throbber
Downloaded from
`
`jpet.aspetjournals.org
`
` at ASPET Journals on January 16, 2017
`
`0022-3565/00/2941-0323$03.00/0
`THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS
`Copyright © 2000 by The American Society for Pharmacology and Experimental Therapeutics
`JPET 294:323–332, 2000 /2243/834819
`
`Vol. 294, No. 1
`Printed in U.S.A.
`
`Tissue Distribution and Clinical Monitoring of the Novel
`Macrolide Immunosuppressant SDZ-RAD and Its Metabolites
`in Monkey Lung Transplant Recipients: Interaction with
`Cyclosporine1
`
`NATALIE SERKOVA, BERNARD HAUSEN, GERALD J. BERRY, WOLFGANG JACOBSEN, LESLIE Z. BENET,
`RANDALL E. MORRIS, and UWE CHRISTIANS
`
`Department of Biopharmaceutical Sciences, School of Pharmacy, University of California, San Francisco, California (N.S., W.J., L.Z.B., U.C.);
`and Transplantation Immunology, Department of Cardiothoracic Surgery (B.H., R.E.M., U.C.), and Department of Pathology (G.J.B.), Stanford
`University Medical School, Stanford, California
`Accepted for publication March 27, 2000
`
`This paper is available online at http://www.jpet.org
`
`ABSTRACT
`We report the tissue distribution and clinical monitoring of the
`novel macrolide immunosuppressant SDZ-RAD [40-O-(2-hy-
`droxyethyl)-rapamycin] and its metabolites in monkey lung trans-
`plant recipients as well as its interaction with cyclosporine as the
`Neoral formulation. After left unilateral lung transplantation, cyno-
`molgus monkeys received by oral administration either 1) 1.5
`mg/kg/day SDZ-RAD (n 5 4); 2) 100 mg/kg/day cyclosporine (n 5
`4); 3) 0.3 mg/kg/day SDZ-RAD 1 100 mg/kg/day cyclosporine
`(n 5 6); 4) 1.5 mg/kg/day SDZ-RAD 1 50 mg/kg/day cyclosporine
`(n 5 5); or 5) SDZ-RAD and cyclosporine doses adjusted accord-
`ing to trough blood concentration measurements (n 5 6). At the
`end of the observation period (usually 29 days after transplanta-
`tion), and 24 h after the last doses, tissue samples were collected
`and analyzed with HPLC/mass spectrometry. Gall bladder, pan-
`creas, the transplant lung, cerebellum, kidneys, and spleen had
`the highest SDZ-RAD concentrations. Coadministration of cyclo-
`
`sporine increased SDZ-RAD concentrations in most tissues as
`well as tissue-to-blood distribution coefficients. In contrast, SDZ-
`RAD had only a small effect on cyclosporine blood and tissue
`concentrations. Rejection in lung grafts in monkeys treated with
`either of the cyclosporine/SDZ-RAD combinations was signifi-
`cantly less than in the monotherapy groups (P , .002). Histolog-
`ical rejection scores were inversely correlated with SDZ-RAD con-
`centrations in blood (r 5 20.68; P , .001; n 5 24), lymph nodes
`(P 5 20.58; P , .003; n 5 24), thymus (r 5 20.63; P , .001; n 5
`23) and transplant lung tissue (r 5 20.58; P , .003; n 5 24). We
`conclude that, in addition to the synergistic pharmacodynamic
`interaction, a pharmacokinetic interaction resulting in higher SDZ-
`RAD tissue concentrations contributed to the significantly better
`immunosuppressive efficacy when both drugs were combined
`compared with monotherapy.
`
`The novel macrolide SDZ-RAD [40-O-(2-hydroxyethyl)-
`rapamycin] is currently in phase II/III clinical trials as an
`immunosuppressant coadministered with microemulsion cy-
`closporine (Neoral;
`international nonproprietary name:
`ciclosporin) after organ transplantation. SDZ-RAD is a semi-
`synthetic derivative of rapamycin (international nonpropri-
`etary name: sirolimus). Although not yet proven, it is as-
`sumed that SDZ-RAD has the same molecular actions as
`rapamycin. The rapamycin/FK-binding protein (FKBP) com-
`plex, and probably also the SDZ-RAD/FKBP complex, binds
`
`Received for publication November 1, 1999.
`1 This study was supported in part by the Alexander von Humboldt-Foun-
`dation, Grant V-3-FLF-1052812 (to N.S.); the Deutsche Forschungsgemein-
`schaft Grants Ch 95/6-2 (to U.C.) and Ha 1967/2-1 (to B.H.); National Insti-
`tutes of Health Grants CA72006 and GM26691 (to L.Z.B.); the Ralph and
`Marian Falk Medical Research Fund; the Hedco Foundation; and a grant from
`Novartis Pharma AG, Basel, Switzerland (to R.E.M.).
