throbber
CENTER FOR DRUG EVALUATION AND
`
`RESEARCH
`
`I
`
`APPLICA TION NUMBER:
`
`21-880
`
`PHARMACOLOGY REVIEW(S)
`
`

`

`
`
`DEPARTMENT OF HEALTH AND HUMAN SERVICES
`PUBLIC HEALTH SERVICE
`FOOD AND DRUG ADMINISTRATION
`CENTER FOR DRUG EVALUATION AND RESEARCH
`
`PHARMACOLOGY/TOXICOLOGY REVIEW AND EVALUATION
`
`NDA NUMBER:
`
`SERIAL NUMBER:
`
`21—880
`
`001
`
`DATE RECEIVED BY CENTER:
`
`04/07/2005
`
`DRUG NAME:
`
`INDICATION:
`
`SPONSOR:
`
`Revlimid®
`
`Myelodysplastic syndrome (MDS)
`
`Celgene Corporation
`
`86 Morris Avenue, Summit, NJ 07901
`
`DOCUMENTS REVIEWED:
`
`Electronic submission
`
`REVIEW DIVISION:
`
`I
`
`Division of Drug Oncology Products
`
`PHARM/TOX REVIEWER:
`
`M. Anwar Goheer, PhD.
`
`(HFD-150)
`
`Kimberly Benson, Ph.D.
`
`PHARM/TOX SUPERVISOR:
`
`John K. Leighton, Ph.D., D.A.B.T.
`
`ACTING DIVISION DIRECTOR:
`
`Robert Justice, MD, MS.
`
`PROJECT MANAGER:
`
`Carl Huntley, R. Ph., MBA.
`
`Date of review submission to Division File System (DFS):
`
`5 October 2005
`
`

`

`TABLE OF CONTENTS
`
`EXECUTIVE SUMMARY ...................................................................................... 4
`
`2.6
`
`PHARMACOLOGY/TOXICOLOGY REVIEW ............................................ 7
`
`2.6.1
`
`INTRODUCTION AND DRUG HISTORY ............................................................. 7
`
`2.6.2
`
`PHARMACOLOGY ............................................................................................... 9
`
`2.6.2.2 Primary pharmacodynamics ...................................................................................... 9
`2.6.2.3 Secondary pharmacodynamics ............................................................................... 10
`
`2.6.4 PHARMACOKINETICSITOXICOKINETICS ....................................................... 10
`
`2.6.4.2 Methods of Analysis ................................................................................................... 10
`2.6.4.3 Absorption ..................................................................................................................... 1]
`2.6.4.7 Pharmacokinetic drug interactions 11
`
`2.6.6 TOXICOLOGY ......................................................................'.............................. 1 1
`
`2.6.6.3 Repeat-dose toxicity ................................................................................................... 11
`2.6.6.6 Reproductive and developmental toxicology ............................................................... 12
`
`2.6.3 PHARMACOLOGY ............................................................................................. 13
`
`2.6.2.2 Primary pharmacodynamics .................................................................................... 13
`2.6.2.4 Safety pharmacology .................................................................................................. 13
`
`2.6.4 PHARMACOKINETICS/TOXICOKINETlCS ....................................................... 13
`
`2.6.4.3 Absorption ..................................................................................................................... 13
`2.6.4.5 Metabolism .................................................................................................................... 13
`
`2.6.67 TOXICOLOGY .................................................................................................... 14
`
`2.6.6.2 Single-dose toxicity .................................................................................................... 14
`2.6.6.3 Repeat-dose toxicity ................................................................................................... 14
`6.6.6.4 Genetic toxicology ...................................................................................................... 14
`
`2.6.2
`
`PHARMACOLOGY ............................................................................................. 15
`
`2.6.2.1 Brief summary........................................ 15
`2.6.2.2 Primary pharmacodynamics .................................................................................... 16
`2.6.2.3 Secondary pharmacodynamics ............................................................................... 39
`2.6.2.4 Safety pharmacology.................................................................................................. 41
`
`2.6.3
`
`PHARMACOLOGY TABULATED SUMMARY .................................................. 42
`
`2.6.4
`
`PHARMACOKINETICSITOXICOKINETICS ....................................................... 43
`
`2.6.4.1 Brief summary ............................................................................................................. 43
`2.6.4.2 Methods of Analysis
`............................................................................................... 43
`2.6.4.3 Absorption ..................................................................................................................... 44
`2.6.4.5 Metabolism .................................................................................................................... 53
`2.6.4.6 Excretion....................................................................................................................... 54 .
`
`2.6.4.7 Pharmacokinetic drug interactions............................................................................... 54
`2.6.4.8 Other Pharmacokinetic Studies .................................................................................... 62
`
`

