ArticlePDF Available

Abstract and Figures

l-type amino acid transporter 1 (LAT1) is an amino acid transporter that is overexpressed in several types of cancer and, thus, it can be a potential target for chemotherapy. The objectives of this study were to (a) synthesize LAT1-targeted chlorambucil derivatives and (b) evaluate their LAT1-mediated cellular uptake as well as antiproliferative activity in vitro in the human breast cancer MCF-7 cell line. Chlorambucil was conjugated to l-tyrosine—an endogenous LAT1 substrate—via either ester or amide linkage (compounds 1 and 2, respectively). While chlorambucil itself did not bind to LAT1, its derivatives 1 and 2 bound to LAT1 with a similar affinity as with l-tyrosine and their respective cellular uptake was significantly higher than that of chlorambucil in MCF-7. The results of our cellular uptake study are indicative of antiproliferative activity, as a higher intracellular uptake of chlorambucil derivatives resulted in greater cytotoxicity than chlorambucil by itself. LAT1 thus contributes to intracellular uptake of chlorambucil derivatives and, therefore, increases antiproliferative activity. The understanding gained from our research can be used in the development of LAT1-targeted anticancer drugs and prodrugs for site-selective and enhanced chemotherapeutic activity.
Content may be subject to copyright.
International Journal of
Molecular Sciences
Article
Tyrosine–Chlorambucil Conjugates Facilitate Cellular
Uptake through L-Type Amino Acid Transporter 1
(LAT1) in Human Breast Cancer Cell Line MCF-7
Piman Pocasap 1,2, Natthida Weerapreeyakul 1,2, * , Juri Timonen 3, Juulia Järvinen 3,
Jukka Leppänen 3, Jussi Kärkkäinen 3and Jarkko Rautio 3, *
1Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University,
Khon Kaen 40002, Thailand; piman.pocasap@kkumail.com
2Human High Performance and Health Promotion Research Institute, Khon Kaen University,
Khon Kaen 40002, Thailand
3School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627,
FI-70211 Kuopio, Finland; juri.timonen@uef.fi (J.T.); juulia.jarvinen@uef.fi (J.J.); jukka.leppanen@uef.fi (J.L.);
jussi.karkkainen@uef.fi (J.K.)
*Correspondence: natthida@kku.ac.th (N.W.); jarkko.rautio@uef.fi (J.R.)
Received: 20 February 2020; Accepted: 19 March 2020; Published: 20 March 2020


Abstract:
l-type amino acid transporter 1 (LAT1) is an amino acid transporter that is overexpressed
in several types of cancer and, thus, it can be a potential target for chemotherapy. The objectives
of this study were to (a) synthesize LAT1-targeted chlorambucil derivatives and (b) evaluate their
LAT1-mediated cellular uptake as well as antiproliferative activity
in vitro
in the human breast cancer
MCF-7 cell line. Chlorambucil was conjugated to l-tyrosine—an endogenous LAT1 substrate—via
either ester or amide linkage (compounds
1
and
2
, respectively). While chlorambucil itself did
not bind to LAT1, its derivatives
1
and
2
bound to LAT1 with a similar anity as with l-tyrosine
and their respective cellular uptake was significantly higher than that of chlorambucil in MCF-7.
The results of our cellular uptake study are indicative of antiproliferative activity, as a higher
intracellular uptake of chlorambucil derivatives resulted in greater cytotoxicity than chlorambucil
by itself. LAT1 thus contributes to intracellular uptake of chlorambucil derivatives and, therefore,
increases antiproliferative activity. The understanding gained from our research can be used in
the development of LAT1-targeted anticancer drugs and prodrugs for site-selective and enhanced
chemotherapeutic activity.
Keywords: AT1; cancer; chlorambucil; cellular uptake; antiproliferative; MCF-7
1. Introduction
l-type amino acid transporter 1 (LAT1), commonly referred to as the large neutral amino acid
transporter 1, is a Na
+
-independent amino acid transporter responsible for the transport of mainly
large and neutral amino acids from extracellular fluids into cells. LAT1 substrates include several
essential amino acids, e.g., leucine, isoleucine, valine, tryptophan, methionine, histidine, tyrosine,
and phenylalanine [
1
]. In addition, LAT1 is also known to mediate the transport of thyroid hormones
and prescription drugs (e.g., melphalan, gabapentin, and l-dopa). LAT1 has also been demonstrated to
transport amino acid-containing prodrugs, in which amino acids as promoieties have been linked with
non-substrate parent drugs. The prodrug strategy has been exploited, for example, to utilize LAT1 as a
drug carrier to increase drug permeability through the blood–brain barrier by conjugating various
drugs (e.g., dopamine, ketoprofen, and valproic acid) to various amino acids [24].
Int. J. Mol. Sci. 2020,21, 2132; doi:10.3390/ijms21062132 www.mdpi.com/journal/ijms
Int. J. Mol. Sci. 2020,21, 2132 2 of 15
High levels of LAT1 are expressed in many cancerous tissues [
5
], the overexpression being a result
of the higher amounts of essential amino acids needed to support the growth and development of
the malignancy [
6
]. LAT1 is thus an intriguing target for improved and targeted tumor therapy [
7
].
Some LAT1 inhibitors have, on the other hand, been developed to thwart the supply of amino acids,
impeding protein synthesis and cancer cell proliferation [
8
10
]. Recently, a LAT1 inhibitor JPH203
displayed promising results in a clinical phase I study, especially in patients with advanced solid
tumors expressing high levels of LAT1 [
11
,
12
]. The results emphasized LAT1 as a potential target for
cancer treatment. On the other hand, only a very few anticancer agents have been modified to take
advantage of LAT1-targeted cancer cell uptake [13].
The chemotherapeutic agent chlorambucil is mainly used in the treatment of chronic lymphocytic
leukemia as well as some types of lymphoma; however, the cancer resistance mechanisms, i.e., eux
by MRP1 as well as drug metabolism by glutathione (GSH) conjugation and
β
-oxidation, reduce
chlorambucil intracellular levels and hinder its eectiveness [
14
,
15
]. The objective of the present study
was to modify the structure of chlorambucil so that it resembles LAT1 substrates, thereby enhancing
its intracellular uptake through LAT1-mediated transport, and thus increasing its ecacy against
cancer cells. Based on the LAT1 model [
16
], chlorambucil derivatives
1
and
2
(Figure 1) were designed:
chlorambucil was linked from its carbonyl group with an endogenous LAT1 substrate, l-tyrosine,
or its amine derivative by either an ester or amide bond, respectively. The anity of chlorambucil
and its derivatives to LAT1, their cellular uptake, and antiproliferative activity were determined in
a LAT1-expressing human breast adenocarcinoma cell line (MCF-7). The obtained results support
rational drug/prodrug design to increase drug uptake into cancer cells via a specific transporter and,
thereby, enhance chemotherapeutic eciency.
Int
.
J
.
Mol
.
Sci
.
2020,21,xFORPEERREVIEW2of15
example,toutilizeLAT1asadrugcarriertoincreasedrugpermeabilitythroughtheblood–brain
barrierbyconjugatingvariousdrugs(e.g.,dopamine,ketoprofen,andvalproicacid)tovarious
aminoacids[2–4].
HighlevelsofLAT1areexpressedinmanycanceroustissues[5],theoverexpressionbeinga
resultofthehigheramountsofessentialaminoacidsneededtosupportthegrowthand
developmentofthemalignancy[6].LAT1isthusanintriguingtargetforimprovedandtargeted
tumortherapy[7].SomeLAT1inhibitorshave,ontheotherhand,beendevelopedtothwartthe
supplyofaminoacids,impedingproteinsynthesisandcancercellproliferation[8–10].Recently,a
LAT1inhibitorJPH203displayedpromisingresultsinaclinicalphaseIstudy,especiallyinpatients
withadvancedsolidtumorsexpressinghighlevelsofLAT1[11,12].TheresultsemphasizedLAT1as
apotentialtargetforcancertreatment.Ontheotherhand,onlyaveryfewanticanceragentshave
beenmodifiedtotakeadvantageofLAT1targetedcancercelluptake[13].
Thechemotherapeuticagentchlorambucilismainlyusedinthetreatmentofchronic
lymphocyticleukemiaaswellassometypesoflymphoma;however,thecancerresistance
mechanisms,i.e.,effluxbyMRP1aswellasdrugmetabolismbyglutathione(GSH)conjugationand
βoxidation,reducechlorambucilintracellularlevelsandhinderitseffectiveness[14,15].The
objectiveofthepresentstudywastomodifythestructureofchlorambucilsothatitresemblesLAT1
substrates,therebyenhancingitsintracellularuptakethroughLAT1mediatedtransport,andthus
increasingitsefficacyagainstcancercells.BasedontheLAT1model[16],chlorambucilderivatives1
and2(Figure1)weredesigned:chlorambucilwaslinkedfromitscarbonylgroupwithan
endogenousLAT1substrate,Ltyrosine,oritsaminederivativebyeitheranesteroramidebond,
respectively.TheaffinityofchlorambucilanditsderivativestoLAT1,theircellularuptake,and
antiproliferativeactivityweredeterminedinaLAT1expressinghumanbreastadenocarcinomacell
line(MCF7).Theobtainedresultssupportrationaldrug/prodrugdesigntoincreasedruguptake
intocancercellsviaaspecifictransporterand,thereby,enhancechemotherapeuticefficiency.
Figure1.Structuresofchlorambucilderivatives1and2.
2.Results
2.1.SynthesisofChlorambucilDerivatives
Compounds1and2weredesignedtolinkchlorambucilwithtyrosineoritsaminederivative
througheitheranesteroranamidebond,respectively.Compound1wassynthesizedby
conjugatingchlorambucil,3,tocommerciallyavailableBocLtyrosineOtBu,5,usingEDC/DMAP
andthenremovingoftheprotectinggroups(BocandOtBu)bytrifluoroaceticacid.Incompound2
synthesis,theaminoacidpart(Boc4aminoLphenylalaninemethylester,6)wasfirstsynthesized
aspreviouslydescribed[16]withmethylesterificationofBoc4nitroLphenylalanine,4,using
dimethylsulfateinthepresenceofK2CO3.Thereductionofthenitrogroupwasdoneby
Figure 1. Structures of chlorambucil derivatives 1and 2.
2. Results
2.1. Synthesis of Chlorambucil Derivatives
Compounds
1
and
2
were designed to link chlorambucil with tyrosine or its amine derivative
through either an ester or an amide bond, respectively. Compound
1
was synthesized by conjugating
chlorambucil,
3,
to commercially available Boc-l-tyrosine-OtBu,
5,
using EDC/DMAP and then
removing of the protecting groups (Boc and OtBu) by trifluoroacetic acid. In compound
2
synthesis,
the amino acid part (Boc-4-amino-l-phenylalanine methyl ester,
6
) was first synthesized as previously
described [
16
] with methyl esterification of Boc-4-nitro-l-phenylalanine,
4,
using dimethyl sulfate in
the presence of K
2
CO
3
. The reduction of the nitro group was done by hydrogenation in the presence
of 10% Pd-C. Then, peptidic coupling of chlorambucil with amino derivative
6
was catalyzed by
Int. J. Mol. Sci. 2020,21, 2132 3 of 15
EDC/DMAP. Finally, deprotection of the protecting groups, i.e., methyl ester and Boc, using LiOH and
trifluoroacetic acid, respectively, aorded chlorambucil derivative 2(Scheme 1).
Int
.
J
.
Mol
.
Sci
.
2020,21,xFORPEERREVIEW3of15
hydrogenationinthepresenceof10%PdC.Then,peptidiccouplingofchlorambucilwithamino
derivative6wascatalyzedbyEDC/DMAP.Finally,deprotectionoftheprotectinggroups,i.e.,
methylesterandBoc,usingLiOHandtrifluoroaceticacid,respectively,affordedchlorambucil
derivative2(Scheme1).
Scheme1.Syntheticpathwayforchlorambucilderivatives.Reagentsandconditions:(a)K2CO3,
(CH3O)2SO2,acetone,overnight,and(b)10%PdC,H2,MeOH,overnight;(c)EDC,DMAP,CH2Cl2,
overnight;(d)TFA,CH2Cl2,6h;(For2)(e)LiOH,THF,1.5h,and(f)TFA,CH2Cl2,2h;allconditions
wereconductedatroomtemperature.
2.2.InvitroConversion
Theinvitroconversionofcompounds1and2wasdeterminedbothinaqueousbuffersolution
(pH7.4)andinhumanlivermicrosomes.Bothcompoundsreleasedtheparentdrug,chlorambucil,
inbothexperiments.Therespectivechemicalconversionprofilesofcompounds1and2resultedin
halflives(T1/2)of167and179min(Figure2A),suggestingnodifferenceinthenonenzymatic
conversionratebetweentheesterandamidederivatives.Bycontrast,theenzymaticconversionin
humanlivermicrosomesresultedinmorerapidconversionofcompound1(T1/2:5min)comparedto
compound2(T1/2:52min)(Figure2B).