`
`in T lymphocytes to mTOR, the mammalian target of rapa-
`mycin. The results are inhibition of the interleukin-2-stimu-
`lated phosphorylation activation of p70-kd S6 protein kinase
`and blockade of cell cycle progression at the G1-S-interface
`(Schuler et al., 1997; Bo¨hler et al., 1998). While the cal-
`cineurin inhibitor cyclosporine inhibits interleukin-2 synthe-
`sis, SDZ-RAD inhibits interleukin-2-mediated T-lymphocyte
`proliferation. It is generally thought that inhibition of the
`subsequent steps of T-lymphocyte proliferation is the main
`reason for the synergistic immunosuppressive interaction of
`cyclosporine and SDZ-RAD observed in in vitro and in vivo
`studies (Schuler et al., 1997; Schuurman et al., 1997, 1998).
`Major side effects of cyclosporine include nephrotoxicity, hep-
`atotoxicity, neurotoxicity, and hypertension (Kahan, 1989).
`The side effect pattern of SDZ-RAD in patients can be ex-
`pected to be similar to that of rapamycin (Murgia et al.,
`
`ABBREVIATIONS: FKBP, FK-binding protein; CYP, cytochrome P450; MS, mass spectrometry; amu, atomic mass units.
`
`323
`
`West-Ward Exhibit 1091
`Serkova 2000
`Page 001
`
`

`

`Downloaded from
`
`jpet.aspetjournals.org
`
` at ASPET Journals on January 16, 2017
`
`324
`
`Serkova et al.
`
`1996): thrombocytopenia, hyperlipidemia, and gastrointesti-
`nal disorders.
`Access of cyclosporine to the various body compartments is
`governed by cytochrome P450 (CYP)3A, ATP-binding cas-
`sette protein transporters, and binding to immunophilins
`(Ryffel et al., 1991; Wacher et al., 1998). Like cyclosporine,
`SDZ-RAD is a substrate of both CYP3A and the ATP-binding
`cassette protein transporter p170-glycoprotein (Crowe and
`Lemaire, 1998; Crowe et al., 1999). Because many of the
`factors that determine cyclosporine pharmacokinetics also
`are involved in SDZ-RAD pharmacokinetics, we hypothe-
`sized that cyclosporine might have a significant effect on
`SDZ-RAD tissue distribution, thus influencing its immuno-
`suppressive efficacy and its tolerability.
`Due to the poor oral bioavailability of microemulsion cyclo-
`sporine in cynomolgus monkeys (Schuurman et al., 1996),
`significantly higher doses than in humans are required to
`maintain cyclosporine trough blood concentrations in the
`target range proposed for clinical lung transplantation. The
`cyclosporine (100 mg/kg) and SDZ-RAD (1.5 mg/kg) doses
`used in our study were based on those used in the same
`animal species by Schuurman et al. (1998). These doses re-
`sulted in trough blood concentrations in the therapeutic
`range for patients and were found to be effective in prevent-
`ing rejection of life-supporting kidney transplants for .50
`days. Because coadministration of 100 mg/kg cyclosporine
`and 1.5 mg/kg SDZ-RAD was tolerated poorly (Hausen et al.,
`2000), doses of 50 mg/kg cyclosporine and 1.5 mg/kg SDZ-
`RAD (for comparison with the SDZ-RAD monotherapy group)
`and of 100 mg/kg cyclosporine and 0.3 mg/kg (for comparison
`with the cyclosporine monotherapy group) were combined.
`These doses were based on the results of tolerability studies
`(Hausen et al., 2000). In addition to these fixed-dose drug
`regimens, we also included a study group in which, as in the
`clinical practice, doses were adjusted according to blood
`trough concentrations (20–40 mg/l for SDZ-RAD and 100–
`200 mg/l for cyclosporine). The cyclosporine target concentra-
`tions were based on those used in patients (Oellerich et al.,
`1995).
`
`Experimental Procedures
`Materials. SDZ-RAD, cyclosporine, and cyclosporin D were pro-
`vided by Novartis Pharma AG (Basel, Switzerland). Acetonitrile
`(HPLC grade), sulfuric acid (American Chemical Society grade),
`methanol, and methylene chloride were obtained from Fisher Scien-
`tific (Fairlawn, NJ). Zinc sulfate, formic acid, and sodium formate
`(all American Chemical Society grade) were purchased from Sigma
`Chemical Co. (St. Louis, MO). Extraction columns (bonded phase
`C18; 1 ml) were from Varian Sample Preparation Products (Harbor
`City, CA). The internal standard used for quantification of SDZ-
`RAD, 28-,40-O-diacetyl rapamycin, was synthesized as described by
`Streit et al. (1996). Analytical columns (250 3 4 mm) filled with
`Hypersil (Shandon, Chadwick, UK) C8, 3-mm material were from
`Keystone Scientific (Bellefonte, PA). HPLC micro vials, 100-ml in-
`serts, and Teflon screw caps were purchased from Hewlett Packard
`(Palo Alto, CA).
