throbber
ANTICANCER RESEARCH 29: 3777-3784 (2009)
`
`In Vivo Imaging of Human Colorectal Cancer Using
`Radiolabeled Analogs of the Uroguanylin Peptide Hormone
`DIJIE uu1.s, DOUGLAS OVERBEY1, LISA D. WATKINSON1.5, SAID DATBES-FIOUEROA 1·-",
`TIMOTHY J. HOFFMAN1.4.5, LEONARD R. FOIITE 1•3.S, WYNN A. VOLKERT1•2•5 and MICHAEL F. GIBLIN1•2•5•*
`
`1Research Service, Harry S, Truman Memorial Veterans' Administration Hospital, Columbia, MO 65201;
`Departments of 2Radiology, 3Medical Pharmacology and Physiology, and 4Tnternal Medicine, and
`5The Radiopharmaceutical Sciences Institute, Univen·ity of Missouri-Columbia, Columbia, MO 65211, U.SA.
`
`Abstract. Background: Uroguanylin is an endogenous
`peptide agonisi that binds ra the guanylate cyclase C
`is overexpressed
`receptor (GC-C). GC-C
`in human
`colorectal cancer (CRC), and exposure of GC-C-expressing
`cells to GC·C agonists results in cell cycle arrest and/or
`apoptosis, highlighting the therapeutic potential of such
`compounds. This study describes the first use of radiolabeled
`uroguanylin analogs for in vivo detection of CRC. Materials
`and Methods: The peptides uroguanylin and IP-uroguanylin
`were N-terminally labeled with the DOTA chelating group
`via NHS ester activation and characterized by RP-HPLC,
`ESl-MS, and GC-C receptor binding assays. The purified
`conjugates were radiolabeled with ln-111 and uud for in
`vivo biodistribution and SPECT imaging studies. In vivo
`experiments were carried out using SC!D mice bearing T84
`human colorectal cancer
`tumor xenografts. Results:
`Alteration of the position 3 aspartate residue to glutamate
`resulted in increased affinity for GC-C, with IC50 values of
`5.0±03 and 9.6±2.9 nM for IP-uroguanylin and DOTA-E3-
`111 Jn-DOTA-E3-
`uroguanylin,
`respectively.
`In
`vivo,
`uroguanylin demonstrated tumor uptake of 1.17±0.23 and
`0.61 ±0.07%JD/g at 1 and 4 h post injection, respectively.
`The specificity of tumor localization was demonstrated by co(cid:173)
`injection of 3 mg/kg unlabeled E3-uroguanylin, which
`reduced tumor uptake by 69% . Uptake in kidney, however,
`was dramatically higher for the uroguanylin peptides than
`for previously characterized radiolabeled E. coli heat-stable
`enterotoxin ( STh) analogs targeting GC-C, and was also
`
`Correspondence to: Dr. Michael F. Giblin, Harry S. Truman
`Memorial VA Hospital, Research Service Room A004, 800 Hospital
`Drive, Columbia, MO 65201, U.S.A. Tel: +l 5738146000 ell!,
`53669, Fax: + I 5738821663, e-mail: gibllnm@health.missouri.edu
`
`Key Words: Uroguanylin, E.coli heat-stable enterotox!n, guanylyl
`cyclase C, single photon-emitting computed tomography (SPECT),
`colorectal cancer, in vivo imaging.
`
`inhibited by coinjection of unlabeled peptide in a fashion not
`previously observed. Conclusion: Use of uroguanylin(cid:173)
`targtting vectors for in vivo imaging of colorectal cancers
`expressing GC-C resulted in tumor uptake that paralleled
`that of higher affinity heat-stable enterotoxin peptides, but
`also resulted in increased kidney uptake in vivo.
`
`Guanylate cyclase C (GC-C) is a type I transmembrane
`glycoprotein expressed on brush border membranes of
`intestinal epithelial cells, as wen as on transformed human
`colon cancer cell lines such as the T-84 cell line (1, 2). In
`the normal intestinal mucosa, GC-C receptors are expressed
`within the apical (luminal) face of epithelial cell membranes,
`and are therefore isolated from the bloodstream by cell-cell
`tight junctions (3-6). GC-C expression is maintained in
`transformed cells throughout the process of colorectal
`cHrcinogenesis, while expression of the endogenous GC-C
`ligands guanylin and uroguanylin is typically lost (7-9).
