throbber
Biochimica et Biophysica Acta 1537 (2002; 258 275
`
`Review
`
`Bl(')T‘HlMlCJ\ ET R}(1PH"i'.'\‘t(_A ACTA
`
`
`
`www.bba-direct.con.1
`
`New developments in anti—HIV chemotherapy
`
`Regrr In.sr1'mtefnr 31-i'(.’dr'r.'cz.' Rmtaarrr/1,
`
`Erik De Clercq*
`I\;'crtr‘1r'Jr'i(a/re Ur1iver.s'r'tr».'.r'r .I.(:‘ll1-'(‘P1. Mincirerhr'rJecl'er:s.m'.t1uI‘ IU, B—3l’}Ut'} 1'.reL4ven, Belgiitrpl
`
`Received 24 January 2002; accepted 24 January 2002
`
`
`
`ELSEVIER
`
`Abstract
`
`the treatment of human
`for
`trials,
`the compounds that are currently used, or are subject of advanced clinical
`Virtually all
`immunodeficiency virus (HIV) infections. belong to one of the following classes: (i) nueleoside/nucleotide reverse transcriptase inhibitors
`(NRTIs): i.e. zidovudine (AZT), didanosine (ddl), zalcitabine (ddC), stavudine (d4T). lamivudine (3TC). abacavir (ABC), emtricitahine
`[( — )FTCj,
`tenofovir disoproxil
`fumarate;
`(ii) non-nucleoside reverse transcriptase inhibitors (NNRTIs):
`i.e. nevirapine. delavirdine,
`efavircnz. ernivirine; and (iii) protease inhibitors (Pls): i.e. saquinavir, ritonavir, indinavir. nelfinavir. amprenavir and lopinavir. In addition to
`the reverse transcriptase (RT) and protease reaction, various other events in the HIV rcplicative cycle can be considered as potential targets for
`chemotherapeutic intervention: (i) viral adsorption, through binding to the viral envelope glycoprotein gp120 (polysulfates. polysulfonates,
`polycarboxylates, polyoxometalates, polynueleotides. and negatively charged albumins); (ii) viral entry,
`through blockade of the viral
`coreceptors CXCR4 [bicyclarn (AM1')3l()t)) derivatives] and CCR5 (TAK-779 derivatives); (iii) virus—cell fusion. through binding to the
`viral envelope glycoprotein gp4l
`(T-20. T-1249); (iv) viral assembly and disassembly.
`through NCp7 zinc finger-targeted agents [2.2’ -
`dithiobisbenzamides ('DIBAs). azadicarbonaniidc (ADA')];
`(V) proviral DNA integration. through integrase inhibitors such as 4-aryl-2.4-
`dioxobutanoic acid derivatives; (vi) viral mRNA transcription, through inhibitors of the transcription (transactivation) process (flavopiridol,
`fluoroquinolones). Also. various new NRTIs. NNRTIs and Pls have been developed that possess. respectively: (i) improved metabolic
`characteristics (ie. phosphorarnidate and r,j:c:r'nsaligenyl pronucleotides by-passing the first p}4ospho1"ylation step of the NRTIs), (ii) increased
`activity [“second“ or “third" generation NNRTls (i.e. TMC-I25, DPC-083)] against those lIlV strains that are resistant to the “first"
`generation NNRTls. or (iii) as in the case of Pls. a different, nonpeptidic scaffold [i.e. cyclic urea (mozcnavir). 4-hydroxy-2-pyrone
`(tipranavir}]. Nonpeptidic Pls may be expected to i.nhibit HIV mutant strains that have become resistant to pcptidomimetic Pls. Given the
`multitude of molecular targets with which anti-HIV agents can interact, one should be cautious in extrapolating the mode of action of these
`agents from cell-free enz_vn1atic assays to intact cells. Two examples in point are L-chicoric acid and the nonapeptoid CGP64222. which were
`initially described as an integrase inhibitor or Tat antagonist, respectively, but later shown to primarily act as virus adsorption/entry inl1ibitors.
`the latter through blockade of CXCR4. © 2002 Elsevier Science B.V. All rights reserved.
`
`Ke_vwonIs.' Human irnnrunodeficiency virus (HIV); Reverse transcriptase {HIV}: Protease (HIV); CXCR4 (I-IIV}: (‘CR5 (HIV); Integrase (HIV); Fusion (I-IIVI;
`Transcription (HIV)
`
`Abbre\'t'utz'or1.s'.' HIV, human innnunodeiiciency virus; _\'RTIs. nucleoside/nucleotide reverse transcriptase inhibitors; NNRTIS. non-nucleoside reverse
`transcriptase inhibitors; Pls. protease inhibitors; DIBA, 2.2’-ditlriobisbeiwainide; ADA, azadicarbonannde; AIDS, acquired innnune deficiency syndrome; I-ISV.
`herpes simplex virus; STD. sexually transmitted disease; MII‘-lot and -1|}. macrophage inflannnatory proteins; SDF-l. stronial-cell derived factor; PBMCS.
`peripheral blood mononuelear cells; TM4. transrnenibrane segment; SI. syncytiunzt-inducing; NSI, non-syncytiI.uJ:i—inducing'. NOBA, 3-nitrosobenzainide; AZT.