(A)(B)
 
Figure2.Timecourses(mean,n=2)forchlorambucilderivatives1and2duringchemical
conversioninphosphatebuffer(pH7.4)(A)andenzymaticconversioninhumanmicrosomes(pH
7.4)(B)at37°C.
Scheme 1.
Synthetic pathway for chlorambucil derivatives. Reagents and conditions: (
a
) K
2
CO
3
,
(CH
3
O)
2
SO
2
, acetone, overnight, and (
b
) 10% Pd-C, H
2
, MeOH, overnight; (
c
) EDC, DMAP, CH
2
Cl
2
,
overnight; (
d
) TFA, CH
2
Cl
2
, 6 h; (For
2
) (
e
) LiOH, THF, 1.5 h, and (
f
) TFA, CH
2
Cl
2
, 2 h; all conditions
were conducted at room temperature.
2.2. In Vitro Conversion
The
in vitro
conversion of compounds
1
and
2
was determined both in aqueous buer solution
(pH 7.4) and in human liver microsomes. Both compounds released the parent drug, chlorambucil,
in both experiments. The respective chemical conversion profiles of compounds
1
and
2
resulted
in half-lives (T
1/2
) of 167 and 179 min (Figure 2A), suggesting no dierence in the non-enzymatic
conversion rate between the ester and amide derivatives. By contrast, the enzymatic conversion in
human liver microsomes resulted in more rapid conversion of compound
1
(T
1/2
: 5 min) compared to
compound 2(T1/2: 52 min) (Figure 2B).
Int
.
J
.
Mol
.
Sci
.
2020,21,xFORPEERREVIEW3of15
hydrogenationinthepresenceof10%PdC.Then,peptidiccouplingofchlorambucilwithamino
derivative6wascatalyzedbyEDC/DMAP.Finally,deprotectionoftheprotectinggroups,i.e.,
methylesterandBoc,usingLiOHandtrifluoroaceticacid,respectively,affordedchlorambucil
derivative2(Scheme1).
Scheme1.Syntheticpathwayforchlorambucilderivatives.Reagentsandconditions:(a)K2CO3,
(CH3O)2SO2,acetone,overnight,and(b)10%PdC,H2,MeOH,overnight;(c)EDC,DMAP,CH2Cl2,
overnight;(d)TFA,CH2Cl2,6h;(For2)(e)LiOH,THF,1.5h,and(f)TFA,CH2Cl2,2h;allconditions
wereconductedatroomtemperature.
2.2.InvitroConversion
Theinvitroconversionofcompounds1and2wasdeterminedbothinaqueousbuffersolution
(pH7.4)andinhumanlivermicrosomes.Bothcompoundsreleasedtheparentdrug,chlorambucil,
inbothexperiments.Therespectivechemicalconversionprofilesofcompounds1and2resultedin
halflives(T1/2)of167and179min(Figure2A),suggestingnodifferenceinthenonenzymatic
conversionratebetweentheesterandamidederivatives.Bycontrast,theenzymaticconversionin
humanlivermicrosomesresultedinmorerapidconversionofcompound1(T1/2:5min)comparedto
compound2(T1/2:52min)(Figure2B).
(A)(B)
 
Figure2.Timecourses(mean,n=2)forchlorambucilderivatives1and2duringchemical
conversioninphosphatebuffer(pH7.4)(A)andenzymaticconversioninhumanmicrosomes(pH
7.4)(B)at37°C.
Figure 2.
Time courses (mean, n=2) for chlorambucil derivatives
1
and
2
during chemical conversion
in phosphate buer (pH 7.4) (
A
) and enzymatic conversion in human microsomes (pH 7.4) (
B
) at 37
C.
Int. J. Mol. Sci. 2020,21, 2132 4 of 15
2.3. LAT1 Binding Anity
The binding abilities of compounds
1
and
2
to LAT1 were investigated in MCF-7 cells at 10
µ
M in
a competitive inhibition assay with the endogenous LAT1 substrate, l-leucine. Chlorambucil, which is
not a LAT1 substrate, displayed no binding anity to LAT1 as it does not have the physicochemical
facets to inhibit [
14
C]-l-leucine uptake. The chlorambucil derivatives
1
and
2
, however, exhibited
binding anity to LAT1, evidenced by the significantly decreased cellular uptake of [
14
C]-l-leucine to
61.5%
±
2.7% and 49.6%
±
5.3%, respectively. The endogenous LAT1 substrate, l-tyrosine, was able to
decrease the cellular uptake of [14C]-l-leucine to 48.6% ±7.9% (Figure 3).
Int
.
J
.
Mol
.
Sci
.
2020,21,xFORPEERREVIEW4of15
2.3.LAT1BindingAffinity
Thebindingabilitiesofcompounds1and2toLAT1wereinvestigatedinMCF7cellsat10μM
inacompetitiveinhibitionassaywiththeendogenousLAT1substrate,Lleucine.Chlorambucil,
whichisnotaLAT1substrate,displayednobindingaffinitytoLAT1asitdoesnothavethe
physicochemicalfacetstoinhibit[14C]Lleucineuptake.Thechlorambucilderivatives1and2,
however,exhibitedbindingaffinitytoLAT1,evidencedbythesignificantlydecreasedcellular
uptakeof[14C]Lleucineto61.5%±2.7%and49.6%±5.3%,respectively.TheendogenousLAT1
substrate,Ltyrosine,wasabletodecreasethecellularuptakeof[14C]Lleucineto48.6%±7.9%
(Figure3).
Figure3.Abilityofchlorambucilanditsderivatives1and2(10μM)toinhibituptakeof
[14C]Lleucine(0.157μM)inMCF7cells.Alldatapresentedasmean±SD(n=3).*p<0.05,
significantdifferencecomparedtountreatedcontrol.
2.4.InvitroCellularUptake
Therespectivecellularuptakeofcompounds1and2aswellastheparentdrug,chlorambucil,
wereevaluatedinMCF7cellstodeterminetheexposureeffectofbothtimeandconcentrationon
cellularuptake.Chlorambucilanditsderivatives1and2werealltakenupintoMCF7cellsinbotha
timeandconcentrationdependentmanner.
Inthetimedependentstudy,theuptakeofcompound1wassignificantlygreaterthanthatof
chlorambucilatallfivetimepointsbetween5and45min.Compound2showedsignificantlygreater
cellularuptakethanchlorambucilafter10minofincubation(Figure4A).Thecellularuptakeof
compoundsvariedasfollows:chlorambucil0.160.29μmol/mgofprotein;compound10.270.45
μmol/mgofprotein,andcompound20.180.47μmol/mgofprotein.
Figure 3.
Ability of chlorambucil and its derivatives
1
and
2
(10
µ
M) to inhibit uptake of [
14
C]-l-leucine
(0.157
µ
M) in MCF-7 cells. All data presented as mean
±
SD (n=3). * p<0.05, significant dierence
compared to untreated control.
2.4. In Vitro Cellular Uptake
The respective cellular uptake of compounds
1
and
2
as well as the parent drug, chlorambucil,
were evaluated in MCF-7 cells to determine the exposure eect of both time and concentration on
cellular uptake. Chlorambucil and its derivatives
1
and
2
were all taken up into MCF-7 cells in both a
time- and concentration-dependent manner.
In the time-dependent study, the uptake of compound
1
was significantly greater than that of
chlorambucil at all five time points between 5 and 45 min. Compound
2
showed significantly
greater cellular uptake than chlorambucil after 10 min of incubation (Figure 4A). The cellular
uptake of compounds varied as follows: chlorambucil 0.16–0.29
µ
mol/mg of protein; compound
10.27–0.45 µmol/mg of protein, and compound 20.18–0.47 µmol/mg of protein.
According to time-dependent data, the uptake of both compounds
1
and
2
reached their maximum
at 15 min. Therefore, 15 min was selected for the concentration-dependent uptake study as it displayed
unsaturated uptake with maximum values. The respective uptakes of compounds
1
and
2
(2.5
±
0.35 and
2.61
±
0.10
µ
mol/mg of protein) were significantly higher than that of chlorambucil (1.61
±
0.14
µ
mol/mg
of protein) at concentrations of 80 µM and higher, at 15 min incubation time (Figure 4B).
Int. J. Mol. Sci. 2020,21, 2132 5 of 15
Int
.
J
.
Mol
.
Sci
.
2020,21,xFORPEERREVIEW5of15
(A)(B)
 
Figure4.Cellularuptakeofchlorambucilanditsderivatives1and2atdifferentincubationtimes(20
μM)(A)anddifferentconcentrations(15min)(B)inMCF7cells.Valuesexpressedasmean±SD(n
=3).*p<0.05,significantdifferencecomparedtochlorambucilatrelevanttimesandconcentrations.
Accordingtotimedependentdata,theuptakeofbothcompounds1and2reachedtheir
maximumat15min.Therefore,15minwasselectedfortheconcentrationdependentuptakestudy
asitdisplayedunsaturateduptakewithmaximumvalues.Therespectiveuptakesofcompounds1
and2(2.5±0.35and2.61±0.10μmol/mgofprotein)weresignificantlyhigherthanthatof
chlorambucil(1.61±0.14μmol/mgofprotein)atconcentrationsof80μMandhigher,at15min
incubationtime(Figure4B).
2.5.AntiproliferativeActivity
Theantiproliferativeactivityofchlorambucilanditsderivatives1and2werestudiedinMCF7
cellsinvitro.Thecellcytotoxicitywasdeterminedaftertreatmentwithallcompoundsatvarious
concentrationsfor24,48,and72h.Allcompoundsdisplayedtimedependentcellcytotoxicity
(Figure5A).Therewasnocelldeathinanyofthetreatmentgroupsat24hincubation.At48h,
compounds1and2displayedcellcytotoxicityinaconcentrationdependentmanner,whilenocell
deathwasdetectedinthechlorambuciltreatedcells(6.7%±2.0%,15.5%±3.1%,and(1.1%)±2.5%
cellcytotoxicityfor1,2,andchlorambucilat120μM,respectively).Allcompoundsincreasedcell
cytotoxicityinaconcentrationdependentmannerat72hincubationwiththehighestcell
cytotoxicityobservedat120μMconcentration.Therespectivecellcytotoxicitywas25.8%±3.2%for
2,18.0%±2.5%for1,and10.6%±1.3%forchlorambucil(Figure5B).Insummary,compounds1and
2possessedhigherantiproliferativeactivitythanchlorambucilinbothatime‐ and
concentrationdependentmanner.ToinvestigatethecontributionofLAT1onthechlorambucil
derivativeactivity,BCH,astandardLAT1inhibitor[17],wascoincubatedwithcompound1or2
(Figure5C).BCHwasreportedtoinhibitthefunctionofLAT1around80%–100%withno
cytotoxicityataminimumconcentrationof10mM[18,19].ThatmeanslowerBCHconcentrations
(<10mM)werenotsufficienttoinhibitLAT1function,whilehigherconcentrations(>10mM)could
becytotoxictothecellandinterferewiththeresults.Thereporteddataagreedwithourstudyinthat
BCHhadnosignificantcytotoxicitytoMCF7(4.3%±3.4%)attheconcentrationused(10mM,72h).
Therefore,thisconcentrationwasusedincoincubationwithcompounds1and2.Ourresultshowed
thatBCHwasnotabletoreducecytotoxicityofcompound1significantly,possiblyduetothe
insensitivityofMCF7tothechlorambucilderivative.Aprofoundresultwashoweverdemonstrated
inthecoincubationofBCHwithcompound2,sinceBCHsignificantlyreduceditscytotoxicity
(15.7%±2.7%).
Figure 4.
Cellular uptake of chlorambucil and its derivatives
1
and
2
at dierent incubation times
(20
µ
M) (
A
) and dierent concentrations (15 min) (
B
) in MCF-7 cells. Values expressed as mean
±
SD
(n=3). * p<0.05, significant dierence compared to chlorambucil at relevant times and concentrations.
2.5. Antiproliferative Activity
The antiproliferative activity of chlorambucil and its derivatives
1
and
2
were studied in MCF-7
cells
in vitro
. The cell cytotoxicity was determined after treatment with all compounds at various
concentrations for 24, 48, and 72 h. All compounds displayed time-dependent cell cytotoxicity
(Figure 5A). There was no cell death in any of the treatment groups at 24 h incubation. At 48 h,
compounds
1
and
2
displayed cell cytotoxicity in a concentration-dependent manner, while no cell
death was detected in the chlorambucil-treated cells (6.7%
±
2.0%, 15.5%
±
3.1%, and (
1.1%)
±
2.5%
cell cytotoxicity for
1
,
2
, and chlorambucil at 120
µ
M, respectively). All compounds increased cell
cytotoxicity in a concentration-dependent manner at 72 h incubation with the highest cell cytotoxicity
observed at 120
µ
M concentration. The respective cell cytotoxicity was 25.8%
±
3.2% for
2
, 18.0%
±
2.5%
for
1
, and 10.6%
±
1.3% for chlorambucil (Figure 5B). In summary, compounds
1
and
2
possessed higher
antiproliferative activity than chlorambucil in both a time- and concentration-dependent manner.