`Normally expressed at high levels within the lumen of the
`gut, GC-C is expressed on virtually all histologically
`confirmed primary and metastatic colorectal
`tumors
`examined in human patients, while normal tissues and other
`types of cancer express minimal or no GC-C receptors (4-
`6). GC-C receptors on colorectal tumors retain their ligand(cid:173)
`binding capacity, and expression of GC-C receptors does not
`vary as a function of metastatic site or grade of these tumors
`(5). The unique expression of GC-C by metastatic cells of
`colorectal origin within lymph nodes of patients undergoing
`staging for colorectal cancer (CRC) forms the basis for a
`PCR-based diagnostic lest that is currently undergoing
`clinical trials (10). GC-C expression has also formed the
`basis of development of ligand-based molecular agents for
`in vivo detection and therapy of colorectal cancer (9, 11-20).
`Uroguanylin is a 16 amino acid peptide with two disulfide
`bonds, and has mmornolar affinity for the GC-C receptor (21,
`22). Secretion of the endogenous peptides guanylin and
`uroguanylin into the lumen of the gut by enterochromaffm
`cells plays a role in regulation of ion and fluid homeostasis
`
`0250-7005/2009 $2 .00+ .40
`
`3777
`
`Bausch Health Ireland Exhibit 2046, Page 1 of 7
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`ANTICANCER RESEARCH 29; 3777-3784 (2009)
`
`by activation of the cystic
`transmembrane
`fibrosis
`conductance regulator ( CFTR), generating net efflux of
`sodium, chloride, bicarbonate, and water into the lumen of
`the intestine (23), In nature, the E. coli heat-stable
`enterotoxin (STh) is expressed by enterotoxigenic strains of
`to co-opt the endogenous
`E. coli bacteria in order
`uroguanylin/GC-C ligand-receptor system. Bacteria such as
`E. coli have evolved heat-stable peptides which differ
`st1ucturally from uroguanylin family peptides in that they
`third disulfide bond, which is presumably
`possess a
`responsible for both increased resistance to heat/enzymatic
`degradation as well as superagonist activity. These
`guanylin/uroguanylin mimics are also the highest affinity
`ligands known for the GC-C receptor (22, 23).
`We have developed numerous analogs of the E.coli heat(cid:173)
`stable enterotoxin for the purpose of developing an optimal
`molecular imaging vector for colorectal cancer ( 11, 13-1.5).
`Such imaging constructs could enable noninvasive imaging
`of CRC patients, and help to define patient groups that could
`benefit from treatment with GC-C agonists. Here, we
`examine the consequences of using the structurally less
`complex uroguanylin molecule on targeting efficiency. ln
`vivo targeting was assessed using peptides with N-terminal
`DOTA macrocyclic chelators, labeled with the radionuclide
`In-111. Two analogs of the uroguanylin peptide have been
`compared with respect to in vitro binding affinity and in vivo
`biodistribution patterns in SCID mice bearing T84 human
`co1orectal cancer tumor xenogrnfts.
`
`Materials and Methods
`
`All solvents were either ACS certified or HPLC grade solvents
`obtained from Fischer Scientific and used as received, DOfA-NHS
`ester was purchased from Macrocyclics (Dallas, TX, USA). 111InCl3
`was obtained from Mallinckrodt Medical, Inc (St. Louis, MO, USA)
`as a 0,05 N HCI solution. Wild-type human uroguanylin was
`obtained from the American Peptide Company, and E3-uroguanylin
`was kindly provided by Dr. Kunwar Shailubhai at Callisto
`Pharmaceuticals, All other reagents were purchased from Aldrich
`Chemical Company. Human colon carcinoma T-84 cells were
`obtained from the American Type Culture Collection (ATCC) and
`maintained and grown for use in these studies in the University of
`Missouri Cell and Immunology Core facilities. MALDI-TOF mass
`spectral analyses were performed by the proteornics core facility at
`the University of Missouri-Columbia,
`
`High performance liquid chromatography ( HPLC). High performance
`liquid chromatography (HPLC) analyses were performed on a
`Shimadzt, system equipped with an SPD-20A lN detector imd an in(cid:173)
`line sodium iodide crystal radiometric detector, HPLC solvents
`consisted of H2O containing 0 .1 % trifluoroacetic acid ( solvent A)
`and acetoni tril.e containing O. I % trifluoroacetic acid ( solvent B).
`Conditions: A Phenomenex Jupiter C-18 (5 µrn, 300 A, 4.6x250 mm)
`column was used with a flow rate of i .5 ml/min, Gradient purification
`of compounds is achieved during a linear 30 minute ramp from 23%
`B to 33% B, followed by colunm rinse and re-equilibration.