`

`2.6.4.9 Discussion and Conclusions ................................................................................... 62
`2.6.4.10Tables and figures to include comparative TK summary: ......................................... 63
`
`2.6.5
`
`PHARMACOKINETICS TABULATED SUMMARY ...........................'................. 63
`
`2.6.6 TOXICOLOGY .................................................................................................... 64
`2.6.6.1 Overall toxicology summary..................................................................................... 64
`2.6.6.2 Single-dose toxicity .................................................................................................... 67
`2.6.6.3 Repeat-dose toxicity ................................................................................................... 67
`6.6.6.4 Genetic toxicology ...................................................................................................... 75
`2.6.6.5 Carcinogenicity ............................................................................................................. 75
`2.6.6.6 Reproductive and developmental toxicology ...................................................... 76
`2.6.6.7 Local tolerance........................................................................................................... 127
`2.6.6.8 Special toxicology studies ...................................................................................... 127
`2.6.6.9 Discussion and Conclusions
`................................... 127
`
`2.6.7 TOXICOLOGY TABULATED SUMMARY ........................................................ 128
`
`OVERALL CONCLUSIONS AND RECOMMENDATIONS .......................................... 131
`
`APPENDIX A ................................................................................................................ 132
`
`APPENDIX B .............................................................................................................. 158
`
`ADpears ThIS W0
`
`

`

`Reviewer:
`
`Anwar Goheer, Ph.D.
`
`NDA No.
`
`21—880
`
`|.
`
`Recommendations
`
`EXECUTIVE SUMMARY
`
`A. Recommendation on approvability: The non-clinical studies submitted to
`this NDA provide sufficient information to support the use of lenalidomide
`(Revlimid®) in patients with transfusion-dependent anemia due to low- or
`intermediate—1—risk myelodysplastic syndromes (MDS) associated with a
`deletion 5q cytogenetic abnormality with or without additional cytogenetic
`abnormalities.
`
`B. Recommendation for nonclinical studies: Adequate reproductive toxicity
`assessment, specifically embryo—fetal developmental toxicity in two species,
`needs to be conducted.
`
`C. Recommendations on labeling:
`
`A separate review will be conducted.
`
`II.
`
`Summary of nonclinical findings
`
`Lenalidomide (3-(4’aminoisoindoline-1—
`Brief overview of nonclinical findings:
`A.
`one)-1—piperidine—2, 6-dione; CC-5013; lMiD—3 and Revlimid®) is a thalidomide
`analogue.
`It is a racemic mixture of S (-) and R (+) forms. The in vitro and in vivo
`characterization of pharmacological properties of lenalidomide had demonstrated that
`the drug inhibits the secretion of pro-inflammatory cytokines (TNF—a,
`lL—1B, lL-6 and IL-
`12) and increases the secretion of anti—inflammatory cytokine (IL-10) from peripheral
`blood mononuclear cells (PBMC), induces T—cell proliferation (IL-2, lFN—y), inhibits cell
`proliferation (MM, Burkitt’s lymphoma) and inhibits angiogenesis (Knight—R, Semin
`Oncol 2005; 32:24-30 & Dredge et al., Microvasc Res. 2005; 69:56—63). Lenalidomide
`inhibits the expression of cyclooxygenase-2 (COX-2) but did not affect COX—1 in vitro.
`in
`This may translate into adverse effects that need to be fully explored in clinical trials.
`addition to these immune effects, there is evidence that thalidomide and its analogues
`' may act directly on tumor cells, via inducing apoptosis or G1 growth arrest.
`
`The oral administration of lenalidomide at dose levels of 3, 6 and 12 g/m2 produced no
`effects on behavior or general activity in male rats.
`Intravenous administration of the
`drug at doses up to 400 mg/m2 did not produce any significant effect on cardiovascular
`and respiratory systems of the anesthetized dog.
`In vitro, lenalidomide inhibited the
`cloned human potassium channel (hERG) current by 8% only at the highest
`concentration tested (787 pM).
`
`Lenalidomide did not inhibit or induce any of the major cytochrome P450 isozymes in
`vitro and in vivo. indicating limited potential for P450-related drug-drug interactions.
`Distribution of radioactivity in the fetal tissues of pregnant rat was low after oral
`administration but fetal brain showed more activity than maternal brain. The highest
`
`