`Samples were analyzed on an HPLC/electrospray-mass spectrom-
`etry (MS) system consisting of a series 1100 HPLC system (G1322A
`degasser, G1312A binary pump, G1313A autosampler, and G1316A
`column thermostate), a 59887A electrospray interface equipped with
`an Iris hexapole ion guide (Analytica of Brandford, Brandford, CT),
`and a 5989B MS (Hewlett-Packard, Palo Alto, CA).
`
`Vol. 294
`
`Lung Transplantation in Cynomolgus Monkeys. The animals
`received humane care in compliance with the Principles of Labora-
`tory Animal Care (National Society for Medical Research) and the
`Guide for Care and Use of Laboratory Animals (National Academy of
`Sciences, published by the National Institutes of Health). The Insti-
`tutional Animal Care and Use Committee of Stanford University
`granted approval.
`imported by the
`Cynomolgus monkeys, Macaca fascicularis,
`Charles River Biomedical Research Foundation (Houston, TX) from
`Mauritius, were free of herpes B virus and Sendai virus. All animals
`were quarantined for a minimum of 2 months at Charles River and
`for 7 weeks at Stanford. While in quarantine, blood was drawn on
`each donor/recipient pair of animals for a mixed lymphocyte reaction
`assay after the protocol kindly provided by Dr. B. A. Cosimi (Harvard
`Medical School, Boston, MA). Donor and recipient monkeys were
`blood-group matched and mixed lymphocyte reaction mismatched to
`obtain a stimulation index of at least 2.5. After quarantine, left
`unilateral lung transplantation was carried out as described by
`Cooper (1989). For the first 24 postoperative hours, the monkeys
`were placed into intensive care cages supplied with oxygen and
`warm, humid air. They had free access to food and water 4 h after
`transplantation and were transferred to the regular animal room 1
`day later. The animals were assigned to one of five treatment groups
`after transplantation: 1) 1.5 mg/kg/day SDZ-RAD (group 1.5RAD;
`n 5 4); 2) 100 mg/kg/day cyclosporine (group 100Cs; n 5 4); 3) 0.3
`mg/kg/day SDZ-RAD 1 100 mg/kg/day cyclosporine (group 0.3RAD 1
`100Cs; n 5 6); 4) 1.5 mg/kg/day SDZ-RAD 1 50 mg/kg/day cyclospor-
`ine (group 1.5RAD 1 50Cs; n 5 5); and 5) concentration-controlled
`dose adjustments based on SDZ-RAD and cyclosporine trough blood
`concentrations (C24 h, group CCRAD 1 Cs; n 5 6). In the groups with
`100 mg/kg/day cyclosporine, the monkeys initially received 150 mg/
`kg/day. On postoperative day 7, the dose was lowered to 100 mg/kg/
`day and remained unchanged for the remainder of the study period.
`In the concentration-controlled dosing group (CCRAD 1 Cs), doses
`were adjusted to maintain SDZ-RAD and cyclosporine trough blood
`concentrations in target concentration ranges of 20 to 40 mg/l and 100
`to 200 mg/l, respectively. During the study period, the mean 6 S.D.
`daily SDZ-RAD dose in this group was 0.6 6 0.1 and 38 6 1 mg/kg for
`cyclosporine. Immunosuppressive therapy was started immediately
`after surgery (day 0). Cyclosporine (Neoral) and SDZ-RAD (in meth-
`ylcellulose vehicle) were given daily as a single oral dose. In addition,
`animals received a single dose of methylprednisolone (4 mg/kg i.v.) at
`the day of surgery, and the antibiotics cefazolin (25 mg/kg i.m.) and
`gentamycin (3 mg/kg s.c.) for the first 4 days after surgery. At either
`the end of the observation period (29 days after surgery) or when the
`animals’ health status seriously deteriorated, and 24 h after the last
`dose, the animals were sacrificed and the following samples were
`collected for measurement of drug tissue distribution: blood, brain
`stem, cerebellum, cerebrum, colon, duodenum, fat, gall bladder,
`heart, ileum, jejunum, kidney, liver, transplant lung, native lung,
`lymph nodes, pancreas, spleen, stomach, testis, and thymus. The
`tissues were frozen in liquid nitrogen immediately after collection
`and stored at 280°C until HPLC/MS analysis. Samples were ana-
`lyzed within 20 days.
`Extraction of Tissue Samples. Tissue samples were thawed,
`weighed, and homogenized with 2 ml of KH2PO4 buffer [pH 5 7.4
`(1 M)] with a Teflon-glass manual homogenizer. One milliliter of
`homogenate was taken for analysis. Cyclosporin D and 28-,40-di-
`acetyl sirolimus (in acetonitrile/sulfuric acid; pH 5 3; 90:10 v/v) were
`added as internal standards, resulting in final concentrations of 100
`mg/l of each. After addition of 2 ml of methanol/0.4 M ZnSO4 (80:20
`v/v) for protein precipitation, the samples were vortexed for 30 s and
`centrifuged at 1500g for 3 min. The organic supernatant was loaded
`on C18 extraction cartridges by drawing the samples through the
`columns with a 210 mm Hg vacuum. The extraction columns had
`previously been primed with 3 ml of water and 3 ml of acetonitrile.
