throbber
Future of Depot Neuroleptic Therapy:
`Pharmacokinetic and Pharmacodynamic Approaches
`LARRY ERESHEFSKY, Pharm.D., STEPHEN R. SAKLAD, Pharm.D., MICHAEL W. JANN, Pharm.D.,
`CHESTER M. DAVIS, Ph.D., ANN RICHARDS, Pharm.D., and DONALD R. SEIDEL, M.D.
`
`The future of depot neuroleptic therapy is discussed
`in terms of pharmacokinetic and pharmacodynamic re-
`search opportunities. Analytic methods for neuroleptic
`assays, including chromatographic, radioreceptor, nu-
`clear magnetic resonance, and radioimmunoassay tech-
`niques, are briefly reviewed. Elucidation of depot
`neuroleptic multicompartment kinetics utilizing nonlin-
`ear mixed-effects modeling and the usefulness of these
`data in interpreting plasma levels are discussed. The
`clinical significance of plasma monitoring of depot
`fluphenazine, including the development of dosage con-
`version guidelines, is presented. The relationships be-
`tween haloperidol and its metabolite reduced
`haloperidol (RH) are discussed in terms of dosage form
`and response. Clinical advantages resulting from the
`availability of more depot neuroleptics are discussed.
`(J Clin Psychiatry 45 [5, Sec. 21:50-59, 1984)
`
`One of the most common reasons for the readmission of
`psychiatric patients to the hospital stems from the lack of
`medication compliance. The introduction of injectable de-
`pot neuroleptics in the early 1960s provided clinicians with
`a method of accurately assessing medication compliance in
`the mentally ill patient. Several studies have suggested that
`depot neuroleptics reduce the frequency of relapse or length
`of hospital stay when compared to oral agents." However,
`other investigators have reported that depot neuroleptics did
`not reduce the number of relapses among schizophrenics
`when compared with oral medications.' It is beyond the
`scope of this article to review all the comparative studies;
`the reader is referred to the articles by Kane and Johnson in
`this issue:6
`The major perceived use of depot neuroleptics currently
`is in cases of patient refusal or failure to take oral medica-
`tion as prescribed.' Injectable depot therapy bypasses ab-
`sorption variability, gut wall metabolism, and first pass
`extraction by the liver. This method of drug administration
`is useful with patients who absorb inadequate amounts of
`
`From the Departments of Pharmacology and Psychiatry, University of
`Texas Health Science Center at San Antonio; the College of Pharmacy,
`University of Texas at Austin; and San Antonio State Hospital and The
`Texas Research Institute for the Mental Sciences, Houston, TX.
`The authors acknowledge the contributions of Charles A. Harrington,
`Ph.D., and Jeffrey L Browning.
`Reprint requests to: Larry Ereshefsky, Pharm. D., Department of
`Pharmacology, University of Texas Health Science Center of San Antonio,
`7703 Floyd Curl Drive, San Antonio, TX 78284.
`
`50
`
`drug from oral therapy, resulting in subtherapeutic plasma
`levels.'
`Assessment of the action of drugs in psychiatric disor-
`ders is complicated because drug effects are only one of
`several inputs to the brain. Many environmental factors,
`including drugs, affect the brain's biochemical responses.
`Behavior is the outcome of a complex interplay of genetics,
`prior skills, events stored as memories, and biochemical
`factors. When a drug is added to this complicated set of
`interactions, there may be initially only a minor change in
`behavior. Substantial changes in behavior, however, can oc-
`cur over extended periods of time.
`The longitudinal course of drug response is partially de-
`pendent on individual pharmacodynamic and pharmaco-
`kinetic factors. Pharmacokinetic studies of drugs are
`needed to investigate how bioequivalence, therapeutic effi-
`cacy, and drug distribution characteristics influence the pa-
`tient's response. Failure to consider pharmacokinetics
`explains much of the disparity in results found among clini-
`cal studies. These principles and their applications will play
`an important role in the future use of long-acting neurolep-
`tic drugs.
`
`OVERVIEW OF DEPOT NEUROLEPTICS
`
`The synthesis of long-acting neuroleptics is accom-
`plished by an esterification of fluphenazine or other hy-
`droxyl-containing neuroleptics to a long-chain fatty acid.