`zidovudinc; ddl, didanosinc: drl(', zalcitabineg d4T,
`stavudinc; 3T(f,
`Iamivudinc; ABC. ahacavir: bis(P()M}-P_\r1l*I.A. his(pivaloyloxymcthyl)-9—(2-
`phosphonylinetlroxyethyhadenine, adefovir dipivoxyl; bis(POC')-PMPA, bis(isopropyloxycarbonyloxyinethylHR]-9-(2-phosphonylmethoxypiopylladenine.
`te11ofovi1' disoproxil; dOTC, ii I2’-deoxy-3’-oxa-4-thiocytidine;
`(
`lFTC‘, enitricitabine; DAPD. Luntloxovir,
`( VH3»-D-2.6-diauninopurine dioxolane:
`bis(S/\TE)ddAMP, bis(S—acetyl-2—tl1ioethyllphosphotriester of ddA
`Proceedings of the 8th l_nterr1ational Syinpositnn on Molecular Aspects of Chemotherapy. Gdansk. Poland. 5 9 September 2001.
`l Tel; -32—16—33'/341; fax: +32-l6—33734U.
`Email address.‘ erilt.declercq@rega.l€.uleuven.ac.be (E. De Clercq).
`
`0925-4439/02xs — see front matter cc: 2002 Etsevier Science av. All rights reserved.
`*
`Pl]: S(]925—4439(U2 }UU(l89—3
`
`Case No. 2: 1 0-ev-D5954
`
`J°"5*?°” F_''°‘.‘”_°*‘~‘i ‘-9 °‘“'~
`V. Lupin Limited, at al.
`
`PTX79
`
`Janssen Ex. 2018
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`|PR2015-01030
`
`(Page 1 of 18)
`
`

`
`E. De Ck-.'J'r.'q /Bior'h1'im'('u er Bfr1pI1}'sir:a Acta 1587 (20U_’) 258-2 75
`
`259
`
`1. Introduction
`
`three anti-
`least
`Combination therapy, comprising at
`human immunodeficiency virus (HIV) drugs, has become
`the standard treatment of acquired immune deficiency
`syndrome (AIDS) or HIV-infected patients. Virtually all
`drugs that have been licensed for clinical use (or made
`available through expanded access programmes) for the
`treatment of HIV infections fall into one of the following
`three categories: (i) nucleoside/nucleotide reverse transcrip-
`lase inhibitors (NRTIS), that, following two phosphorylation
`steps (tenofovir) or three phosphorylation steps [zidovudine
`("AZ-T). didanosine (ddl), zalcitabine (ddC), stavudine (d4T),
`lamivudine (3TC). abacavir (ABC)]. act, as chain termina-
`tors, at the substrate binding site ofthe reverse transciiptase
`(RT);
`(ii) non-nucleoside reverse transcriptase inhibitors
`(NNRTIS) that interact with the RT at an allosteric. non-
`substrate binding site (nevirapine, delavirdine, efavirenz);
`and (iii) protease inhibitois (PIS) that specifically inhibit, as
`peptidomimetics. the vi1'us—associated protease (saquinaviii,
`ritonavir,
`indinavir, nelfinavir, amprenavir.
`lopinavir).
`Guidelines to the major clinical trials with these compounds
`have been recently published [1].
`Although the long—tenn goal of eradicating the virus from
`latently and chronically infected cells remains forbidding [2],
`the advent of so many new compounds other than those that
`
`have been formally approved. for the treatment of HIV
`infections. will undoubtedly improve the prognosis of
`patients with AIDS and AIDS-associated diseases. Here. I
`will primaiily address those new anti—HlV compounds that (i)
`have emerged as promising anti-HIV drug candidates during
`the last few years, that (ii) are in preclinical or early—clinical
`development. and that (iii) are targeted at well-defined steps
`in the HIV replicative cycle.
`
`2. Virus adsorption (gpl2t)) inhibitors
`
`A great variety of polyanionic compounds have been
`described to block HIV replication through interference with
`virus adsorption (or binding) to the cell surface: i.e. poly-
`sulfates, polysulfonates. polycarboxylates, polyphosphates.
`polyphosphonates, polyoxoinetalates, etc. This class of
`compounds also comprises the cosalane analogues ('1) con-
`taining the polycarboxylate pharmacophore [3]. as well as
`the sulfated polysaccharides extracted from sea algae [4].
`All these compounds, whether synthetic or of natural origin,
`are assumed to exert their anti-HIV activity by shielding off
`the positively charged sites in the V3 loop of the viral
`envelope glycoprotein (gp]20) [5,6]. which is necessary for
`virus attachment
`to the cell surface heparan sulfate, a
`primary binding site. before a more specific binding occurs
`
`
`
`ll
`
`Compound 1
`Cosataue alzatogue
`
`Janssen Ex. 2018
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`IPRZO15-01030
`
`(Page 2 of 18)
`
`

`
`260
`
`E. De Cleruq / Bfr)cii1'm1'c‘n at Biczpirv.sir.'a Acfu 1.1787 (2002) 258-2 7:?