`
`Peptide synthesis and radiolabeling. Fl9-STh(l-19) was iodinated
`by a modified lactoperoxidase method. Briefly, 2 µg peptide was
`suspended in 50 µJ 100 mM sodium phosphate buffer, pH 7 .5,
`containing 2 µg lactopemxidase and 0.25-1.0 mCi Na12~I. The
`reaction was initiated by addition of 2 µI of a l: 10,000 dilution of
`30% H20 2• The reaction wru, incubated 30 min at room temperature
`with occasional mixing, then diluted with dH20 and purified to
`homogeneity by RP-HPLC.
`DOfA labeling of the folded urogmmylin peptides proceeded using
`a l 00-fold molar excess of !he DITTA-NHS ester. The reactions were
`incubated in 150 mM HEPES ut 4'C overnight, quenched with TRIS
`buffer and purified by Cl8 RP-HPLC. HPLC purified DOTA-peptides
`were subsequently characteri7,ed by ESI-MS and in vitro cell binding
`assay. For the synthesis of ll!Jn-labeled compounds, aliquots of
`1 l 1InCl3 (0.2-2.5 mCi, 4-50 µ!) were added to solutions of 50 µg of
`respective peptides in 0.2 M ammonium acetate (200 µl). The pH of
`reaction mixtures was adjusted lo 5.8, and reactions were incubated
`for 1 hour at 80'C. After 1 hour, 2 mM EDTA (50 µl) was added to
`complex unreacted 111In+3. The resulting conjugates were purified to
`homogeneity by RP-HPLC. The 111In-metallated conjugates eluted
`between 1.1-1.9 minutes before the associated non-metallated species
`enabling collection of high-specific activity, no cllITier-added I llfo(cid:173)
`peptide conjugates. All purified 111Jn-peptido conjugates were then
`concentrated by passing through a 3M Empore C-18 HD high
`pcrformruice extraction disk (7 mm/3 ml) cartridge and eluting with
`50% ethanol in 0.1 M NaH2P04 buffer (500 µ!). The concentrated
`fraction was then reduced in volume under a stream of N2, and finally
`diluted with 0.1 M NaH2P04 buffer, pH 7 .0, to a final activity of
`approximately 2 µCi/JOO pl.
`
`In vitro eel/ binding swdies, Peptide IC50s were determined by a
`competitive displacement cell binding assay using 12SI-F19-STh(l(cid:173)
`l9). Briefly 3xl06 cells suspended
`in DMEM/F-12 media
`containing 15 mM MES and 0.2% BSA, were incubated at 37'C for
`I hour in presence of approximately 20,000 cpm 125I-tracer and
`increasing concentration of the DITTA-peptide conjugates. After the
`incubation, the reaction medium was aspirated and cells were
`washed three times with media. The rndioaclivily bound to the cells
`was counted in a Packard Ria.star gamma counting system. The %
`12'I-F19-STh(1-19) bound to cells was plotted vs. increasing
`concentrations of DOTA-peptides to determine the respective IC50
`values. For statistical considerations, duplicate in vitro cell binding
`experiments with each analog were performed. IC50 calculations
`were performed using the 4 parameter logistic model within !he
`program grafil (Erithacus Software, Ltd),
`
`in vivo pharmacokinetic studies of 111 ln-peptide analogs in SCID
`mice. Four- to 5-wcck old female ICR SCID (severe combined
`immunodeficient) outbrnd mice were obtained from Taconic
`(Germantown, NY). The mice were housed four animals per cage
`in sterile micro isolator cages in a temperature- and hnmidity(cid:173)
`controlled room with a 12-hour ligh!/12-hour dark schedule. The
`animals were fed sterile rodent chow (Ralston Purina Company, St.
`Louis, MO, USA) and wuter ad libitum. Animals were housed one
`week prior to inoculation of tumor cells and anesthetized for
`injections with isoflurnne (Baxter Healthcare Corp., Deerfield, IL,
`USA) at II rate of 2.5% with 0.41 oxygen through a non-rebreathing
`anesthesia vaporizer.
`Human colon cancer T-84 cells were injected on the bilateral
`subcutaneous (s,c.) flank with N5x106 cells in a suspension of JOO
`
`3778
`
`Bausch Health Ireland Exhibit 2046, Page 2 of 7
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`Liu el al: Radiolabeled Uroguanylin Analogs
`
`A
`
`B
`"'N-,-1'.)- S,-b.,.;E,-L-O-V;..,N ... V;_A~.6-i-G-O'-L
`I
`. ·1
`
`Figure I. Struciures of(A) a11d uroguanylin (BJ E3-uroguanylln,
`
`Table I. Calc11/a1ed and observed (M+lfJ+ ya/11es and ICso values(~
`SD) for characterized peptidu.
