the next reaction in the synthesis in the
`same way as
`the desired intermediate,
`to form process
`impurities,
`Impurity 1 and Impurity 2.
`
`IPR2020-00770
`United Therapeutics EX2021
`Page 7 of 59
`
`

`

`884
`
`F. Qiu and D. L. Norwood
`
`Figure 2. By-product impurities formed through side reactions during preparation of
`the drug substance LY297802 (adapted from ref. 46).
`
`Process Impurities Originating from Solvents
`
`Residual solvents are organic volatile impurities in drug substances and drug
`products which are used or produced in the manufacturing process of the drug
`substance (e.g., solvents for chemical reaction, separation, and crystalliza-
`tion), excipients, and drug products (e,g., solvents for wet granulation and
`coating). Residual solvents are expected impurities; therefore, identification
`of residual solvents is relatively straightforward. Recent progress in analysis
`of residual solvents was reviewed by Camarasu et al.[15]
`
`Impurities Originating from Degradation of the Drug Substance
`
`Degradation of the drug substance is one of the main sources of impurities in
`both bulk drug and formulated product. Degradation of the drug substance is
`caused by chemical instability of the drug substance under the conditions (e.g.,
`heat, humidity, solvent, pH, light, etc.) of manufacturing, isolation, purifi-
`cation, drying, storage, transportation, and interactions with other chemical
`entities in the formulation (e.g., excipients and coating materials). Chemical
`stability is an inherent property of a drug substance and is a reflection of
`the chemical properties of all functional groups in the drug molecule. In the
`following sections, various impurities originating from degradations of drug
`substances are discussed based on their formation mechanisms.
`
`Impurities Formed from Hydrolysis of the Drug Substance
`
`Hydrolysis of the drug substance is a chemical reaction between the drug
`substance and water. Obviously, the presence of water is a prerequisite for
`
`IPR2020-00770
`United Therapeutics EX2021
`Page 8 of 59
`
`

`

`Identification of Pharmaceutical Impurities
`
`885
`
`the hydrolysis of the drug substance. In a drug substance or drug product,
`water can be introduced as residual water from the manufacturing process
`of the drug or excipient, or absorbed from the environment. Most hydrolytic
`degradations are catalyzed by both acid and base. In some cases, it can
`follow other mechanisms as well. For example, it was reported that metal
`ions can catalyze the hydrolysis of acrylate esters and amides.[47]
`A variety of functional groups are subject to hydrolysis under acidic or
`basic conditions.[50] Common functional groups that are susceptible to
`hydrolysis and some real world examples are summarized in Table 5.
`Depending on the structure of the drug substance, hydrolysis can either
`break the drug molecule into two pieces or break a cyclic structure (e.g., in
`lactones and lactams) into a linear structure. Esters and amides (including
`cyclic esters and amides) are the most common functional groups in drugs
`that are susceptible to hydrolysis. The hydrolysis of esters and amides is
`mostly catalyzed by acid and base. In most cases, drugs with these functional
`groups give the same hydrolytic degradation products under acidic or basic
`conditions. Sometimes, different hydrolytic degradation pathways can be
`observed. For example, penicillin undergoes different hydrolytic pathways
`under acidic and basic conditions to give two different primary hydrolytic
`degradation products, i.e., penicillenic acid under acidic condition, and peni-
`cilloic acid under basic condition, respectively.[58] Hydrolysis of carbonyl-
`containing functional groups, such as esters, amides, and carbamates, etc.,
`usually takes place at the acyl-O(N) bond. Exceptions can be found where
`the alkyl-O(N) bond cleaves.[78] In this hydrolytic pathway, the alkyl groups
`are usually those functional groups that can stabilize the carbonium ion
`formed by alkyl-O(N) cleavage, such as a tertiary carbon or a benzyl group.
`Hydrolysis of the drug substance is often followed by further degradation.
`Primary hydrolytic degradation products can further undergo dehydration,
`decarboxylation, cyclization, or rearrangement, etc., to form the final degra-
`dation products.[57,78]
`
`Impurities Formed from Oxidative Degradation of the Drug Substance
`
`In organic chemistry, oxidation is a class of chemical reactions that leads to an
`increase in oxidation state of an element. In the pharmaceutical industry,
`oxidative degradation is confined to chemical reactions of drug substances
`that increase the oxidation state of C and, in some cases, N, S, P, etc., via
`addition of oxygen or abstraction of hydrogen. Autoxidation is the reaction
`of organic compounds with elemental oxygen under mild conditions (e.g.,
`drug storage conditions). Autoxidation involves ground state elemental
`oxygen; thus, under normal storage conditions, it is the most important
`oxidative degradation pathway of drugs. Autoxidation is a free radical
`reaction;
`typically it requires a free radical
`initiator to start
`the chain
`reaction. Autoxidation initiators are often trace level impurities in the drug
`substance or excipients, such as peroxide or metal ions. These impurities
`
`IPR2020-00770
`United Therapeutics EX2021
`Page 9 of 59
`
`