`These esters are usually dissolved in sesame seed oil, coco-
`nut oil, or Viscoleo. Medications such as fluspirilene are
`not esterified, but are formulated as an aqueous microcrys-
`talline suspension with slow release properties.' The medi-
`cation, in oil or suspension, when injected into muscle,
`forms a reservoir of slowly released drug. The esterified
`compounds are reportedly taken up into fat stores in various
`body areas, except the brain and spinal cord, within days.'
`The drug is slowly and steadily released from the depot and
`hydrolyzed by plasma esterases to the parent compound,'
`although preliminary reports suggested that fluphenazine
`decanoate was released "erratically, in spurts," from the
`injection site.'
`At the present time, fluphenazine enanthate and deca-
`noate are the only long-acting depot neuroleptics available
`in the United States. An increase in the number of depot
`neuroleptics available will lead to potentially improved pa-
`tient care for the following reasons:
`1. Easier conversion from oral to depot medication without
`switching drug class.
`
`Mylan v. Janssen (IPR2020-00440) Ex. 1020 p. 001
`
`

`

`J CLIN PSYCHIATRY 45:5 (Sec. 2) MAY 1984
`
`PHARMACOKINETICS AND PHARMACODYNAMICS
`
`TABLE 1. Comparison of Depot Neuroleptics
`
`Neuroleptics
`Fluphenazine
`Fluphenazine
`Haloperidol
`Flupenthixol
`Clopenthixol
`Perphenazine
`Pipotiazine
`
`Fluspirilene
`
`Fatty Acid Chain
`Decanoate
`Enanthate
`Decanoate
`Decanoate or palmitate
`Decanoate
`Enanthate
`Palmitate
`Undecylenate
`None
`
`Vehicle
`Sesame oil
`Sesame oil
`Sesame oil
`Viscoleo
`Viscoleo
`Sesame oil
`Sesame oil
`Sesame oil
`Aqueous
`suspension
`
`Dosage and Interval
`12.5-100 mg, 1-4 weeks
`12.5-100 mg, 1-2 weeks
`20-400 mg, 4 weeks
`10-50 mg, 2-4 weeks
`50-600 mg, 1-4 weeks
`50-200 mg, 1-4 weeks
`25-600 mg, 4 weeks
`2-200 mg, 1-4 weeks
`2-30 mg, 1-4 weeks
`
`Plasma
`Levels
`0.5-3.0
`NA
`3.0-10.0
`0.5-2.0
`0.5-9.0
`2.0-10.0
`NA
`NA
`NA
`
`Single-Dose Kinetics
`Time to Peak
`lialfrUfe
`(daysl
`(days-
`0.3-1.5
`6-9
`2
`3.5-4.0
`3-9
`21
`1 1 -17
`17
`4-7
`19
`2-3
`3.5-4.5
`NA
`NA
`NA
`NA
`NA
`NA
`
`2. Flexible selection of depot agent to minimize side ef-
`fects.
`3. Increased selections for alternative therapies of the re-
`fractory patient.
`4. Decreased costs of depot therapy secondary to competi-
`tion in the marketplace.
`5. More widespread use of depot therapy, leading to in-
`creased clinical experience and to decreased morbidity
`and health care costs.
`Table 1 summarizes the studies examining the pharma-
`cokinetics of depot neuroleptics available worldwide. m-16
`Most of the clinical studies with the various depot neurolep-
`tics have compared their side effect profile to that of
`fluphenazine decanoate or simply reported the various sed-
`ative, autonomic, and extrapyramidal symptoms (EPS) oc-
`curring in the study population. Fluphenazine enanthate
`was reported to cause increased extrapyramidal and motor
`side effects compared to fluphenazine decanoate during a 7-
`month double-blind study with 50 schizophrenic outpa-
`tients." Haloperidol decanoate was reported to cause
`sedation in 4 of 38 patients.18 In a larger study of haloperi-
`dol decanoate involving 239 chronic schizophrenic pa-
`tients, 27% had autonomic side effects or EPS.19
`Fluspirilene and clopenthixol decanoate were compared to
`fluphenazine decanoate, and their sedative effects ("som-
`nolence," "drowsiness") were found to be similar.2°'21 In a
`report on 10 schizophrenics, 5 displayed akathisia and
`pseudoparkinsonism after administration of perphenazine
`enanthate. 22 Extrapyramidal symptoms with pipotiazine
`palmitate and undecylenate were reported to range between
`10% and 43% among the patients in the various published
`studies!'