`
`lll'IO" 1x1dr . 1X1b" · i x1.tJ•
`,1x1·0-:~ 1x10-•• 1X!O'" 1~1.0~
`[peptidel(M}
`
`Figure 2. JC50 ,matyscs ofuroguanyl/n airaJog Ji,p/aceme,r/ of 125/-F19.
`STh( 1-19) from TS4 human colortc/111 cancer cel/1.
`
`Peptide
`
`(M+H)+ Cale.
`
`(M+H}·Obs. ICso (nM)
`
`Results
`
`Uroguonylin
`DOTA•uroguanylin
`El•urognanylin
`DOTA-E3-urog11any\in
`
`1667.6
`2053.6
`1681.6
`2067.6
`
`1667.7
`2053.9
`1681.6
`2067.9
`
`39.8:1:14.9
`34.5±3.3
`S.0±0.3
`9.6:t2.9
`
`µI 3:1 PBS:Matrigel (BO Biosciences, Bedford, MA, USA) per
`injection site. T-84 cells were allowed to grow In vivo four to si,c
`weeks post inoculation, developing tumors ranging in sizes from
`0.06·0.59 grams. The biodlstributioa and uptalce of lllJn-DOTA(cid:173)
`uroguanylin and 111Jn-DOTA-E3.uroguanylin in tumor bearing
`SCIO mice was studied following randomization of animnls such
`that no significant (p<0.05) differences existed with respect to
`tumor sizes between test groups. The mice (average weight, 25 g)
`were injected with nliquots (50- tOO µI) of the radiolabelcd peptide
`solution (1-3 µCi) in each animal via the tail v.ein. Tissues, organs
`and tumors were excised from animals sacrificed at 1 hour and 4
`hour post-iajectlon (p.l.), weighed. and counted. Radioactivity was
`measured in a NaI counler and tile percent-injected dose per organ
`and the percent-injected dose per gram tissue were calculated.
`Animal studies were conducted in accordance willl the highest
`standards of care as outlined in the NIH guide for Core and Use of
`Laboratory Animals and the Policy and Procedures for Animal
`Research at the Harry S. Truman Memorial Veterans' Hospital and
`according to approved protocols.
`
`SPECTICT imaging. A combined micro-SPECT/CT unit (microCAT
`IT, Siemens Medical Systems) was employed for Single Photon
`Emitting Computed Tomography/ Computed Tomography imaging
`studies. One SCID mouse bearing T84 human colorectal cancer
`tumor l\enografts was injected intravenously with 220 11Ci (100 µI)
`111In-DOTA-uroguanylin solution and sacrificed at I hour p.i.
`Micro-SPECT scans of 60 projections were performed using a
`symmetrical 20% photopenk discriminating window. Volumetric
`data from SPECT nnd CT was visualized and image fused using
`Amira 3.1 (TGS, San Diego, CA, USA).
`
`Two DOTA-Jabeled uroguanylin nnulogs were synthesized in
`this work. and their characteristics as agents targeting the
`GC-C receptor were compared to tl1ose of previously
`synthcsiz:ed analogs of the E. coli heat-stable enterotoxin
`(STh) ( 13, 14). Each of these peptides is structurally rel.ated,
`and the two urogttanylin analogs differ only in a conservative
`D3E substitution (Figure 1). Uroguanylin peptides share
`significant sequence homology with STh, including absolute
`conservation of four cysteine residues that form a conserved
`array of disu]fide bonds (Figure 1). Analogs of the E. coli
`heat-stable enterotoxin also possess a third disulfide bond,
`lending these bacterial uroguanylin mimics exceptional
`stability and affinity for the GC-C receptor.
`Peptides were purified by RP-HPLC and characterized by
`MALDI-TOP MS and by a competitive displacement
`receptor binding assay utilizing T84 human colorectal cancer
`cells and 125I- labeled F 19-STh(l-19) (Table I, Figure 2) .
`Observed JC50 values for uroguanylin and DOTA(cid:173)
`uroguanylin (39.8:t 14.9 and 34.5±3.3 oM, respectively) were
`significantly higher than those of previously characterized
`STh analogs. as expected from previous studies of this class
`of peptide agonists (13-15, 22). A conservative D3E
`substitution, however, significantly increased the binding
`affinities of the resultant uroguaoylin analogs, with measured
`JC50 values for E3- urogunnylin and DOTA- E3-uroguanylin
`of 5.0±0.3 and 9.6±2.9 nM. respectively.