`Immunosuppressants, metabolites, and internal standards were
`washed on the columns with 3 ml of water. The extraction columns
`
`West-Ward Exhibit 1091
`Serkova 2000
`Page 002
`
`

`

`Downloaded from
`
`jpet.aspetjournals.org
`
` at ASPET Journals on January 16, 2017
`
`2000
`
`were dried by drawing air for 5 min. Samples were eluted with 1.5 ml
`of methylene chloride. The samples were evaporated to dryness
`under a stream of nitrogen at 50°C. The residues were reconstituted
`in 120 ml of acetonitrile/0.1% formic acid (75:25 v/v) and were trans-
`ferred into micro HPLC vials with conical 100-ml inserts and sealed
`with Teflon screw caps.
`HPLC/Electrospray-MS Analysis. Extracts (100 ml) were in-
`jected onto a 250- 3 4-mm analytical column filled with Hypersil
`C8-material of 3-mm particle size. SDZ-RAD, cyclosporine, their me-
`tabolites, and the internal standards were eluted from the analytical
`column with methanol and 0.1% formic acid supplemented with 1
`mmol/l sodium formate. The following linear gradient was used: 0
`min, 70% methanol; and 30 min, 90% methanol. The column was
`reequilibrated to starting conditions for 5 min before injection of the
`next sample. The flow rate was 0.4 ml/min and the column temper-
`ature was 65°C. The drying gas in the electrospray source was
`adjusted to a value of 42 (arbitrary units) and heated to 350°C. The
`pressure of the needle spray gas was 80 psi. The quadrupole was
`heated to 120°C. The mass spectrometer was run in the positive
`mode and the multiplier voltage was set to 1750 V and the X-ray
`voltage to 210 kV. For single ion detection, the mass spectrometer
`was focused on the [M 1 Na]1 of SDZ-RAD [980 atomic mass units
`(amu)], hydroxy SDZ-RAD (996 amu), desmethyl SDZ-RAD (966
`amu), the internal standard 28-,40-diacetyl sirolimus (1020 amu),
`cyclosporine (1224 amu), hydroxy cyclosporine (1240 amu), dihy-
`droxy cyclosporine (1256 amu), desmethyl cyclosporine (1210 amu),
`and the internal standard cyclosporin D (1238 amu). The dwell time
`per ion was 100 ms.
`Assay Validation and Quantification. The assay was validated
`for SDZ-RAD and cyclosporine in blood with the procedures de-
`scribed in detail by Segarra et al. (1998). The HPLC/MS assay had
`the following specifications for SDZ-RAD determined in blood: linear
`range 0.1 to 100 mg/l (y 5 0.96x 1 0.05; r2 5 0.99), mean intra-assay
`variability 6.9% (n 5 10), intraday accuracy 16.8% (n 5 10), inter-
`assay variability 8.0% (n 5 6; 3 days), and mean analytical recovery
`83%. In-process stability, freeze-thaw stability, dilution integrity,
`and partial volume verification were established and have been
`reported by Segarra et al. (1998). The specifications for cyclosporine
`were as follows: linear range 1 to 1000 mg/l (y 5 0.93x 1 15.1; r2 5
`
`SDZ-RAD Tissue Distribution
`
`325
`
`0.98), intra-assay variability 7.7% (n 5 10), intraday precision
`23.7% (n 5 10), interassay variability 9.8% (n 5 5; 3 days), and mean
`analytical recovery 86%. Abbreviated assay validations were carried
`out for each of the tissues, including analytical recovery, lower and
`upper limit of quantitation, linearity, interassay variability, and
`accuracy. Samples from tissues of untreated donor monkeys were
`collected. Tissue samples were homogenized and cyclosporine and
`SDZ-RAD were added and incubated at 37°C for 30 min to allow for
`distribution and protein binding. The following samples were pre-
`pared: blanks (n 5 3/tissue), calibration controls (four concentra-
`tions; n 5 3/concentration), precision controls (three concentrations;
`n 5 3/concentration), samples for determination of the lower (SDZ-
`RAD, 0.5 mg/l and cyclosporine, 10 mg/l; n 5 5) and upper limit of
`quantitation (SDZ-RAD, 100 mg/l and cyclosporine, 1000 mg/l; n 5 5).
`The samples were extracted and analyzed as described above. Re-
`coveries were calculated from the quality control samples (n 5 3 for
`each concentration). The mass spectrometer responses of the ex-
`tracted samples were compared with the response after injection of
`corresponding amounts of internal standard or with standard solu-
`tions of the immunosuppressants (in methanol/0.1% formic acid; 9:1
`v/v) directly on the analytical column. The lowest concentration that
`met the following criteria was accepted as the lower limit of quanti-
`tation: 80% of the samples analyzed had to be within 620% of the
`nominal value, and precision and accuracy variation had to be less
`than 20%. The upper limit of quantitation was determined similarly.