`In several studies, all of the extrapyramidal side effects
`were adequately treated with antiparkinsonian agents.
`There were no reports of abnormal effects on liver, renal,
`or other organ systems, as measured by routine laboratory
`analysis. The most serious problem that can occur with a
`long-acting neuroleptic is the neuroleptic malignant syn-
`drome (NMS). Because of the apparently long half-life of
`the injected drug, it can be difficult to effectively treat pa-
`tients in whom this syndrome develops. Of the 12 deaths
`reported from NMS secondary to neuroleptics, 6 were in
`
`patients treated with fluphenazine decanoate." Finally, it
`has been reported that the relative occurrence of tardive
`dyskinesia is not specifically related to any particular
`neuroleptic agent.25 Careful epidemiologic studies to accu-
`rately assess the effects of drug product selection and
`plasma levels on the incidence of tardive dyskinesia have
`not been performed to date. Additional carefully controlled
`studies that consider pharmacokinetic and pharmacody-
`namic variables need to be performed as new depot agents
`become available.
`
`NEUROLEPTIC ASSAY
`
`A prerequisite to the study of the pharmacokinetics of
`neuroleptic agents is the ability to measure the clinically
`relevant subnanomolar quantities of drug in biological flu-
`ids. In general, the best correlation between drug at the site
`of action within the central nervous system and a peripheral
`measure is the free (unbound) level of drug within the
`plasma. Free drug levels are more useful than total drug
`levels because they measure the drug that is available to
`diffuse across the blood brain barrier to the site(s) of
`action.' Some studies suggest that models of the amount of
`drug within the central nervous system have produced bet-
`ter correlations when based on red blood cell levels.27-29
`Several low-affinity binding sites in plasma are located
`on albumin. Cholesterol and alpha-1-acid glycoprotein are
`high-affmity binding sites!'-'' Alpha-1-acid glycoprotein
`levels in plasma are important because of the dramatic in-
`crease in the concentration of this protein found in patients
`during stressful events!' Perhaps higher total levels of
`drug, with free levels relatively unchanged, are required
`during an acute stressful event to maintain a constant
`amount of drug at the site of action.
`Pharmacokinetic descriptions of neuroleptic drugs in-
`volve the use of multicompartment models. From the
`plasma, usually termed the central compartment, unbound
`drug may be actively or passively transported into
`"deeper" compartments (e.g., skeletal muscle, brain tis-
`sue, or fat). Until these deeper compartments equilibrate
`with the free drug in the central compartment, the medica-
`tion will not achieve steady-state concentrations at the site
`
`51
`
`Mylan v. Janssen (IPR2020-00440) Ex. 1020 p. 002
`
`

`

`J CLIN PSYCHIATRY 45:5 (Sec. 2) MAY 1984
`
`ERESHEFSKY ET AL.
`
`of action. Furthermore, when the administration of the drug
`is discontinued, these deep compartments will leach the ac-
`tive drug out through the central compartment and back to
`the site of action:4 Therefore, effects secondary to oral or
`depot neuroleptic therapy can persist for weeks and months
`after the discontinuation of routine therapy.
`As a general rule, three drug plasma levels must be ob-
`tained in a standard kinetic analysis to define each compart-
`ment. For a neuroleptic drug that has at least three
`significant compartments, nine levels drawn at appropriate
`times would be required to define one individual's pharma-
`cokinetic parameters.34 Analysis of the kinetics of depot
`neuroleptics is complicated, since prolonged studies are re-
`quired. One method of obtaining more information in a
`shorter time with fewer levels is to use a nonlinear mixed
`effects model (NONMEM) for analysis of the data. NON-
`MEM requires knowledge of how much drug was given,
`and the times of administration and plasma level sam-
`pling."'"