`

`Reviewer:
`
`Anwar Goheer, Ph.D.
`
`NDA No.
`
`21-880
`
`concentrations were found in the kidney (cortex and medulla), liver, spleen and the
`mucosa of the Gl tract of rats.
`
`During traditional toxicity assessment, lenalidomide was administered to rodents (mice,
`rats) and non rodents (monkeys) for 1, 7, and 28 days and 13, 26, and 52 weeks.
`Single dose administration of lenalidomide up to 6 g/m2 in mice and 12 g/m2 in rats did
`not cause any adverse effects. Daily oral administration of lenalidomide at 6 g/m2 to
`rats for 28 days was associated with moderate to severe tubular nephropathy/nephritis,
`which was attributed to precipitation of the lenalidomide in the kidney. Once daily oral
`administration of lenalidomide to rats at doses of 450, 900 or 1800 mg/mzlday for 26
`weeks was mainly associated with reduced body weight gain (12% ~L) for high dose
`males and reversible pelvic mineralization in the kidney of all treated animals.
`
`Oral administration of lenalidomide to cynomolgus monkeys at dose levels of 12, 24, 48,
`or 72 mg/mzlday for 52 weeks was associated with hemorrhage in multiple organs,
`gastrointestinal tract inflammation and lymphoid and bone marrow atrophy. Dosing at
`48 and 72 mg/m2/day was discontinued after 20 weeks of treatment due to toxicity and
`mortalities. A reversal of the macroscopic and microscopic findings seen in decedent
`and the terminal sacrifice was noted in 7 week treatment-free recovery animals.
`It is
`clear that this species is much more sensitive to lenalidomide than rodents.
`
`Lenalidomide did not induce mutation in the Ames test, chromosome aberrations in
`cultured human peripheral blood lymphocytes, or mutation at the thymidine kinase (tk)
`locus of mouse lymphoma L5178Y cells. Lenalidomide did induce micronuclei in the
`polychromatic erythrocytes of the bone marrow of male rats.
`
`Reproductive studies were conducted
`Reproductive and developmental toxicity:
`with lenalidomide, examining the effects on fertility and early embryo development,
`embryo-fetal development, and pre-and post—natal development. Only the embryo-fetal
`development studies are required for drugs with oncologic indications. These studies
`have not been adequately conducted at this time. The first study, conducted in a rat,
`showed very slight maternal toxicity and no fetal malformations. The rat, however, is
`not an adequate species for the full assessment of lenalidomide's developmental
`effects, given the structural similarity to thalidomide. Historical data indicates that the
`rat is not sensitive to the full range of thalidomide's teratogenic effects.
`'
`
`An additional developmental study was conducted in the rabbit, with a concurrent
`thalidomide dose group. This study had a confounding variable with some rabbits not
`eating prior to the study and all these rabbits had a negative outcome in the study.
`Additionally, the highest dose tested did not meet the standard criteria for sufficient drug
`exposure.
`
`

`

`Reviewer:
`
`Anwar Goheer, Ph.D.
`
`NDA No.
`
`21—880
`
`Both lenalidomide and thalidomide have been
`Pharmacologic activity:
`B.
`shown to increase the secretion of anti-inflammatory cytokine lL—1O from LPS-stimulated
`PBMC, stimulates T—cells proliferation and production of lL—2 and IFN—y. Both inhibit the
`secretion of pro-inflammatory cytokines TNF—d,
`lL—1B, and lL-6.
`In addition to these
`immune effects, there is evidence that-thalidomide and its analogues may act directly on
`tumor cells, via inducing apoptosis or 61 growth arrest. Exact mechanisms of action
`however remain unknown.
`
`Inflammation of the
`Nonclinical safety issues relevant to clinical use:
`C.
`gastrointestinal tract and atrophy of the bone marrow, thymus, and lymphoid tissues
`were observed during repeat dose toxicity studies (up to 12 months) in cynomolgus
`monkeys. Embryo-fetal developmental toxicity has not been adequately addressed.
`The structural similarity of lenalidomide to thalidomide, a known human teratogen,
`suggests developmental risk. Lenalidomide also inhibits expression of COX-2 in vitro
`but not COX-1. This finding should be fully explored in clinical trials.
`
`APPEARS TillS WAY
`0N ORlGlNAL
`
`