`No interferences with the assay were detected when blank tissues
`were analyzed. Recoveries of SDZ-RAD and cyclosporine from all
`tissues were .60%. Validation results are summarized in Table 1.
`The validation results in all tissues were similar and did not signif-
`icantly differ from those in blood. Therefore, tissue and blood con-
`centrations of the immunosuppressants in the study samples were
`calculated with external calibration curves prepared in blood and
`were corrected with the internal standards. When tissue sample
`concentrations exceeded the upper limit of quantification, samples
`were diluted with acetonitrile/0.1% formic acid (75:25 v/v) and rean-
`alyzed.
`Cyclosporine and SDZ-RAD metabolites were identified by com-
`
`TABLE 1
`Validation of the HPLC/MS assay for cyclosporine and SDZ-RAD in monkey organ tissues
`Tissue samples were collected from untreated monkeys. Tissues were homogenized and cyclosporine or SDZ-RAD was added and incubated at 37°C for 30 min to allow for
`distribution and protein binding. The samples were extracted as described in Experimental Procedures. A concentration was accepted as the lower or upper limit of
`quantitation when four of five samples (80%) were within 620% of the nominal concentration and the coefficient of variation of all samples was #20%. The calibration curve
`consisted of four different concentrations (n 5 3 for each concentration). Interassay coefficient of variation (CV) and accuracy (Acc) were measured at three different
`concentrations (n 5 3 per concentration). Means (n 5 9) are presented.
`
`Tissue
`
`Brainstem
`Cerebellum
`Cerebrum
`Kidney
`Liver
`Stomach
`Colon
`Ileum
`Duodenum
`Jejunum
`Pancreas
`Heart
`Lung (right)
`Lung (left)
`Spleen
`Testes
`Thymus
`Fat
`Lymph node
`Gall bladder
`
`Linear
`Range
`
`mg/l
`50–1000
`50–1000
`50–1000
`50–1000
`50–1000
`50–1000
`50–1000
`50–1000
`50–1000
`50–1000
`50–1000
`50–1000
`50–1000
`50–1000
`50–1000
`50–1000
`50–1000
`50–1000
`50–1000
`50–1000
`
`Cyclosporine
`
`SDZ-RAD
`
`Regression Analysis
`
`CV
`
`Acc
`
`y 5 1.02x 2 7.5, r 5 0.996
`y 5 1.04x 2 9.5, r 5 0.997
`y 5 0.98x 2 4.6, r 5 0.998
`y 5 1.01x 2 4.1, r 5 0.995
`y 5 0.95x 2 4.6, r 5 0.992
`y 5 0.99x 2 1.5, r 5 0.993
`y 5 1.01x 1 3.9, r 5 0.993
`y 5 1.01x 2 5.7, r 5 0.988
`y 5 1.02x 1 0.8, r 5 0.990
`y 5 0.91x 1 0.8, r 5 0.995
`y 5 0.96x 1 5.1, r 5 0.998
`y 5 1.07x 2 5.9, r 5 0.996
`y 5 1.02x 2 2.6, r 5 0.999
`y 5 1.06x 1 2.2, r 5 0.998
`y 5 1.07x 2 2.1, r 5 0.997
`y 5 1.04x 2 2.7, r 5 0.995
`y 5 1.06x 2 5.3, r 5 0.996
`y 5 1.02x 2 1.4, r 5 0.996
`y 5 1.04x 2 1.5, r 5 0.995
`y 5 1.08x 2 7.8, r 5 0.999
`
`3.0
`5.7
`5.9
`9.8
`13.9
`4.1
`6.9
`6.5
`11.8
`6.0
`9.2
`6.8
`7.4
`6.0
`8.0
`4.4
`6.3
`3.6
`5.5
`10.9
`
`%
`
`20.8
`21.3
`23.9
`10.2
`210.3
`11.1
`16.6
`20.9
`110.2
`26.6
`26.6
`15.5
`10.4
`18.6
`17.2
`14.7
`13.5
`13.0
`12.7
`12.1
`
`Linear
`Range
`
`mg/l
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`0.5–100
`
`Regression Analysis
`
`CV
`
`Acc
`
`y 5 0.91x 1 0.22, r 5 0.999
`y 5 0.91x 2 0.31, r 5 0.999
`y 5 1.05x 2 0.25, r 5 0.995
`y 5 1.05x 2 0.03, r 5 0.997
`y 5 1.04x 2 0.37, r 5 0.999
`y 5 1.05x 2 1.38, r 5 0.997
`y 5 0.93x 1 1.02, r 5 0.997
`y 5 0.96x 1 0.28, r 5 0.997
`y 5 1.01x 1 0.42, r 5 0.999
`y 5 0.95x 2 0.91, r 5 0.997
`y 5 1.06x 2 1.67, r 5 0.996
`y 5 1.00x 2 0.69, r 5 0.999
`y 5 1.03x 2 0.02, r 5 0.997
`y 5 0.90x 1 1.75, r 5 0.997
`y 5 1.03x 2 0.46, r 5 0.999
`y 5 0.99x 2 1.25, r 5 0.999
`y 5 0.96x 2 0.5, r 5 0.998
`y 5 0.95x 2 0.43, r 5 0.996
`y 5 0.98x 6
`0, r 5 0.999
`y 5 1.11x 2 0.47, r 5 0.999
`
`8.3
`4.7
`7.1
`12.3
`9.8
`7.9
`11.5
`7.9
`11.5
`7.7
`15.4
`9.0
`7.6
`4.5
`4.3
`3.6
`8.0
`5.1
`3.8
`9.3
`
`%
`
`25.5
`22.4
`11.4
`16.0
`110.8
`21.1
`22.8
`12.8
`22.8
`22.7
`26.1
`25.6
`22.7
`23.7
`20.8
`22.3
`13.0
`23.4
`24.3
`28.1
`
`West-Ward Exhibit 1091
`Serkova 2000
`Page 003
`
`

`

`Vol. 294
`
`Downloaded from
`
`jpet.aspetjournals.org
`
` at ASPET Journals on January 16, 2017
`
`Fig. 1. Distribution of cyclosporine (A), SDZ-RAD (B), and their main
`metabolites in tissues of lung transplant monkeys. Data are presented as
