throbber
MYLAN - EXHIBIT 1058
`Mylan Pharmaceuticals Inc. et al. v. Allergan, Inc.
`IPR2016-01127, -01128, -01129, -01130, -01131, & -01132
`
`

`

`CONSENSUS REPORT ON CYCLOSPORIN
`
`643
`
`7. In the immediate posttransplant period, the
`recommended frequency of monitoring is
`once-every 24 to 48 h.
`8. The laboratory should be able to provide
`same-day turnaround during the early post-
`transplant period.
`9. CsA concentrations need to be interpreted in
`conjunction with other laboratory data, clin-
`ical considerations, and concomitant immu—
`nosuppressive therapy.
`10. In the majority of clinical situations, the mon-
`itoring of CsA metabolites is not warranted.
`11. There is a need to develop assay systems ca-
`pable of measuring the individual patient
`state of immunosuppression.
`
`SAMPLE-COLLECTION TIME AND
`PHARMACOKINETIC MONITORING
`
`Whole blood with EDTA as anticoagulant is the
`preferred matrix for CsA measurement. Whenever
`possible, peripheral venopuncture sampling should
`be preferred over sampling from a resident central
`venous line or capillary testing. It is also important
`to note that CsA displays a circadian variation, eve-
`ning trough levels being significantly lower than
`morning trough levels (3).
`The marked intra— and interindividual variation in
`
`cyclosporin pharmacokinetics (4,5) complicates
`effective immunosuppressive therapy. There is a
`poor correlation between CsA dose and whole
`blood trough concentration. Early investigations of
`the therapeutic window of CsA documented that
`CsA trough levels far outside a therapeutic range
`tended to predict adverse events (4). The relation
`between cyclosporin trough concentration and im-
`munosuppressive efficacy or toxicity after organ
`transplantation has been widely studied (6), and
`currently most centers use trough CsA blood con-
`centrations as a guide for the dosage of this drug.
`However, patients displaying trough concentra-
`tions within a putative CsA therapeutic range are
`not always spared from either rejection or nephro—
`toxicity (7). In an attempt to understand the phar-
`macokinetic factors critical to outcome, a concen-
`tration-controlled strategy (8) based on serial con—
`centration—time profiles was applied to determine
`appropriate drug doses. Because of variations in-
`trinsic to the conventional oral liquid and gel cap
`formulations of CsA,
`this strategy was based on
`establishing a range of drug concentrations during
`continuous intravenous infusion and after oral ad-
`
`ministration. This range of concentrations allowed
`identification of optimal targets during therapy:
`steady-state concentrations of 400 ug/L during iv
`and initial l-month average concentrations of 550
`ug/L, tapering to a target of 400 pug/L from 6 to 12
`months (9). Strategies using 3-point samplings seem
`to yield reasonable estimates of actual AUC (10)
`and in practice have proven equally efficient as full
`7-point profiles (0, 2, 4, 6, 10, 14, and 24 h for q.d.
`and 0, 2, 4, 6, 8, 10, and 12 h for b.i.d.). The avail-
`ability of the new microemulsion formulation of
`CsA (Neoral) may streamline the endeavor, be-
`cause with this formulation, trough levels show an
`improved correlation with AUC (r2, 0.81 versus
`0.50 with a standard nonmicroemulsion formula-
`
`tion) and achieve good estimates of full AUC with
`only a 2— and 6-h sampling during a 12-h dosing
`interval (11). The use of the new microemulsion for-
`
`mulation is associated with reduced variability in
`CsA kinetics, implying that single trough levels will
`be more consistent. With this formulation, CsA ab-
`sorption is less dependent on bile acids and not af-
`fected by food intake (12). It
`is hoped that the
`greater reproducibility of CsA pharmacokinetic
`profiles when using the new microemulsion formu-
`lation (11,13) will facilitate dosage adjustment to
`compensate for inter— and intraindividual differ-
`ences and will facilitate the incorporation of phar-
`macokinetic approaches into transplant care. The
`major practical limitation, however, for AUC mon-
`itoring is the necessity for blood collections to be
`made at precisely timed intervals after oral dosing.
`So far, comparative studies on AUC versus
`trough-level monitoring have not consistently
`shown overall superiority of either method (9).
`Well—designed, preferably randomized prospective
`clinical trials, including the new microemulsion for-
`mulation and employing specific, well-validated as-
`says to measure CsA, will help to resolve this issue.
`In such studies, assay specificity and precision
`must be documented. There is widespread consen-
`sus that at present, trough-concentration monitor-
`ing is the most appropriate and practical (Table 1).
`
`FREQUENCY OF MONITORING
`
`The frequency of blood CsA concentration mon—
`itoring should depend on the time elapsed since
`transplantation, intercurrent illnesses, and concom-
`itant therapy with drugs affecting CsA metabolism.
`The immediate posttransplant period is character—
`ized by unstable graft function, extreme inter- and
`
`Ther Drug Monit, Vol. 17, No. 6, 1995
`
`