`To determine the effects of affinity differences on in vivo
`tumor localization, DOTA peptides were labeled with 111In ,md
`purified by RP-HPLC (Figure 3). Addition of N-tennioal DOTA
`moieties resulted in a 0.9-1.1-minute shift to earlier retention
`times for each uroguanylin peptide. Further 1.1-1.9-minute shifts
`to earlier retention times were ob~erved upon coordination of
`lllin by DOTA peptides. lllJn.Jabeled uroguanylin analogs were
`
`3779
`
`Bausch Health Ireland Exhibit 2046, Page 3 of 7
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`ANTICANCER REsEARCH 29: 3777-3784 (2009)
`
`l,,,._ ____ ,. ,: l _ _
`
`Figure 3. RP-HPLC chromarogrmns of pur/fi'-d m1n-DOTA-E3-
`urog110nyUn (top), 111/n-.DOTA· urogua,.ylin (bottom).
`
`Figure 4, BlodistrlbUJion analysis of /lJ /11-DOTA-l'J-uroguonylin with
`and without co-injection of 70 µg unlabeled E1,uroguanylin, and I llJn(cid:173)
`DaTA-uroguanylin at I hr pi (%/Dig, N=4).
`
`found to posse~ chromatographic properties cotTesponding to
`those of previously cha:acterized 111In-labeled STo peptides,
`requiring similar acetonitrile concentrations for elution from a
`Cl8 RP-HPLC column. RP-HPLC purification of each lllJn.
`labeled peptide resulted in high specific activity radiotmcers that
`were subsequently tested in animal models.
`ln vivo, tumor uptake of 111hi-DOTA-E3-uroguanylin at l
`hour p.l. trended higher than that of 111In-DOI'A-uroguanylh1
`(1.04±0.07 and 0.88±033 %ID/g, respectively), although the
`difference did not achieve the p<0.05 significance level (Figure
`4). 111In-DCTfA-E3-uroguanylin demonstrated significant
`(p<0.001) specific tumor uptake in T84 human colorecta\
`cancer tumor xenografts, as uptake of this compound was
`reduced to 0.32±0.06 % ID/g at 1 hour p.i. by co-injection of
`saturating concentrations of unlabeled E3-uroguanylio (Figure
`4). Uptake in tumor at 1 hour p.i. was higher than for all other
`tissues, with
`the exception of kidney. Tumor/blood,
`tumor/muscle, and tumor/liver ratios at this timepoint wete 4.7,
`20.8, and 6.1, respectively. At 4 hours p.i., tumor uptake of
`111In-DITTA-E3-uroguanylin decreased to 0 .61±0.07 % ID/g.
`However, activity also rapidly washed out of nontarget tissues,
`resulting in tumor/blood, tumor/muscle, and tumor/liver ratios
`of 6l, 61, and 4.4, respectively at 4 hours p.1. These
`target:nontarget ratios compare favorably in certain respects
`with those obtained previously at the same time p.i. using STh
`peptides such as 1 t!In-DOI'A-R l.4 ,F19-STh(l-19) (21, 55, and
`4.4, respectively) (13), although tumor uptake was generally
`higher for 111Io-DOTA-labeled STh peptides ( 1.64 % ID/g at
`4 hour p.i. for 11 1In-DOl'A-R1.4,F 19-STh(l-19)) (13, 14) .
`The most significant difference between the In vivo
`biodistribution of uroguanylin analogs and previously
`characterized STh peptides related to uptake in kidney. While
`the observed kidney uptake of lllJn. DOTA-labeled STh
`analogs at 4 hours p.i. has previously been observed in the
`(13-15), 111In-DOI'A-E3-
`range of 2.2-4.3 %ID/g
`
`3780
`
`1111'>-COT/\·8-
`01 hr
`Urog"""y)l'l
`• I hr '"il,00)'A•R1,◄•
`Ftfl.SThf1·11l)
`.,tfir"'il-OOTA-et,
`u-.,g..,,y:t,
`JH t,t ' "11,COTA,Rt,4-
`F1M!Th(MG)
`
`~,
`Figure 5. Kidney up1ake of 111Jn-DOTA-l'J-urog1Wnylin al I and 4
`hour., post injectio,1 compared with kld,1ey uptake of 111Jn-DOTA-
`11'·'.f19.STh( 1-19)11 at the Sll/M ti~ points.
`
`uroguanylin kidney uptake at this timepoint was greater than
`10-fold higher (Figure 5). This high kidney uptake was
`blocked by co-injection of excess unlabeled E3-uroguanylin,
`decreasing from 28.3±10.6 %ID/g to 14.9±3.0 %ID at 1
`hour p .l. Despite this higher kidney uptake however, in vivo
`SPECT/CT i.mGgiog clearly showed specific uptake of l 11In(cid:173)
`DOTA-uroguanylin in T84 tumor xenografts (Figure 6). Toe
`tumor:kidney ratio of 1111n-DOTA-E3-uroguanylin is lower
`than previously observed for l llJn-labeled STh analogs (13-
`15). However, uptake in other nontarget tissues is also low,
`demonstrating the utility of radiolabeled uroguanylin pepti<k
`analogs for the locali7..ation of GC-C-expres~ing cells in vivo.