`mean 6 S.E. (n 5 4) and are given in nanograms per gram of wet tissue
`weight except for blood, which is given in micrograms per liter. The
`animals in A received 100 mg/kg/day (150 mg/kg the first week) cyclo-
`sporine (Neoral) and the animals in B received 1.5 mg/kg/day SDZ-RAD
`as a single daily oral dose for 4 weeks. TX, transplant.
`
`the median blood concentration of cyclosporine was 221 mg/l
`(range 131–255 mg/l; n 5 4). Cyclosporine concentrations in
`the native and transplant lung were similar (Fig. 1). The
`cyclosporine metabolites AM1 and AM9 were present in all
`tissues, except AM9 was not detectable in brain tissues.
`Median cyclosporine metabolite concentrations, except in
`gall bladder and cerebrum, were lower than cyclosporine
`concentrations. The concentrations of cyclosporine and its
`metabolites exceeded those in blood in most tissues (Table 2).
`Again, mainly due to the high variability, only cyclosporine
`concentrations in fat tissue, kidney, spleen, lymph nodes, gall
`bladder, and colon were significantly higher than in blood
`(P , .05).
`Effect of Cyclosporine on Tissue Distribution of
`SDZ-RAD (Group 1.5RAD 1 50Cs). Compared with the
`administration of each drug alone, combination of SDZ-RAD
`and cyclosporine affected the tissue concentrations of both
`drugs. As shown in Fig. 1B, SDZ-RAD concentrations had the
`tendency to increase in most tissues when coadministered
`with 50 mg/kg/day cyclosporine (group 1.5RAD 1 50Cs) com-
`pared with tissues from monkeys in the corresponding SDZ-
`RAD monotherapy group (group 1.5RAD). In contrast to most
`other organs, the average SDZ-RAD tissue concentrations in
`
`326
`
`Serkova et al.
`
`parison of the mass spectra and HPLC retention times with those of
`authentic standard material. Cyclosporine metabolites were isolated
`and their structures identified as previously described (Christians et
`al., 1991). SDZ-RAD was incubated with human liver microsomes
`and an NADPH-generating system. The metabolites were isolated by
`HPLC and their structures identified by MS and analysis of the
`fragment pattern after induction of nozzle-skimmer fragmentation
`as described by Vidal et al. (1998). Validation results for the quan-
`tification of metabolites were similar to those of the parent com-
`pounds.
`Clinical Monitoring of Transplant Animals. Laboratory
`screening was performed three times a week and included differen-
`tial blood counts, blood chemistry (serum lipid patterns, serum pro-
`tein, liver and kidney function parameters), and serum electrolytes.
`Lung transplant function and rejection was monitored by 1) chest
`radiographs (two times per week), 2) lung function tests with a
`Bicore CP-100 pulmonary monitor (Bicore Monitoring Systems, Ir-
`vine, CA), 3) bronchoscopy (day 4 and day 14 after surgery), and 4)
`open lung biopsies 2 and 4 weeks after transplantation. The histo-
`logical grade of rejection was classified according to the International
`Society for Heart and Lung Transplantation (A0, no rejection; A1,
`minimal rejection; A2, mild rejection; A3, moderate rejection; and
`A4, severe rejection; Yousem et al., 1996).
`Data Analysis. Data were processed with ChemStation software
`revision A04.02 for the HPLC system and C.03.00 for the electros-
`pray interface and MS (all from Hewlett-Packard). Concentrations of
`the immunosuppressants were calculated from an external standard
`curve and corrected on the basis of the internal standards. SPSS
`software, version 9.0, was used for statistical analysis (SPSS Inc.,
`Chicago, IL). Because data were not normally distributed, tissue
`concentrations in the combination therapy and respective control
`groups were compared with the nonparametric Mann-Whitney U
`test and results of distribution statistics are reported as median and
`range (minimum–maximum). Clinical chemical and biochemical
`data among groups were compared by multivariate ANOVA. Corre-
`lation (Pearson correlation coefficients, two-tailed test of signifi-
`cance) and stepwise regression analysis (probability-of-F-to-enter,
`.05) were based on the data of all study groups (n 5 25).
