ArticlePDF Available

Human Serum Albumin Conjugated Nanoparticles for pH and Redox-Responsive Delivery of a Prodrug of Cisplatin

Wiley
Chemistry - A European Journal
Authors:

Abstract and Figures

Platinum anticancer drugs are particularly in need of controlled drug delivery because of their severe side effects. Platinum(IV) agents are designed as prodrugs to reduce the side effects of platinum(II) drugs; however, premature reduction could limit the effect as a prodrug. In this work, a highly biocompatible, pH and redox dual-responsive delivery system is prepared by using hybrid nanoparticles of human serum albumin (HSA) and calcium phosphate (CaP) for the Pt(IV) prodrug of cisplatin. This conjugate is very stable under extracellular conditions, so that it protects the platinum(IV) prodrug in HSA. Upon reaching the acidic and hypoxic environment, the platinum drug is released in its active form and is able to bind to the target DNA. The Pt-HSA/CaP hybrid inhibits the proliferation of various cancer cells more efficiently than cisplatin. Different cell cycle arrests suggest different cellular responses of the Pt(IV) prodrug in the CaP nanocarrier. Interestingly, this delivery system demonstrates enhanced cytotoxicity to tumor cells, but not to normal cells.
Content may be subject to copyright.
&
DrugDelivery |Hot Paper|
Human Serum Albumin Conjugated Nanoparticles for pH and
Redox-Responsive Delivery of aProdrug of Cisplatin
Hongdong Shi, Qinqin Cheng,Siming Yuan, Xin Ding, and YangzhongLiu*[a]
Abstract: Platinum anticancer drugs are particularly in need
of controlled drug delivery because of their severe side
effects. Platinum(IV)agentsare designed as prodrugs to
reduce the side effects of platinum(II) drugs;however, pre-
mature reduction could limit the effect as aprodrug. In this
work, ahighly biocompatible, pH and redox dual-responsive
delivery system is prepared by using hybrid nanoparticles of
human serum albumin(HSA) and calcium phosphate (CaP)
for the PtIV prodrug of cisplatin. This conjugate is very stable
under extracellular conditions, so that it protects the
platinum(IV) prodrug in HSA. Upon reaching the acidic and
hypoxicenvironment, the platinum drug is released in its
active form and is able to bind to the target DNA.The Pt–
HSA/CaPhybrid inhibits the proliferation of various cancer
cells more efficiently than cisplatin. Different cell cycle ar-
rests suggest different cellularresponses of the PtIV prodrug
in the CaP nanocarrier.Interestingly,this delivery system
demonstrates enhanced cytotoxicity to tumor cells, but not
to normal cells.
Introduction
Cisplatinisafirst-line chemotherapeutic agent effective for
alarge varietyofsolid cancers. It is well known that cisplatin
targets DNA, and hence,leads to cell apoptosis.[1,2] Due to the
nonspecific reaction of cisplatin, only asmall portion of cellular
cisplatin forms DNA cross-links.[3] Nonspecific reactions reduce
the drugefficacy andare also associated with the side effects
of cisplatin.[1, 4] To solve this problem, many delivery systems
have been designed to improve the drug efficacy of cisplatin,
such as the use of liposomes,[5] polymers,[6] inorganic nano-
materials,[7–9] and metal–organic frameworks.[10, 11]
Human serum albumin(HSA) is the mostabundant plasma
protein (35–50gL
¢1human serum),and is essential for the
transport and metabolism of fatty acids.[12] HSA accumulates
highly in activatedcells, such as those in malignant and in-
flamed tissues,tocover their increased need for amino acids
and energy.[13] Such accumulationresults from the enhanced
retention of macromolecules in tumor tissue, primarily caused
by alack of lymphatic drainage due to an impaired or absent
lymphatic system.[13] In addition, the enhanced permeability of
the vascular system and reduced clearanceinsolid tumors are
also responsible for the accumulationofmacromolecules over
40 kDa.[14] Therefore, HSA is proposed as an effective drug
carriertosites of inflammationormalignancy.[13]
Platinum anticancerdrugs are particularly in need of con-