`

`886
`
`F. Qiu and D. L. Norwood
`
`Table 5. Functional groups susceptible to hydrolysis in drugs
`
`Functional group
`
`Basic structure
`
`Example drugs
`
`Ester
`
`Lactone
`
`Amide
`
`Lactam
`
`Imide
`
`Imine
`
`Carbamic ester
`
`Phosphate
`
`Ether
`Thioether
`Nitrile
`
`Acetal/ketal
`
`Halides
`Sulfonamide
`
`Aspirin,[48] cisatracurium besylate,[51]
`nicergolin,[52] lovastatin,[53] and
`atropine[68]
`
`Lovastatin[53] and warfarin[69]
`
`Acetaminophen,[49] indinavir,[54] and
`indomethacin[55]
`
`Amoxicillin,[56,57] penicillin,[58] cepha-
`losporin,[58] bromazepam,[67]
`diazepam[70]
`
`Phenobarbital[59]
`
`Adinazolam,[60] famotidine,[61]
`diazepam,[70]
`
`Benzimidazole anthelmintics,[62] zol-
`mitriptan,[63] loratadine[77]
`
`Rivastigmine,[64] triamcinolone aceto-
`nide 21-phosphate[74]
`
`Diphenhydramine hydrochloride,[65]
`duloxetine[79]
`Penicillin[58]
`Danazol,[66] cimetidine[71]
`Erythromycin,[72] ECyd acetal deriva-
`tives,[73] triamcinolone[75]
`
`Chlorambucil[76]
`Sulfamethazine[78]
`
`IPR2020-00770
`United Therapeutics EX2021
`Page 10 of 59
`
`

`

`Identification of Pharmaceutical Impurities
`
`887
`
`can be brought into the drug product from the manufacturing process or
`formed from degradation of formulation components (e.g., PEG and PVP)
`in the drug product. Autoxidation can be self initiated because the ground
`state oxygen molecule is a biradical; however, this reaction is expected to
`be slow because the ground state of the majority of organic molecules is
`singlet; thus the reaction is spin-forbidden. A typical autoxidation mechanism
`is shown below:
`
`†
`
`R
`
`ROO
`
`2ROO
`
`RH þ ln
`† !R† þ lnHðIntiation StepÞ
`
`† þ O2!ROOH
`† ðPropagation StepÞ
`† þ RH!ROOH þ R
`†!Non-radical moleculeðTermination StepÞ
`† þ R
`†!Non-radical moleculeðTermination StepÞ
`† þ R
`†!Non-radical moleculeðTermination StepÞ
`
`ROO
`
`R
`
`The non-radical molecules formed in the propagation and termination steps are
`likely to appear in the drug substance or drug product as impurities.
`Autoxidation can be catalyzed by heavy metals. These metals usually
`have two readily accessible oxidation states differing by one unit (e.g.,