`
`to the CD4 receptor of the CD4' cells. and to the CXCR4
`coreceptor of the CXCR-4+ cells (the latter in the case of X4
`and dual
`tropic X4/‘R5 HIV strains). Heparan sulfate is
`widely expressed on animal cells and, as it is involved in
`the virus-cell binding of a broad spectrum of enveloped
`viruses, including herpes simplex virus (HSV) [7], dengue
`virus [8] and other flaviviruses (i.e. Japanese encephalitis
`virus) [9]. it also explains why polysulfates have a broad-
`spectrum antiviral activity against HIV, HSV and various
`other enveloped viruses [10].
`
`ligand. namely SDF-1 (“strom-a|—cell derived factor”) has
`been identified. Of these chemokines, the LD78t3 isoform of
`MIP-10: has emerged as the most potent chemokine for
`inhibiting HIV-1 infection in peripheral blood mononuclear
`cells (PBMCS) [14,15] as well as monocytes/‘macrophages
`[16].
`TAK-779. a quaternary ammonium derivative (2) is the
`first nonpeptidic molecule that has been described to block
`the replication of M—tropic R5 HlV—l strains at the CCR5
`level [17].
`
`
`
`Compound 2
`TAK-779
`
`The major role of polysulfates or polyanionic substances
`in general in the management of HIV infections may reside
`in the prevention of sexual transmission of HIV infection,
`these compounds. if applied as a vaginal formulation. may
`successfully block HIV infection through both virus—to-cell
`and cell-to—cell contact. These compounds therefore merit
`being pursued as vaginal microbicides. The fact
`that
`in
`addition to their anti-HIV activity. these polyanjonic sub-
`stances, as demonstrated, for example, for poly(sodium(4-
`styrene)sulfonate). also inhibit other sexually 11'flI‘lSlTllI‘lTBCl
`disease (STD) pathogens,
`i.e. HSV, Neisseria grmorrheae
`and C'l2lmnydr'a tmclzornatis [l I],
`further adds to their
`potential therapeutic. and preventive value.
`
`3. Viral coreccptor antagonists
`
`To enter cells. following binding with the CD4 receptor.
`the HIV-1 particles must interact, again through the viral
`envelope glycoprotein gp120_. with the CXCR4 coreceptor
`[12] or CCR5 coreceptor [13]. CXCR4 is the coreceptor for
`HIV-1 strains that infect T—cells (T—tropic or X4 strains), and
`CCR5 is the coreceptor for HIV-1 strains that infect macro-
`phages (M—tropic or R5 str'ains). CXCR4 and CCR5 have
`not evolved simply to act as coreceptors for HIV entry; they
`normally act
`receptors for chemokines (chemoattractant
`cytokines). The normal
`ligands for CCR5 are RANTES
`(“regulated upon activation, nonnal T-cell expressed and
`secreted”) and MIP—1cx and -16 (“macrophage inflamma-
`tory proteins”), whereas for CXCR4, only one natural
`
`A binding site for TAK-779 has been identified within
`the transmernbrane helices I, 2, 3 and 7 of CCR5 [18].
`TAK-779 has been found to inhibit R5 HIV-1 strains in the
`
`nanomolar concentration range, while not affecting X4
`HIV-1 strains at
`l0,000—tbld higher concentrations [17].
`TAK-7'79 is not a “pure” CCR5 antagonist. as it also
`demonstrates some antagonism towards CCR2b. Unlike
`RANTES, TAK-779 does not
`induce internalization of
`
`CCR5. The clinical potential of TAK-779 and its conge-
`ners [19]
`in the therapy and/or prophylaxis of HIV-1
`infections remains to be further explored. Meanwhile.
`several new CCR5 antagonists have been reported
`[2D.21]_. and a lead clinical candidate (SCH C) for further
`development has been identified.
`i.e.
`three compounds.
`Almost simultaneously [22—24]_.
`the bicyclarn AMD310O [22], [Tyr-5,12.Lys-7]polyphemu-
`sin or T22 [23] and the nonapeptide (D—A1'g)y 01' ALX40—
`4C [24] were announced as CXCR4 antagonists. blocking
`the replication ot"T-tropic X4. but not M-tropic R5, HIV-1
`strains through selective antagonism of CXCR4. The
`bicyclams are the most specific and most potent CXCR4
`antagonists that have been described to date [25,26]. The
`bicyclams had been known as potent and selective HIV
`inhibitors for a number of years
`[27.28]. before their
`target of action was identified as the CXCR4 coreceptor
`[22.29,30]. The bicyclam AMD3l00 (3)
`inhibits the
`replication of X4 HIV-1 strains within the nanomolar
`concentration range [28]. As it
`is not
`toxic to the host
`cells at concentrations up to 500 uM, its selectivity index.
`or ratio of 50% cytotoxic concentration (CC5.3)
`to 50%
`
`Janssen Ex. 2018
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`IPRZO15-01030
`
`(Page 3 of 18)
`
`

`
`E. De Ck-.'J'r.'q / 13‘iot'ltt'rm'('u er Bfr1pI1}'s.I'r:a Auto 1587 (20t3_’) 258-2 75
`
`261
`
`antivirally effective concentration (EC_r,c.) can be estimated
`at > 100.000.