trolled drug delivery because of their nonspecific reactions and
relatedsevere side effects. Platinum(IV) prodrugs exhibit ad-
vantages in platinum drug delivery because they are kinetically
more inert in coordination substitution than cisplatin. However,
these platinum(IV) compounds could suffer from the
disadvantage of premature reduction prior to cell uptake,[15]
resultinginundesired bindingtoplasma protein molecules,
and thus, limiting their effectiveness as prodrugs. HSA hasalso
showntoprotect platinum(IV) prodrugs in the reducing
environment of the blood, resulting in enhanced circulation
time in the blood.[15] Severalrecent works indicate that HSA
also significantly improves drug targetingtosolid tumors.[16–20]
Cell-responsive delivery is an attractive strategy for en-
hancing drug efficacy. In recent years, various cell-responsive
delivery systems, such as redox active, pH dependent,and en-
zymes, have been intensively investigated. Glutathione(GSH),
which is acellular reductant, is presentmuch more abundantly
in cells (1–10 mm)than in plasma ( 2mm).[21] Some cellular
environments, such as endosomes (pH 5.0) and lysosomes
(pH4.5), are relatively acidic.[22] In addition, solid tumors are
known to have an alteredredox state in comparison with
normaltissues due to tumor hypoxia, lower pH, and elevated
levels of GSH.[23, 24] Therefore, cellular environment responsive
delivery systemscould enhancedrug efficacy and specificity.
Nevertheless, complicated chemical syntheses are often
neededtogenerate responsive delivery systems, andthe
biological functions of thesechemical groups are barely
predictable.
Herein, we designed and synthesized abiocompatible
system,which was sensitivetopHand cellular reductants, for
[a] H. Shi,+Q. Cheng,+S. Yuan, X. Ding,Prof. Y. Liu
CAS Key LaboratoryofSoft Matter Chemistry
CAS High Magnetic Field Laboratory
DepartmentofChemistry
UniversityofScience and Technology of China
Hefei, Anhui,230026(P.R. China)
E-mail:liuyz@ustc.edu.cn
[+
+]These authors contributed equally to this work.
Supportinginformation for this article is available on the WWW under
http://dx.doi.org/10.1002/chem.201502756.
Chem. Eur.J.2015,21,16547 –16554 Ó2015 Wiley-VCH Ve rlag GmbH &Co. KGaA, Weinheim16547
Full PaperDOI:10.1002/chem.201502756
the deliveryofthe PtIV prodrug of cisplatin. The PtIV prodrug
was linked to HSA to form aPt–HSA complex. Then the Pt–
HSA complex was conjugated to calcium phosphate (CaP)
nanoparticles with sizes of about 100 nm. It is knownthat
nanoparticles of such sizes can be internalized into cells
throughendocytosis.[25] Upon cellular uptake,C
aP is de-
composed with weak acidity (pH <6.0) in the endosome;thus
Pt–HSA is released from the conjugate.The PtIV prodrug is dis-
sociated from HSA in the form of cisplatin upon reduction in
cells (Scheme 1). Thus, this conjugatepossesses both pH and
redox dual-responsive properties in the cellular environment.
Results and Discussion
Preparation of the PtIV–HSA prodrug loaded CaP
nanoparticles
The PtIV prodrug loadedHSA (Pt–HSA) was prepared by the re-
action of cis,cis,trans-[Pt(NH3)2Cl2(OH)(O2CCH2CH2CO2H)] (1)
with HSA (see the Supporting Information for details). MALDI-
TOF mass spectra showedthe mass increaseofHSA after bind-
ing of the platinum complex (Figure S3 in the Supporting In-
formation).The protein-conjugatednanoparticle (Pt–HSA/CaP)
was prepared by the formation of CaP nanoparticles in the
presenceofPt–HSA.
The preparation of protein-loaded CaP nanoparticles was
optimized with the addition of surfactants and changes to the
reactionconditions. The well-dispersed and protein-rich spheri-
cal particleswere obtained in the presence of polyethylene
glycol (PEG). The use of other surfactants, such as cetrimonium
bromide (CTAB), bis(2-ethylhexyl) sulfosuccinate sodium salt
(AOT), or Tween-80, led to alow protein loading capacity and/
or poor particlemorphology (Figure S4 in the Supporting Infor-
mation).The uniform size of Pt–HSA/CaP nanoparticles can be
obtained at arange of low temperatures (25–40 8C, see
Figure S5 in the Supporting Information).