To investigate the contribution of LAT1 on the chlorambucil derivative activity, BCH, a standard
LAT1 inhibitor [
17
], was co-incubated with compound
1
or
2
(Figure 5C). BCH was reported to
inhibit the function of LAT1 around 80%–100% with no cytotoxicity at a minimum concentration
of 10 mM [
18
,
19
]. That means lower BCH concentrations (<10 mM) were not sucient to inhibit
LAT1 function, while higher concentrations (>10 mM) could be cytotoxic to the cell and interfere
with the results. The reported data agreed with our study in that BCH had no significant cytotoxicity
to MCF-7 (4.3%
±
3.4%) at the concentration used (10 mM, 72 h). Therefore, this concentration was
used in co-incubation with compounds
1
and
2
. Our result showed that BCH was not able to reduce
cytotoxicity of compound
1
significantly, possibly due to the insensitivity of MCF-7 to the chlorambucil
derivative. A profound result was however demonstrated in the co-incubation of BCH with compound
2, since BCH significantly reduced its cytotoxicity (15.7% ±2.7%).
Int. J. Mol. Sci. 2020,21, 2132 6 of 15
Int
.
J
.
Mol
.
Sci
.
2020,21,xFORPEERREVIEW6of15
(A)(B)
(C)
Figure5.Antiproliferativeactivityofchlorambucilderivatives1and2atdifferentincubationtimes
(120μM)(A),differentconcentrations(72h)(B),anddifferentconcentrationswithBCH(10mM)
(C)inMCF7cells.Resultsexpressedasmean±SD(n=3).*p<0.05,significantdifference
comparedtochlorambucil(A),untreatedcontrol(B),andcoincubatedwithBCH(C).
3.Discussion
StructuralfeaturesforLAT1recognitionarepronetoresemblemoietiesbelongingto
endogenousLAT1substrates.ThepreferablefeaturesforLAT1bindinginclude(1)freeaminoand
carboxylicacidgroups;(2)alargeandneutralsidegroup(referringtoanaminoacidsidechain);
and,(3)anHbondacceptorbehindtheaminoacidsidechain[20–22].Nevertheless,thefeaturesare
notobligatoryasderivatizationorreplacementofthefunctionalgroupscanmaintainsomeaffinity
toLAT1[1,21].
Toensureadequatebindingaffinity,compounds1and2weredesignedaccordingtoour3D
quantitativerelationship(3dQSAR)modeloftheLAT1bindingsitesoastocontain:(1)freeamino
andcarboxylicacidgroups;(2)anaromaticsidechain;and,(3)anester/amidelinkage(Hbond
acceptor).Whileboththeester1andamide2bondscanundergobioconversion,thechlorambucil
derivatives1and2weresupposedtobeactivewithoutbondcleavagebecauseofthepresenceofthe
unconjugatedN,Nbis(2chloroethyl)aminemoiety.Themoietyactsastheactivefunctionalgroup
tointeractwithDNAandexertitspharmacologicalactivity.Thesecompoundsare,therefore,
terminologicallynotprodrugsthatpossessnopharmacologicalactivity[23],eventhoughtheycan
releasechlorambucilandactlikeprodrugs.
SincetheabilityofthechlorambucilderivativestobindtoLAT1dependsontheaminoacid
promoiety,thestabilityoftheesteroramidebondsignificantlyaffectsLAT1bindingproperties.In
Figure 5.
Antiproliferative activity of chlorambucil derivatives
1
and
2
at dierent incubation times
(120
µ
M) (
A
), dierent concentrations (72 h) (
B
), and dierent concentrations with BCH (10 mM) (
C
) in
MCF-7 cells. Results expressed as mean
±
SD (n=3). * p<0.05, significant dierence compared to
chlorambucil (A), untreated control (B), and co-incubated with BCH (C).
3. Discussion
Structural features for LAT1 recognition are prone to resemble moieties belonging to endogenous
LAT1 substrates. The preferable features for LAT1 binding include (1) free amino and carboxylic acid
groups; (2) a large and neutral side group (referring to an amino acid side chain); and, (3) an H-bond
acceptor behind the amino acid side chain [
20
22
]. Nevertheless, the features are not obligatory as
derivatization or replacement of the functional groups can maintain some anity to LAT1 [1,21].
To ensure adequate binding anity, compounds
1
and
2
were designed according to our 3D
quantitative relationship (3d-QSAR) model of the LAT1 binding site so as to contain: (1) free amino and
carboxylic acid groups; (2) an aromatic side chain; and, (3) an ester/amide linkage (H-bond acceptor).
While both the ester
1
and amide
2
bonds can undergo bioconversion, the chlorambucil derivatives
1
and
2
were supposed to be active without bond cleavage because of the presence of the unconjugated
N,N-bis-(2-chloroethyl)-amine moiety. The moiety acts as the active functional group to interact with
DNA and exert its pharmacological activity. These compounds are, therefore, terminologically not
prodrugs that possess no pharmacological activity [
23
], even though they can release chlorambucil and
act like prodrugs.
Since the ability of the chlorambucil derivatives to bind to LAT1 depends on the amino acid
promoiety, the stability of the ester or amide bond significantly aects LAT1 binding properties. In our
results, there was no dierence in the nonenzymatic conversion rates between the ester and amide
Int. J. Mol. Sci. 2020,21, 2132 7 of 15
derivatives, as the two compounds displayed a comparable half-life. Both compounds displayed
chemical stability with a respective half-life being around 170 min, meriting further study; however,
they underwent faster bioconversion in human liver microsomes, resulting in a dramatic reduction
in half-life. The half-life of the bioconversion of the amide derivative
2
was up to 10 times higher
compared with that of the ester
1
. As with other reports, the result indicates that the amide bond is less
sensitive to enzymatic hydrolysis than the corresponding ester bond [
2
,
24
]; the ester derivative is more
susceptible to enzymatic bioconversion, limiting its ability to reach the target site intact in vivo.
While chlorambucil demonstrated no binding anity to LAT1 in MCF-7 cells, its derivatives
1
and
2
bound to LAT1 with an anity comparable to the endogenous LAT1 substrate L-tyrosine. Nonetheless,
the high-binding anity is not obligatory to the ecient LAT1-mediated cellular uptake [
25
]. Smaller
compounds seem to be taken up by LAT1 faster than the larger ones, at the expense of specificity
and anity [
26
]. By contrast, the larger molecules with high lipophilicity can bind more eciently
to LAT1, at the expense of transport capacity, resulting in compounds acting as inhibitors rather
than substrates [
27
]. Consequently, the cellular uptake profiles as well as binding anity data are
crucial to determining whether the LAT-utilizing compound act as either an inhibitor or a substrate.
Chlorambucil derivatives
1
and
2
were taken up into the MCF-7 cells with a higher intracellular
concentration than chlorambucil itself in the time-dependent study. This result is consistent with the
concentration-dependent study. We hypothesize that carrier-mediated transport is participating in
improving the uptake of the designed derivatives via LAT1 into cancer cells. The computational analysis
(Table 1) of the physicochemical properties indicates that chlorambucil is a small lipophilic molecule with
a low polar surface area (PSA) that can eectively cross the cell membrane by passive diusion [
28
,
29
],
which has been demonstrated in the ascites sarcoma cell [
30
] and in situ rat intestine [
31
]. In contrast,
derivatives
1
and
2
are larger molecules (in both weight and size) with a high PSA, making these
derivatives less susceptible to passive diusion across cell membranes. The higher cellular uptake of
chlorambucil derivatives
1
and
2
is, thus, more likely attributable to other transportation processes
than to passive diusion. Taken together, the LAT1 binding study and the cellular uptake profile
indicate the roles of derivatives 1and 2as LAT1 substrates.
Table 1. Physicochemical properties of chlorambucil and its derivatives 1and 2.
Computational Parameter Compound
Chlorambucil 1 2
Log P 4.1 4.7 4.1
Molecular weight (g/mol) 304.2 467.4 466.4
Molecular accessible area (Å2)558.3 798.7 802.6
Polar surface area (Å2)40.5 92.8 95.7
Due to a more ecient cellular transport, compounds
1
and
2
accumulated in the MCF-7
cells more than did the parent drug, supporting the hypothesis that the antiproliferative activities
of derivatives
1
and
2
have higher cytotoxicity than those of chlorambucil in both a time- and
concentration-dependent manner. The incubation with BCH, a LAT1 inhibitor, could also decrease
the chlorambucil derivative cytotoxicity, especially compound
2
, suggesting the contribution of
LAT1 in antiproliferative activity. Our results demonstrate that the increased intracellular uptake
attributed to LAT1 transport of derivatives
1
and
2
is positively related to their higher antiproliferative
activity. Derivative
2
exhibited a higher intracellular concentration and cytotoxicity than derivative
1
.
The metabolic process in the MCF-7 cells was probably engaged; since the amide derivative
2
was less
sensitive to enzymatic bioconversion than the ester form
1
, the higher intracellular accumulation was
detected after the treatment of
2
. The chlorambucil derivatives are pharmacologically active on their
own and require no bioconversion, like a prodrug, so their intracellular concentrations relate directly
to their pharmacological activity (i.e., antiproliferation). In accordance with the antiproliferative study,
the MCF-7 cells were not as sensitive to chlorambucil, as cell cytotoxicities were not significantly
Int. J. Mol. Sci. 2020,21, 2132 8 of 15
increased until the concentrations of compounds
1
and
2
reached 80
µ
M. Nonetheless, our result
demonstrated that the LAT1-targeting approach could increase cellular drug uptake, leading to the
improvement of drug eciency, and thereby indicating the possibility of using this approach in other
LAT1-expressing cell lines.
The issues with drug stability, resistance, and selectivity limit clinical usage of chlorambucil [
12
].
The consequence of instability as well as drug resistance reduces chlorambucil intracellular levels,
leading to inecient treatment. Our results demonstrate that the chlorambucil derivatives
1
and
2
accumulate inside cells at a higher level and display a greater eciency than their parent drug.
In addition, the slow release of chlorambucil from its ester/amide derivatives could possibly retard
metabolic and/or resistance deactivation [32].
The lack of selectivity, a major concern, hinders the clinical usage of chlorambucil due to serious
adverse eects, i.e., myelosuppression and an increased risk of developing secondary neoplasm in
normal tissues [
33
,
34
]. We propose conjugating chlorambucil to LAT1 substrate so as to increase drug
selectivity. Whereas chlorambucil enters the cells nonspecifically by passive diusion, its amino acid
conjugates bypass the cell membrane using more specific active transport. Since LAT1 is upregulated in
many cancers, specific transport using LAT1 as a carrier is conceivable to accumulate the chlorambucil
derivatives in cancer cells at a higher level than in normal cells. Therefore, the higher selectivity
could possibly lead to the reduction of nonselective side eects. In addition, the binding anity to
LAT1 could also be enhanced by structural modifications. The meta-conjugation of l-phenylalanine
(or l-tyrosine) to a parent drug has been shown to display higher binding anity to LAT1 than
para-conjugation [
2
,
26
]. In our experiment, chlorambucil conjugation to l-tyrosine derivatives at the
meta-position was highly acid sensitive, and both the ester and amide bonds were cleaved during the
deprotection process (unpublished data).
LAT1 has been reported to share its substrates with other transporters such as LAT2 [
35
],
monocarboxylate transporter 8 and 10 (MCT8/10) [
36
], and organic anions transporting polypeptides
(OATPs) [
21
]. Whereas the OATPs transport a broad range of amphipathic substrates and are not
specific to only amino acids, the MCT8/10 specifically uptakes thyroid hormone derivatives and
displays low anity to aromatic amino acids such as tyrosine and tryptophan (K
m
~ 5 mM in
Xenopus oocyte) [
37
]. On the other hand, LAT1 preferentially transports large and neutral amino
acids, while LAT2 displays a broader range of substrate selectivity including smaller amino acids.
In general, LAT1 binds to its substrates with higher anity (K
m
~ 10–20
µ
M in rat) than LAT2 to its
substrates (K
m
~ 30-300
µ
M in rat) [
38
]. Therefore, it is reasonable to target LAT1 for improved cancer
drug uptake using aromatic amino acids, such as tyrosine, as the promoiety. Based on the results,
LAT1 contributed to cellular uptake of chlorambucil derivatives. However, the contribution of other
amino acid transporters (OATPs, MCTs, and LAT2) should not be ruled out as they share overlapping
substrates. While some of the other transporters can be found in normal tissues, the unspecific
binding among these transporters could complicate LAT1 utilization of compounds by impairing their
selectivity to cancer cells. The o-target eects are also possible and should be taken into consideration
in further in vivo studies.