`
`Discussion
`
`Research into the development of peptide ligands targeting
`the GC-C receptor has resulted in the generation of
`numerous analogs with a diverse array of incorporated
`structural alterations. The development of GC-C targeted
`
`Bausch Health Ireland Exhibit 2046, Page 4 of 7
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`Liu et al: Radiolabeled Uroguanylin Analogs
`
`affinity for GC-C only two- to three-fold lower than that of
`full-length STh analogs (13-15).
`Differences in hydrophobicity between uroguanylin and
`STh peptides did little to alter the pharrnacokinetic behavior
`of the compounds in vivo with respect to paran_ieters such as
`hepatic excretion. In vivo, lllln-DOTA-labeled uroguanylin
`peptides cleared rapidly from the bloodstream via the
`renal/urinary route, with >83% of injected activity excreted
`into urine at I hour p.i. Both uroguanylin analogs however
`had lower urinary excretion and higher retention in kidney
`than 111In-labeled STh peptides. Although DOTA-E3-
`uroguanylin demonstrated significantly higher binding
`affinity than DOTA-uroguanylio In vitro, this change did not
`result in significantly (p<0.05) increased tumor-specific
`uptake (Figure 4, Table I). At l hour p.i., the tumor
`specificity of 111 In-DOTA-E3-uroguanylin was demonstrated
`by a 69% drop in tumor specific uptake upon coinjection of
`a blocku1g dose of unlabeled uroguanylin peptide (Figure 4).
`Surprisingly, 111In-D0fA-E3-uroguanylin also demonstwted
`a high degree of uptake in kidney (Figure 5) . The high
`kidney uptake observed at l hour p.i. (28.3±10.6 %ID)
`incrensed even further at 4 hours p.i. to 49.4±3.2 % ID, a
`value more thnn IO-fold higher than was observed previously
`for 111In-DOTA-STh peptides at 4 hours p.i. (13, 14).
`Comparable uptake in kidney has never previously been
`observed for labeled heat-stable enterotoxin analogs, and
`suggests the possibility of a novel uroguaoylin receptor existing
`in kidney that can discrimlnate between uroguanylin and heat(cid:173)
`stable enterotoxin ligands. Several experimental findings have
`contributed to the belief that a receptor for uroguanylin family
`peptides dislinct from the well-characterized GC-C receptor
`exists. First, induction of natriuresis and kaliuresis by peptides
`in the uroguanylin family is observed in kidneys of GC-C
`knockout mice (24). Second, cellular responses to uroguanylin
`in immortalized human kidney epithelial (IHKE-1) cells were
`detected io whole cell patch clamp experiments that were
`distinguishable from responses to an STh peptide (2S),
`suggesting involvement of an unknown pertussis toxin-sensitive
`a protein in binding of uroguanylin in kidney cells. Thi.rd,
`uroguanylin knockout mice demonstrated a decrease in Na+
`excretion following oral salt loads when compared to wild-type
`controls, while GC-C knockout mice demonstrated 110 such
`impairment (26). In this stody, kidney uptake of 111In-DarA(cid:173)
`E3-Urogaanylin at 1 hour p.i. was reduced 47% by colnjection
`of 70 µg unlabeled E3-Uroguanylin. However, In vitro binding
`studies utilizing partially purified kidney membranes
`ll1Jn-DOTA-E3-
`demonstrated no specific binding of
`urogunnylin (Data not shown). Therefore, the question of
`whe1her the increased kidney uptake of uroguaoylin peptides
`observed here is due to ao as yet u11characteri1.ed uroguanylin(cid:173)
`speeific receptor in kidney, or alternatively is due to a structural
`property of the uroguanylin peptide such as its ac.idic N(cid:173)
`terminus, remains to be determined.
`
`3781
`
`Figure 6. SPECTICT image of a SCJD mouse bearing blla1eral hind
`}lank T84 human colorectal cancer tumor xenografts I hour p .i. of 220
`µCi IIIJn-DOTA-11,-oguanylin. Arrows indicale locations of tumors.