`Assays of the neuroleptics are usually performed either
`by physical detection of the drug by high performance liq-
`uid chromatography (HPLC),37'" gas liquid chromatogra-
`phy (GLC):94° high performance thin layer chromatogra-
`phy (HPTLC)," or by radioimmunoassay (RIA).38.42'43 An in
`vitro measure of neuroleptic activity can also be approx-
`imated by determining the total amount of dopamine-
`blocking activity by its competition with a radioactively la-
`belled ligand bound to dopamine receptors (radioreceptor
`assay, RRA):64446 Alternately, biological responses in vivo
`have been measured by assaying prolactin.""
`In addition to a reliable, sensitive, and specific assay
`method, blood collection procedures must be stringently
`controlled. Because these drugs are present in such low
`concentrations, small changes in the collection procedure
`may result in a dramatic change in the assay result. One
`example of a poorly controlled collection procedure is to
`draw a sample into a vacuum collection tube capped with a
`plastic stopper. These stoppers, however, contain a plastici-
`ser that causes redistribution of the drug into erythrocytes,
`which will be spun out of the sample when centrifuged. A
`second example is photo-oxidation of the drug by exposure
`to ultraviolet light prior to assay. Both examples result in
`lower drug levels measured than actually exist in the sam-
`ple.
`Once these sampling procedures have been perfected,
`reproducible separation of the drug from contaminants must
`be performed with a high extraction efficiency, monitored
`by the use of an internal standard. A clinically usable assay
`should have a coefficient of variation (both intra- and in-
`terassay) of less than the minimum change in plasma level
`that might yield a clinical effect (e.g., 10%-20%). Many
`methods of detection, such as electron capture, scintilla-
`tion, and nitrogen or fluorescent detection, have been em-
`ployed; the small quantity of drug present in clinical
`samples makes this a critical procedure. Our laboratory
`currently employs an HPTLC assay for fluphenazine sepa-
`
`52
`
`ration, followed by paraffin oil treatment of the chromatog-
`raphy plates to permit increased resonance within the
`molecule, then ultraviolet development and fluorescent
`scanning with computerized quantification." We have
`found this to be an extremely reliable and efficient method
`of detecting subnanomolar quantities of fluphenazine." An
`advantage of HPTLC over GLC or HPLC is that many
`samples may simultaneously be run on a single plate, pro-
`viding excellent speed and efficiency. An additional advan-
`tage is that HPTLC is a closed system and the plates may be
`stored for future reanalysis; in contrast, GLC and HPLC
`are open systems, in which the sample is lost following
`chromatographic separation and detection.'
`RIAs potentially offer the most efficient detection of
`drug, but there are still problems with specificity. The ap-
`propriate antibody must be produced to measure only the
`drug to be examined, and not the metabolites or other con-
`taminants in the sample. To do this, one must have a reli-
`able, sensitive, and specific assay (such as HPTLC or
`HPLC) to standardize the RIA.38'42'43
`The neuroleptic RRA, although a substantial improve-
`ment over many of the quantitative assays in terms of mea-
`surement of total biological effects, has numerous
`limitations. These include low sensitivity, nonspecific pro-
`tein binding limiting the sample aliquot, and the use of cau-
`date tissue instead of limbic tissue, the presumed site of
`neuroleptic action. Maximum sensitivity achieved with the
`RRA usually is > 1 ng/ml for neuroleptic agents. In the
`case of fluphenazine, because of its high affinity binding to
`dopamine receptors, a sensitivity of 0.5 ng/ml is achievable
`(personal communication, Charles L. Bowden, January 10,
`1983). This approximates the sensitivity of most quantita-
`tive analytic processes. Another approach, the measure-
`ment of prolactin increase as a function of the plasma
`concentration of a neuroleptic, has not provided consistent
`correlations with clinical effect.'" Further development of
`analytic techniques should allow us to clarify the role of
`neuroleptic metabolites and drug response:I'52
`Another area in which dramatic progress in assay tech-
`nology is being made is in the use of quantitative nuclear
`magnetic resonance (QNMR). QNMR is a technique using
`a strong magnetic field and a perpendicular radio frequency
`field to cause the fluorine nuclei in a neuroleptic to absorb
`energy quantifiably. QNMR may permit us to measure the
`drug at the site of action. This might be done by a device
`that looks similar to a computerized axial tomography
`(CAT) scanner. Refinement of this technique may permit
`quantification and detection of the molecular interaction at
`the site of action in the brain, e.g., protein, tissue, or recep-
`tor binding. Improvements in QNMR resolution might
`yield the capability to produce sectional images of the
`drug's distribution and binding within the body (personal
`communication from David Young, December 5, 1983)."