In short, our study demonstrated a drug-design approach to increase the eciency of chlorambucil,
as its derivatives displayed both higher intracellular accumulation and cytotoxicity against cancer,
which compensate for the issues of drug instability and resistance that reduce intracellular concentration
and eciency. As LAT1 is overexpressed in several cancers, the structural modification of the drug
could possibly increase drug selectivity and reduce nonselective side eects. We suggest that the
(pro)drug design approach might be applied to other chemotherapeutic agents that contain available
functional group(s) to conjugate to the LAT1 substrate, rectifying permeability and selectivity issues.
Further studies are also needed to elucidate transporter selectivity and structural modifications for
enhancing the selectivity of chlorambucil derivatives. The pharmacokinetic profile using an animal
model should also be addressed since metabolism, unbound/bound factions, and distribution [
39
]
could determine the eciency of the derivatives in vivo.
Int. J. Mol. Sci. 2020,21, 2132 9 of 15
4. Materials and Methods
4.1. General Synthetic Procedure
The amino acid component of compounds
1
and
2
were synthesized from
Boc-l-tyrosine-OtBu (Combi-Blocks, San Diego, CA, USA) and Boc-4-nitro-l-phenylalanine
(Combi-Blocks), respectively. Chlorambucil was obtained from TCI (Tokyo, Japan).
N-Ethyl-N
0
-(3-dimethylaminopropyl)carbodiimide (EDC), 4-(dimethylamino)pyridine (DMAP),
and Pd (10% on activated charcoal), potassium carbonate (K
2
CO
3
), dimethyl sulfate ((CH
3
O)
2
SO
2
),
and celite were purchased from Sigma-Aldrich (St. Louis, MO, USA).
Thin-layer chromatography (TLC) with aluminum sheets coated with silica gel 60 F
245
(0.24
mm) (Merck, Darmstadt, Germany) was used with suitable visualization to monitor reactions. Flash
chromatography with the Sepacore Flash system, coupled with Sepacore flash cartridge (Buchi AG,
Flawil, Switzerland), was used for purification. Nuclear magnetic resonance (NMR) spectra were
recorded by Bruker Avance 500 spectrometer (Bruker Biospin, Fällanden, Switzerland), operating at
500.13 MHz for
1
H and at 125.75 MHz for
13
C using tetramethylsilane (TMS) as an internal standard.
Mass spectra were characterized using a Finnigan LCQ quadrupole ion trap mass spectrometer
(Finnigan MAT, San Jose, CA, USA) coupled with an electrospray ionization source. Purity of final
products was confirmed by quantitative
1
H NMR (qHNMR) using maleic acid as an internal standard.
Boc-4-amino-l-phenylalanine methyl ester 6:
Boc-4-nitro-l-phenylalanine
4
(400 mg, 1.28 mmol)
was dissolved in 10 mL acetone after which K
2
CO
3
(884 mg, 6.4 mmol) and (CH
3
O)
2
SO
2
(242
µ
L, 2.56
mmol) were added. The reaction was stirred at room temperature overnight. The reaction mixture was
subsequently evaporated under reduced pressure to get a dry residue of Boc-4-nitro-l-phenylalanine
methyl ester (378 mg).
1
H NMR (MeOD):
δ
ppm 1.38 (s, 9H), 3.03–3.07 (m, 1H), 3.27–3.31 (m, 1H), 3.74
(s, 3H), 4.46 (dd, J
1
=5.3 Hz, J
2
=9.2 Hz), 7.49 (d, J=8.4 Hz, 2H), 8.18 (d, J=8.4 Hz, 2H). Pd (10% on
activated charcoal) (10 mg) was carefully added to the solution of Boc-4-nitro-l-phenylalanine methyl
ester (378 mg, 0.58 mmol) in methanol (30 mL). The reaction was stirred in a H
2
-saturated atmosphere
(344.7 kPa (50 psi)) overnight. The mixture was then filtered through celite and evaporated under
reduced pressure. The residue was purified by flash chromatography in gradient mode (petroleum
ether/ethyl acetate=1:99 to 99:1) to get compound
6
(165 mg, 43%), a brown solid.
1
H NMR (MeOD):
δ
ppm 1.41 (s, 9H), 2.79–2.83 (m, 1H), 2.94–2.98 (m, 1H), 3.69 (s, 3H), 4.27–4.30 (m, 1H), 6.67–6.69
(d, J=8.2 Hz, 2H), 6.94–6.96 (d, J=8.2 Hz. 2H).
Chlorambucil (Boc-OtBu-phenylalaninate) ester 7:
Chlorambucil
3
(200 mg, 0.66 mmol) was
dissolved in 5 mL dichloromethane (DCM) with DMAP (154 mg, 0.8 mmol) and Boc-l-tyrosine-OtBu
5
(270 mg, 0.8 mmol). EDC (98 mg, 0.8 mmol) in 5 mL DCM was then added dropwise into the mixture
and the reaction stirred at room temperature overnight. Ethyl acetate (40 mL) was subsequently
added to the reaction mixture. The combined solvent was washed with 1 M hydrochloric acid (HCl)
(10 mL
×
2), saturated sodium bicarbonate (NaHCO
3
) (10 mL
×
2), and H
2
O (10 mL), respectively.
The organic solvent was dried over anhydrous sodium sulfate (Na
2
SO
4
), filtrated, and evaporated
under reduced pressure. The reaction yielded compound
7
(270 mg, 88%), an o-white solid.
1
H NMR
(CDCl
3
):
δ
ppm 1.40 (s, 9H), 1.43 (s, 9H), 2.03 (quin, J=7.5 Hz, 2H), 2.55 (t, J=7.5 Hz, 2H), 2.64 (t,
J=7.5 Hz, 2H), 3.04 (d, J=5.7 Hz, 2H), 3.61-3.72 (m, 8H), 4.43 (m, 1H), 5.00 (d, J=8.5 Hz, 1H), 6.65
(d, J=8.4 Hz, 2H), 6.99 (d, J=8.4 Hz, 2H), 7.11 (d, J=8.4 Hz, 2H), 7.17 (d, J=8.4 Hz, 2H).
13
C NMR
(CDCl
3
):
δ
ppm 26.73 (1
×
C), 27.96 (3
×
C), 28.33 (3
×
C), 33.67 (1
×
C), 33.94 (1
×
C), 40.49 (2
×
C), 53.66
(2
×
C), 54.80 (1
×
C), 79.75 (1
×
C), 82.20 (1
×
C), 112,29 (2
×
C), 121.39 (2
×
C), 129.77 (2
×
C), 130.47 (2
×
C),
134.07 (1×C), 144.37 (1×C), 149.54 (1×C), 170.80 (1×C), 172.01 (1×C).
Chlorambucil (Boc-methyl-phenylalaninate) amide 8:
Chlorambucil
3
(100 mg, 0.33 mmol) was
dissolved in 5 mL DCM with DMAP (77 mg, 0.4 mmol) and compound
6
(130 mg, 0.4 mmol). EDC
(49 mg, 0.4 mmol) in 5 mL DCM was then added dropwise into the mixture and the reaction stirred
at room temperature overnight. Ethyl acetate (40 mL) was then added into the reaction mixture.
The combined solvent was washed with 1 M HCl (10 mL
×
2), saturated NaHCO
3
(10 mL
×
2), and H
2
O
Int. J. Mol. Sci. 2020,21, 2132 10 of 15
(10 mL), respectively. The organic solvent was dried over anhydrous Na
2
SO
4
, filtrated, and evaporated
under reduced pressure. The reaction yielded compound
8
(118 mg, 77%), a brown solid.
1
H NMR
(CDCl
3
):
δ
ppm 1.42 (s, 9H), 2.02 (quin, J=7.3 Hz, 2H), 2.33 (t, J=7.5 Hz, 2H), 2.62 (t, J=7.3 Hz, 2H),
3.00–3.11 (m, 2H), 3.61–3.72 (m, 11H), 4.55 (m, 1H), 4.95 (d, J=7.7 Hz, 1H), 6.63 (d, J=8.5 Hz, 2H),
7.04–7.10 (m, 4H), 7.42 (d, J=8.5 Hz, 2H).
13
C NMR (MeOD):
δ
ppm 19.44 (1
×
C), 27.25 (3
×
C), 33.34
(1
×
C), 35.91 (1
×
C), 40.29 (2
×
C), 51.19 (1
×
C), 53.17 (2
×
C), 60.12 (1
×
C), 79.23 (1
×
C), 112,16 (2
×
C), 119.83
(2
×
C), 129.17 (2
×
C), 129.26 (2
×
C), 130.31 (1
×
C), 132.62 (1
×
C), 144.62 (1
×
C), 172.78 (1
×
C), 173.00 (1
×
C).
Chlorambucil phenylalanine ester 1:
Compound
7
(255 mg, 0.55 mmol) was dissolved in DCM
(4 mL) and trifluoroacetic acid (2 mL) was added dropwise. The reaction was stirred for 6 h at room
temperature and the solvent evaporated. The residue was purified by flash chromatography in gradient
mode (DCM/MeOH=1:99 to 99:1) to get compound
1
, an o-white solid in trifluoroacetic salt (2X)
form (134 mg, 35%).
1
H NMR (MeOD):
δ
ppm 2.00 (quin, J=7.2 Hz, 2H), 2.58 (t, J=7.3 Hz, 2H),
2.64 (t, J=7.4 Hz, 2H), 3.10–3.14 (dd, J
1
=8.2 Hz, J
2
=14.8 Hz, 1H), 3.32–3.34 (m, 1H), 3.65–3.75 (m,
8H), 4.08 (dd, J
1
=8.2 Hz, J
2
=5.1 Hz, 1H), 6.71 (d, J=8.7 Hz, 2H), 7.06 (d, J=8.5 Hz, 2H), 7.11 (d,
J=8.6 Hz, 2H), 7.17 (d, J=8.5 Hz, 2H).
13
C NMR (MeOD):
δ
ppm 26.43, 32.88, 33.55, 35.58, 40.30 (2
×
C),
53.17 (2
×
C), 54.69, 112.17 (2
×
C), 122.01 (2
×
C), 129.30 (2
×
C), 129.99, 130.22 (2
×
C), 132.61, 144.75, 150.34,
170.72, 172.80. MS: calcd. for C23 H28Cl2N2O4466.14 [M]+, found 467.12 [M+H]+, 97% purity.
Chlorambucil phenylalanine amide 2:
Compound
8
(100 mg, 0.21 mmol) was dissolved in
tetrahydrofuran (5 mL) and lithium hydroxide (20 mg, 0.8 mmol) in H
2
O (5 mL) added dropwise.
The reaction was stirred for 1.5 h and the solvent evaporated. The intermediate compound was then
dissolved in DCM (4 mL) and trifluoroacetic acid (2 mL) was added dropwise. The reaction was
stirred for 2 h at room temperature and the solvent evaporated. The residue was purified by flash
chromatography in gradient mode (DCM/MeOH=1:99 to 99:1) and recrystallized using MeOH to get
compound
2
, a light-brown solid in trifluoroacetic salt (2X) form (24 mg, 16%).
1
H NMR (MeOD):
δ
ppm 1.98 (quin, J=7.4 Hz, 2H), 2.38 (t, J=7.5 Hz, 2H), 2.61 (t, J=7.4 Hz, 2H), 2.98–3.02 (dd, J
1
=14.4
Hz, J
2
=8.8 Hz, 1H), 3.28–3.33 (dd, J
1
=4.1 Hz, J
2
=14.9 Hz, 1H), 3.65–3.75 (m, 8H), 3.77–3.79 (dd,
J
1
=4.1 Hz, J
2
=8.4 Hz, 1H), 6.70 (d, J=8.6 Hz, 2H), 7.10 (d, J=8.6 Hz, 2H), 7.27 (d, J=8.4 Hz, 2H), 7.52
(d, J=8.4 Hz, 2H).
13
C NMR (MeOD):
δ
ppm 27.34, 33.82, 35.86, 36.24, 40.28 (2
×
C), 53.17 (2
×
C), 56.10,
112.17 (2
×
C), 120.48 (2
×
C), 129.24 (2
×
C), 129.39 (2
×
C), 130.28, 131.43, 137.72, 144.65, 172.28, 173.19. MS:
calcd. for C23H29Cl2N3O3465.16 [M]+, found 466.09 [M+H]+, 95% purity.
4.2. HPLC Analysis
The analytical and biological samples were injected into a reversed-phase C18 analytical column
Zorbax SB-C18 (3
×
150 mm, 3.5
µ
m) coupled with guard-column Zorbax SB-C18 (Agilent Technologies,
Little Falls Wilmington, DE, USA). The solutions were eluted in isocratic mode with 50% acetonitrile
and deionized water (with 0.1% formic acid) as eluents at a flow rate of 0.8 mL/min. The sample
peaks were monitored by Agilent 1100 series HPLC (Agilent Technologies, Waldbronn, Karlsruhe,
Germany) with UV detection at 254 nm. The retention time of chlorambucil derivatives (
1
and
2
) and
chlorambucil (3) were 2.4, 1.6, and 5.0 min, respectively.