`
`.T1“©”Vf“.3.
`l:i\li{ HN:l
`l:1‘\iil
`H1'*«l:l
`l\,/l
`K)
`
`Compound 3
`AMD31fl{i
`
`A close correlation has been found, over a concentration
`range of 01-1000 ng/ml, between the AMD3l00 con-
`centrations required to inhibit (i) HIV-1 NL4-3 replication,
`(ii) monoclonal antibody (mAb 12G5) binding to the
`CXCR4 coreceptor, and (iii) SDF-1-induced signal trans-
`duction (Cazlfltix). suggesting an intimate relationship
`between these three parameters [29.30]_ The inhibitory
`effects of AMD3100 on the T—tropic HIV-1 NL4—3 strain
`have been demonstrated in a wide variety ofcells expressing
`CXCR4,
`including PBMCS; and, vice versa. various T-
`tropic and dual-tropic, but not M-tropic, HIV-1 strains have
`proven sensitive to AMD3100 in PBMC.
`Negatively charged amino acid (i.e. aspartic acid) resi-
`dues in the extracellular regions of CXCR4 must be
`involved in its interaction with both AMD3100 and SDF-
`
`I, and the V3 loop of X4 HIV gp12t), which are all three
`highly basic. Substitutions of a neutral amino acid residue
`for aspaitic acid in the second extracellular loop generated
`resistance to AMD31Cl0 [31].
`in particular,
`the aspartate
`residues at positions 171 and 262,
`located close to the
`extracellular sides of the transmembrane segments TM4
`
`and TM6. may represent crucial sites of interaction with
`the bicyclam AMD3100 [32].
`When the bicyclam AMD3100 was added to PBMC
`infected with clinical HIV isolates displaying the syncy-
`tium-inducing (SI) phenotype, these strains reverted to the
`non—syncytium-inducing (N81) phenotype, and, concomi-
`tantly, these strains switched from CXCR4 to CCR5 cor-
`eceptor use [33]. These findings indicate that selective
`blockade of CXCR4 by AMD3100 may prevent the switch
`from the less pathogenic M-tropic R5 to the more patho-
`genic T—tropic X4 strains of HIV, which in vivo heralds the
`progression to AIDS. AMD310O has proved efficacious,
`alone and in combination with other anti—HlV drugs,
`in
`achieving a marked reduction in viral load in the SCID-hu
`Thy/Liv mouse model [34]. Following a phase I clinical trial
`for safety in normal healthy volunteers [35], AMDBIOO
`recently entered phase II clinical
`trials in HIV-infected
`individuals.
`
`Given their high potency and selectivity as CXCR4
`antagonists, bicyclams, such as AMD3100. may not only
`have great potential for the therapy and/or prophylaxis of
`X4 HIV infections. but also other pathologic processes,
`such as breast cancer metastasis, which are at least partially
`dependent of, or mediated by, signaling through CXCR4
`[36].
`
`4. Viral fusion (gp41) inhibitors
`
`The interaction of the X4 or R5 HIV-1 envelope gly-
`coprotein gp120 with the coreceptor CXCR4 or CCR5,
`respectively.
`is followed by a spring—loaded action of the
`viral glycoprotein gp4l
`[normally covered by the bulkier
`gpl20), which then anchors through its amino terminus
`(the “fusion peptides”) into the target cell membrane. This
`initiates the fusion of the two lipid bilayets,
`that of the
`
`Feséttt)
`
`i_.ettr:.i.oe Zipper
`‘"3-;35l€7’1l
`
`:\»le2rti}r'at3e
`
`‘-%‘i»‘%’Im5=»i3.l.£.
`W-‘éflm
`
`25.? :_m*z;'.<2:'r r:.t._'a'r;".-3'
`
`
`
`...,.,'»'~'*§<.;:<.)«:;::t—t
`
`‘Flt?
`
`lf}}‘~ l. (T?
`
`.....—r-*“’"M
`
`_____,,...,..r---
`
`~'_....,,,_
`
`“-«--...,__
`
`- -M.
`____“_--......__
`[Est.z;as.i...ter:soait;iQa.ss:Naoatmaz.at<was.z..vas'a*it
`
`{.1oraptmnti 4
`'1’-2% tgtsrariizifissittii}
`
`Janssen Ex. 2018
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`IPRZO15-01030
`
`(Page 4 of 18)
`
`

`
`262
`
`E. De C(E?I‘L‘£f / Bfr)cii1'irtica er Biczpizy.vit.'a Acfu 1.1787 (2002) 258-2 7:?
`
`viral envelope with that of the cellular plasma membrane
`[37]. At
`the onset of the fusion process,
`the hydropho-
`bic grooves on the surface of the N36 coiled coil in the
`gp41 ectodotnain become available for binding with ex-
`traneous inhibitors, such as DP-178 (T—20), a 36-residue
`peptide.
`that binds to the hydrophobic groove of N36
`[37].
`is a synthetic, 36—amino acid
`(4)
`T—20 (pentafuside)
`peptide corresponding to residues 127-162 of the ectodo—
`main of gp4l
`(or residues 643—678 in the gpl60 pre-
`cursor).