`Because of the specialization and sophistication of the
`personnel and equipment needed to perform these types of
`quantitative analysis, we envision regional analytic centers
`
`Mylan v. Janssen (IPR2020-00440) Ex. 1020 p. 003
`
`

`

`J CLIN PSYCHIATRY 45:5 (Sec. 2) MAY 1984
`
`PHARMACOKINETICS AND PHARMACODYNAMICS
`
`FIGURE 2. Idealized plasma level versus time curve for a single
`oral dose of a neuroleptic, illustrating absorption phase (a), dis-
`tribution into equilibrating compartments (b), metabolic elimi-
`nation (c), and leakage back into plasma from equilibrating
`compartments (d).
`
`d
`
`20
`
`25
`
`b
`
`a
`
`Log Plasma Concentration
`
`0
`
`5
`
`10
`15
`Time (Hours)
`
`rate of elimination. This "flip-flop" model is useful for the
`understanding and application of depot neuroleptic pharma-
`cokinetics to patient care. It also must be reemphasized that
`these highly lipophilic compounds do not possess simple
`one-compartment pharmacokinetic characteristics. Drugs
`that are highly ionized or hydrophilic usually follow a sin-
`gle-compartment kinetic model, where the kinetics can be
`characterized over the entire concentration time curve as a
`biexponential function (absorption and elimination). For
`depot neuroleptics and other lipophilic drugs, multicom-
`partment kinetic models are necessary. The measured decay
`phase half-life of neuroleptic agents is longer during studies
`performed at steady state (once plateau concentrations have
`been reached) than during single-dose studies. An early il-
`lustration of multicompartment effects was the description
`of chlorpromazine urinary metabolite detection after cessa-
`tion of chronic therapy for up to 6 months!'
`
`FIGURE 3. Detail of a patient's plasma level versus time curve
`following the injection of a single 25 mg dose of fluphenazine
`decanoate.
`
`1.5
`oo
`• 1.4
`• 1.3
`0
`
`o .9
`C.)
`•
`.8
`E
`.7
`g .6
`0
`ar
`.—c .4
`
`a)
`.c
`Q
`
`=8days
`
`10 20 30 40 50 60 70
`Time (Hours)
`
`I
`I
`I
`i
`r
`90 100 110 120 130
`
`53
`
`FIGURE 1. Pharmacokinetic variables that affect the amount of
`active drug able to bind to the site of action.
`
`Receptor
`Binding
`
`Cerebrospinal
`Fluid
`
`Nonspecific
`Binding
`
`Blood Brain1Barrier
`
`Esterified Drug
`In Plasma
`
`Esterase
`
`Unbound
`Drug
`In Plasma
`
`Metabolism
`
`Plasma Protein
`Binding
`
`Esterified Drug
`In Depot Oil
`
`Rena and
`Biliary
`Elimination
`
`Multicompartment
`Tissue Binding
`
`as a cost-effective solution to the growing demand for psy-
`chotropic drug assay. These centers should utilize methods
`that have a large sample capability and rapid turnaround
`time.
`
`PHARMACOKINETIC AND
`PHARMACODYNAMIC APPLICATIONS
`
`Pharmacokinetics is the study of drug absorption, distri-
`bution throughout the body, metabolism, and eventual elim-
`ination. 54 Pharmacodynamics is the study of the
`biochemical and physiological effects of drugs and, most
`importantly, their mechanisms of action. A clearer under-
`standing of the unique advantages and disadvantages of de-
`pot neuroleptics versus oral therapy requires further study
`of the interplay between drug concentration at the site of
`action over time. Drug effects on receptors, such as kin-
`dling phenomena, denervation hypersensitivity, and down
`regulation, also need further study.