`

`Reviewer:
`
`Anwar Goheer, PhD.
`
`NDA No.
`
`21—880
`
`2.6
`
`PHARMACOLOGY/TOXICOLOGY REVIEW
`
`2.6.1 INTRODUCTION AND DRUG HISTORY
`
`NDA number:
`Review number:
`
`21-880
`1
`
`Sequence number/date/type of submission: 001 / 12—22-2004 / NDA
`Information to sponsor:
`Yes (X) No ( )
`Sponsor and/or agent:
`Celgene Corporation
`86 Morris Avenue, Summit
`New Jersey 07901.
`
`Manufacturer for drug substance:
`
`/
`
`i
`
`_
`
`Reviewer name:
`
`Division name:
`
`HFD #:
`
`M. Anwar Goheer, Ph.D.
`
`Division of Oncology Drug Products
`
`HFD—150
`
`Review completion date:
`
`September 27, 2005
`
`Drug:
`
`Trade name:
`Generic name:
`Code name:
`Chemical name:
`
`REVLIMID®
`Lenalidomide
`CC-5013, CDC—501, lMiD 3
`3—(4’-amino—1,3-dihydro-1—oxo-2H—isoindol—2-
`yl)-2,6-piperidinedione,
`3-(4’—amino-1—oxo—1 ,3-dihydro—2H—isoindol—2-
`yl) piperidine-2,6—dione.
`191732—72-6
`C13H13N303/ 259.25
`-
`
`CAS registry number:
`Molecular formula/molecular weight:
`Structure:
`
`0 0
`
`HN
`
`Niko
`
`NH2.
`
`..
`DMF Numbers: '
`Relevant _lNDs/NDAs/DMFs:
`“a
`lND numbers: 60,100,
`Immunomodulator/ anti—angiogenesis
`
`Drug class:
`
`

`

`Reviewer:
`
`Anwar Goheer, PhD.
`
`NDA No.
`
`21-880
`
`Indication:
`
`Transfusion-dependent anemia due to low— or intermediate-1-risk
`myelodysplastic syndromes (MDS) associated with a deletion 5q
`cytogenetic abnormality with or without additional cytogenetic
`abnormalities.
`Clinical formulation:
`Composition of lenalidomide capsules
`
`
`
`10 mg
`5 mg
`
`Capsule
`Capsule
`
` Ingredient
`Quality
`
`Theoretical Weight per
`
`Standard
`Function
`
`
`Capsule (mg)
`Lcnalidomidcfl
`
`
`
`
`
`Lactose Anhydrousb
`
`Microcn-‘smlline Cellulose
`
`._'—
`
`C‘rosczu‘mcllose Sodium
`
`
`Magnesium Stearate
`.=
`
` _—
`400.0
`Total Fill Weight
`
`
`
`
`White Capsule Shells (Size 2)
`
`Imprinted with Black Ink°
`
`
`
`
`
`
`
`
`In-housc
`--
`
`Pale Yellow Body'Bluc Green
`
`I (‘upsulc
`Cap Capsule Shells (Size 0)
`
`
`Imprinted with Black Ink‘
`
`:
`
`"/
`The capsule shells are supplied by
`gelatin in the capsule shells isprovided in PA.
`
`lnfommtion pertaining to the components and source of
`
`(Excerpted from the sponsor’s submission)
`
`Route of administration:
`
`Oral
`
`“Lenalidomide (10 mg daily) is. indicated for the treatment of
`Proposed use:
`patients with transfusion-dependent anemia due to low- or intermediate-1-risk
`myelodysplastic syndromes associated with a deletion 5q cytogenetic abnormality
`with or without additional cytogenetic abnormalities.”
`
`Disclaimer: Tabular and graphical information are constructed by the reviewer unless
`cited otherwise.
`'
`
`Data reliance : Except as specifically identified below, all data and information
`discussed below and necessary for approval of NDA 21—880 are owned by Celgene
`Corporation or are data for which Celgene has obtained a written right of reference.
`Any information or data necessary for approval of NDA 21-880 that Celgene does not
`own or have a written right to reference constitutes one of the following: (1) published
`literature, or (2) a prior FDA finding of safety or effectiveness for a listed drug, as
`
`

`

`Reviewer:
`
`Anwar Goheer, PhD.
`
`NDA No-
`
`21—880
`
`described in the drug’s approved labeling. Any data or information described or
`referenced below from a previously approved application that Celgene does not own (or
`from FDA reviews or summaries of a previously approved application) is for descriptive
`purposes only and is not relied upon for approval of NDA 21-880.
`
`APPEARS T’rllS WAY
`0N ORlGlNAL
`
`Studies reviewed within this submission:
`
`2.6.2 PHARMACOLOGY
`
`2.6.2.2
`
`Primary pharmacodynamics
`
`Mechanism of action:
`
`1.
`2.
`
`3.
`
`4.
`
`5.
`
`6.
`
`9°.“
`
`9.
`
`10.
`
`11.
`
`12.
`
`Amino- Substituted thalidomide analogs: Potent inhibitors of TNF—a production.
`Inhibition of TNF-a production by PBMC and elevation of lL—2 and 'MlP-3a
`production by T cells by CC—4047, CC—5013, and C-11006 in vitro.
`Inhibition of tumor necrosis factor alpha (TNF-a) production by 004047, CC-
`5013, and CC—11006 from human and rat whole blood stimulated with
`Iipopolysaccharide.
`Elevation of lL—2 production by CC-4047, CC—5013, and CC-11006 from human
`and rat whole blood stimulated with Concanavalin A.
`Cytokine profiling for five classes of lMiDs in primary human PBMCs and CD4+ T
`lymphocytes.
`Anti- Inflammatory effects of CC~4047, CC-5013 and CC-11006 on G-CSF, lL-10,
`and COX-2 Expression by LPS—stimulated PBMC.
`Effect of the lMiD CC-5013 on Akt phosphorylation in the *Jurkat T cell line.
`Effect of the PDE4 Inhibitors CC—10004, CC—10082 (cilomilast), CC—11050 and
`CC—14064 (roflumilast), and the lMlD CC-5013 on lL-6 production by human, rat,
`mouse, and monkey whole blood stimulated with LPS in vitro.
`lmmunomodulatory drugs (lMiDsTM) inhibit expresSion of cyclooxygenase—2 from
`TNF—a, lL-1l5 and LPS stimulated human PBMC in a partially lL-10- dependent
`manner.
`
`lmmunomodulatory analogs of thalidomide inhibit growth of Hs Sultan cells and
`angiogenesis in vivo.
`Thalidomide and its analogues inhibit lipopolysaccharide-mediated induction of
`cyclooxygenase—Z.
`Anti-proliferative activity and mechanism of action of thalidomide, CC-4047, CC—
`5013 and CC-11006 in chromosome 5 deleted cells N-amalwa and KG-1 and
`control cell lines MUTZ—5 and UT—7 in vitro.
`
`