`
`radiopharmaceutical agents has until now centered on the use
`of tri-disu1fide analogs of the heat-s1able enterotoxin molecule,
`due to their having the highest affinity known for the GC-C
`receptor. In this work, we have compared the properties of
`such a peptide with those of structurally related uroguanylin
`peptides. Uroguanylin peptides are the endogenous ligands for
`the GC-C receptor, have lower affinity for the GC-C receptc>r,
`and Jack one of the three disalfide bonds present in heat-stnble
`enterotoxin peptides, thereby simplifying their chemical
`synthesis. Comparison of such peptides in vitro and in vivo
`makes possible the analysis oft.he effects of small variations in
`receptor binding affinities on tumor localiz.ation, and could
`also lead to the imaging of GC-C in vivo \\-'.ith o structurally
`simpler class of peptide ligands.
`In this study, we have synthesized two modified analogs
`of uroguanylin, and compared their in vitro and In vivo
`properties with previously characterized E. coll beat-stable
`entcrotoxin analogs possessing an array of linker sequences
`to N-tenninal DarA moieties. Each uroguanylin peptide also
`possessed an N-terminal DOTA chelating moiety, which is
`capable of complexing a wide range of trivalent imaging and
`therapeutic radionuclides, including 90y+3, 111 Jn+J, l49pm+3,
`153sm+3 , 166Ho+3, and 177Lu+3 . Each DCYI'A-peptide was
`synthesized and purified by C 18 RP-HPLC. Both DOTA(cid:173)
`urogunnylio and DOTA-E3-uroguanylin were shown 10 have
`lower affinity for the GC-C receptor than previously
`characterized DOTA-STh peptides (13-15). For example,
`DOTA-uroguanylin showed approximately a 10-fold lower
`affinity than DOTA-R1•4 ,F19-STh(l-19) in an
`in vitro
`competitive receptor binding assay. A conservative D3B
`substitution in wild-type human uroguanylin was shown to
`increase the affinity of the peptide for GC-C three- to four(cid:173)
`fold, resulting in a uroguanylin-based imaging probe with
`
`Bausch Health Ireland Exhibit 2046, Page 5 of 7
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`ANTICANCER RESEARCH 29: 3777-3784 (2009)
`
`Peptides in the uroguanylin family are currently under
`active investigation as phannacologicel agents with potential
`applications in the treatment of colorectal cancer (9, 16-20,
`27-30). Such therapeutic use of uroguanylin peptides relies
`on the function of uroguanylin analogs as GC-C agonists,
`eliciting intracellular production of cGMP (23). Early
`investigations into the function of uroguunylin peptides
`demonstrated that bacterial ST peptides functioned as
`superagonists for the GC-C receptor when compared with
`uroguanylin analogs, having higher binding affinity as well
`as increased potency in assays meaYuring both cGMP
`synthesis and stimulation of short circuit current resulting
`from c1- efflux from GC-C-expressing cells (23). Subsequent
`demonstrations by other researchers that expression of GC-C
`is maintained or even up-regulated in colorectal cancer
`metastases ( 4-6), while expression of the endogenous
`guanylin ligand is lost (7, 8), led to the hypothesis that the
`endogenous ligands could function in a tumor suppressor role.
`Experiments lo address this hypothesis have been carried out
`in a number of different laboratories, and have confirmed the
`growth-inhibitory signaling elicited by peptides in
`the
`uroguunylin family (9, 16-20, 27-29). Several studies have
`demonstrated either cell cycle arrest or apoptosis in response
`to exposure of GC-C-expressing cells to STh ligands (9, 19,
`27-29), and treatment of APCmin mice with therapeutic levels
`of uroguanylin resulted in marked reduction of intestinal
`polyp formation in this in vivo model (9). Together, these
`results point to a possible therapeutic role for uroguanylin
`peptides in and of themselves in the treatment of colorectal
`cancer, either alone or in combination with phosphodiesterase
`inhibitors such as exisulind.
`The results presented here have implications both for the
`molecular imaging and therapy of colorectal cancer as well
`as for the function of uroguanylin as a specific effector of
`natriuretic and kaliuretic responses in the kidney. With
`respect to molecular imaging of colorectal cancer in vivo, the
`DOIA-uroguanylin peptides described here have been used
`to successfully image GC-C-expressing human colorectal
`cancer xenografts in vivo, with rapid clearance from the
`blood pool and minimal uptake in nontarget tissues other
`than kidney (Figure 6). Given their ease of synthesis relative
`to STh analogs and similar tumor:nontarget tissue ratios,
`uroguanylin analogs labeled with In-l 11 or other imaging
`radionuclides (e.g. F-18, Tc-99m, Ga-68, Cu-64) are worthy
`of further study. The ability to image GC-C-expressing
`malignancies in vivo would provide a useful adjunct lo the
`development of GC-C ligands as cancer therapeutics, The far
`higher kidney retention of radiolabeled uroguanylin however
`necessarily reduces contrast between tumor tissue and this
`organ. Although potentially acceptable in the context of
`imaging studies, such high kidney uptake would preclude the
`use of labeled uroguanylin analogs as radiotherapeutic agents
`in favor of STh-based peptide-receptor radiotherapy (PRRT)
`
`constructs. With respect to use of uroguanylin peptides for
`the treatment of disseminated colorectal malignancies, work
`is currently underway to further elucidate the nature of the
`kidney uptake observed in this study.