`Depot neuroleptic pharmacokinetic variables that inter-
`relate with brain effects are illustrated in Figure 1," which
`shows that once the drug is absorbed from its injection site,
`redistribution and extensive binding to tissue and protein
`occur. As stated, the fraction of the free drug in plasma is
`the only portion that can equilibrate with cerebral spinal
`fluid and brain tissue. Therefore, it is essential in any study
`where only total plasma levels are measured that the protein
`binding be constant over the entire concentration range for
`the drug. Nonlinear protein binding, as with valproic acid,
`decreases the usefulness of total plasma levels.' Figures 2
`and 3 depict the relevant pharmacokinetic parameters ob-
`tained from the plasma level versus time curves for both
`oral and depot neuroleptic therapy. As Figure 3 shows, for
`the depot neuroleptic, absorption from the injection site
`may be the rate-limiting step. The typical "decay phase" of
`the depot curve provides an estimate of the rate of absorp-
`tion of other deep compartment rate constants and not the
`
`Mylan v. Janssen (IPR2020-00440) Ex. 1020 p. 004
`
`

`

`J CLIN PSYCHIATRY 45:5 (Sec. 2) MAY 1984
`
`ERESHEFSKY ET AL.
`
`The valid study of the relationships between plasma lev-
`els and clinical effects depends on the methods used for
`analysis of the drug, the pharmacokinetic derivation of the
`individual parameters, clinical assessments, statistical anal-
`ysis, and the nature of the psychiatric illness. The response
`that is the best measure of neuroleptic effectiveness is rela-
`tive improvement, rather than absolute severity of symp-
`toms. For this reason, it is recommended that correlations
`be based on the difference between the patient's baseline
`severity of illness and severity at the time of the plasma
`level determination. This is especially important in evaluat-
`ing drug effects in a population in which large numbers of
`minimal and nonresponders are anticipated. If a patient's
`psychotic symptoms were considered extremely severe on
`admission and have improved to a "moderately ill" rating,
`then a definite clinical benefit has resulted from the treat-
`ment. On the other hand, if a patient was moderately ill on
`admission and remains moderately ill, no clinical benefit
`has resulted. Both patients are considered moderately ill,
`but one has responded to treatment, while the other has
`not.58 Acutely psychotic patients, although more difficult to
`enroll in studies of plasma level-response relationships, are
`the best candidates in whom to obtain clinical correlations.
`Chronically ill patients with multiple dependencies and per-
`sonality characteristics related to institutionalization are
`less suitable for clinical response studies. A response study
`should use a fixed-dose design, in which patients are ran-
`domly assigned to various dosages that result in a uniform
`distribution from subtherapeutic to supratherapeutic levels.
`In the past, most of the plasma level response studies were
`technically flawed by the use of a variable dose titration
`scheme. However, the ethical difficulties in using acutely
`psychotic patients as subjects in this type of fixed-dose de-
`sign are apparent.
`A parallel strategy for the elucidation of clinical re-
`sponse, adverse effects, and plasma level relationships is
`the use of large numbers of patients in naturalistic environ-
`ments, rather than a few patients in a clinical research cen-
`ter. Many of the flaws and biases present in small
`populations (e.g., inadequate statistical power and lack of a
`uniform distribution of patients who have drug levels above
`and below the therapeutic range) are easily avoided by us-
`ing large populations of patients. The problems inherent in
`the analysis of data obtained from a naturalistic environ-
`ment are overcome by the use of a NONMEM technique to
`develop response and kinetic relationships. We are using
`NONMEM-assisted analysis and subnanomolar quantifica-
`tion of psychotropic drugs in a naturalistic environment to
`develop models of response. The application of these tech-
`niques is illustrated below by data on kinetics for fluphena-
`zine decanoate and metabolite relationships for haloperidol.
`
`METABOLITE RELATIONSHIPS
`FOR HALOPERIDOL
`
`The development of our RIA for haloperidol and the
`
`54
`
`reduced metabolite (hydroxyhaloperidol) shows the benefit
`of collaboration between members of our interdisciplinary
`research group." Our initial RIA procedure did not dis-
`criminate between the parent and metabolite moiety. Clini-
`cal response studies indicated that two compounds were
`being detected on the basis of the pharmacokinetic profile.