4.3. In Vitro Conversion
The rates of chemical conversion of compounds
1
and
2
were studied in aqueous phosphate
buer solutions (50 mM, pH 7.4) at 37
C. The solutions of the chlorambucil derivatives were prepared
by adding 80
µ
L of stock solution (0.5 mg/mL in 5% HP-
β
-cyclodextrin (Carvasol, Wacker Chemie,
Munich, German) in phosphate buer) to 720
µ
L of the preheated buer. At predetermined time
intervals, aliquots (50
µ
L) were sampled, mixed with MeOH (50
µ
L), and analyzed for the remaining
compound by HPLC.
Compounds
1
and
2
were incubated with the human liver microsomes as an indication of the
susceptibility to bioconversion in the liver. The reactions were initiated by adding 3.0
µ
L of compounds
(10 mM in DMSO) to a preheated solution consisting of 747
µ
L phosphate buer (20 mM, pH 7.4),
Int. J. Mol. Sci. 2020,21, 2132 11 of 15
200
µ
L NADPH (1 mM), and 50
µ
L human liver microsomes (20 mg/mL). The mixture was incubated
thermostatically at 37
C, and the 50
µ
L aliquots were withdrawn at predetermined time intervals.
The aliquots were then immediately mixed with 100
µ
L of ice cold MeOH and centrifuged at 14,000
×
g
for 5 min to precipitate proteins. The clear supernatants were analyzed for the remaining compound
by HPLC. The respective pseudo-first-order half-lives (T
1/2
) for the conversion rate of the compounds
were calculated from the slope of the linear portion of the remaining compound logarithm plots
against time.
4.4. Cell Line and Culture Condition
The human breast adenocarcinoma cell line MCF-7 was purchased from the American Type
Culture Collection (ATCC#HTB-22) (Manassas, VA, USA). The MCF-7 cells were cultured in Dulbecco’s
Modified Eagle Medium (DMEM) supplemented with 2 mM l-glutamine, 10% fetal bovine serum
(FBS), 50 units/mL penicillin, and 50% streptomycin. The cell line was incubated in a humidified
incubator at 37
C with 5% CO
2
, and the cells were used after reaching approximately 80% confluency.
4.5. LAT1 Binding Anity
The expression and function of LAT1 and LAT2 in MCF-7 cells was characterized in our recent
study [
26
]. The study indicated that LAT1 is predominant in both expression and function compared to
LAT2, implying its role as a major transporter responsible for the uptake of LAT1 substrates. The ability
of compounds to bind to LAT1 was determined as previously described [
40
]. In brief, MCF-7 cells
were seeded into collagen-coated, 24-well plates at a density of 1.5
×
10
5
cells/well. The seeded
cells were incubated overnight in the incubator before the anity experiment. The cells were then
pre-incubated in 500
µ
L of warm HBSS (Hank’s balance salt solution) for 10 min. After the removal
of HBSS, the cells were incubated for 5 min in 250
µ
L of 5% HP-
β
-cyclodextrin in HBSS, containing
0.157
µ
M of [
14
C]-l-leucine (PerkinElmer, Waltham, MA, USA) and 10
µ
M of compound: chlorambucil,
its derivatives (
1
and
2
), or l-tyrosine (Fluka, Buchs, Switzerland). The cells were washed twice
with ice-cold HBSS (500
µ
L) on an ice bath and lysed by 250
µ
L of 0.1 M sodium hydroxide (NaOH)
for 1 h at room temperature. The lysate was then mixed with 1 mL of an emulsifier-safe cocktail
(PerkinElmer) and the radioactivity recorded by liquid scintillation counter (Wallac 1450 MicroBeta,
Wallac Oy, Finland).
4.6. In Vitro Cellular Uptake
Determination of the intracellular concentration of compounds was performed following a
previous report [
25
]. Briefly, MCF-7 cells were seeded and treated as in LAT1 anity studies.
The uptake of chlorambucil and its derivatives was studied in 250
µ
L of 5% HP-
β
-cyclodextrin in
HBSS by incubating the MCF-7 cells for varying incubation times (5–45 min, 20
µ
M) or concentrations
(10–120
µ
M, 15 min). The cells were washed twice with ice-cold HBSS (500
µ
L) in an ice bath, then lysed
by 1% perchloric acid (250
µ
L) for 0.5 h at room temperature. The suspension was removed to an
Eppendorf tube and centrifuged at 14,000
×
g, 4
C, for 5 min. The supernatant (200
µ
L) was analyzed
by HPLC. The respective standard curve for the intracellular concentration of compounds (
1
,
2
, or
3
) was performed by spiking known amounts of the compounds into cell lysate of untreated cells.
For protein determination, the cells were lysed using 250
µ
L of 0.1 M NaOH for 0.5 h instead of 1%
perchloric acid and were determined using Bio-Rad protein assay (Bio-Rad, CA, USA) as described by
the manufacturer, using serum albumin (Sigma-Aldrich) as the standard. The protein concentrations
in samples were recorded using an EnVision multimode plate reader (PerkinElmer). The final results
were expressed as the intracellular concentration normalized by its protein content.
4.7. Antiproliferative Activity
Cell cytotoxicity was assessed using MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium
bromide) assay as previously described [
41
]. MCF-7 cells were seeded at a density of
6×103cells/well
Int. J. Mol. Sci. 2020,21, 2132 12 of 15
into a 96-well plate and incubated overnight in an incubator. Thereafter, chlorambucil and its
derivatives were added and incubated for 24, 48, and 72 h with a final concentration ranging from 10
to 120
µ
M. At predetermined times, MTT (Amresco, Solon, OH, USA) solution, in phosphate buer
saline, was added to each well to achieve a final concentration of 0.5 mg/mL. The cells were then
incubated for 3 h in an incubator before discarding the media, after which 100
µ
L of DMSO were
added to each well to solubilize the formazan. The absorbance of solubilized formazan was measured
at 570 nm (reference wavelength: 650 nm) using an EnSight multimode plate reader (PerkinElmer).
BCH (2-aminobicyclo-[2,2,1]-heptane-2-carboxylic acid) (Sigma-Aldrich) was used as a standard LAT1
inhibitor in the co-incubation assay. DMSO in 5% HP-
β
-cyclodextrin was used as the dispersing agent,
which had no cytotoxicity at the respective concentrations at any of the predetermined time points.
4.8. Computational Analysis
The chemical structures were analyzed using CS ChemBio3D Ultra (version 12.0; Cambridge
Soft Corporation, Waltham, MA, USA). For each molecule, energy minimization was performed by
molecular mechanics (MM2) with a root-mean-square of 0.001. The property server was used to
compute the log P, molecular weight, molecular accessible area (Å
2
) (as per the Connolly method),
and polar surface area (PSA) (Å2).
4.9. Statistical Analysis
The data were analyzed for normality of distribution (Shapiro–Wilk test) and homogeneity of
variance (Levene’s test) (p>0.05). Dierences among treatments were assessed using a one-way
ANOVA followed by a Tukey’s multiple comparison post hoc tests using SPSS 19.0 software for
Windows
®
(SPSS Inc, Chicago, IL, USA). Respective data were presented as a mean
±
SD and any
dierences with a pvalue <0.05 were considered statistically significant.
5. Conclusions
Structural modifications of a chemotherapeutic drug by conjugating it to LAT1 substrate can
increase its eectiveness against cancer. A synthetic method for conjugating chlorambucil and tyrosine
via either an ester or amide bond was developed to obtain derivatives
1
and
2
. The MCF-7 cell line was
used as a cancer cell model to determine binding ability, intracellular uptake, and antiproliferative
activity
in vitro
. The chlorambucil derivatives bound to LAT1 in a manner comparable to l-tyrosine.
The respective intracellular concentration after treatment with derivatives
1
and
2
was higher and their
respective antiproliferative activity had a higher cytotoxicity than that of chlorambucil. By exploiting
LAT1 as a carrier-mediated transporter, derivatives
1
and
2
eciently permeated the cell membrane,
accumulated, and exerted their cytotoxicity
in vitro
more eciently than did chlorambucil. Our results
support rational drug design to elevate the intracellular drug level using a specific transporter as well
as enhancing its chemotherapeutic eciency.
Author Contributions:
Conceptualization, P.P., N.W., J.R; methodology, P.P., J.T., J.J., J.L., J.K., J.R., N.W.; formal
analysis, P.P., J.T., J.J., J.L., J.K., J.R., N.W.; investigation, P.P.; validation, P.P., J.T., J.J., J.L., J.K., J.R., N.W.;
resources, N.W., J.R.; data curation, P.P., J.R., N.W.; visualization, P.P., J.R., N.W.; writing—original draft, P.P., N.W.;
writing—review and editing, P.P., J.R., N.W.; funding acquisition, N.W., J.R.; project administration, N.W., J.R.;
All authors have read and agreed to the published version of the manuscript.
Funding:
This work has received funding under the Post-Doctoral Training Program (Grant No. PD2563-09)
from Khon Kaen University, Thailand, for financial support to P.P. and N.W., and the Academy of Finland
(Grant No. 308329) for financial support to J.R.
Acknowledgments:
The authors thank Tarja Ihalainen for technical assistance with the
in vitro
conversion studies
and Bryan Roderick Hamman for assistance with the English-language presentation.
Conflicts of Interest: The authors declare no conflict of interest.
Int. J. Mol. Sci. 2020,21, 2132 13 of 15
References
1.
Chien, H.C.; Colas, C.; Finke, K.; Springer, S.; Stoner, L.; Zur, A.A.; Venteicher, B.; Campbell, J.; Hall, C.;
Flint, A.; et al. Reevaluating the substrate specificity of the L-type amino acid transporter (LAT1). J. Med.
Chem. 2018,61, 7358–7373. [CrossRef] [PubMed]
2.
Peura, L.; Malmioja, K.; Laine, K.; Leppanen, J.; Gynther, M.; Isotalo, A.; Rautio, J. Large amino acid
transporter 1 (LAT1) prodrugs of valproic acid: New prodrug design ideas for central nervous system
delivery. Mol. Pharm. 2011,8, 1857–1866. [CrossRef] [PubMed]
3.
Rautio, J.; Gynther, M.; Laine, K. LAT1-mediated prodrug uptake: A way to breach the blood-brain barrier?
Ther. Deliv. 2013,4, 281–284. [CrossRef] [PubMed]
4.
Gynther, M.; Peura, L.; Vernerova, M.; Leppanen, J.; Karkkainen, J.; Lehtonen, M.; Rautio, J.; Huttunen, K.M.
Amino acid promoieties alter valproic acid pharmacokinetics and enable extended brain exposure. Neurochem.
Res. 2016,41, 2797–2809. [CrossRef]
5.
Zhao, Y.; Wang, L.; Pan, J. The role of L-type amino acid transporter 1 in human tumors. Intractable Rare Dis.
Res. 2015,4, 165–169. [CrossRef]
6.
Hayashi, K.; Anzai, N. Novel therapeutic approaches targeting L-type amino acid transporters for cancer
treatment. World J. Gastrointest. Oncol. 2017,9, 21–29. [CrossRef]
7.
Häfliger, P.; Charles, R.P. The L-type amino acid transporter LAT1—An emerging target in cancer. Int. J. Mol.
Sci. 2019,20, 2428.
8.
Oda, K.; Hosoda, N.; Endo, H.; Saito, K.; Tsujihara, K.; Yamamura, M.; Sakata, T.; Anzai, N.; Wempe, M.F.;
Kanai, Y.; et al. L-type amino acid transporter 1 inhibitors inhibit tumor cell growth. Cancer Sci.
2010
,101,
173–179. [CrossRef]
9.
Huttunen, K.M.; Gynther, M.; Huttunen, J.; Puris, E.; Spicer, J.A.; Denny, W.A. A selective and slowly
reversible inhibitor of L-Type amino acid transporter 1 (LAT1) potentiates antiproliferative drug ecacy in
cancer cells. J. Med. Chem. 2016,59, 5740–5751. [CrossRef]
10.
Singh, N.; Scalise, M.; Galluccio, M.; Wieder, M.; Seidel, T.; Langer, T.; Indiveri, C.; Ecker, G.F. Discovery of
potent inhibitors for the large neutral amino acid transporter 1 (LAT1) by structure-based methods. Int. J.
Mol. Sci. 2019,20, 27. [CrossRef]
11.
Okano, N.; Kawai, K.; Yamauchi, Y.; Kobayashi, T.; Naruge, D.; Nagashima, F.; Endou, H.; Furuse, J.
First-in-human phaseIstudy of JPH203 in patients with advanced solid tumors. J. Clin. Oncol.
2018
,36, 419.
[CrossRef]
12.