`
`Acknowledgements
`
`This material is the result of work supported with resources and the
`use of facilities at the Harry S Truman Memorial Veterans' Hospital,
`Columbia, MO 65201, and the University of Missouri-Columbia
`School of Medicine Deparlmenl of Radiology, Columbia, MO
`65211. This work was funded -by a United States Department of
`Veterans' Affairs Biomedical Laboratory Research and Development
`Service VA Merit Award, and a National Cancer Institute center
`grant (l P50 CA103130-0l).
`
`Refe:rences
`
`Guarino A, Cohen M, Thompson M, Dha.rmsathaphorn K and
`Giannella R: T84 cell receptor binding and guanyl cyclase
`activation by Escherichia coli heat-stable toxin. Am J Physiol
`253: G775-G780, 1987.
`2 Vaandrager AB, Schulz S, De Jonge HR and Garbers DL:
`Guanylyl cyclase C is an N-linked glycopro!ein receptor that
`accounts for multiple heat-stable enterotoxin-binding proteins in
`the intestine. J Biol Chem 268: 2174-2179, 1993.
`3 Guarino A, Cohen M, Overmann G, Thompson Mand Giannella
`R: Binding of E. coli heat-stable enterotoxin to rat intestinal
`brush border and basolateral membranes. Dig Dis Sci 32: 1017-
`1026, 1987.
`4 Carrithers SL, Parkinson SJ, Goldstein S, Park P, Robertson DC
`and Waldman SA: Escherichia coli heat-stable toxin receptors in
`humwi colllnic tumors. Gastroentero1ogy 107: 1653-1661, 1994.
`5 Carrithers SL, Parkinson SJ, Goldstein SD, Park P, Urbanski RW
`and Waldman SA: Escherichia coli heal-stable enterotoxin
`receptors: A novel marker for colorectal tumors. Dis Colon
`Rectum 39: 171-181, 1996.
`6 Carrithers SL, Barber MT, Biswas S, Parkinson SJ, Park PK,
`Goldstein SD and Waldman SA: Guanylyl cyclase C is a
`selective marker for metastatic colorectal tumors in human
`extra.intestinal tissues. Proc Natl Acad Sci USA 93: 14827-
`14832, 1996.
`7 Cohen MB, Hawkins JA and Witte OP: Guanylin RNA
`expression in human intestine and colorec!al adenocarcinoma.
`Lab Invest 78: 101-108, 1998.
`8 Steinbrecher KA, Tuohy TM, Heppner Goss K, Scott MC, Witte
`DP, Groden J and Cohen MB: Expression of guanylin is down(cid:173)
`regulated in mouse and human intestinal adenomas. Biochem
`Biophys Res Commun 273: 225-230, 2000.
`9 Shailubhai K, Yu HH, Karunanandaa K, Wang JY, Eber SL,
`WangY, Joo NS, Kim HD, Miedema BW,Abbas SZ,Boddupalli
`SS, Currie MG et a/: Uroguanylin treatment suppresses polyp
`formation in the ApcMin/+ mouse and induces apoptosis in
`human colon ndenocarcinoma cells via cyclic GMP. Cancer Res
`60:5151-5157,2000.
`10 Waldman SA, Hyslop T, Schulz S, Barkun A, Nielsen K, Haaf
`J, Bonaccorso C, Li Y and Weinberg DS: Association of
`GUCY2C expression in lymph nodes with time to recurrence
`and disease-free survival in pN0 colorectal cancer, JAMA 301:
`745-752, 2009.
`
`3782
`
`Bausch Health Ireland Exhibit 2046, Page 6 of 7
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`Liu et al: Radiolabeled Uroguanylin Analogs
`
`11 Gali H. Sieckman GL, Hoffman TJ, Kiefer GE, Chin DT, Forte
`LR and Volkert WA: Synthesis and in vitro evaluation of an
`11 lJn-labeled ST-peptide enterotoxio (ST) analog for specific
`targeting of guanylin receptors on human colonic cancers.
`Anticancer Res 21: 2785-2792, 2001.
`12 Wolfe HR, Mendizabal M, Lleong E, Cuthbertson A, Desai V,
`Pullan S, Fujii DK, Morrison M, Pithcr R and Waldman SA: In
`imagin

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