`

`644
`
`M. OELLERICH ET AL.
`
`TABLE 1. Therapeutic ranges for CsA obtained from a survey of transplant centers during 1994/1995
`Immuno-
`
`Analytic
`Dosing
`suppression
`Therapeutic
`Transplant
`
`Center [No.]
`method
`interval
`protocol
`ranges (pg/L)
`type
`
`HPLC
`
`b.i.d.
`
`University of
`Pennsylvania
`Medical Center,
`Philadelphia, PA,
`U.S.A. [1]
`
`mFPIA
`
`Oklahoma
`Transplantation
`Institute, Oklahoma
`City, OK, U.S.A.
`[2]
`Georg—August-Universitat, EMIT
`Gottingen, FRG [3]
`
`b.i.d.
`
`b.i.d.
`
`b.i.d.
`
`b.i.d.
`
`m 125I-RIA
`
`b.i.d., t.i.d.
`
`mFPIA
`
`b.i.d., t.i.d. in
`children <24
`mo
`
`St. Christophers
`Hospital for
`Children,
`Philadelphia, PA,
`U.S.A. [4]
`
`University of
`Cincinnati Medical
`Center, Cincinnati,
`OH, U.S.A. [5]
`
`Universitats
`Krankenhaus
`Eppendorf,
`Hamburg, FRG [6]
`Hospital for Sick
`Children, Toronto,
`ON, Canada [7]
`
`111
`
`111
`111
`111
`III
`III
`
`II
`
`111
`
`II + ALA
`
`IV
`
`111
`
`111; I, 12—18 mo
`
`III
`
`IV
`
`IV
`
`IV
`
`100—250 <3 mo
`80—125 >3 mo
`200—300
`200—300 <12 mo
`100-150 >12 mo
`250—350 <12 mo
`200—300 >12 mo
`
`400—500 <6 mo
`200—400 >6 mo
`
`150—200 <3 mo
`100—150 >3 mo
`150—200 <3 mo
`100—150 >3 mo
`250—350 <3 mo
`150—250 >3 mo
`
`100—200 <3 mo
`75—150 >3 mo
`250—350 <3 mo
`150—250 >3 mo
`250—350 <3 m0
`100—200 >3 mo
`
`250-375 <6 mo
`100—250 >6 mo
`350—450 <1 mo
`250—350 2—6 mo
`170—240 >6 mo
`300—420 <6 wks
`180—300 6—12 wks
`120—180 >12 wks
`
`mFPIA
`
`b.i.d.
`
`II, III in some
`patients
`
`200—250 <3 mo
`150—250 >3 mo
`
`HPLC
`
`b.i.d.
`
`III
`
`111
`
`III
`
`175—225 <3 mo
`150—175 3—12 mo
`100-125 >12 mo
`300—400 <3 mo
`250—300 3-12 mo
`200—250 12—18 mo
`80-200 >18 mo
`250—325 <6 mo
`200—250 6—12 mo
`150—200 >12 mo
`
`K
`
`L
`H
`Lu
`
`L
`
`K
`
`H
`
`K
`
`H
`
`K & K-Panc
`
`L
`
`H
`
`L
`
`K
`
`L
`
`H
`
`Ped K
`
`b.i.d.
`
`111
`
`University of
`California Los
`Angeles (UCLA)
`Medical Center, Los
`Angeles, CA,
`U.S.A. [8]
`
`HPLC and
`pFPIA
`Pediatric K
`only. The
`higher
`concentrations
`pertain to
`pFPIA
`
`250—375
`800—1,000 <1 mo
`225—300
`700—900 1—2 mo
`200-250
`500—750 2—3 mo
`125—200
`400—600 3—4 mo
`100—175
`350—500 4—6 mo
`100—150
`325—425 >1 year
`
`Ther Drug Monit, Vol. 17, No. 6, 1995
`
`