SEM images revealed that the particlesizes were about 90–
110nm(Figure 1A). The high-resolution TEM image indicates
that the particles are nearly spherical;the contrast difference
within the particles is consistentwith the protein loading. The
DLS measurements showedthat the average hydrodynamic
radius in solutionwas 150 nm with anarrow size dispersion
(polydispersion index (PDI)=0.12;Figure 1B). The HSA content
in the hybrid nanoparticles was measured by means of TGA
with heating to 800 8C. Amass loss of 35.7 %was observed on
Pt–HSA/CaP,whereas only 15 %massloss was observed on
CaP withoutHSA (Figure 1C). These data indicatethat about
24%HSA was loaded in the Pt–HSA/CaP hybrid nanoparticles,
which corresponded to 0.27 %Ptinthe particles. The XRD
resultsfor HSA/CaP show abroad band at 2q308,which
indicates theamorphous phase of CaP in the nanoparticles
(Figure S6 in the Supporting In-
formation).
Stability of the nanoparticles
The stability of Pt–HSA/CaP was
analyzed in water and in DMEM
containing 10 %FBS. DLS meas-
urements showedthat the nano-
particles were stable for only 6h
in water and the size increased
considerably over 64 h(Fig-
ure 1D). However,the nanoparti-
cles exhibited high stabilityin
DMEM/FBS medium andthe par-
ticle sizes remained unchanged
for six days. The lack of either
PEG or HSA in the preparation
considerably reduces the stabili-
ty of CaP nanoparticles (Figure 1E). DMEM/FBS medium is
amimic of plasma and widelyused in cell cultures.The high
stabilityofPt–HSA/CaP particles in this mediumsuggests that
this hybrid is suitable for applications in drug delivery.
CD spectra were recorded on the protein released from the
nanoparticles to verify structure perturbation of the protein
during the preparation of nanoparticles. The CD spectrum of
native HSA showsanegative band between 208 and 222 nm
and apositive band between195 and 200 nm ;these indicate
well-folded protein with the formation of secondary structures.
The CD spectra werenot changed with the platinum binding.
In addition, the proteins in CaP were obtained by dissolving
the nanoparticles in MES buffer at pH 4.5 (see next section).
The protein releasedfrom CaP had aCDspectrum nearly the
same as that of the native HSA protein (Figure 1F). Thisresult
indicates that the mild preparation process does not perturb
the secondary structure of the protein.
Cellular uptake and distribution of CaP nanoparticles
The cellular uptake of HSA/CaP was analyzed on hepatocellular
carcinoma(HepG2) cells. The HSA was labeled with rhodamine
B(RhB), so that the uptake of nanoparticlescould be
visualized by fluorescencemicroscopy.The time-dependent
measurements showed that relativelyless red fluorescence
was observed in cells at 0.5 hand it increased greatlyduring
4h of incubation (Figure S7 in the Supporting Information).
Scheme1.Schematicillustration of the synthesis Pt–HSA/CaP nanoparticles and the release of cisplatin in
response to low pH and cellular reductants.
Chem. Eur.J.2015,21,16547 –16554 www.chemeurj.org Ó2015 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim16548
Full Paper
The results indicate that the hybrid nanoparticles can efficient-
ly enter cells in atime-dependent manner.Confocal laser scan-
ning microscopy (CLSM) clearly shows that the majority of pro-
tein is located in the cytosol(Figure 2A). The cellular uptake of
RhB–HSA/CaP nanoparticleswas also measured by meansof
flow cytometry on HepG2 and HL7702 cells;this showedthat
cellular uptake increased with increasing concentration of
RhB–HSA/CaP (Figure 2B). In addition, the platinum content in
the cells was directly measured by meansofinductively cou-
pled plasma mass spectrometry (ICP-MS). The result shows
that similar amounts of Pt were detected with the treatment
of cisplatin andPt–HSA, whereas
the treatment of Pt–HSA/CaP (in
the same amount as that of Pt)
resultedinabout 70%more
uptake of platinum in cells(Fig-
ure S8 in the Supporting Infor-
mation). Although cytotoxicity
dependsonmany different
properties of drugs, these data
confirm that the Pt–HSA/CaP
particles can be efficiently inter-
nalized into cancer cells.