`

`Reviewer:
`
`Anwar Goheer, Ph.D.
`
`NDA No.
`
`21-880
`
`13.
`
`14.
`
`15.
`
`16.
`17.
`
`18.
`
`19.
`
`Effects of lMiDs on proliferation of breast cancer, NSCLC, CML and NHL cell
`lines in vitro.
`
`Anti- proliferative activity of CC-4047, CC—5013, CC—5079, and CC-10004 against
`the non-Hodgkin’s B lymphoma cell line Farage in vitro.
`Effect of CC—10004 and CC-5013 on proliferation of the mouse CLL line LNC,
`alone and in combination with vincristine.
`'
`Inhibition of endothelial cell migration by thalidomide, 00-4047, and CC-5013.
`Effect of CC-5013 on HlF—1 alpha expression and VEGF production in PC-3
`cells.
`
`Lenalidomide inhibits angiogenesis in vitro and reduces lung metastasis of
`mouse melanoma cells in an animal model.
`Novel thalidomide analogues display anti—angiogenic activity independently of
`immunomodulatory effects.
`
`2.6.2.3
`
`Secondary pharmacodynamics
`
`1.
`
`2.
`
`3.
`
`4.
`
`5.
`
`Addition of immunomodulatory drugs CC-5013 or CC-4047 to Rituximab
`enhances anti-tumor activity in a severe combined immunodeficiency (SCID)
`mouse lymphoma model.
`Use of lmiD3, a thalidomide analog, as an adjunct to therapy for experimental
`tuberculous meningitis.
`Thalidomide and its analogs overcome drug resistance of human multiple
`myeloma cells to conventional therapy.
`lMiDs augment fetal hemoglobin synthesis and can be used for the treatment of
`hemoglobin disorders like sickle cellanemia and B-thalassemia.
`Thalidomide and thalidomide analogue drug costimulate virus- specific CD8+ T
`cells in vitro.
`
`See previous reviews for details; a brief
`2.6.2.4 Safety Pharmacology
`summary of these studies is presented in this section.
`
`2.6.4 PHARMACOKINETICSITOXICOKINETICS,
`
`2.6.4.2
`
`Methods of Analysis
`
`1.
`
`2.
`
`Validation for the determination of CC-5013 in rat plasma (Heparin anticoagulant)
`using
`_
`“J
`for sample preparation and liquid chromatography
`with mass spectrometric detection.
`Validation of an analytical procedure for the determination of the enantiomers of
`CC-5013 in rat plasma (Heparin) using solid phase extraction and liquid
`chromatography with 5-—
`. mass spectrometric detection.
`
`lO
`
`