`

`CONSENSUS REPORT ON CYCLOSPORIN
`
`645
`
`TABLE 1. Continued
`
`Immuno-
`Transplant
`Therapeutic
`Dosing
`Analytic
`suppression
`method
`interval
`type
`Center [No.]
`protocol
`ranges (ug/L)
`350—450
`mFPIA and
`III
`b.i.d.
`BOO—1,000 <3 mo
`pFPIA. The
`100—150
`higher
`200—350 >3 mo
`concentrations
`280—300
`pertain to
`500—800 <3 mo
`pFPIA
`200—400
`600—800 <3 mo
`100—200
`400—600 >3 mo
`
`Adult K
`
`Adult L
`
`Adult H
`
`III
`
`III
`
`St George’s Hospital,
`The Medical School,
`London, UK. [9]
`
`Mayo Clinic,
`Rochester, MN,
`U.S.A. [10]
`
`m 125I-RIA
`
`b.i.d.
`
`HPLC
`
`b.i.d.
`
`III
`Steroids stopped
`at 3 mo
`
`III
`
`III
`
`IV
`
`III
`
`III <3 mo
`11 >3 mo
`
`300—350 <6 wk
`200—250 6 wk—6 mo
`150—200 6 mo—l year
`80—120 >1 year
`150—250 <2 wk
`150—200 <2 mo
`100—150 >4 mo
`350
`<2 wk
`250—350 <2 mo
`150—250 <4 mo
`100—150 >4 mo
`250—350 2 wks—2 mo
`75—125 >2 mo
`
`120—200 <3 mo
`100—160 >3 mo
`250—300 <3 mo
`200—250 3—12 mo
`100—150 >12 mo
`
`<1 mo
`550
`2—3 mo
`500
`3—6 mo
`450
`6—12 mo
`400
`12—24 mo
`350
`>24 mo
`300
`800—1,200 <1 mo
`700—1,200 >1 mo
`150—250
`200—300
`150—250
`200—350 <2 mo
`150—250 >3 mo
`
`200—250 <1 mo
`150—200 1—2 mo
`100—150 2—3 mo
`75—100 >3 mo
`200
`
`K, Panc
`
`Panc
`
`For H only,
`pFPIA and
`plasma/37°C
`HPLC
`
`b.i.d.
`
`mFPIA
`
`pFPIA
`
`mFPIA
`
`Dosing based on
`pharmacokinetic
`studies to
`achieve
`steady-state
`conc.
`b.i.d.
`
`b.i.d.
`
`mFPIA
`
`b.i.d.
`
`II
`
`II
`
`III
`III
`III
`III
`
`IV
`
`IV
`
`Princess Alexandra
`Hospital, Brisbane,
`Australia [1 1]
`
`University of Texas
`Health Science
`Center, Medical
`Center, Houston,
`TX, U.S.A. [12]
`
`University of Virginia
`Medical Center,
`Charlottesville, VA,
`U.S.A. [13]
`
`St. Johns Hospital &
`Medical Center,
`Detroit, Michigan,
`U.S.A. [14]
`
`University of Alberta
`Hospitals,
`Edmonton, AL,
`Canada [15]
`
`mFPIA
`
`b.i.d.
`
`III
`
`III
`
`300—400 <2 wks
`250—300 2—4 wks
`200—250 1—3 mo
`150—200 3—6 mo
`100—150 6—12 mo
`100-125 >12 mo
`300—350 <30 days
`250—350 30—60 days
`250—300 60—90 days
`200—250 90—180 days
`175—225 180 days—l year
`150—175 >1 year
`III
`350—500 <90 days
`
`300—350 >90 days
`
`

`

`646
`
`M. OELLERICH ET AL.
`
`TABLE 1. Continued
`Immuno-
`Analytic
`Dosing
`suppression
`Therapeutic
`Transplant
`
`Center [No.]
`method
`interval
`protocol
`ranges (pug/L)
`type
`111
`400—500 <90 days
`H-Lu
`400—500 >90 days
`
`Clin-Tox Associates,
`Germantowu, TN,
`U.S.A. [16]
`
`St. Vincent‘s Hospital,
`Darlinghurst, NSW,
`Australia [17]
`
`pFPIA
`Serum
`
`mFPIA
`
`b.i.d.
`
`b.i.d.
`
`West Virginia
`University
`Hospitals,
`Morgantown, WV,
`U.S.A. [18]
`
`University of North
`Carolina Hospitals,
`Chapel Hill, NC,
`U.S.A. [19]
`
`mFPIA
`
`b.i.d.
`
`mFPIA
`
`b.i.d.
`
`Queen Elizabeth
`Hospital,
`Birmingham, U.K.
`[20]
`
`Huddinge Hospital,
`Stockholm, Sweden
`[21]
`
`mFPIA
`
`b.i.d.
`
`m 125I—RIA
`
`b.i.d.
`
`Neues Allgemeines
`Krankenhaus,
`Vienna, Austria [22]
`Northwestern
`University Medical
`School, Chicago, IL,
`U.S.A. [23]
`
`mFPIA
`
`mFPIA
`
`HPLC
`
`b.i.d.
`
`b.i.d.
`
`b.i.d.
`
`III
`
`III
`
`111
`
`111
`
`III
`
`111
`
`IV
`
`IV in children.
`Prednisone
`stopped after
`3 mo
`IV
`
`111
`
`111
`
`III
`
`IV
`
`II
`
`II + ALA
`111
`
`IV
`
`175~225 <30 days
`125—175 30—90 days
`75—125 >90 days
`
`250-375 <6 mo
`100—250 >6 mo
`350—450 >2 mo
`300—400 2—3 mo
`250—300 3—6 mo
`200-300 6—12 mo
`150—200 >12 mo
`
`250—375 <6 mo
`100—250 >6 mo
`
`~200 initial post Tx
`150—200 >3 mo
`350
`>6 mo
`300
`6—12 mo
`200450 <12 the
`400—500 <1 wk
`250—350 2~3 wks
`200—300 3—4 wks
`180—280 >4 wks
`350
`<6 mo
`300
`6—12
`250
`>12
`
`400—500 <1 wk
`250—350 >2 wks
`
`100—300
`
`250—350 <1 mo
`200—300 1—2 mo
`150—250 2—3 mo
`70—150 >3 mo
`350—450 <9 days
`250—300 9 days—3 mo
`200—250 3—4 mo
`150—200 >4 mo
`300—400 <1 mo
`200-300 1—2 mo
`100—200 2—3 mo
`~100 >3 mo
`
`1254250 <3 mo
`100—200 >3 mo
`125—250
`250—400 <6 mo
`250—300 >6 mo
`200
`< 6 mo
`200
`<6 mo
`
`H
`
`K
`
`H, H-Lu
`
`K
`
`L
`
`H-Lu
`
`Panc
`
`L
`
`K
`
`L
`
`K-Panc, Pane
`
`K
`
`L
`K
`
`L
`Panc
`
`Univ.-Klinikum Rudolf
`Virchow, Berlin,
`PRO [24]
`Academisch
`Ziekenhuis,
`Groningen, Holland
`[25]WW—
`
`mFPIA
`
`HPLC
`
`b.i.d.
`
`b.i.d.
`
`IV
`
`III
`
`2004300 <4 wks
`100—200 >4 wks
`
`200—250 <4 wks
`100—150 >4 wks
`
`L
`
`L
`
`