`Cu2þ/Cu
`and Fe3þ/Fe2þ
`). Peroxides react with both the upper and lower
`oxidation states of these metals to generate more free radicals.[80] As a
`result, metal ions can greatly enhance the effectiveness of peroxides as
`initiators.
`
`Fe2 þ ROOH!FeðOHÞ2þ þ OR
`FeðOHÞ2þ þ ROOH!Fe2þ þ HOH þ ROO
`
`†
`
`†
`
`Drug substance oxidative degradation can also occur through an electron
`transfer mechanism to form reactive anions or cations. Amines, sulfides, and
`phenols are susceptible for electron transfer oxidation to give N-oxides,
`hydroxylamine, sulfones, sulfoxides, etc.[84] C55C double bonds may react
`with hydrogen peroxide and peroxy acid to form epoxides via a similar
`ionic mechanism.[81] In addition, heavy metals such as Fe3þ
`and Cu2þ
`can
`also be directly involved in the oxidation of drug substances through an
`electron transfer mechanism.
`To some extent, the susceptibility of a drug substance to oxidative degra-
`dation can be predicted from the functional groups in its chemical structure.
`Functional groups with a labile hydrogen, such as benzylic carbon, allylic
`carbon, tertiary carbon, or a-positions of a hetero atom[100] are often suscep-
`tible to oxidation to form hydroperoxide, hydroxide, or ketone.[84] Functional
`groups without a labile hydrogen, such as a tertiary amine or thioether, can
`undergo oxidative degradation to form N-oxide[86] or sulfoxide.[88] Common
`functional groups in drugs that are sensitive to oxidation and examples of
`drugs that contain these functional groups are summarized in Table 6.
`
`IPR2020-00770
`United Therapeutics EX2021
`Page 11 of 59
`
`

`

`888
`
`F. Qiu and D. L. Norwood
`
`Table 6. Functional groups susceptible to oxidative degradation in drugs
`
`Functional group
`
`Basic structure
`
`Example drugs
`
`Benzyl
`
`Ph-CH2-
`
`-CH55CH-CH2-
`
`C55C
`Ar-OH
`R-OH
`0
`R-O-R
`0
`R-S-R
`
`Allylic
`
`Tertiary C
`
`Olefins
`Phenol
`Alcohol
`Ether
`Thioether
`
`Tertiary Amine
`
`Primary/secondary
`amine
`
`Tipranavir[83] methoxamine
`hydrochloride,[89] impipramine
`hydrochloride[90]
`Tetrazepam,[82] Reserpine,[85]
`
`Tipranavir[83]
`
`L-tryptophan[87]
`Epinephrine[91]
`Lovastatin[92]
`Ragaglitazar[98]
`Pergolidemesylate,[93] fluphena-
`zine enanthate,[94] tipredane[99]
`
`Pipamperone[86], dibucaine
`hydrochloride,[96] raloxifene
`hydrochloride[97]
`Brinzolamide[95]
`
`Abstraction of protons from functional groups, such as CH2-CH2,
`CH-OH, or CH-NH in the drug molecule, to form unsaturated bonds C55C,
`C55O or C55N increases the oxidation state of C, and N;
`therefore,
`dehydrogenation is also an oxidative degradation. Dehydrogenation can
`occur in both solid state and in liquid phase. Typical types of dehydrogenation
`include aromatization, conversion of alcohols to ketones or aldehydes,
`conversion of amine to imine or nitriles, etc. For example, L-1,4-cyclohexa-
`diene-1-alanine hydrate degrades to L-phenylalanine in solid state through
`dehydrogenation;[101] nifedipine can convert to dehydrogenated degradation
`products in solution within a few hours under photo irradiation.[102]
`
`Impurities Formed from Photolytic Degradation of the Drug Substance
`
`When exposed to light, many types of organic molecules can undergo photo-
`chemical reactions and form photolytic degradation products.[103] There are
`two main types of photochemical reactions that are relevant to degradations
`of drugs. The first type is a non-oxidative photochemical reaction, which
`includes light induced isomerization, cyclization, dimerization, rearrange-
`ment, hydrolysis, decarboxylation, and homolytic cleavage of X-C hetero
`bonds, such as halogen bond, ether bond, N-alkyl bond in amine (dealkylation
`irradiation, cis-/trans
`or deamination), SO2-C bond, etc. Under
`light
`
`IPR2020-00770
`United Therapeutics EX2021
`Page 12 of 59
`
`