Kaira, K.; Sunose, Y.; Ohshima, Y.; Ishioka, N.S.; Arakawa, K.; Ogawa, T.; Sunaga, N.; Shimizu, K.;
Tominaga, H.; Oriuchi, N.; et al. Clinical significance of L-type amino acid transporter 1 expression as a
prognostic marker and potential of new targeting therapy in biliary tract cancer. BMC Cancer
2013
,13, 482.
[CrossRef]
13.
Wu, W.; Dong, Y.; Gao, J.; Gong, M.; Zhang, X.; Kong, W.; Li, Y.; Zeng, Y.; Si, D.; Wei, Z.; et al.
Aspartate-modified doxorubicin on its N-terminal increases drug accumulation in LAT1-overexpressing
tumors. Cancer Sci. 2015,106, 747–756. [CrossRef] [PubMed]
14.
Millard, M.; Gallagher, J.D.; Olenyuk, B.Z.; Neamati, N. A Selective Mitochondrial-Targeted Chlorambucil
with Remarkable Cytotoxicity in Breast and Pancreatic Cancers. J. Med. Chem.
2013
,56, 9170–9179. [CrossRef]
15.
Pathak, R.K.; Wen, R.; Kolishetti, N.; Dhar, S. A prodrug of two approved drugs, cisplatin and chlorambucil,
for chemo war against cancer. Mol. Cancer Ther. 2017,16, 625–635. [CrossRef]
16.
Ylikangas, H.; Malmioja, K.; Peura, L.; Gynther, M.; Nwachukwu, E.O.; Leppänen, J.; Laine, K.; Rautio, J.;
Lahtela-Kakkonen, M.; Huttunen, K.M.; et al. Quantitative insight into the design of compounds recognized
by the L-type amino acid transporter 1 (LAT1). ChemMedChem 2014,9, 2699–2707. [CrossRef] [PubMed]
17.
Kim, C.S.; Cho, S.H.; Chun, H.S.; Lee, S.Y.; Endou, H.; Kanai, Y.; Kim, D.K. BCH, an inhibitor of system L
amino acid transporters, induces apoptosis in cancer cells. Biol. Pham. Bull.
2008
,31, 1096–1100. [CrossRef]
18.
Shennan, D.B.; Thomson, J.; Gow, I.F.; Travers, M.T.; Barber, M.C. L-leucine transport in human breast cancer
cells (MCF-7 and MDA-MB-231): Kinetics, regulation by estrogen and molecular identity of the transporter.
BBA Biomembr. 2004,1664, 206–216. [CrossRef]
19.
Van Geldermalsen, M.; Quek, L.E.; Turner, N.; Freidman, N.; Pang, A.; Guan, Y.F.; Krycer, J.R.; Ryan, R.;
Wang, Q.; Holst, J. Benzylserine inhibits breast cancer cell growth by disrupting intracellular amino acid
homeostasis and triggering amino acid response pathways. BMC Cancer 2018,18, 689. [CrossRef]
Int. J. Mol. Sci. 2020,21, 2132 14 of 15
20.
Ylikangas, H.; Peura, L.; Malmioja, K.; Leppanen, J.; Laine, K.; Poso, A.; Lahtela-Kakkonen, M.; Rautio, J.
Structure-activity relationship study of compounds binding to large amino acid transporter 1 (LAT1) based
on pharmacophore modeling and in situ rat brain perfusion. Eur. J. Pharm. Sci.
2013
,48, 523–531. [CrossRef]
21.
Nagamori, S.; Wiriyasermkul, P.; Okuda, S.; Kojima, N.; Hari, Y.; Kiyonaka, S.; Mori, Y.; Tominaga, H.;
Ohgaki, R.; Kanai, Y. Structure-activity relations of leucine derivatives reveal critical moieties for cellular
uptake and activation of mTORC1-mediated signaling. Amino Acids.
2016
,48, 1045–1058. [CrossRef]
[PubMed]
22.
Uchino, H.; Kanai, Y.; Kim, D.K.; Wempe, M.F.; Chairoungdua, A.; Morimoto, E.; Anders, M.W.; Endou, H.
Transport of amino acid-related compounds mediated by L-type amino acid transporter 1 (LAT1): Insights
into the mechanisms of substrate recognition. Mol. Pharmacol. 2002,61, 729–737. [CrossRef] [PubMed]
23.
Rautio, J.; Kumpulainen, H.; Heimbach, T.; Oliyai, R.; Oh, D.; Jarvinen, T.; Savolainen, J. Prodrugs: Design
and clinical applications. Nat. Rev. Drug Discov. 2008,7, 255–270. [CrossRef] [PubMed]
24.
Gynther, M.; Pickering, D.S.; Spicer, J.A.; Denny, W.A.; Huttunen, K.M. Systemic and brain pharmacokinetics
of perforin inhibitor prodrugs. Mol. Pharm. 2016,13, 2484–2491. [CrossRef] [PubMed]
25.
Huttunen, K.M.; Huttunen, J.; Aufderhaar, I.; Gynther, M.; Denny, W.A.; Spicer, J.A. L-type amino acid
transporter 1 (LAT1)-mediated targeted delivery of perforin inhibitors. Int. J. Pharm.
2016
,498, 205–216.
[CrossRef] [PubMed]
26.
Kärkkäinen, J.; Gynther, M.; Kokkola, T.; Petsalo, A.; Auriola, S.; Lahtela-Kakkonen, M.; Laine, K.; Rautio, J.;
Huttunen, K.M. Structural properties for selective and ecient l-type amino acid transporter 1 (LAT1)
mediated cellular uptake. Int. J. Pharm. 2018,544, 91–99. [CrossRef] [PubMed]
27.
Augustyn, E.; Finke, K.; Zur, A.A.; Hansen, L.; Heeren, N.; Chien, H.C.; Lin, L.; Giacomini, K.M.; Colas, C.;
Schlessinger, A.; et al. LAT-1 activity of meta-substituted phenylalanine and tyrosine analogs. Bioorg. Med.
Chem. Lett. 2016,26, 2616–2621. [CrossRef]
28.
Hou, T.; Wang, J.; Zhang, W.; Wang, W.; Xu, X. Recent advances in computational prediction of drug
absorption and permeability in drug discovery. Curr. Med. Chem. 2006,13, 2653–2667. [CrossRef]
29.
Palm, K.; Luthman, K.; Ungell, A.L.; Strandlund, G.; Artursson, P. Correlation of drug absorption with
molecular surface properties. J. Pharm. Sci. 1996,85, 32–39. [CrossRef]
30.
Hill, B.T. Studies on the transport and cellular distribution of chlorambucil in the Yoshida ascites sarcoma.
Biochem. Pharmacol. 1972,21, 495–502. [CrossRef]
31.
Adair, C.G.; McElnay, J.C. Studies on the mechanism of gastrointestinal absorption of melphalan and
chlorambucil. Cancer Chemother. Pharmacol. 1986,17, 95–98. [CrossRef] [PubMed]
32.
Giraud, I.; Rapp, M.; Maurizis, J.C.; Madelmont, J.C. Synthesis and
in vitro
evaluation of quaternary
ammonium derivatives of chlorambucil and melphalan, anticancer drugs designed for the chemotherapy of
chondrosarcoma. J. Med. Chem. 2002,45, 2116–2119. [CrossRef] [PubMed]
33.
Singh, R.K.; Kumar, S.; Prasad, D.N.; Bhardwsaj, T.R. Therapeutic journery of nitrogen mustard as alkylating
anticancer agents: Historic to future perspectives. Eur. J. Med. Chem.
2018
,151, 401–433. [CrossRef]
[PubMed]
34.
Reda, G.; Fattizzo, B.; Cassin, R.; Orofino, N.; Flospergher, E.; Iurlo, A.; Barcellini, W.; Cortelezzi, A. Secondary
malignancies in chronic lymphocytic leukemia: A single centre retrospective analysis of 514 cases. Blood
2015,126, 5279. [CrossRef]
35.
Wang, Q.; Holst, J. L-type amino acid transport and cancer: Targeting the mTORC1 pathway to inhibit
neoplasia. Am. J. Cancer Res. 2015,5, 1281–1294.
36.
Kinne, A.; Schülein, R.; Krause, G. Primary and secondary thyroid hormone transporters. Thyroid Res.
2011
,
4, S7. [CrossRef]
37.
Kim, D.K.; Kanai, Y.; Chairoungdua, A.; Matsuo, H.; Cha, S.H.; Endou, H. Expression cloning of a
Na+-independent aromatic amino acid transporter with structural similarity to H+/monocarboxylate
transporters. J. Biol. Chem. 2001,276, 17221–17228. [CrossRef]
38.
del Amo, E.M.; Urtti, A.; Yliperttula, M. Pharmacokinetic role of L-type amino acid transporters LAT1 and
LAT2. Eur. J. Pharm. Sci. 2008,35, 161–174. [CrossRef]
39.
Puris, E.; Gynther, M.; Huttunen, J.; Petsalo, A.; Huttunen, K.M. L-type amino acid transporter 1 utilizing
prodrugs: How to achieve eective brain delivery and low systemic exposure of drugs. J. Control. Release
2017,261, 93–104. [CrossRef]
Int. J. Mol. Sci. 2020,21, 2132 15 of 15
40.
Rautio, J.; Kärkkäinen, J.; Huttunen, K.M.; Gynther, M. Amino acid ester prodrugs conjugated to the
α
-carboxylic acid group do not display anity for the L-type amino acid transporter 1 (LAT1). Eur. J. Pharm.
Sci. 2015,66, 36–40. [CrossRef]
41.
Pocasap, P.; Weerapreeyakul, N.; Barusrux, S. Cancer preventive eect of Thai rat-tailed radish (Raphanus
sativus L var. caudatus Alef). J. Funct. Foods 2013,5, 1372–1381. [CrossRef]
©
2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... The location of the phenol hydroxyl group was essential in the development of novel Chlorambucil-tyrosine hybrid drugs. Thus, meta-, ortho-, and para-substitutions were explored during the synthesis of these analogues by Brasseur and Descôteaux [30]. Figure 6) [33]. The antiproliferative activity of these compounds was eval on MCF-7 breast cancer cell lines [33]. ...
... Thus, meta-, ortho-, and para-substitutions were explored during the synthesis of these analogues by Brasseur and Descôteaux [30]. Figure 6) [33]. The antiproliferative activity of these compounds was eval on MCF-7 breast cancer cell lines [33]. Their antiproliferative activity was time-conc tion-dependent, with compounds 11a and 11b exhibiting higher antiproliferative a and cell viability than Chlorambucil [33]. ...
... The antiproliferative activity of these compounds was eval on MCF-7 breast cancer cell lines [33]. Their antiproliferative activity was time-conc tion-dependent, with compounds 11a and 11b exhibiting higher antiproliferative a and cell viability than Chlorambucil [33]. The presence of a free carboxylic and a group, an ester/amide linkage, and an aromatic side chain were responsible for th proved chemotherapeutic effect of the compounds [33]. ...
Article
Full-text available
Increasing cases of cancer have been a primary concern in recent decades. Developing new chemotherapeutics is challenging and has been faced with limitations, such as multidrug resistance, poor specificity, selectivity, and toxicity. The aforementioned factors contribute to treatment failure. Hybrid compounds have features that can overcome the limitations mentioned above. Chlorambucil, an anticancer drug that is used to treat prostate and breast cancer, suffers from poor aqueous solubility and specificity, a short half-life, and severe side effects, including anaemia and bone marrow suppression. It compromises the immune system, resulting in treatment failure. Hence, its combination with other pharmacophores has been reported to result in effective anticancer agents with fewer side effects and high therapeutic outcomes. Furthermore, this review gives an update (2010 to date) on the developments of chlorambucil hybrid compounds with anticancer activity, and the structure-activity relationship (SAR), and also highlights future strategies for developing novel anticancer agents.
... Melphalan and acivicin are chemotherapeutic agents known to be transported via the LAT1 protein [20]. Chlorambucil conjugated with L-tyrosine is a successful example of prodrug development that targeted LAT1 to increase intracellular uptake and selectively transport it into cancer cells [23]. Thus, LAT1-mediated prodrug delivery is a promising mechanism to enhance both intracellular uptake and the antiproliferation effect. ...
... LAT1-targeted prodrugs can be formed using hydrolysable bonds, such as ester, carbamate, and amide bonds [23][24][25]. Compared to the ester bond, the amide bond is more stable for chlorambucil conjugated with L-tyrosine [23]. ...
... LAT1-targeted prodrugs can be formed using hydrolysable bonds, such as ester, carbamate, and amide bonds [23][24][25]. Compared to the ester bond, the amide bond is more stable for chlorambucil conjugated with L-tyrosine [23]. Carbamate bonds (urethanes) have also provided good chemical stability for prodrug molecules [26]. ...