`

`Reviewer:
`
`Anwar Goheer, Ph.D.
`
`NDA No.
`
`21-880
`
`3.
`
`4.
`
`5.
`
`Validation of an analytical procedure for the determination of CC—5013 in rabbit
`plasma (Heparin) using
`_ I
`i and liquid chromatography with
`—— mass spectrometric detection.
`Validation for the determination of CC-5013 in dog plasma (Heparin
`anticoagulant) using
`/ or sample preparation and liquid
`chromatography with mass spectrometric detection.
`.
`Validation for the determination of CC—5013 in primate plasma (Heparin
`anticoagulant) using
`_ V
`"for sample preparation and liquid
`chromatography with mass spectrometric detection.
`
`2.6.4.3
`
`Absorption
`
`1.
`
`2.
`
`[14C] CC-5013: A study of absorption and excretion following oral and
`intravenous administration to the rat.
`[14C] CC—5013: A study of absorption, excretion and metabolism following oral
`and intravenous administration to the cynomolgus monkey.
`
`2.6.4.4
`
`Distribution
`
`1.
`
`[14C] CC-5013: Quantitative whole-body autoradiography following a single oral
`administration (150 mg/kg) to the rat.
`
`2.6.4.7
`
`Pharmacokinetic drug interactions
`
`1.
`
`2.
`
`3.
`
`4.
`
`5.
`
`N93
`
`CC- 5013: Effect on cytochrome P450 and related parameters in the male and
`female Sprague Dawley rat following oral (gavage) administration at 0, 75, 150
`and 300 mg/ kg/ day for 26 weeks.
`'
`CC— 5013: Effect on cytochrome P450 and related parameters in the male and
`female cynomolgus monkey following oral (gavage) administration at 0, 1 and 2
`mg/ kg/ day for 52 weeks.
`identification of the cytochrome P450 enzymes responsible for the in-vitro
`metabolism of (14C)—CC-5013 in human liver microsomes.
`Identification of human P450 isozymes involved in the metabolism of CC-1088
`and CC-5013.
`
`Effects of CC—1088 and CC-5013 on selected cytochrome P450 activities in
`human liver microsomes:_Prediction of drug interactions.
`Metabolism of (14C)—CC—5013 in isolated human hepatocytes.
`Comparison of chemical degradation pathways of Ienalidomide and thalidomide.
`
`2.6.6 TOXICOLOGY
`
`2.6.6.3
`
`Repeat-dose toxicity
`
`1-.
`2.
`
`CC— 5013: 7- day oral (gavage) administration range—finding study in the mouse.
`CC- 5013: 7 day oral (gavage) range-finding toxicity study in the rat.
`
`ll
`
`

`

`Reviewer:
`
`Anwar Goheer, Ph.D.
`
`NDA No.
`
`21-880
`
`3.
`4.
`5.
`
`CC— 5013: 28 day oral (gavage administration) toxicity study in the rat.
`CC— 5013: 13 week oral (gavage administration) toxicity study in the rat.
`CC— 5013: 26 week oral (gavage) administration toxicity study in the rat with a 4
`week treatment— free period.
`
`2.6.6.6
`
`Reproductive and developmental toxicology
`
`Fertility and early embryonic development
`
`1.
`
`CC- 5013: Oral (gavage) study of fertility and early embryonic development in the
`rat (Segment 1).
`
`Embryofetal development
`
`1.
`
`.wzv
`
`4.
`
`5.
`
`CC- 5013: Oral (gavage) range-finding study of embryo-foetal development in the
`rat.
`'
`
`CC- 5013: Oral (gavage) study of embryo—foetal development in the rat.
`Preliminary study of CC- 5013 embryo-foetal development (Segment ll) in the
`non—pregnant New Zealand white rabbit.
`CC— 5013: Oral (gavage) range—finding study of embryo-foetal development in the
`rabbit.
`_
`CC— 5013: Oral (gavage) study of embryo-foetal development in the rabbit.
`
`Prenatal and postnatal development
`
`1.
`
`CC— 5013: Oral (gavage) study of pre—and postnatal development in the rat.
`
`MREEXRS nits writ
`Etta deleted.
`
`12
`
`

`

`Reviewer:
`
`Anwar Goheer, Ph.D.
`
`NDA No.
`
`21-880
`
`See Appendix A for studies reviewed by Dr.
`Studies Previously Reviewed:
`Anwar Goheer on 4/27/2000, 6/2/2000, & 2/14/2001 and Appendix B for
`studies reviewed by Dr. Anthony Proakis on 6/06/01, 10/29/02, 12/23/02,
`& 3/31/05,
`.
`
`2.6.3 PHARMACOLOGY
`
`2.6.2.2
`
`Primary pharmacodynamics
`
`Mechanism of action:
`
`1.
`2.
`
`3.
`
`Amino-substituted thalidomide analogs: Potent inhibitors of TNF-a production.
`Differential cytokine modulation and T cell activation by two distinct classes of
`thalidomide analogues that are potent inhibitors of TNF-a.
`Thalidomide and its analogs overcome drug resistance of human multiple
`myeloma cells to conventional therapy.
`
`2.6.2.4
`
`Safety pharmacology
`
`Neurological effects:
`
`1.
`
`CC-5013: Effects on general activity and behaviour in the rat following oral
`administration.
`
`Cardiovascular effects:
`
`1.
`2.
`
`Effects of CC—5013 on cloned hERG channels expressed in mammalian cells.
`CC— 5013: Cardiovascular and respiratory effects in the anaesthetised dog
`following intravenous administration.
`
`2.6.4 PHARMACOKINETICSITOXICOKINETICS
`
`2.6.4.3
`
`Absorption
`
`1.
`
`2.
`3
`
`CC- 5013: A study to determine the oral bioavailability in the rat, dog and
`monkey.
`.
`CC- 5013: In vitro binding to plasma proteins in rat, rabbit, monkey and human.
`00-1088, CC—4047, CC-5013 and CC~7025: Comparative absorption by the
`Caco-2 cell line.
`
`2.6.4.5
`
`Metabolism
`
`1.
`
`Metabolism of (14C)—CC-5013 in isolated human hepatocytes.
`
`l3
`
`