`T—20_. previously called DP-178. was modeled
`after a specific domain (within gp41) predictive of ot-
`helical secondary structure: DP—178 consistently afforded
`100% blockade of vims-mediated cell-cell fusion (syncy—
`tium formation) at concentrations ranging from 1 to 10 ng/
`ml,
`i.e. 104- to 105—fold lower than the cytotoxic concen-
`tration [38]. An initial clinical trial has been carried out
`with T—20 at four doses (3. 10. 30 and 100 mg twice daily,
`intravenously. for 14 days) in 16 HIV-infected adults; at
`the highest dose (100 mg, twice daily). T-20 achieved by
`the 15th day a 1.5- to 2.0-fold reduction in plasma HIV
`RNA [39]. These data provide proof-of-concept that HIV
`fusion inhibitors are able to reduce virus replication in
`VIVO.
`
`is the
`The betulinic acid derivative RPR 103611 (5)
`only nonpeptidic low-molecular-weight compound that has
`been reported to block HIV-1 infection through interaction
`with gp4l:
`this liiterpene derivative has been found to
`inhibit the infectivity of a number of HIV-1 strains in the
`10 nM concentration range [41], apparently through inter-
`ference with a post—binding. envelope—dependent step
`involved in the fusion of the virus with the cell plasma
`membrane.
`The exact mode of action of RPR 103611 remains to be
`
`elucidated. Sequence analysis of RPR103611—resistant inu-
`tants indicated that a single amino acid change. 184$.
`in
`HIV-1 gp41 is sufficient
`to confer drug resistance [42].
`However, this I848 mutation did not occur in sotne of the
`
`naturally RPRl036l l -resistant HIV-1 strains such as NDK.
`More recently, the action ofRPR103611 has been thought to
`depend on the accessibility of gp4l
`[43]. and for the
`isomeric betulinic acid derivative IC 9564. HIV-1 gp120_.
`rather than gp41, has been proposed as the prime target
`(based on the mutations G23"/‘R and 1{252K emerging in
`gp120 of drug-resistant mutants) [44]. YK-FH312. a betu-
`linic acid derivative unrelated to RPI-(103611 or IC 9564.
`
`was reported to block the assembly andtor budding of HIV
`particles [45].
`
`
`
`Compound 5
`RPR [0361]
`
`
`
`Meanwhile, T-20 has proceeded to phase 1]:"lII clinical
`trials. and phase 1 clinical trials have been initiated with T-
`1249, a 39-amino acid peptide derived fi‘om DP-107 (which
`is a 38—amino acid peptide corresponding to residues 558-
`595 of gp160); T—1249 would be 10-fold more potent than
`T-20 when evaluated in vitro against HIV under the same
`conditions [40].
`
`5. Nuclcocapsid protein (NCp7) Zn finger-targeted
`agents
`
`The two zinc fingers [Cys-X3-Cys-X4-His-X4-Cys
`(CCHC). whereby X=any amino acid] in the nucleocapsid
`(NCp7) protein [46] comprise the proposed molecular target
`for zinc—ejecting compounds such as 3-nitrosobenzamide
`
`Janssen Ex. 2018
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`IPRZO15-01030
`
`(Page 5 of 18)
`
`

`
`E. De Ck-.'J't.'q /13‘ior'l'1irm't'u er Bfr1;>I1}'s1'(:a Auto 1587 (2()tl_’) 258-2 75
`
`263
`
`already equipped with a phosphonate group, and therefore
`only need two phosphorylation steps to be converted to the
`active metabolite [58]. From PMEA and PMPA.
`the oral
`prodrug forms [bis(pivaloyloxymethyl)—9-(2-phosphonyIme-
`thoxyethynadenine (bis(POM)-PMEA) or adefovir dipi—
`voxyl (7). and bistisopropyloxycarbonyloxymethyl)—(R)—9—
`(2-phosphonylmethoxypropyl)adenine (bis(POC)-PMPA) or
`tenofovir disoproxil (8) funiarate. respectively] have been
`prepared. The former is in advanced phase Lll clinical trials
`for the treatment of hepatitis B virus (HBV) infections.
`whereas the latter has completed phase 111 clinical trials for
`the treatment of HIV infections. A new drug application
`(NDA) and market authorization application (MAA) has
`been recently filed for tenofovir disoproxil fumarate with
`the FDA (US) and EMEA (EU). respectively.
`In rhesus
`macaques infected with the highly pathogenic chimeric virus
`SHIV. tenofovir treatment initiated 1 week post-infection. at
`a time when disseminated infection and extensive viral
`
`replication had already been established and CD4 ‘ T-cell
`loss had begun. led to prompt. virtually complete suppres-
`sion of viral replication and long—term stabilization of CD4 ‘
`T-cell levels. which were sustained. ever: after withdrawal of
`
`tenofovir (after 12 weeks of treatment) [59].
`
`i“H—i
`NN
`is >i
`
`i
`K}.