`

`CONSENSUS REPORT ON CYCLOSPORIN
`
`647
`
`TABLE 1. Continued
`
`Immuno-
`Analytic
`Dosing
`suppression
`Therapeutic
`Transplant
`
`Center [No.]
`method
`interval
`protocol
`ranges (ptg/L)
`type
`
`Hopital Paul Brousse,
`Villejuif, France [26]
`
`mFPIA
`
`b.i.d.
`
`The Liver Unit, The
`Children’s Hospital,
`Birmingham, UK.
`[27]
`Addenbrooke’s
`Hospital,
`Cambridge, UK.
`[28]
`
`Papworth Hospital,
`Cambridge, UK.
`[29]
`
`University of
`Michigan, Ann
`Arbor, MI, U.S.A.
`[30]
`
`HPLC
`
`m 12SI-RIA
`
`EMIT
`
`HPLC
`
`King’s College,
`London, UK. [31]
`
`mFPIA
`
`Virginia
`Commonwealth
`University,
`Richmond, VA,
`U.S.A. [32]
`Hospital das Clinicas,
`Universidade de Sao
`Paulo, Brasil [33]
`University of
`Nebraska Medical
`Center, Omaha, NE,
`U.S.A. [34]
`
`’
`
`mFPIA
`
`m 125I-RIA
`
`pFPIA
`
`b.i.d.
`
`b.i.d.
`
`b.i.d.
`
`b.i.d.
`
`b.i.d.
`
`b.i.d.
`
`q.d.
`b.i.d.
`
`t.i.d.
`
`t.i.d.
`b.i.d.
`
`b.i.d.
`
`b.i.d.
`
`b.i.d.
`
`b.i.d.
`
`b.i.d.
`
`111
`
`III
`
`III
`
`III
`
`III
`
`III
`
`111
`
`IV
`IV
`
`IV
`
`III
`IV
`
`111
`
`111
`
`IV
`
`111
`
`II
`
`L
`
`L
`
`K
`
`Adult L
`
`Ped L
`
`H
`
`H-Lu
`
`K
`L
`
`H
`
`Lu
`Panc
`
`Adult L
`
`Ped L
`
`L
`
`K
`
`L
`
`400—500 1—15 days
`300—450 15—30 days
`250—300 30—90 days
`150—200 >12 mo
`
`<41/2 mo
`>250
`150—200 41/2—12 mo
`80—120 >12 mo
`
`200—250
`175—250
`200-250
`175—250
`150—250
`
`i.v.
`oral
`i.v.
`oral
`
`300—400 <3 mo
`200—300 3—12 mo
`100—250 >12 mo
`300—500 <3 mo
`200—300 3—12 mo
`100—250 > 12 mo
`
`100—150
`<3 mo
`150
`100—150 >3 mo
`200—250 <6 mo
`150—200 6—12 mo
`100—150 12—24 m0
`75—100 224 mo
`100—300
`225—275 <1 mo
`175—225 1—2 mo
`125—175 >2 m0
`
`150—250 <3 wks
`100—200 3—8 wks
`<125
`>12 wks
`150—250 <6 mo
`100—150 6—12 mo
`50—100 > 12 mo
`
`400
`300
`200
`
`<6 mo
`6—12 mo
`>12 mo
`
`160—200 <6 mo
`100—150 >6 mo
`
`900—1,100 >2 mo
`600—800 2—4 mo
`500—600 4—6 mo
`400—500 6—12 mo
`300—400 >12 mo
`
`Medizinische
`Hochschule,
`Hannover, FRG [35]
`
`EMIT
`
`b.i.d.
`
`100—200 <3 mo
`50—150 >3 mo
`250—300 <12 mo
`200—250 >12 mo
`L
`100—200 <3 mo
`11 + ALA
`50—150 >3 m0
`
`IV or
`II + ALA
`IV
`
`K
`
`H, H-Lu
`
`Except where listed otherwise, all centers use whole blood specimens, obtained just before the next dose, for CsA determinations.
`HPLC, high—performance liquid chromatography; mFPIA, monoclonal antibody Abbott TDx fluorescence polarization immunoassay;
`pFPIA, polyclonal antibody Abbott TDX fluorescence polarization immunoassay; m 125I-RIA, monoclonal antibody INCSTAR radio-
`immunoassay; EMIT, monoclonal antibody Syva enzyme-multiplied immunoassay; b.i.d., twice-daily dosing; t.i.d., three times daily
`dosing; I, CsA monotherapy; 11, “double therapy“: CsA + prednisone; III, “triple therapy”: CsA + prednisone + azathioprine; IV,
`“quadruple therapy,” induction therapy in which anti-lymphocyte antibody (ALA) is used as part of initial immunosuppression until
`good kidney function is achieved <14 days after surgery. Many centers use quadruple therapy in transplant patients with poor initial
`renal function only. This use of IV is not specifically defined for each center. Only when IV is used in all patients for a particular
`transplant type is this protocol noted; K, kidney; H, heart; L, liver; Lu, lung; Pane, pancreas; Ped, pediatric.
`
`