Article
Full-text available
Sesamol is a compound reported to have anti-melanogenesis and anti-melanoma actions. Sesamol, however, has low intracellular drug concentration and fast excretion, which can limit its benefits in the clinic. To overcome this drawback and increase intracellular delivery of sesamol into the target melanoma, research has focused on L-type amino acid transporter 1 (LAT1)-mediated prodrug delivery into melanoma cells. The sesamol prodrug was designed by conjugating sesamol with L-phenylalanine at the para position with a carbamate bond. LAT1 targeting was evaluated vis-à-vis a competitive [14C]-leucine uptake inhibition. The sesamol prodrug has a higher [14C]-leucine uptake inhibition than sesamol in human LAT1-transfected HEK293 cells. Moreover, the sesamol prodrug was taken up by LAT1-mediated transport into SK-MEL-2 cells more effectively than sesamol. The sesamol prodrug underwent complete hydrolysis, releasing the active sesamol at 72 h, which significantly exerted its cytotoxicity (IC50 of 29.3 µM) against SK-MEL-cells more than sesamol alone. Taken together, the strategy for LAT1-mediated prodrug delivery has utility for the selective uptake of sesamol, thereby increasing its intracellular concentration and antiproliferation activity, targeting melanoma SK-MEL-2 cells that overexpress the LAT1 protein. The sesamol prodrug thus warrants further evaluation in an in vivo model.
... The majority of the SLC transporters are not considered to be drug transporters, and thus, for their exploitation for anticancer drug delivery, prodrug and nanoparticle technologies have been developed (Table 2), which are discussed below. L-phenylalanine prodrug of melphalan, threoninederivative of gemcitabine, aspartate derivative of doxorubicin, liposomes of L-Dopa functionalized amphiphile, lysine-conjugated methotrexate, Land D-Dopa conjugated anisotropic gold nanoparticles, lysine-and aromatic amino acid-mustards, αmethyl-L-tyrosine conjugate of Astatine-211, tyrosine-conjugated liposomes, L-tyrosine esterand amide-conjugates of chlorambucil [35,164,[290][291][292][293][294][295][296][297][298][299] SLC1A5 ASCT2 ...
... Moreover, the distribution and efficacy of the developed liposomes was not compared to irinotecan itself. In another study, Ltyrosine ester-and amide-conjugates of chlorambucil were synthetized and demonstrated binding to LAT1 in the human breast cancer MCF-7 cell line, while the distribution in vivo of these derivatives has not been evaluated [294]. ...
Article
Full-text available
Transporter-mediated drug resistance is a major obstacle in anticancer drug delivery and a key reason for cancer drug therapy failure. Membrane solute carrier (SLC) transporters play a crucial role in the cellular uptake of drugs. The expression and function of the SLC transporters can be down-regulated in cancer cells, which limits the uptake of drugs into the tumor cells, resulting in the inefficiency of the drug therapy. In this review, we summarize the current understanding of low-SLC-transporter-expression-mediated drug resistance in different types of cancers. Recent advances in SLC-transporter-targeting strategies include the development of transporter-utilizing prodrugs and nanocarriers and the modulation of SLC transporter expression in cancer cells. These strategies will play an important role in the future development of anticancer drug therapies by enabling the efficient delivery of drugs into cancer cells.
... This mechanism aligns with findings in the referenced study, where the tyrosinechlorambucil conjugate utilized LAT1-mediated transport to achieve higher intracellular drug concentrations in MCF-7 cells, resulting in a stronger cytotoxic effect [71]. For F 2 BGly, the glycine residues appear to play a similar role, potentially facilitating selective membrane transport via amino acid transport proteins and thereby increasing compound concentration within cancer cells. ...
Article
Full-text available
Difluorinated sulfonamide porphyrin (F2PGly) and bacteriochlorin (F2BGly), modified by glycine residues, were synthesized and evaluated for photodynamic therapy (PDT). F₂PGly exhibits superior stability and singlet oxygen generation efficiency but features a low-intensity band in the red range (λmax = 639 nm). In contrast, F2BGly shows a favorable, red-shifted absorption spectrum (λmax = 746 nm) that aligns well with phototherapeutic window, facilitating deeper tissue penetration. Moreover, it demonstrates reasonable photostability, necessary for the efficient generation of both singlet oxygen (type II) and oxygen-centered radicals (type I mechanism) which contributes to enhanced therapeutic efficacy. Importantly, the glycine modifications in F2BGly enhance its uptake in MCF-7 cells, known for their resistance to PDT due to efflux transport proteins like LAT1, showing great potential in the cancer cell-targeted PDT. The glycine groups potentially enable F2BGly to bypass these barriers, resulting in increased intracellular accumulation and more effective Reactive Oxygen Species (ROS) generation under illumination. In vivo studies indicated promising vascular-targeted PDT results, with real-time fluorescence imaging used to monitor photosensitizer distribution prior to irradiation. These findings suggest that F2BGly is a promising photosensitizer candidate with enhanced cancer cell selectivity and photodynamic efficiency, meriting further exploration in targeted PDT applications for multiple types of cancers.
... On the other hand, compounds that act as LAT1 inhibitors bind to LAT1 and block nutrient supply to the cells, leading to cellular death due to nutrient deprivation. In previous studies, co-treatment of compounds with LAT1 inhibitors (BCH) has been used to determine the role of amino acid conjugates on LAT1 activity [48]. Hence, in the present study, THC-di-Phe was coincubated with BCH, an inhibitor of LAT1, to determine the role of THC-di-Phe on LAT1. ...
Article
Full-text available
Glioblastoma, a fatal brain cancer with limited treatments and poor prognosis, could benefit from targeting the L-type amino acid transporter I (LAT1). LAT1 is essential for cancer cells to acquire necessary amino acids. Tetrahydrocurcumin (THC), a key curcumin derivative, shows potential for glioblastoma treatment. However, its effectiveness is hindered by poor physicochemical and pharmacokinetic properties. Therefore, this study aims to improve the therapeutic efficacy of THC against glioblastoma by chemically modifying it to target LAT1. A novel series of THC-amino acid conjugates were synthesized by conjugating five amino acids: glycine, leucine, isoleucine, and phenylalanine to THC via carbamate bonds. The therapeutic efficacy of THC-amino acid conjugates was further examined in C6 glioma cells, including the role of LAT1 in their therapeutic effects. Among the conjugates tested, THC conjugated with two phenylalanines (THC-di-Phe) showed remarkably higher cytotoxicity against C6 glioma cells (35.8 μM) compared to THC alone (110.7 μM). THC-di-Phe induced cellular death via necrosis and apoptosis, outperforming THC. Additionally, THC-di-Phe inhibited C6 cell proliferation and migration more effectively than THC. Co-incubation of THC-di-Phe with the LAT1 inhibitor 2-Aminobicyclo-(2,2,1)-heptane-2-carboxylic acid (BCH) further increased cellular death. THC-di-Phe also significantly inhibited the P70SK/S6 pathway, regulated by LAT1 inhibitors, more effectively than THC and displayed a similar binding mode with both JX-075 and BCH to the active site of LAT1. Findings suggest the potential role of THC-di-Phe as a LAT1 inhibitor and provide novel insight into its use as a potent antitumor agent in glioma with increased therapeutic efficacy.
... Additionally, the cytotoxicity of tyrosine-conjugated chlorambucil is greater than that of chlorambucil alone due to its increased intracellular absorption. L-type amino acid transporter 1 (LAT1), which is overexpressed in a number of cancer types, enhances the antiproliferative activity of the chlorambucil compound and aids in its intracellular uptake [18]. It is overexpressed in a number of cancer types [19]. ...
Article
Full-text available
Natural compounds have demonstrated good biological activity when combined with certain amino acids. For example, a glycine-conjugated glycyrrhetinic acid exhibits heightened efficiency against MCF7 cancer cells. Consequently, a molecular modeling analysis is conducted to construct glycine-conjugated α-mangostins and investigate their potential. According to pharmacophore modeling using the ligand-based drug design technique, only two glycine-conjugated α-mangostins conform to the pharmacophore features. The docking simulation results show that the Am1Gly conjugate can interact with the estrogen receptor-α (ERα) with a binding energy of −10.91 kcal/mol. This interaction is further supported by molecular dynamics simulations performed over a 200 ns timeframe. Based on molecular dynamics modeling using the MMPBSA method, the binding affinity of Am1Gly (ΔGTotal = −48.79 kcal/mol) is determined. The results of this analysis indicate that Am1Gly might function as an antagonist to estrogen receptors.
Article
Full-text available
Tumor cells are known for their ability to proliferate. Nutrients are essential for rapidly growing tumor cells. In particular, essential amino acids are essential for tumor cell growth. Tumor cell growth nutrition requires the regulation of membrane transport proteins. Nutritional processes require amino acid uptake across the cell membrane. Leucine, one of the essential amino acids, has recently been found to be closely associated with cancer, which activate mTOR signaling pathway. The transport of leucine into cells requires an L-type amino acid transporter protein 1, LAT1 (SLC7A5), which requires the 4F2 cell surface antigen heavy chain (4F2hc, SLC3A2) to form a heterodimeric amino acid transporter protein complex. Recent evidence identified 4F2hc as a specific downstream target of the androgen receptor splice variant 7 (AR-V7). We stressed the importance of the LAT1-4F2hc complex as a diagnostic and therapeutic target in urological cancers in this review, which covered the recent achievements in research on the involvement of the LAT1-4F2hc complex in urinary system tumors. In addition, JPH203, which is a selective LAT1 inhibitor, has shown excellent inhibitory effects on the proliferation in a variety of tumor cells. The current phase I clinical trials of JPH203 in patients with biliary tract cancer have also achieved good results, which is the future research direction for LAT1 targeted therapy drugs.
Article
Full-text available
Chronic proliferation is a major hallmark of tumor cells. Rapidly proliferating cancer cells are highly dependent on nutrients in order to duplicate their cell mass during each cell division. In particular, essential amino acids are indispensable for proliferating cancer cells. Their uptake across the cell membrane is tightly controlled by membrane transporters. Among those, the L-type amino acid transporter LAT1 (SLC7A5) has been repeatedly found overexpressed in a vast variety of cancers. In this review, we summarize the most recent advances in our understanding of the role of LAT1 in cancer and highlight preclinical studies and drug developments underlying the potential of LAT1 as therapeutic target.
Article
Full-text available
The large neutral amino acid transporter 1 (LAT1) is a promising anticancer target that is required for the cellular uptake of essential amino acids that serve as building blocks for cancer growth and proliferation. Here, we report a structure-based approach to identify chemically diverse and potent inhibitors of LAT1. First, a homology model of LAT1 that is based on the atomic structures of the prokaryotic homologs was constructed. Molecular docking of nitrogen mustards (NMs) with a wide range of affinity allowed for deriving a common binding mode that could explain the structure−activity relationship pattern in NMs. Subsequently, validated binding hypotheses were subjected to molecular dynamics simulation, which allowed for extracting a set of dynamic pharmacophores. Finally, a library of ~1.1 million molecules was virtually screened against these pharmacophores, followed by docking. Biological testing of the 30 top-ranked hits revealed 13 actives, with the best compound showing an IC50 value in the sub-μM range.
Article
Full-text available
Background: Cancer cells require increased levels of nutrients such as amino acids to sustain their rapid growth. In particular, leucine and glutamine have been shown to be important for growth and proliferation of some breast cancers, and therefore targeting the primary cell-surface transporters that mediate their uptake, L-type amino acid transporter 1 (LAT1) and alanine, serine, cysteine-preferring transporter 2 (ASCT2), is a potential therapeutic strategy. Methods: The ASCT2 inhibitor, benzylserine (BenSer), is also able to block LAT1 activity, thus inhibiting both leucine and glutamine uptake. We therefore aimed to investigate the effects of BenSer in breast cancer cell lines to determine whether combined LAT1 and ASCT2 inhibition could inhibit cell growth and proliferation. Results: BenSer treatment significantly inhibited both leucine and glutamine uptake in MCF-7, HCC1806 and MDA-MB-231 breast cancer cells, causing decreased cell viability and cell cycle progression. These effects were not primarily leucine-mediated, as BenSer was more cytostatic than the LAT family inhibitor, BCH. Oocyte uptake assays with ectopically expressed amino acid transporters identified four additional targets of BenSer, and gas chromatography-mass spectrometry (GCMS) analysis of intracellular amino acid concentrations revealed that this BenSer-mediated inhibition of amino acid uptake was sufficient to disrupt multiple pathways of amino acid metabolism, causing reduced lactate production and activation of an amino acid response (AAR) through activating transcription factor 4 (ATF4). Conclusions: Together these data showed that BenSer blockade inhibited breast cancer cell growth and viability through disruption of intracellular amino acid homeostasis and inhibition of downstream metabolic and growth pathways.