`
`N
`
`W
`
`o
`‘
`P/'
`(c:rr;.)3c—c—o—Crr;—o” “V” \-
`I
`
`I0
`
`Compound 7
`bis(POM)-PMEA
`Adefovir dipivoxyl
`
`ii
`(ca )3CH'-——-0-—~i:~——-t)-—-- CH -—~——o
`3
`2
`
`yo
`:~.: Pg0
`
`CH:
`
`lO
`
`Compound 8
`bis{POC)-PMPA
`'i'enofovi:' disoproxil
`
`Janssen Ex. 2018
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`IPRZO15-01030
`
`(Page 6 of 18)
`
`(NOBA). 2,2’—dithiobisben7.amide (DIBA). SRR—SB3
`(cyclic DIBA)
`[47]. 1.2—ditl1iane-4,5-diol.l.1-dioxide (di-
`thiane) [48] and azadicarbonamide (ADA) [49,50]. These
`compounds should be able to interfere with both early
`(uncoating. disassernbly) and late phases (packaging, assem-
`bly) of retrovirus replication. Their effect at the early phase
`(disassembly) may also be ascribed to cross-linkage among
`adjacent zinc fingers. The DIBAS are able to enter intact
`virions and the c1'oss—linkage of NCp7 in virions correlates
`with loss of intectivity and decreased proviral DNA syn-
`thesis during acute infection [51]. Electron microscopically,
`the effect bestowed by DIBAs on virus morphology ‘tould
`be described as “core—freezing" [52].
`
`HN
`
`“it
`
`0
`
`ENH
`
`2
`
`)1”
`
`0
`
`Compotmdti
`ADA (azesiicarbonaniide)
`
`Although NOBA, DIBA. dithiane and ADA have been
`shown to dock nicely on the NC137 Zn finger domains [53]
`and are believed to selectively target
`these Zn fingers
`without affecting the cellular Zn finger proteins,
`their
`selectivity indexes [ratio of CC;;{. (50% cytotoxic concen-
`tration) over EC“. (50% effective concentration)] are not
`that
`impressive [53]. Of the NCp7-targeted compounds,
`ADA (6) has been the first to proceed to phase I/II clinical
`trials in advanced AIDS patients. Some preliminary evi-
`dence of efficacy was witnessed with add-on ADA in
`patients failing current antiretroviral
`therapy [54];
`these
`studies should be further extended. Although ADA is an
`HIV NCp7 Zn-finger inhibitor, its action in vivo is likely to
`be multipronged. ADA may well interact with a variety of
`targets and. certainly.
`its inhibitory effects on T-cell
`responses in vitro and in vivo [55] can hardly be attributed
`to a.n action targeted at the HIV NCp7 Zn fingers.
`
`6. RT inhibitors targeted at the substrate binding site
`
`The substrate (dNTP) binding site of the HIV-1 RT is the
`target for a large variety of NRTI analogues, which have for
`several years [56] been recognized as efficacious drugs for
`the treatment of HIV infections: i.e. AZT. ddl. ddC, d4T,
`3TC, ABC. and the yet experimental drug emtricitabine
`[(—)FTC]. Fozivudine tidoxil
`is a thioether lipid AZT
`conjugate that has recently passed phase 11 clinical trials
`[57] and should be as effective as. and potentially better
`tolerated than, AZT. As a rule, all these compounds must be
`phosphorylated to their 5’-triphosphate form, before they can
`act as competitive inhibitors/substrate analogues/chain ter-
`minators at the RT level. In contrast to the nucleoside
`
`analogues. the nucleotide analogues PMEA and PMPA are
`
`

`
`264
`
`E. De C[E?I‘L‘£f / Bfr)cii1'1rtica er Biczpizy.vir.'a Acfu I.1787 (2002) 258-2 7:?
`
`NH2
`
`N
`
`<’
`N
`
`r
`
`I
`
`\
`
`N
`,-/A
`N
`
`NH;
`
`Compound 10
`Amtloxovir
`
`IDAPD, (—}-[5-D-2.6-diantinopu tine dioxolane]
`
`In addition to 3TC and (- )FTC, the stntcturally related
`(i )2’-deoxy-3'—oxa-4'—lhiocylidine (BCI-I-10652.dOTC)
`[60], the dioxolane purine nucleoside analogues [61], the
`methylenecyclopropane nucleoside analogues (and their
`phospl:ioro—L-alaninate diesters) [62.63] and the 4’—ethynyl
`nucleoside analogues [64] have recently been described as
`new anti-I-[IV agents. [( — )FTC] (9) is in phase III trials for
`HIV and phase I/II trials for HBV; it is considered for use
`in the multidrug combination therapy of HIV-1 and HBV
`infections. Amdoxovir [DAPD. (-)-[3-n-2,6-diarninopur-
`ine dioxolane]
`(10), which is converted by adenosine
`deaminase to dioxolane guanine (DXG), has proven active
`against AZ—T- and 3TC—resistant HIV-1 strains and has
`proceeded to phase I/II clinical studies [65.66]. BCH-
`10652 (dOTC)
`(11) has demonstrated activity against
`HIV-1 in the SCID-hu ThyfLiv model. Despite its struc-
`tural similaiity to 3TC. dOTC proved also active against
`3TC—resistant HIV-1 (M184V)_. albeit at a relatively high
`dosage level (400 mg/kg/day) [67]. Also in vitro, dOTC and
`its (+) and (—) enanliomers still retained, albeit reduced.