pH-responsive release of HSA/
CaP
Protein releasefrom HSA/CaP
nanoparticles wasstudied by
measuring the fluorescence of
HSA in solution.After the incu-
bation of HSA/CaP with buffer at
differentpHvalues, the nanopar-
ticles were removed and the
fluorescenceofHSA in solution
was measured. The result
showedthat the hybrid was
stable in neutralsolutions
(pH 7.0 and 7.4), whereas weak
acidity (pH6.0) led to the com-
plete releaseofprotein from the
nanoparticles (Figure 3). The
treatment of 1 mNaCl caused
very little HSA release from the
nanoparticles. This result implies
that the hybrid nanoparticles are
stable in extracellular media
(0.15mNaCl, pH 7.4) ;hence the
protein and PtIV prodrug are ef-
fectively protected in body
fluids. However,upon cellular
uptake by endocytosis, the hy-
brids in endosomes/lysosomes
(pH 4–6) can be disassembled
and protein is thus released.
Redox-responsive release of PtII drug and the bindingof
DNA
The PtIV–HSA complex released from CaP particles is in the
form of aprodrug, which is inert to DNA binding. Cellular re-
ductionisthe crucial step for the activation of PtIV prodrugs
prior to bindingtotarget DNA. Therefore, the reduction and
DNA binding of the Pt–HSA prodrug was investigatedbythe
reactionofPt–HSA with DNA in the presence of acellular re-
ductantascorbic acid (AsA). The platination of DNA was
analyzed by fluorescencemeasurementswith EtBr as aprobe.
Platinum binding prevents intercalation of EtBr into DNA,
Figure 1. Characterization of Pt–HSA/CaP nanoparticles.A)SEM image of Pt–HSA/CaP nanoparticles (scale bar
200 nm). The inset shows aHR-TEMimage (scale bar 20 nm). B) Size distribution of Pt–HSA/CaP nanoparticles
measured by dynamiclight scattering (DLS). C) TGA of HSA, CaP, HSA/CaP,and Pt–HSA/CaP samples underanair
atmosphere. D) The time-dependent size variation of Pt–HSA/CaP nanoparticles measured in water or Dulbecco’s
modified eagle medium (DMEM) containing10% fetal bovine serum (FBS). E) The stability of CaP nanoparticles
prepared with HSA and/orPEG. The particles were dispersed in DMEM/FBS solution and centrifuged at 3000 rpm
for 3min. F) CD spectraofnative HSA, Pt–HSA, and the protein released from nanoparticles.The proteins in CaP
particles were obtained by dissolution in 2-(N-morpholino)ethanesulfonic acid (MES) buffer (pH 4.5).All samples
were measuredin10mmphosphate bufferat258C, pH 7.4.
Chem. Eur.J.2015,21,16547 –16554 www.chemeurj.org Ó2015 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim16549
Full Paper
which results in the stoichiometric quenching of fluorescence
from the EtBr–DNA complex.[26] The controlexperiment on cis-
platin confirmed that DNA platination caused aslow decrease
in the fluorescence(Figure 4). Pt–HSA and PtIV complex 1did
not alter the fluorescenceinthe absence of reductants. The
presence of 5mMAsA reduced the fluorescenceofDNA/EtBr
complextoanextent similar to that of treatment with cispla-
tin. This resultconfirmed the reduction of PtIV in the Pt–HSA
conjugate and subsequent DNA binding in the presenceofcel-
lular concentrationsofAsA (Figure 4). This result is consistent
with areport in the literature on the reduction of PtIV prodrugs
in the cellular environment,[27] and suggests that Pt–HSA can
functionasaprodrug of cisplatin and respondtothe reducing
agent AsA.