`Subsequent review of our analytic techniques revealed that
`our antibody, prepared from the oximoconjugate of halo-
`peridol, was co-specific for both haloperidol and reduced
`haloperidol. We then developed a procedure utilizing selec-
`tive succinylation of the hydroxyl group, followed by silica
`gel chromatographic separation and reassay."
`Our studies with oral haloperidol indicate that reduced
`haloperidol concentrations range between zero and twice
`the parent level.' When patients are given intravenous hal-
`operidol in a cardiac intensive care unit, no detectable re-
`duced haloperidol is found. We speculate that the use of
`haloperidol decanoate may similarly result in a different
`parent-to-metabolite relationship. The dopaminergic antag-
`onist activity of reduced haloperidol is 20% and 25% of
`that of haloperidol, as measured by apomorphine-induced
`stereotypy in rats and in calf caudate RRA, respectively. No
`reduced haloperidol was observed after intraperitoneal in-
`jection of haloperidol in these rats. In addition, the effects
`on weight gain, appetite, and spontaneous motor activity
`were different in rats administered these two compounds
`intraperitoneally." The differing biological activities of re-
`duced haloperidol and the parent compound may partially
`explain our initial findings in schizophrenic patients who
`were poor responders to oral haloperidol therapy."
`The NONMEM analysis of our data on 41 schizo-
`phrenic patients is now in progress. This retrospective data
`set includes 66 plasma levels and clinical global impres-
`sions rating scales.52 Pearson product-moment correlation
`analysis yielded the following: dose vs. the summed plasma
`concentrations of haloperidol + reduced haloperidol, r =
`0.69 (t = 5.42, df = 64, p < .01); dose vs. plasma con-
`centration of haloperidol, r = 0.76 (t = 5.99, p < .01);
`and dose vs. plasma concentration of reduced haloperidol, r
`= 0.57 (t = 4.60, p < .01). A log-linear correlation analy-
`sis showed weaker relationships, with the exception of dose
`vs. plasma concentration of reduced haloperidol, r = 0.71
`(t = 5.59, p < .01). Two groups of patients were identified
`based on our earlier work": those with reduced-haloperi-
`dol/haloperidol ratios > 1 (N = 17, Group 1) and reduced-
`haloperidol/haloperidol ratios < 1 (N = 24, Group 2).
`Age, sex, and diagnosis were not found to differ signifi-
`cantly between the two groups. Significant differences
`identified between Groups 1 and 2, respectively, were as
`follows: haloperidol dosage = 44.5 ± 22.9 vs. 35.4 ±
`17.6 mg/day (grouped Student's t-test, (t = 2.20, df = 64,
`p < .05); reduced haloperidol plasma concentration =
`40.3 + 24.9 vs. 10.6 ± 14.3 ng/ml (t = 2.75, p < .01);
`severity of illness = 4.50 ± 1.18 vs. 3.68 ± 1.11, t =
`2.75, p < .01); and therapeutic effect = 3.36 ± 0.95 vs.
`2.81 ± 1.08 t = 2.00, p < .05). Haloperidol plasma con-
`
`Mylan v. Janssen (IPR2020-00440) Ex. 1020 p. 005
`
`

`

`J CLIN PSYCHIATRY 45:5 (Sec. 2) MAY 1984
`
`PHARMACOKINETICS AND PHARMACODYNAMICS
`
`TABLE 2. Demographics and Descriptive Information for Population Studied
`Fluphenazine Dosage Form
`
`Variable
`Sex
`Male
`Female
`Age
`Range
`Mean ± SD
`Weight (kg)
`Range
`Mean ± SD
`Diagnosis
`
`Decanoate (N=39)
`
`Hydrochloride (N=22)
`
`29
`10
`
`20-64
`36.2±13.2
`
`15
`7
`
`18-59
`33.0+11.5
`
`45-114
`71.0±16.2
`Chronic undifferentiated schizophrenia, 35.9%;
`paranoid schizophrenia, 30.8%; schizoaffective, 15.4%;
`schizophreniform, 7.7%; disorganized schizophrenia,
`5.1%; bipolar manic, 2.6%; OBS with psychosis, 2.6%
`
`50-108
`76.6+14.6
`Paranoid schizophrenia, 45.4%; schizoaffective,
`22.7%; chronic undifferentiated schizophrenia, 18.2%;
`schizophreniform, 9.1%; bipolar manic, 4.5%
`
`same rater, although numerous raters were used throughout
`the 2-year period of the review.