`activity against 3TC-resistant M]S4V and M1841 HIV-I
`mutants [68].
`
`H0
`
`0
`
`Compound 11
`(:)2*—deoxy—3’—oxa-4’—thiocytidine (dOTC)
`
`Compound 9
`Emtricitabine [(—)F'I‘Cj
`
`The bottleneck in the metabolic pathway leading from
`AZT and the other 2’,3’-dideoxynucleoside (ddN) ana-
`logues to their active 5’-triphosphate form is
`the ['n'st
`phosphorylation step. Therefore. attempts have been made
`at constructing 2’,3"—dideoxynuc1eotide (‘ddNMP) prodrugs
`that, once they have been taken up by the cells, deliver
`the nucleotide (ddNMP) form. This approach has proven
`particularly successful
`for a number of NRTIS such as
`2'3’-dideoxyadenosine (ddA) and d4T. Thus.
`the bis(S-
`acetyI-2-thioethyl)phosphotriester of ddA [bis('SATI:‘]d—
`dAMP] (12) was synthesized and found to be 1000-fold
`more potent against HIV than the parent compound ddA
`[69]. Similarly, aryloxyphosphoramidate derivatives of
`d4T [i.e. S0324, a d4T-MP prodrug containing at
`the
`
`phosphate moiety a phenyl group and the methylesler of
`alanine linked to the phosphate group through a phos-
`phoramidate linkage] have been constructed [70—72].
`After the d4T aryloxyphosphoramidate (13) has been
`taken up by the cells. d4TMP is released intlacellularly
`and then processed onto its active metabolite d4TTP
`[73]. This “thymidine kinase bypass” explains the high
`anti-HIV activity of d4T aryloxyphosphoramidate deriva-
`tives in thymidine kinase deficient cells and resting mono-
`cytes/macrophages [74]. The thymidine kinase (in the
`case of d4T) and the adenosine deaminase (in the case
`of ddA) can also be bypassed by using the cyclic sali-
`genyl approach [75,76]. Cy:-losaljgenyl pronucleotides of
`d4T and ddA deliver exclusively the nucleotides d4TMP
`and ddAMP_. not only under chemical—simulated hydrol-
`ysis conditions but also under
`intracellular conditions
`[77.78]. This has been convincingly shown for the q,vclo—
`saligenyl derivative of d4TMP (14) in a number of cell
`lines [79].
`
`Janssen Ex. 2018
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`IPRZO15-01030
`
`(Page 7 of 18)
`
`

`
`E. De Clemq /Biot'him1'('u er Bfr1;>I1}'s1'(:a Acta 1587 (20f3_’) ."55’—2 75
`
`265
`
`N
`\
`
`‘>
`
`N
`
`NH;
`
`N/
`
`isN
`
`I
`
`\n-5-:1
`
`0 I
`
`E
`CEi3--—-'C~-"-S"-~-CH2“--Cllzmm-OK /,0
`/ ‘P \
`\ O
`
`cH;,—~— C-~-- s---~ct-12-~-—-Cit,-mi
`I
`
`E0
`
`Conspou ml 1.2
`bis(S—acetyl—2—thioethyhpliosphotriester of ddA [bis(SATE}ddAMP}
`
`[“=:H;‘:v::
`
`H3C0-fi-~ cam as
`0 CH3
`
`x’’0
`
`__...........l
`
`Compound 13
`(HT aryluxyphosphoramidate
`
`O\P’/Ox‘
`\0/ \0 \i/“I0
`
`\ ____
`
`Compound 14
`cyclosaligenyi ai4TMP
`
`7. RT inhibitors targeted at the allusteric, nonsuhstrate
`binding site
`
`More than 30 structurally different classes of com-
`pounds have been identified as NNRTIS, viz. compounds
`that are specifically inhibitory to HIV-1 replication and
`targeted at a nonsubstrate binding site of the RT [80].
`Three NNRTls (nevirapine, delavirdine and efavirenz)
`have so far been formally iicensed fo1' clinical use in the
`
`treatment of HIV-1 infections. ernivirine (MKC-442) (I5)
`is in advanced (phase 111) clinical trials. and others are in
`preclinical or early clinical development. The NNRTIS
`interact with a specific “pocket” site of the HIV-1 RT
`[81], which is closely associated with, but distinct
`fi‘om.
`the substrate binding site. NNRTIS are notorious for
`rapidly eliciting resistance [82], resulting from mutations
`at the amino acid residues that surround the NNRTl-bind-
`
`ing site of HIV-1 RT. However, emergence of NNRTl—
`
`Janssen Ex. 2018
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`|PR2015-01030
`
`(Page 8 of 18)
`
`

`
`266
`
`E. De ClE?I‘L‘£f / Bfr)cli1'm1'cr1 at Biczpizy.vit.'a Acfu I.1787 (2002) 258-2 7:?
`
`resistant HTV strains can be prevented it‘ the NNRTIS are
`combined with NRTls and used from the beginning at
`sufficiently high concentrations [80].