`

`Reviewer:
`
`Anwar Goheer, Ph.D.
`
`NDA No.
`
`21-880
`
`2.6.6 TOXICOLOGY
`
`2.6.6.2
`
`Single-dose toxicity
`
`1.
`
`2.
`
`3.
`
`4.
`
`CC— 5013: Single dose oral toxicity study in the mouse (approximation of the
`minimum lethal dose level).
`CC- 5013: Single dose intravenous toxicity study in the mouse (approximation of
`the minimum lethal dose level).
`CC— 5013: Single dose oral toxicity study in the rat (approximation of the
`minimum lethal dose level).
`CC— 5013: Single dose intravenous toxicity study in the rat (approximation of the
`minimum lethal dose level).
`
`2.6.6.3
`
`Repeat-dose toxicity
`
`.U‘F‘P’Nr"
`
`6.
`7.
`
`CC—5013: 7 day oral (gavage) range— inding toxicity study in the rat.
`CC-5013: 28 day oral (gavage administration) toxicity study in the rat.
`CC—5013: 13 week oral (gavage administration) toxicity study in the rat.
`CC-5013: 28 day oral (gavage administration) toxicity study in the monkey.
`CC-4047 & CC-5013: 28 day oral (gavage administration) toxicity study in the
`monkey.
`CC-5013: 13 week oral (gavage administration) toxicity study in the monkey.
`CC—5013: 52 week oral (gavage) administration toxicity study in the monkey with
`a 7 week treatment free period.
`
`6.6.6.4
`
`Genetic toxicology
`
`1.
`
`2.
`
`3.
`
`4.
`
`CC-5013: Reverse mutation in four histidine—requiring strains of Salmonella
`typhimurium and two tryptophan-requiring strains of Escherichia coli.
`CC-50-13: Mutation at the thymidine kinase (tk) locus of mouse lymphoma l5178y
`cells (MLA) using the microtitrer fluctuation technique.-
`CC—5013: Induction of chromosome aberrations in cultured human peripheral
`blood lymphocytes.
`CC-5013: lnduction of micronuclei in the bone marrow of treated rats.
`
`2.6.6.6
`
`Reproductive and developmental toxicology
`
`1
`
`Developmental and reproductive toxicity screening study for effects on embryo-
`fetal development in rabbits.
`
`14
`
`

`

`Reviewer:
`
`Anwar Goheer, Ph.D.
`
`NDA No.
`
`21-880
`
`2.6.2 PHARMACOLOGY
`
`2.6.2.1
`
`Brief summary
`
`Structural analogues of thalidomide have been synthesized and'examined for inhibition
`of TNF-a production. The tested compounds can be classified into two classes. One
`class of compounds (selective cytokine inhibitory drugs, SelCleTM) are
`phosphodiesterase 4 inhibitors, inhibit TNF—a production, increase lL-10 production (in
`LPS-induced PBMC), and have little effect on T cell activation. The second class of
`compounds (immunomodulatory drugs, lMiDsTM), similar tothalidomide, are not
`phosphodiesterase 4 inhibitors, markedly stimulate T cell proliferation, and lL-2 and
`lFN-y production.
`In addition to these immune effects, there is evidence that
`thalidomide and its analogues act directly on tumor cells, via inducing apoptosis or G1»
`growth arrest.
`
`Lenalidomide (CC-5013) is a thalidomide analogue.
`R (+) forms.
`
`It is a racemic mixture of S (-) and
`
`0 o
`
`‘
`
`S
`
`NH2
`Lenalidomide
`
`O O
`
`n
`
`0
`Thalidomide
`
`Angiogenesis, the formation of new bloodavesselsby pre- existing endothelial cells
`(EC), depends mainly on proper activation, proliferation, adhesion, migration and
`maturation of EC (Griffioen & Molema, Pharmacol Rev 2000; 52:237—268). Therefore,
`inhibition of EC growth, adhesion and migration, and growth factor expression are
`putative anti—angiogenic targets. A series of preclinical studies had been performed to
`support the clinical evaluationof CC:5013in_.can,c,er patients.
`
`Lenalidomide inhibited TNF—a, lL-1B, lL-6 and lL-12 in LPS—stimulated PBMC (Muller et
`al., Bioorg Med Chem lett 1999; 921625-1630).
`It enhanced lFN-y and lL-2 production in
`anti-CD3 stimulated CD4+ T cells (Schafer et al., J Pharmacol Exp Ther 2003;
`305:1222-1232). Lenalidomide increased fetal hemoglobin in human CD34+ progenitor
`cells.
`It inhibited the expression of COX—2, but not COX-1 protein in LPS—, TNF—d, and
`lL-1B stimulated PBMC. Neutralizing antibody to lL-10 but not lL—1B or TNF—o partially
`reversed the inhibitory effect of CC-5013v on COX—2 expression. These results suggest
`that the anti—tumor effects of lenalidomide may be due to in part to elevation of lL-10
`production and its subsequent inhibition of COX-2 expression. Lenalidomide inhibited
`the growth of Namalwa, cells (ahuman. B celliymphoma cellline with a deletion of one
`chromosome 5) but less effective in inhibiting growth of KG-1 cells (human'myeloblastic
`cell line, also with a deletion of one chromosome 5) and other cell lines without
`'
`chromOsome 5 deletions. Patients with an isolated interstitial deletion of chromosome
`5q31 may benefit from this immunomodUlatory / antiangiogenic drug.
`
`15
`
`