`

`Identification of Pharmaceutical Impurities
`
`889
`
`isomerization and dimerization are very common in drug molecules that
`contain a C55C double bond.[104 – 106,108] Other types of photochemical degra-
`dations of drugs, such as light induced hydrolysis of halogen,[107] rearrange-
`ment,[109,111] dealkylation,[110] and cyclization[112] were also reported.
`Photo-oxidative degradation is the other type of photolytic degradation of
`drugs. Photo-oxidative degradation products can be formed from either a
`triplet oxygen (3O2) or a singlet oxygen (1O2) mechanism, depending on the
`electronic state of the oxygen molecule. The former requires sensitization
`and formation of a free radical of the drug molecule, which then reacts with
`a triplet oxygen molecule to form a peroxide. Singlet oxygen molecules
`react with unsaturated bonds such as alkenes, dienes, polynuclear aromatic
`hydrocarbons,
`etc.,
`to
`form photo-oxidative
`degradation
`products.
`Depending on the structure of the drug molecule, singlet oxygen oxidation
`
`can take place through three pathways, i.e., [2 þ 2] cycloaddition, [4 þ 2]
`
`cycloaddition, and “ene” reaction, as illustrated in Figure 3. For example,
`the C55C double bond in thiothixene undergoes photo-oxidative degradation
`through a [2 þ 2] cycloaddition mechanism.[113] Heterocyclic unsaturated
`
`rings (e.g., thiazole, oxazole, and pyrazole, etc.) are known to be sensitive
`toward light irradiation. For instance, an oxazole containing drug undergoes
`photolytic
`degradation
`through
`a
`proposed
`cycloaddition
`
`[4 þ 2]
`
`Figure 3. Singlet O2 oxidative degradation pathways of drugs that contain unsatu-
`rated bonds.
`
`IPR2020-00770
`United Therapeutics EX2021
`Page 13 of 59
`
`

`

`890
`
`F. Qiu and D. L. Norwood
`
`mechanism.[114] An example of an “ene” reaction was discussed by
`Boccardi.[115] Usually, the peroxides formed through these reactions are
`not stable and are just intermediates to the final photolytic degradation
`products.
`Although rare, photolytic degradation of drugs can also take place
`through a reduction mechanism. A typical example of such degradation is
`the reduction of the nitro group to a nitroso group in nifedipine.[102]
`
`Impurities Formed through Isomerization and Oligomerization of the
`Drug Substance
`
`Isomerization and oligomerization are common degradation pathways
`of drugs. Formation of
`isomers and oligomers of a drug substance
`can occur through a variety of mechanisms; therefore, it is worthwhile
`to have a separate overview of the isomerization and oligomerization of
`drugs.
`Commonly seen isomerizations include photo-induced cis/trans isomer-
`ization of drugs with a C55C double bond,[104,105] other asymmetric double
`bonds (e.g., C55N-OH and C55N-NH2)[116 – 118] and racemization or epimer-
`ization of drug substance with chiral centers. Drug molecules with chiral
`light,[119]
`centers can undergo racemization and epimerization under
`heat,[119] acidic,[120] and basic[121] conditions. Aso et al.[122] found that
`gamma cyclodextrins can facilitate the epimerization and racemization of car-
`benicillin. Rearrangement
`is also a source of isomeric impurities. For
`example,
`fenoprofen calcium can degrade to a mixture of
`isomeric
`biphenyls via a photo-Fries rearrangement.[111] In the presence of water,
`FK506 can readily epimerize to form tautomeric impurities.[123]
`In addition to the photo-cycloaddition of alkenes discussed above,
`other
`types of dimerization or oligomerization can also occur. For
`example,
`thiols can dimerize under oxidative conditions to form disul-
`fides;[124] indoles can dimerize under acidic conditions;[125] nalidixic acid
`dimerizes through a thermo-decarboxylation pathway,[126] and losartan
`dimerization is induced by moisture and acid.[127] It was reported that for-
`maldehyde, which is a potential impurity in some excipients (e.g., PEG
`and PVP), can react with primary or secondary amin

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