`The thiocarboxanilide UC-781 (16) is an exceptionally
`potent inhibitor of HIV-1 replication [80]. It has been found
`to restore the antivi1'al activity of AZT against AZT—resistant
`HIV-1 [83]. UC-781 has been recfignized as a (retro)viru-
`cidal agent, capable of reducing the infectivity of HIV-1
`virions, and. therefore, yielding considerable promise for the
`use in (retro)virucidal formulations to prevent the trans-
`mission of HIV from infected to noninfected individuals
`
`[84]. UC—78l would seem an ideal candidate for application
`as a vaginal microbicide (virucide). i.e. when formulated in
`replens gel [85].
`
`the alkenyldiarylmethane
`urea—PETT derivatives [.97].
`(ADAM) series of compounds [98]. the pyrrolobenzoxaze—
`pinone (PBO) derivatives [99]. the quinoxalinylethylpyridyl
`thioureas (QXPTS) [100] the emivitine (MKC-442) deriv-
`ative SJ-3366 [101] and R165335(TMC125) [102]. As a
`rule, the “new" ("‘second" or “third” generation) NNRTIS
`exhibit higher potency than the “old" (‘‘first’’ generation)
`NNRTIS against wild-type and NNRTI-resistant HIV-l
`[9l_.94—96.99,102]. This is particularly prominent
`for
`DPC 083 (17) and 11165335 (TMC125) (18) that showed
`activity against Ll00l, K.l03N_. Yl8lC, Y188L.
`Kl03N t L100] and KlO3N - Yl8lC RT mutant strains in
`
`the nanomolar concentration range [102]. This makes
`Rl65335 (TMCl25) an excellent candidate for further
`clinical development.
`
`C;
`
`\
`
`.
`
`Compmmd 17
`DPC 083
`
`II
`
`it
`
`t \ t
`/
`
`\\
`
`DY
`Br./lE\[»;«
`
`mg
`
`Compound .18
`R165335 {TMCl25)
`
`Some ofthe new NNRTIS. such as SI-3366 (19). possess
`remarkable features. This compound was reported to inhibit
`HIV-1 replication at a concentration below 1 nM with a
`therapeutic index greater than 4,000,000, and to inhibit HW-
`2 replication (albeit at higher concentrations than those
`required for inhibition of HIV-1) at the viral entry stage
`[10]].
`
`Janssen Ex. 2018
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`IPRZO15-01030
`
`(Page 9 of 18)
`
`O
`
`(EH3
`
`
`
`Compound 15
`Emivirine (MKC-442}
`
`
`
`ocn (‘H"““ C/
`‘ T 2 ' ‘ T“ \
`
`C113
`
`CH,
`
`s H (
`
`,\N
`H
`
`CH4
`
`|
`
`0
`
`I
`
`\
`
`Compound 36
`Thiaearboxanilide UC—78i
`
`To the new classes of NNRTIS that offer potent anti—HlV—
`1 activity belong the thieno[3,4][l ,2,4]thiadiazine derivative
`QM9652l
`[86],
`the quinoxaline GW420S67X [87],
`the
`irnidazole derivative S-1153 (AGl549, capravirine) [88~
`90],
`( — )-6-chloro-2-[(1-fi1ro[2.3-c]pyridin-5-yl-ethyl)thio]-
`4—pyrimidinamine (PNU—l4272l) [91]. N-[2—(2,5-dimeth0x-
`yphenylethyl]-N’-[2-(5-bromopyridyl]-thiourea (HI-236)
`[92], the pyrido[l,2a]indole derivative BCH—1 [93], the 4-
`cyclopropylalkynyl-4-tritluoromethyl-3.4-dihydro—2( 1H)
`quinaaolinones DPC 961 and DPC 963, the 4—cyclop1'opylal—
`kenyl-4-tritluoromethyl-3,4-dihydro-2(lH)quinazoIin0nes
`DPC 082 and DPC 083 [94]. the thiophene-ethylthiourea
`(TET) derivative HI-443 [95], the cyclohexenylethylthiourea
`derivatives I-ll-346 and HI-445 [96],
`the C115‘-cyclopropyl
`
`

`
`E. De Ck-.'J'r.'q / Bior'hirm'('u er Bfr1pI1}'s1'r:a Acta 1587 (20U_’) ."58—2 75
`
`267
`
`0
`
`‘:33
`
`(*)—Calanolide A (22) is the only naturally occtming
`NNRTI: it was [i1st isolated from a tropical tree (C.‘w'uph_vl'~
`Hum lariigermn) and has already been the subject of a phase
`1 clinical study in healthy, HIV-negative individuals [103]
`
`Compound 19
`SJ—336ti
`
`Capravirine (AGIS49) (20) has a favorable profile of
`resilience to many drug resistance mutations. which has been
`attributed to extensive main chain hydrogen bonding involv-
`ing the main chain ofresidues 101, 103, and 236 ofthe p66
`RT subunit [89]. Capraviiine has proceeded to phase H/IH
`clinical trials [90].
`
`
`
`o \o
`
`on
`
`HIM-I“I
`Compound 22
`{+)-Calanolide A
`
`Recently. an unexpected e[l"ect of NNRTls on HIV-1 RT
`dimerization was documented [I04]: several NNRTIS,
`including efavirenz, were found to enhance the association
`between the RT subunits p66 and p51, apparently due to a
`confonnational change in the p66 subunit that resulted in
`

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