`

`Reviewer:
`
`Anwar Goheer, Ph.D.
`
`NDA No.
`
`21-880
`
`2.6.2.2
`
`Primary pharmacodynamics
`
`Mechanism of action:
`
`1.
`
`Amino-substituted thalidomide analogs: Potent inhibitors of TNF- a
`production. Muller et al., Bioorg Med Chem Lett 1999; 9:1625-1630. See
`Appendix A for details.
`
`TNF-or inhibition in LPS stimulated human PBMC and whole blood
`b thalidomide analo-ues.
`
`
`
`
`
`
`480
`100
`74
`00-5013
`
`
`
`
`
`
`Compound
`TNF—or Whole blood
`TNF—or Inhibit (%).
`
`Name
`At 100 MM
`ICSO(nM)
`TNF-CX IC50 (nM)
`
`004047
`95
`13
`25
`
`
`CC—4047
`99
`3.9
`14
`
`
`93
`73
`
`
`
`00—4047
`
`85
`
`2.
`
`Inhibition of TNF-a production by PBMC and elevation of lL-2 and MlP-3a
`production by T cells by CC-4047, CC-5013, and C-11006 in vitro. Study
`Number: 5043— 152- 5119- 172.
`
`Final_lC5o and EC50 values
`
`cc-4047
`cc-so13
`CC-11006
`
`
`
`
`HUT’R‘i‘ch’FCXCdg/gay
`0.013
`0.100
`0.050
`muff—2213:“ if“
`0.010
`0.150
`0.11
`
`
`I
`”JifgaTE‘Z';a???
`0.069
`1 .2
`12
`
`‘
`1
`Human T cell 3-day
`0.0075
`Q15
`15
`lL—2 EC50 (11M)
`(Excerpted from the sponsor’s submission)
`
`’
`
`1
`
`l6
`
`

`

`Reviewer:
`
`Anwar Goheer, Ph.D.
`
`NDA No.
`
`21-880
`
`3.
`
`Inhibition of tumor necrosis factor alpha (TNF-a) production by CC-4047,
`CC-5013, and CC-11006 from human and rat whole blood stimulated with
`lipopolysaccharide. Study Number: 5196— 175.
`
`Blood samples from three individual human donors and three rats were used to
`compare the ability of lMiDs to inhibit TNF-a production by LPS. TNF—a production was
`inhibited in human blood as shown below.
`
`
`
`Compound Human Whole Blood Rat Whole Blood
`
`
`TNF—a IC50 (pM)
`TNF-a IC50 (HM)
`
`
`
` cc—4047 0.14 .12
`CC-5013
`13
`>100
`
`
`CC-l 1006 | 73. 0-53
`
`
`
`(Excerpted from the sponsor’s submission)
`
`leo for lenalidomide (CC-5013) in rat whole blood could not be calculated.
`
`4.
`
`Elevation of lL-2 production by CC-4047, CC-5013, and CC-11006 from
`human and rat whole blood stimulated with Concanavalin A. Study Number:
`5197— 189- 5226— 016.
`
`Blood samples from three individual human donors and three rats were used to
`compare the ability of lMiDs to elevate IL—2 production. Human and rat heparinized
`whole blood was plated on 96 well flat-bottom tissue culture plates. After 1 hour of
`incubat

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