In vitro cytotoxicity of Pt–HSA/
CaP
The cytotoxicityofPt–HSA/CaP
was evaluated by 3-(4,5-di-
methylthiazol-2-yl)-2,5- diphenyl-
tetrazolium bromide (MTT) assay
on different human cells, includ-
ing five cancercell lines (hepato-
cellularcarcinomaHepG2 cells,
lung carcinoma A549 cells, cervi-
cal cancer HeLa cells, human
breast cancerMDA-MB-231cells,
and breast carcinoma MCF-7
cells) and three normal cell lines
(liver HL7702cells, human breast
MCF-10A cells, and human em-
bryonickidney HEK293cells).
The cytotoxicity of cisplatin, PtIV
compound 1,and Pt–HSA was
also measured for comparison
(Table 1). The IC50 valuesshowed
that the Pt–HSA/CaP conjugate
exhibited generally higher cyto-
toxicitythan that of cisplatin
and 1to cancer cells. The lower
IC50 value of Pt–HSA/CaP,compared with that of Pt–HSA, to
cancer cells suggeststhat the CaP carrierenhances the cyto-
toxicity of Pt–HSA. It is notable that cisplatin showed lower cy-
totoxicity to HepG2 cells than to HL7702 cells, with an IC50
ratio (HepG2/HL7702) of 2.1. Nevertheless, the conjugates of
Pt–HSA/CaP showedareverse effect with aratio of 0.33, which
suggested that the HepG2 cells could be more sensitive to Pt–
HSA/CaP than to cisplatin. The same effect wasobserved by
comparing MCF-7 cells to MCF-10Acells. Cisplatin showed
Figure 2. Intracellular uptake and distribution of RhB-labeled HSA/CaP nanoparticles.A)CLSM analysis of HepG2
cells treatedwith RhB–HSA/CaP nanoparticles for 4h.The RhB-labeled HSAshowsred fluorescence, the blue fluo-
rescence from the Hoechst 33258 stain shows the cell nuclei,and the green fluorescence from the Alexa 488 stain
shows the cytoskeleton. The mergedimaging showsthe locationofRhB–HSAprotein in cells. DAPI =4,6-diamidi-
no-2-phenylindole. B) Flow cytometric analyses of the fluorescence intensity in HepG2 and HL7702 cells after the
4hof treatment with RhB–HSA/CaP nanoparticles.
Figure 3. The pH-responsive drug release from Pt–HSA/CaPnanoparticles.
HSA release from HSA/CaPnanoparticles was measured by fluorescence. The
particles were incubated in 100 mmsolution of MES buffer at different pH
values,inwater or in a1msolution of NaCl for 1, 4, and 8h.
Figure 4. Redox-responsive release of PtII drug from Pt–HSA and DNA plati-
nation.The reactionswere conductedonplatinumcomplexes in the ab-
sence or presence of reductant AsA.The platinationofDNA was measured
by fluorescencedecline.All reactionswere conducted with herring sperm
DNA (0.01 mg) at aPt/nucleotide ratio of 1in10mmNaClO4and 10 mm
phosphate buffer (pH 7.4) at 37 8C. EtBr (0.04mg) and 0.4 mNaCl was added
before fluorescence measurements. The excitation wavelengthwas
l=530 nm and the emission was observedatl=592 nm.
Chem.Eur.J.2015,21,16547 –16554 www.chemeurj.org Ó2015 Wiley-VCH Ve rlag GmbH &Co. KGaA, Weinheim16550
Full Paper
high cytotoxicity to MCF-10Acells, whereas Pt–HSA/CaP dem-
onstrated high cytotoxicity to MCF-7 cells. Although various ef-
fects can cause different responses of cells to platinum agents
(such as cellular uptake or redox environmentfor PtIV reduc-
tion), the higher cytotoxicity of HSAconjugates is consistent
with areport that HSA can enhance the efficacy of anticancer
drugs more efficiently to cancer cells than to normalhealthy
cells.[16]
Cell cycle and apoptosis analysis
Cell cycle analysis was conducted on A549 cellswith the treat-
ment of Pt–HSA/CaP for 48 or 72 h. Apoptotic cells with
degraded DNA fragments were accumulated in the sub-G1
region.[28] Cisplatin and Pt–HSA displayed time-dependent S
and G2arrest with very little increase in the sub-G1fraction
(Figure 5). Interestingly,the Pt–HSA/CaP conjugate induced cell
cycle arrest different from that of cisplatin or Pt–HSA. Pt–HSA/
CaP did not cause much G2arrest;however,asignificantly
higher sub-G1fraction was observed for Pt–HSA/CaP (14.08 %
at 48 hand 21.05 %at72h)compared with cisplatin (2.66 %at
48 hand 5.65%at72h)and Pt–HSA (1.11% at 48 hand 1.50 %
at 72 h). This observation indicates differentcell cycle perturba-
tions of Pt–HSA/CaP,which suggests that the presence of CaP
nanoparticles alters the cellular response of platinum drugs.