`2. The CGI severity of illness and side effects subscales
`were rated on a scale of 1 to 7, where 1 indicated the item
`was not present, and 7 indicated an extremely severe pre-
`sentation. The change in severity of illness score (delta se-
`verity) was used to assess therapeutic effect of the drug.
`3. Fluphenazine plasma levels for depot therapy were
`obtained either 7 or 14 days after injection, usually preced-
`ing the next dosage administration.
`4. The fluphenazine plasma samples were analyzed by
`HPTLC. Our methodology included careful screening for
`compliance, elimination of data points in patients who re-
`ceived a fluphenazine hydrochloride dose within 48 hours
`of plasma level samplings, controlling for patients with
`known enzyme-inducing or inhibiting substances, and the
`use of an acutely psychotic population.
`Table 2 gives demographic data for the study popula-
`tion, and Table 3 provides fluphenazine dosage information
`and Table 4 plasma level distributions. The 39 patients on
`fluphenazine decanoate were analyzed for the change in
`their severity of illness and side effects from baseline rat-
`ings to final evaluation, at least 4 weeks later.
`Linear Pearson product-moment correlation analysis of
`
`centrations were not significantly different, 24.4 ± 13.3
`vs. 18.2 ± 14.3 ng/ml (t = 1.70, p = .10). A log-linear
`Pearson product-moment correlation analysis yielded the
`best fit for haloperidol plasma concentration vs. reduced
`haloperidol plasma concentration, r = 0.80 (t = 6.21, df =
`64, p < .01). The log-linear least-squares regression line
`was as follows: In reduced haloperidol plasma concentra-
`tion = 0.073 x haloperidol plasma concentration + 0.81.
`Graphical analysis of the data indicates a nonlinear relation-
`ship for reduced haloperidol vs. haloperidol plasma con-
`centration at haloperidol plasma concentrations > 30
`ng/ml.
`Increasing plasma levels of haloperidol result in a
`greater than linear increase in reduced haloperidol plasma
`levels. If reduced haloperidol is a less potent dopamine an-
`tagonist, then the therapeutic window61'62 observed when
`measuring only haloperidol plasma levels might be partially
`explained by the production of reduced haloperidol in the
`intestinal tract. Further studies are currently in progress to
`elucidate this model.
`
`CLINICAL SIGNIFICANCE OF PLASMA LEVEL
`MONITORING OF FLUPHENAZINE DECANOATE
`
`Our pilot data on the use of fluphenazine plasma level
`monitoring in 39 patients receiving fluphenazine decanoate
`and 22 patients on oral fluphenazine therapy in a naturalis-
`tic setting were obtained retrospectively."'64 An additional 3
`patients had plasma samples obtained serially following an
`injection of fluphenazine decanoate. We have been using
`fluphenazine plasma levels and behavioral assessments in
`the San Antonio State Hospital/University of Texas, Col-
`lege of Pharmacy Clinical Psychopharmacology Program
`since 1980. An illustration of the type of information result-
`ing from the analysis of data obtained from our routine
`plasma level monitoring program is presented here.
`1. Before each drug plasma level was obtained, the
`patient was interviewed by a clinical psychopharmacologist
`who completed an assay request form that included the
`Clinical Global Impressions (CGI) rating scale, diagnosis,
`dosage, and demographic data. Each patient always had the
`
`TABLE 3. Descriptive Information for Fluphenazine Decanoate
`and Oral Fluphenazine
`Item
`Starting dose
`
`i
`
`Oral
`23.2+22.9
`mg/day
`
`Decanoate
`29.7+14.7
`mg/injection'
`
`Final dose
`41.0±20.7
`Mean
`27.3 ± 26.9
`Range
`12.5-100
`0-80
`'Starting injection interval = 7 days; injection interval after dis-
`charge = 14-28 days.
`
`TABLE 4. Relationship Between Steady-State Fluphenazine
`Decanoate Plasma Levels and Dosage Range
`Depot Dose (mg/week)
`Plasma Level (ng/mI) Patient N
`25 mg
`0.76
`32
`37.5 mg
`1.11

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