`
`Results
`Tissue Distribution of SDZ-RAD, Cyclosporine, and Their
`Metabolites
`Tissue Distribution of SDZ-RAD (Group 1.5RAD). Af-
`ter single daily oral SDZ-RAD doses (1.5 mg/kg) for 4 weeks
`and 24 h after the last dose, the blood concentrations of
`SDZ-RAD at the time of sacrifice ranged from 3.7 to 44 mg/l
`(median 5 9.2 mg/l) and those of its metabolites from 0 to 7.1
`mg/l (median 5 2.3 mg/l; n 5 4; Fig. 1). SDZ-RAD and its
`metabolites extensively distributed into tissues (Fig. 1; Table
`2). Median concentrations of SDZ-RAD and median total
`concentrations of its metabolites in most tissues exceeded
`those in blood. However, concentrations varied widely and,
`thus, the only organs that reached significantly higher tissue
`concentrations than in blood were pancreas and gallbladder
`(P , .05). Although there was a tendency to a higher median
`SDZ-RAD concentration in the transplant versus the native
`lung (Fig. 1), this difference was not statistically significant.
`It must be taken into account that due to the relatively small
`number of animals in the study groups, the statistical power
`for this analysis as well as all comparisons of the tissue
`concentrations reported below was less than 35%.
`Tissue Distribution of Cyclosporine (Group 100Cs).
`Cyclosporine was detected in all organ tissues examined in
`this study (Fig. 1). Twenty-four hours after the last oral dose,
`
`West-Ward Exhibit 1091
`Serkova 2000
`Page 004
`
`

`

`Downloaded from
`
`jpet.aspetjournals.org
`
` at ASPET Journals on January 16, 2017
`
`2000
`
`SDZ-RAD Tissue Distribution
`
`327
`
`TABLE 2
`Comparison of tissue-to-blood distribution coefficients of SDZ-RAD and its metabolites with (50 mg/kg, group 1.5RAD 1 50Cs) and without
`cyclosporine (group 1.5RAD) and of cyclosporine and its metabolites with (0.3 mg/kg/day, group 0.3RAD 1 l00Cs) and without SDZ-RAD (group
`100Cs)
`The tissue-to-blood distribution coefficients were calculated by dividing the median concentration in tissue (nanograms per gram) by the median concentration in blood
`micrograms per liter.
`
`SDZ-RAD
`
`Metabolites
`
`Cyclosporine
`
`Metabolites
`
`Brainstem
`Cerebellum
`Cerebrum
`Colon
`Duodenum
`Fat
`Gall bladder
`Heart
`Ileum
`Jejunum
`Kidney
`Liver
`Lung (native)
`Lung (tx)
`Lymph node
`Pancreas
`Spleen
`Stomach
`Testes
`Thymus
`
`2Cs
`
`0.1
`4.3
`0.4
`1.7
`1.4
`0.2
`19.4
`0.8
`1.5
`1.2
`3.2
`0.6
`1.2
`12.6
`2.2
`82.6
`7.7
`0.7
`4.0
`2.3
`
`1Cs
`
`0.5
`1.1
`1.1
`5.8
`1.4
`1.1
`27.8
`1.1
`6.5
`3.2
`9.7
`2.2
`2.1
`1.7
`2.4
`8.8
`10.6
`1.8
`1.7
`1.7
`
`2Cs
`
`0.0
`3.3
`0.0
`5.1
`0.1
`1.7
`140.6
`2.1
`1.8
`1.8
`36.6
`8.6
`1.9
`21.4
`3.2
`551.4
`12.5
`1.3
`10.0
`30.9
`
`1Cs
`
`0.2
`0.2
`0.6
`2.9
`1.4
`0.5
`23.3
`1.0
`4.9
`3.8
`12.2
`2.8
`1.6
`3.5
`0.6
`3.7
`9.1
`0.7
`0.5
`1.2
`
`2RAD
`
`1RAD
`
`2RAD
`
`1RAD
`
`0.3
`0.2
`0.3
`13.5
`0.9
`4.1
`8.6
`1.6
`1.1
`1.6
`4.3
`2.9
`1.4
`2.1
`7.9
`2.8
`4.9
`1.3
`0.5
`2.9
`
`0.5
`0.4
`0.8
`3.5
`2.2
`1.5
`9.9
`1.4
`3.0
`3.5
`3.9
`2.1
`1.0
`2.0
`15.6
`1.9
`2.6
`1.4
`0.4
`2.3
`
`0.3
`0.3
`0.3
`6.1
`2.5
`1.0
`67.6
`3.9
`2.2
`2.1
`2.7
`3.1
`2.1
`3.3
`2.0
`5.5
`6.8
`2.1
`0.8
`2.4
`
`0.2
`0.3
`0.1
`0.8
`0.8
`0.3
`35.1
`0.8
`1.3
`2.1
`3.0
`3.4
`1.0
`2.2
`0.8
`1.5
`4.2
`0.6
`0.4
`0.7
`
`Cs; cyclosporine, RAD; SDZ RAD, tx, transplant.