`

`648
`
`M. OELLERICH ET AL.
`
`intraindividual variability in the pharmacokinetics
`of the drug, and an enhanced risk of acute infection.
`During this early critical period, a monitoring
`schedule of four to seven samples per week would
`appear to be appropriate for kidney, liver, and heart
`transplant recipients (6). In liver transplantation,
`regular monitoring is essential after the start of oral
`CsA to compensate for inter— and intraindividual
`variation in pharmacokinetics at this stage of clini-
`cal and pharmacologic instability. Because the new
`microemulsion formulation of CsA appears to be
`less dependent on bile acids for its absorption, it
`may help to reduce such pharmacokinetic variabil-
`ity and thereby reduce requirements for such fre-
`quent CSA monitoring. After the intensive early
`monitoring, the measurement of cyclosporin blood
`levels can be gradually reduced; for example,
`in
`renal transplant recipients with an uncomplicated
`clinical course, CsA should be monitored once a
`month during the first year and at 1 to 3-month in-
`tervals thereafter. However, there are no hard and
`fast rules, and measurements should be performed
`if the clinical signs of symptoms suggest that dosage
`adjustment might be necessary. Furthermore, addi-
`tional measurements should be performed when the
`patient has to be treated with drugs known to inter-
`act with the metabolism or excretion (or both) of
`CsA. If adjustment is required, this should be fol—
`lowed—up with a second blood-concentration mea-
`surement within the following week. It is important
`that CsA measurements be made available on the
`
`same day if they are to influence therapy. This is
`feasible with the latest generation of semiautomated
`immunoassays, which offer a same-day service
`even with a large workload.
`
`METHODS OF MEASUREMENT
`
`Carefully validated high-performance liquid chro—
`matography (HPLC) methods that measure the par—
`ent drug specifically can be used as reference
`procedures for CsA measurement. A definitive
`method, however, or a reference method according
`to the IFCC criteria (14) is not yet available. Recent
`reports (6,15,16) have noted a trend toward the use
`of semiautomated immunoassay techniques involv—
`ing selective monoclonal antibodies rather than
`HPLC for routine measurement of CsA. Data from
`
`the UK Cyclosporine Quality Assessment Scheme
`(UKCQAS), which is predominantly European
`based, show that most laboratories are using one of
`the following three immunoassays: the INCSTAR
`
`Ther Drug Monit, Vol. 17, No. 6, 1995
`
`CYCLO-Trac SP radioimmunoassay (m 125I-RIA),
`the Abbott TDx monoclonal antibody fluorescence
`polarization immunoassay (mFPIA), and the Syva
`enzyme multiplied immunoassay technique
`(EMIT). For nonspecific measurements, both the
`Abbott TDx polyclonal antibody fluorescence po—
`larization assay (pFPIA) and the Incstar nonspecific
`monoclonal RIA are available. The nonspecific RIA
`detects a broader range of metabolites than the pF-
`PIA. Data from other schemes and a recent survey
`summarized in Table 1 are in broad agreement with
`these findings. A receptor assay approach for the
`quantification of CsA that utilizes the specific bind~
`ing of the latter to certain immunophilins (17) is
`currently being evaluated (18).
`The choice of analytic technique depends on a
`number of factors, including available instrumenta—
`tion,
`the technical expertise available, sample
`throughput, the requirement for after—hours work,
`the clinical indications for the use of cyclosporin,
`and local regulations governing the use of radio-
`chemicals.
`
`In the previous consensus conference (19), the
`following performance characteristics were recom—
`mended for a method to be acceptable for selective
`determination of the parent drug CsA:
`
`1. Imprecision: Coefficient of variation of S10%
`at a CsA concentration of 50 ug/L and of <5%
`at a concentration of 300 ug/L.
`2. Accuracy (method comparison with a vali—
`dated HPLC reference method): Slope of the
`line: s10% from the line of identity, intercept
`<15 ug/L; sy/XzsIS pug/L.
`
`These precision requirements should refer to be-
`tween-days imprecision. For the statistical evalua—
`tion of method comparison, a bivariate procedure
`[e.g., principal component analysis (20) or weighted
`Deming (21)], or a nonparametric rank procedure
`[e.g., Passing/Bablok method (21,22)] should be
`used.
`
`PERFORMANCE CHARACTERISTICS
`
`The performance characteristics of the specific
`semiautomated cyclosporine immunoassays have
`recently been reviewed (6). The coefficients of vari—
`ation at CsA concentrations between 50 ug/L and
`~350 ug/L ranged from 7 to 15% with EMIT, 3 to
`10% with mFPIA, and 5 to 11% with m IZSI-RIA.
`With an improved modification of the EMIT, be—
`tween-days coefficients of variation were 5 and 8%
`
`