Article
Background: chronic lymphocytic leukemia (CLL) is characterized by progressive immunodeficiency with high prevalence of infections, autoimmune phenomena and secondary malignancies. The immune deregulation may be due to the disease itself or it may be a consequence of the treatment performed. Despite the use of highly effective chemo-immunotherapy, CLL remains incurable nowadays, even if the availability of new drugs is improving life expectancy. Aims: to evaluate the incidence of second cancers in CLL patients, and to investigate their relationship with disease features and therapy lines. Methods: 514 CLL patients diagnosed and followed from 1983 until 2014 at our Institution were retrospectively evaluated. Secondary cancers were categorized according to the originating organ or tissue; skin cancers were divided into melanoma and non-melanoma. History of neoplasia preceding CLL diagnosis was also registered. Results: clinical, hematological and biological characteristics at CLL diagnosis are listed in Table 1. During the follow up 88 patients (17%) developed secondary cancers, with a mean time from diagnosis to secondary neoplasia of 9 years. Considering tumor site, we observed 9 hematological malignancies, 9 lung, 5 breast, 19 uro-genital tract (5 kidney, 10 prostate, 4 bladder, 2 uterus, and 2 ovarian), 15 gastro-enteric tract (12 colon, 2 gastric and 1 tongue), 4 pancreas, 3 melanoma and 15 skin cancers other than melanoma. No significant differences were observed according to age, gender, Rai/Binet stage and hematologic parameters in patients with or without secondary tumors (Table 1). Considering prognostic features, no association was found with 13q deletion, chromosome 12 trisomy, VHIG mutational status, or with ZAP-70 and CD-38 positivity. On the contrary, the development of second cancers was associated with the presence of chromosome 17p (8% with secondary neoplasia versus 6% without, p=0.05) and 11q deletions (13% versus 9%, p=0.08). Medical history was positive for malignancies in 70 patients (13%): 2 hematological malignancies, 3 airways (2 lower and 1 upper), 3 breast, 6 uro-genital tract (3 bladder, 3 prostate, 2 uterus and 1 ovarian), 3 gastro-enteric tract, 3 skin cancers other than melanoma, and 3 melanoma. As regards treatment, 46/88 (52.3%) and 219/426 (51.4%) patients with or without secondary cancers, underwent at least one therapy line. Eighty-six patients were treated with fludarabine containing regimens, of whom 11 developed a secondary cancer; 180 patients received chlorambucil and 34 developed a secondary tumor. Among 65 patients who underwent alemtuzumab treatment, 10 were later diagnosed with a second cancer. During the follow up, 121 patients died, 18 with secondary malignancy. Of note, 41 patients died from CLL progression, 2 from thrombotic events, 11 from infections and 8 from secondary malignancy. Conclusions: secondary malignancies are not infrequent in patients with CLL and their occurrence is not clearly related to biologic markers or to the treatment performed. A careful clinical follow up, encompassing sex and age adjusted tumors screening, is advisable for an early diagnosis and appropriate treatment of secondary malignancies in CLL. Table 1. clinical and laboratory features of 514 CLL patients with or without secondary malignancies. Data are expressed as median (range) or absolute number (%). Secondary malignancies Yes (N=88) No (N=426) Age years 64 (39-80) 63 (31-90) Gender M/F 57/31 250/176 Follow-up years 12 (0-30) 12 (1-25) Rai/Binet<C/III> C/III 85 (97) 3 (3) 401 (94) 25 (6) WBC x10e3/mmc 18.74 (3.6-138) 17.3 (3-384) ALC x10e3/mmc 12.63 (1.8-91.8) 11.9 (1.4-381) Hb g/dL 14 (9-17) 14 (8-18) PLT x10e3/mmc 198 (77-608) 184 (58-472) LDH U/L 325 (144-838) 334 (142-795) Beta2microglobulin mg/L 2 (1-23) 2 (0-10) FISH*Del11qDel13qDel17p+12 6 (13) 19 (40) 4 (8) 6 (13) 23 (9) 101 (40) 15 (6) 38 (15) VHIG unmutated** 15 (42) 80 (39) ZAP-70 positive*** 14 (39) 73 (36) CD-38 positive**** 15 (42) 76 (37) *Tested in 48 and 252 patients respectively. **Tested in 36 and 205 patients respectively. ***Tested in 33 and 182 patients respectively. ****Tested in 56 and 291 patients respectively. Disclosures No relevant conflicts of interest to declare.
Article
419 Background: Uptake of amino acids is essential for cancer growth. L-type amino acid transporter-1 (LAT-1) is overexpressed in various cancers, and uptake of LAT-1 substrate amino acids is known to have a critical role in cancer growth. JPH203 is a novel, selective, LAT-1 inhibitor. A first-in-human phase I study of JPH203 was designed to determine the safety, maximum-tolerated dose (MTD) and recommended dose. This study included evaluation of the anti-tumor effect, pharmacokinetics, and pharmacodynamics of JPH203 and analyzed plasma free amino acids. Methods: JPH203 was administered intravenously for 7 days followed by 21 days’ rest at planned doses ranging from 12 to 110 mg/m ² in patients with advanced solid tumors refractory to standard therapy. Before starting this schedule, we confirmed safety of a single dose of JPH203. Dose-limiting toxicity was evaluated during the first cycle, using a 3+3 design. Results: 17 patients were enrolled from January 2015 to August 2016. One patient was discontinued after a single dose of JPH203 because of tumor progression. Dosage was escalated up to 85 mg/m ² . Grade 3 liver dysfunction occurred in 1 of 6 patients at 60 mg/m ² and in the first patient at 85 mg/m ² . Therefore, it was determined that MTD was 60 mg/m ² . Common treatment-related adverse events were increased ALT/AST, malaise, nausea, hypertension and fever of Grade 1 or 2. Partial response was achieved in a patient with biliary tract cancer (BTC) who continued JPH203 for two years without progression. Disease control (PR+SD) was observed in 3 of 5 patients with BTC and 2 of 6 with colorectal cancer. LAT-1 substrate amino acids and branched chain amino acids including LAT-1 substrate amino acids were higher in patients with BTC than in those with other cancers. All patients with disease control had a body mass index more than the median of 20.5 kg/m ² . In exploratory analysis, longer survival was achieved in patients with high inhibition of uptake of LAT-1 substrate amino acids, compared with patients with low inhibition of uptake. Conclusions: JPH203 was well tolerated, resulting in promise against BTC. This phase I study suggested that LAT-1 could be targeted in treatment for advanced BTC, because LAT-1 substrate amino acids in plasma tended to remain high. Clinical trial information: UMIN000016546.
Article
The L-Type Amino Acid Transporter 1 (LAT1, SLC7A5) transports essential amino acids across the blood-brain barrier (BBB) and into cancer cells. To utilize LAT1 for drug delivery, potent amino acid promoieties are desired, as prodrugs must compete with millimolar concentrations of endogenous amino acids. To better understand ligand-transporter interactions that could improve potency, we developed structural LAT1 models to guide the design of substituted analogs of phenylalanine and histidine. Furthermore, we evaluated the structure-activity relationship (SAR) for both enantiomers of naturally occurring LAT1 substrates. Analogs were tested in cis-inhibition and trans-stimulation cell assays to determine potency and uptake rate. Surprisingly, LAT1 can transport amino acid-like substrates with wide-ranging polarities including those containing ionizable substituents. Additionally, the rate of LAT1 transport was generally non-stereoselective, even though enantiomers likely exhibit different binding modes. Our findings have broad implications to the development of new treatments for brain disorders and cancer.
Article
L-type amino acid transporter 1 (LAT1) is a sodium-independent exchanger transporting large neural amino acids and several amino-acid mimicking drugs across the cell membranes. LAT1 is highly expressed at the blood brain barrier (BBB) and in numerous cancer cells and is therefore a potential drug target. However, structural features affecting the ability to bind to LAT1 and the cellular translocation by LAT1 are unclear. In the present study we determined the binding to and transport through human LAT1 of several compounds into the human breast adenocarcinoma cells (MCF-7). We show that the meta-conjugation of L-phenylalanine increases binding to human LAT1 compared to para-conjugation or aliphatic amino acid moiety. Furthermore, large, rigid and aromatic meta-substituted L-phenylalanine derivative enabled selective and efficient LAT1-mediated cellular uptake. Our results also demonstrates that in addition to binding studies, it is of utmost importance to determine the cellular accumulation of compounds. It provides crucial information on transport efficiency and selectivity of transport mechanisms that the compounds are able to utilize. Overall, these structural findings and the methodology used herein are exploitable to design LAT1-utilizing compounds, such as markers for cancer imaging and drug molecules, enabling more effective and safer treatments for cancer in the future.
Article
Cancer is considered as one of the most serious health problems today. The discovery of nitrogen mustard as an alkylating agent in 1942, opened a new era in the cancer chemotherapy. This valuable class of alkylating agent exerts its biological activity by binding to DNA, cross linking two strands, preventing DNA replication and ultimate cell death. At the molecular level, nitrogen lone pairs of nitrogen mustard generate a strained intermediate "aziridinium ion" which is very reactive towards DNA of tumor cell as well as normal cell resulting in various adverse side effects alogwith therapeutic implications. Over the last 75 years, due to its high reactivity and peripheral cytotoxicity, numerous modifications have been made in the area of nitrogen mustard to improve its efficacy as well as enhancing drug delivery specifically to tumor cells. This review mainly discusses the medicinal chemistry aspects in the development of various classes of nitrogen mustards (mechlorethamine, chlorambucil, melphalan, cyclophosphamide and steroidal based nitrogen mustards). The literature collection includes the historical and the latest developments in these areas. This comprehensive review also attempted to showcase the recent progress in the targeted delivery of nitrogen mustards that includes DNA directed nitrogen mustards, antibody directed enzyme prodrug therapy (ADEPT), gene directed enzyme prodrug therapy (GDEPT), nitrogen mustard activated by glutathione transferase, peptide based nitrogen mustards and CNS targeted nitrogen mustards.
Article
L-type amino acid transporter 1 (LAT1) is selectively expressed in the blood-brain barrier (BBB) and brain parenchyma. This transporter can facilitate brain delivery of neuroprotective agents and additionally give opportunity to minimize systemic exposure. Here, we investigated structure-pharmacokinetics relationship of five newly synthesized LAT1-utilizing prodrugs of the cyclooxygenase inhibitor, ketoprofen, in order to identify beneficial structural features of prodrugs to achieve both targeted brain delivery and low peripheral distribution of the parent drug. Besides, we studied whether pharmacokinetics and bioconversion of LAT1-utilizing prodrugs in vivo can be predicted in early stage experiments. To achieve these goals, we compared the in vitro brain uptake mechanism of prodrugs, rate of BBB permeation of compounds using in situ perfusion technique, their systemic pharmacokinetics and release of parent drug in brain, plasma and liver of mice. The results revealed that both excellent LAT1-binding ability and transporter utilization in vitro can be achieved by conjugating the parent drug to aromatic amino acids such as phenylalanine in comparison to prodrugs with an aliphatic promoiety. The presence of an aromatic promoiety directly conjugated in meta- or para-position to ketoprofen led to LAT1-utilizing prodrugs capable of delivering the parent drug into the brain with higher unbound brain to plasma ratio and reduced liver exposure than with ketoprofen itself. In contrast, the prodrugs with aliphatic promoieties and with an additional carbon attached between the parent drug and phenylalanine aromatic ring did not enhance brain delivery of ketoprofen. Furthermore, we have devised a screening strategy to pinpoint successful candidates at an early stage of development of LAT1-utilizing prodrugs. The screening approach demonstrated that early stage experiments could not replace pharmacokinetic studies in vivo due to the lack of prediction of the intra-brain/systemic distribution of the prodrugs as well as the release of the parent drug. Overall, this study provides essential knowledge required for improvement of targeted brain delivery and reduction of systemic exposure of drugs via the LAT1-mediated prodrug approach.
Article
Cancer cells maintain normal mitochondrial glutathione as one of the defense mechanisms to inhibit mitochondrial membrane polarization and hence apoptosis. A combinational therapeutic modality Platin-Cbl, a prodrug of Food and Drug Administration (FDA) approved chemotherapeutic agents, cisplatin and chlorambucil (Cbl), was synthesized and characterized to explore the potential of this compound to initiate chemo war on cancer cells using the active drugs, cisplatin and Cbl, when delivered to the cellular power house mitochondrion using a targeted nanoparticle (NP) designed to get associated with this organelle. Platin-Cbl demonstrated significantly high cytotoxic activity across a number of tumor cell lines as well as in a cisplatin resistant cancer cell line compared to cisplatin or its mixture with Cbl suggesting its unique potency in cisplatin resistant tumors. A mitochondria targeted NP formulation of Platin-Cbl allowed for its efficacious mitochondrial delivery. In vitro studies documented high potency of Platin-Cbl NP formulations. Cisplatin resistant cells cells upon treatment with Platin-Cbl were still able to manage energy production to a certain extent via fatty acid pathway; the advantage of using T-Platin-Cbl-NP is that this NP treatment causes impairment of all metabolic pathways in cisplatin resistant cells forcing the cells to undergo efficient apoptosis. This study highlights a combination of several beneficial effects for a cascade of events to overcome resistance associated with single drug therapy.