`
`pancreas and cerebellum were lower in the presence of cyclo-
`sporine than in the SDZ-RAD monotherapy group (Fig. 2).
`This was because, compared with the other animals in this
`group, one monkey in the SDZ-RAD monotherapy group
`(1.5RAD) had extremely high concentrations in these organs
`(Fig. 1). Compared with the monotherapy group, with con-
`comitant cyclosporine the changes of SDZ-RAD concentra-
`tions in blood, ileum, duodenum, native lung, kidney, and
`brainstem (Fig. 1B) reached statistical significance (all P ,
`.04). As indicated by the tissue-to-blood partition coefficients
`[Ctissue (micrograms per gram)/Cblood (micrograms per liter)],
`the interaction with cyclosporine led to an increase of SDZ-
`RAD concentrations in most tissues, the extent of which was
`not predicted by blood concentrations (Table 2). In most tis-
`sues (Table 2; Fig. 2), cyclosporine increased the SDZ-RAD/
`metabolite ratio. Statistically significant correlations of SDZ-
`RAD trough blood concentrations with tissue concentrations
`were only observed in 10 of 20 tissues examined, and significant
`correlations for blood and tissue concentrations of SDZ-RAD
`metabolites were only observed in 2 of 20 tissues (Table 3).
`Effect of SDZ-RAD on Tissue Distribution of Cyclo-
`sporine (Group 0.3RAD 1 100Cs). SDZ-RAD had a
`smaller impact on cyclosporine tissue distribution than the
`inverse (Fig. 1, A and B, respectively). The Mann-Whitney U
`test did not show any statistically significant changes, in-
`cluding concentrations in blood and the transplant lung.
`Comparing the changes in all tissues, the effect of SDZ-RAD
`on median cyclosporine tissue/blood ratios lacked a consis-
`tent tendency (Table 2). In general, trough blood concentra-
`tions of cyclosporine and its metabolites showed a much
`better correlation with their tissue concentrations than SDZ-
`RAD and its metabolites (Table 3).
`
`Lung Transplant Rejection, Toxicity, and Clinical
`Monitoring (Table 4)
`Group 1.5RAD (1.5 mg/kg/day SDZ-RAD). At the end of
`the observation period, three of four animals exhibited severe
`
`rejection (A4) of the transplant lung. The median weight loss
`at the end of the study was 22% (Table 4). There were no
`signs of anemia or thrombocytopenia. Serum cholesterol con-
`centrations increased during the observation period (Table 4)
`but were not significantly different from the cyclosporine
`control group (group 100Cs).
`Group 100Cs (100 mg/kg/day Cyclosporine). At the
`end of the study, the transplant lungs in two animals showed
`moderate (A3) and in the other two animals severe rejection
`(A4). The median weight loss during the study was 15%.
`Serum cholesterol concentrations increased during the obser-
`vation period but the change was not statistically significant.
`No other changes in clinical chemical and biochemical pa-
`rameters were detected.
`Group 0.3RAD 1 100Cs (0.3 mg/kg/day SDZ-RAD 1
`100 mg/kg/day Cyclosporine). The combination of SDZ-
`RAD and cyclosporine was more effective in preventing re-
`jection of the lung allografts than monotherapy of either
`drug. At the end of the study, the biopsies of five animals
`showed a rejection score of A2 (mild acute rejection) and the
`biopsy of one animal had a rejection score of A3 (moderate
`acute rejection). However, in contrast to the monotherapy
`groups 1.5RAD and 100Cs, the animals developed significant
`anemia. Three animals required erythropoetin treatment
`and two animals required blood transfusions. Platelet counts
`were as low as 37,000/ml. These changes are not reflected in
`Table 4 because the table only includes the last values before
`sacrifice. From postoperative day 15 onward, serum choles-
`terol concentrations were higher in this group than in the
`control groups. However, the differences did not reach sta-
`tistical significance. The median weight loss in this group
`during the observation period was 22% (Table 4). Three of six
`animals had to be sacrificed early due to anemia and renal
`failure (n 5 2; days 20 and 23) or seizures (n 5 1; day 22).
`Group 1.5RAD 1 50Cs (1.5 mg/kg/day SDZ-RAD 1 50
`mg/kg/day Cyclosporine). At the end of the study, the lung
`
`West-Ward Exhibit 1091
`Serkova 2000
`Page 005
`
`

`

`Downloaded from
`
`jpet.aspetjournals.org
`
` at ASPET Journals on January 16, 2017
`
`Vol. 294
`
`end of the observ

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