`

`CONSENSUS REPORT ON CYCLOSPORIN
`
`649
`
`at CsA concentrations of 95 Mg/L and 225 ug/L,
`respectively (23). Precision requirements as pro-
`posed by the previous consensus conference were
`best fulfilled by mFPIA compared to the remaining
`immunoassays (6,24).
`The mean overestimation of CsA concentrations
`
`compared with HPLC due to the presence of cross-
`reactivity with CsA metabolites ranged from 8 to
`30% with EMIT, 24 to 48% with mFPIA, and 22 to
`30% with m 125I-RIA. As the EMIT showed the
`highest specificity and exact calibration, this test
`best fulfilled the proposed performance criteria for
`accuracy. The reasons for the discrepancies be-
`tween the methods include specificity of the anti-
`bodies used in the immunoassays and between-
`method differences in the calibration. Particularly
`in patients who may show an accumulation of CsA
`metabolites (e.g.,
`liver transplant recipients), re-
`sults for the same sample can differ by as much as
`100%, depending on which of the specific assays is
`used. Because CsA metabolites do not significantly
`contribute to overall immunosuppression, the use
`of less specific assays in such patients may result in
`the physician underdosing the patient. Assay-
`related differences contribute to the disparity in tar-
`get concentration ranges quoted by individual cen-
`ters (Table 1). The impact of such discrepancies on
`clinical decision making requires further clarifica—
`tion. One area in which analytic accuracy is of par—
`ticular relevance is in the conduct of randomized
`concentration-control clinical trials (25). Because
`such trials are likely to involve a multicenter design,
`constancy of results, in terms of both accuracy and
`precision, is of importance, particularly if multiple
`laboratories rather than a central facility are used to
`monitor concentrations.
`
`QUALITY ASSESSMENT
`
`Previous consensus documents on CsA monitor—
`
`ing have recommended that laboratories should par-
`ticipate in external quality assessment programs
`(16,19). It is also mandatory that laboratories offer—
`ing a service for measurement of cyclosporin have a
`system in place to verify the day-to—day constancy
`of their results using at least one measure of internal
`quality control. National external quality control
`schemes have been set up in various countries. The
`U.K. CQAS Scheme (6), for example, currently
`covers >280 centers in 38 countries. The most re-
`cent findings from this scheme show that >90% of
`the results reported relate to whole blood samples,
`
`and 89% of these are produced by methods with a
`high specificity for the parent compound (6). Re-
`sults from the Canadian Quality Assurance Program
`confirm that HPLC, RIA, and EMIT methods are
`more specific than FPIA (26). To verify correct cal-
`ibration, whole blood samples that have been
`spiked with standard reference material (Cyclospor-
`ine USP-RS; Cat. No. 15850) should be used. For
`the assessment of the specificity of the applied
`method, samples containing various amounts of the
`major cyclosporin metabolites,
`in addition to the
`parent drug, should be a part of the external quality
`control scheme.
`
`CYCLOSPORIN METABOLITES
`
`The clinical relevance of CsA metabolites is still a
`
`controversial issue (6,27). Some immunosuppres-
`sive activity has been found in vitro for metabolites
`AMI, AM9, and AM4N (28—30). Although overim-
`munosuppression induced by high concentrations
`of cyclosporin metabolites could increase the risks
`of serious infection, the relative toxicities of cyclo-
`sporin metabolites are probably more important
`clinically. There is some evidence that high blood
`concentrations of metabolites AMlc9 and AM19
`
`are associated with nephrotoxicity in the early post-
`operative period after liver transplantation (31).
`From the study design, however, it was not possible
`to ascertain whether high metabolite concentrations
`were the cause or the consequence of renal dys-
`function. In vivo studies in rats receiving subcuta-
`neous doses of AMI, AMIA, AMlc, and AM4N
`could not demonstrate renal or hepatic dysfunction
`after exposure to these metabolites (32). Because of
`the known differences in the metabolization of CsA
`in rats and humans, and interindividual susceptibil-
`ities to CsA toxicity, definite conclusions cannot be
`made as to the contribution of CsA metabolites to
`toxicity in humans.
`Some centers have advocated the parallel use of
`both nonspecific and specific methods to gain an
`insight into the ratio between CsA and its metabo-
`lites in transplant recipients with severely disturbed
`liver function (33). They suggested that concentra—
`tions of parent drug and metabolites >1,200 pug/L,
`as determined with the Incstar nonspecific mono-
`clonal RIA (33), should be avoided. For the large
`majority of clinical situations, however, monitoring
`of CsA metabolites seems not to be warranted. The
`failure of pFPIA to distinguish CsA nephrotoxicity
`from rejection (34,35) would argue against a signif-
`
`Ther Drug Monit, Vol. 17, No. 6, I995
`
`

`

`650
`
`M. OELLERICH ET AL.
`
`icant role of metabolites. From the available data, it
`cannot be decided whether the specific measure—
`ment of individual cyclosporin metabolites will be
`of clinical significance.
`
`THERAPEUTIC RANGES AND
`IMMUNOSUPPRESSION PROTOCOLS
`
`In contrast to the concept of reference intervals
`in clinical chemistry, there is no generally accepted
`concept or protocol on how to establish the thera-
`peutic range of a drug. There is consensus that it
`represents the range of drug concentrations within
`which the probability of the desired clinical re-
`sponse is relatively high and the risk of unaccept-
`able toxicity is relatively low. This implies that a
`given concentration within the therapeutic range is
`not necessarily safe and effective for every patient.
`It is difficult to establish a therapeutic range for
`CsA, because there are no simple parameters for
`the assessment of the immunosuppressive effect.
`On the other hand, the lack of such criteria and the
`marked intra— and interindividual variation in CsA
`
`pharmacokinetics are a strong argument for immu-
`nosuppressive drug monitoring to prevent over- or
`underimmunosuppression. There is general agree—
`ment that CsA monitoring has helped to avoid toxic
`concentrations. On the basis of their own and pub—
`lished experience, transplant centers have derived
`therapeutic ranges empirically for the different
`transplant types. These ranges show substantial
`variability between centers (Table 1). Differences
`relate mainly to the specificity of the analytic meth—
`ods used for measurement of CsA and the various
`immunosuppression protocols employed, including
`center-specific preference in the degree of initial ex-
`posure of patients to CsA.
`To facilitate the establishment of consensus ther-
`
`apeutic ranges for the different transplant types, the
`following points have to be considered: use of se—
`lective assays with standards and samples in whole
`blood matrix, sampling time, and time interval from
`transplantation, biochemical and morphologic crite-
`ria for rejection or toxicity, and influence of immu-
`nosuppression protocol.
`The data obtained by us in a survey conducted in
`1994 through 1995 on the acceptable therapeutic
`ranges for CsA concentration are summarized in
`Table 1. The use of time—dependent target ranges of
`CsA is widespread. Most centers recommend a
`higher range of target concentrations during the
`
`Ther Drug Monit, Vol. 17, No. 6, 1995
`
`early postoperative period. Doses are then tapered
`to a lower maintenance concentration range, usu-
`ally 3 to 6 months after transplantation, although
`some centers extend this tapering period over 12
`months or more.
`The median values for the minimum and maxi-
`
`mum concentrations in pre-dose trough samples ob-
`tained with mFPIA, m 125I-RIA, and HPLC are
`shown in Table 2. As can be expected, with similar
`immunosuppression protocols, centers using mF-
`PIA for monitoring report median upper and lower
`limits that are on average ~28% (range, 0—67%)
`higher than those given by centers using specific
`HPLC methods. Surprisingly, the median m 125I-
`RIA/HPLC ratio of the therapeutic window limits
`was 1.00 (range, 0.72—1.59). The similarity in the
`therapeutic ranges from centers using either HPLC
`or m 125I—RIA is even more astonishing in View of
`the fact that the m 125I—RIA overestimates the CsA
`concentration by 22—30%. The target ranges for
`CsA should preferably be defined on the basis of
`specific measurement of the parent drug. Manufac-
`turers should be encouraged to improve their im-
`munoassays accordingly. However, it is also clear
`(Table 1) that organ- and time-specific therapeutic
`ranges vary widely, even between centers using the
`same analytic procedure to determine CsA and
`comparable immunosuppression protocols. As an
`example, Fig. 1 shows therapeutic CsA ranges used
`in kidney transplant recipients. This possibly re-
`flects the different assessment of CsA toxicity risk
`on the one hand and the consequences of underdos-
`ing (acute or chronic rejection) on the other hand.
`Thus there has been increasing emphasis in some
`centers over the past few years on using higher dos-
`ages and maintaining higher CsA blood concentra-
`tions (36). In a recent publication, Soin et a1. (37)
`found a significantly lower incidence of chronic re-
`jection in those patients who were maintained on
`median CsA whole blood trough levels of 2175
`ug/L in the first 28 days after transplantation.
`A review of the information provided in Table 1
`shows that the concentration ranges are somewhat
`higher for heart transplant than for kidney and liver
`transplant patients. Higher CsA doses are used in
`heart or heart—lung transplant recipients as an extra
`measure of immunosuppression because these or-
`gans are more difficult to replace. The consequence
`of intractable acute rejection in heart or heart—lung
`recipients is usually death. In heart—lung recipients,
`the CsA target whole blood concentrations range
`from 350 to 600 tug/L during induction therapy (ap-
`
`

`

`CONSENSUS REPORT ON CYCLOSPORIN
`
`651
`
`TABLE 2. Therapeutic ranges for cyclosporin stratified according to
`transplanted organ, immunosuppressive regimen, induction/maintenance therapy
`and immunoassay technique
`
`
`Kidney
`Heart
`L‘Ver
`
`Method
`Triple therapy
`Triple therapy
`Triple therapy
`Double therapy
`Induction“
`HPLC
`mFPIA
`m 12sI-RIA
`EMIT
`Maintenance
`HPLC
`mFPIA
`m 125I-RIA
`EMIT
`
`150—225 (5)
`250-375 (6)
`160—200 (5)
`125—200 (2)
`
`100—150 (5)
`100—250 (8)
`75—150 (5)
`75—150 (2)
`
`250—325 (1)
`300—400 (3)
`250—325 (2)
`275—375 (2)
`
`125—175 (2)
`150—250 (5)
`90—160 (2)
`150—250 (3)
`
`225—300 (4)
`250—313 (8)
`250—300 (3)
`
`100—150 (6)
`135—200 (8)
`150—238 (4)
`
`300—375 (2)
`
`125—200 (2)
`
`100—150 (1)
`150—250 (3)
`
`75—150 (2)
`
`The ranges are the median values (pg/L) for the minimum and maximum trough cyclosporin
`concentrations (whole blood) calculated from the data of those centers listed in Table 1 that
`fitted the particular category. The number of contributing centers is given in parentheses.
`HPLC, high—performance liquid chromatography; mFPIA, monoclonal antibody fluorescence
`polarization immunoassay; m ‘ZSI-RIA, monoclonal antibody INCSTAR radioimmunoassay;
`EMIT, enzyme-multiplied immunoassay technique.
`a In some centers, anti-lymphocyte antibodies were also included as part of induction ther-
`apy.
`
`prox. S3 months after transplantation) and from
`200 to 300 pug/L during maintenance therapy.
`A high fluctuation in blood CsA levels was found
`to increase the short—term risk of acute lung or kid-
`ney rejection (38,39). Thus efforts should be di-
`rected to maintaining stable blood CsA concentra-
`tions. The tendency of reducing maintenance immu-
`nosuppression over time to reduce the incidence of
`infection and chronic nephrotoxicity should be
`weighed against the emerging evidence of the ben-
`efit of higher CsA doses and blood levels for long-
`term graft survival (36). CsA dosage individualiza-
`tion is particularly important in the cystic fibrosis
`subgroup of heart—lung transplant recipients. Be-
`cause of poor CsA absorption, these patients re—
`quire 2—3 times the daily dose, usually administered
`in three to four divided doses, to achieve equivalent
`blood CsA conccntrations compared to noncystic
`heart—lung recipients (40).
`For kidney transplant recipients, double (CsA
`and steroids), triple, and quadruple induction treat—
`ment protocols are being used (Table 1). Most are
`designed to introduce CsA slowly, particularly in
`patients with delayed kidney function, while pro-
`viding effective immunosuppressive cover with al-
`ternative agents. Some patients can be managed on
`CsA monotherapy or can be switched to monother—
`apy from other regimens (41). At various centers,
`there is an apparent increase in attempts to elimi—
`nate steroids in selected populations (Table 1). Rou—
`tine CsA monitoring is particularly valuable in kid-
`
`ney transplant recipients, because an increasing se-
`rum creatinine can be the result of either rejection
`or nephrotoxicity. A recent study (7) investigated
`the diagnostic utility of whole blood CsA levels
`measured by the m 125I-RIA within the first 100
`days after renal transplantation in patients receiving
`triple therapy. It was concluded that when using
`low-dose triple—therapy regimens, CsA levels be-
`tween 150 and 400 lug/L were of little diagnostic
`value in acute allograft dysfunction, whereas levels
`outside this range were useful in the clinical diag-
`nosis of CsA nephrotoxicity and acute allograft re-
`jection. A review of the existing data on the use of
`CsA in kidney transplantation, particularly with re-
`gard to monitoring of CsA concentrations, has re—
`cently been published (42).
`In pediatric kidney transplant recipients with a
`CsA—steroid regimen, CsA target whole blood con—
`centrations of 150—250 pug/L and 100—150 ug/L have
`been recommended for induction and maintenance
`
`therapy, respectively (43). In general, therapeutic
`CsA ranges used in pediatric patients are similar to
`those employed in adults (Table 1). However, dos-
`age requirements (mg/kg) to achieve these ranges
`are greater in children.
`Recent investigations in patients with simulta—
`neous pancreas—kidney (SPK) transplants point to
`an increased incidence of acute rejection at CsA
`levels S200 pig/L as determined by HPLC (unpub-
`lished data). From the results of a further study in
`SPK recipients, it

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