This alteration could be attributed to different cell uptake and
metabolism of Pt–HSA/CaP conjugates. The CaP control in-
duced aslight G2phase arrest, probably due to the nonsponta-
neous influx of calcium by the uptake of CaP nanoparticles
throughendocytosis.[29, 30]
The tumormicroenvironmental response is an attractive
strategy for controlled drug delivery and release. These drug-
delivery systems are more stable in blood and normaltissues;
however,the specific microenvironment triggers drug release
in tumors,which results in enhanced drug efficacy andre-
duced side effects.[31] In recent years, varioustypes of pH- and
redox-responsive chemical groups have been introduced into
drug-delivery systems, such as hydrazones, esters, orthoesters,
acetals,imines, and cis-aconityls for pH responsiveness and di-
sulfide bonds for redox responsiveness.[32, 33] Complicated
chemicalsyntheses are typically required to introduce these re-
sponsivegroups into drug-delivery system. The harsh reaction
conditions might not be applicable for protein-associated sys-
tems and chemical modificationscould limit the clinic applica-
tion of these materials. Herein, we prepared apHand redox
dual-responsive delivery system by using highly biocompatible
materials with afacile and mild preparation procedure. CaP is
essential for making teeth and bones,[34] and it forms pH-
tunable particlesfor the intracellular release of drugs.[35] The
biomineralization of cisplatin, which formed with CaP and
carbonate, was found to overcome drug resistance.[36] CaP is
known to have ahigh affinity to protein ;[37] thus, we can take
advantage of this to preparePt–HSA/CaP conjugates for the
delivery of PtIV prodrugs.
To achieve redox-responsivedrug release, we take the
advantage of the PtIV prodrug strategy.Itisknown that PtIV
complexes are reduced in cells, which generates the active
form of the PtII drug for binding to target DNA. However,the
presence of extracellular reductants leads to the premature re-
ductionofPt
IV agents and limits their function as prodrugs.[38]
HSA aloneisknown to protect the PtIV agentsinthe reducing
environment of the blood.[15, 39] The formation of HSA/CaP par-
ticles furtherprotects the platinum compound from the
mediumand enhances the stabilityofplatinum agents. The
conjugation of HSA could also improve drug efficacy against
solid tumors.[40]
Bovine serum albumin (BSA) has also been used as amodel
protein for adelivery system. Thereversibleformation of BSA
particles is applicable for the delivery of therapeutic agents.[41]
The loading of the hydrophobic drug ibuprofen on BSA/CaP
particles in organic solvent re-
sults in the sustained release of
the drug.[42,43]
Herein, the PtIV prodrug conju-
gated to HSA/CaP nanoparticles
allows pH and redox dual-re-
sponsivereleaseofthe drug.
The stabilityofCaP nanoparti-
cles is important for biological
applications. We found that the
addition of PEG significantly im-
provedthe stability of HSA/CaP
nanoparticles. It is worth noting
that all materials used in the
preparation of the delivery
Table 1. Inhibitoryeffect (IC50 in Pt [mm])ofcisplatin, 1,Pt–HSA, and
Pt–HSA/CaPondifferentcells.
Cisplatin 1Pt–HSAPt–HSA/CaP
HepG2 5.340.79 13.452.87 8.29 1.34 2.91 0.56
A549 6.711.06 30.67 5.87 13.742.612.35 0.31
HeLa 5.081.64 8.76 2.477.503 0.851.66 0.12
MCF-79.840.95 72.10 8.31 50.49 6.2113.71 3.14
MDA-MB-231 2.660.54 8.27 1.373.09 0.63 1.360.15
HL7702 2.530.23 8.76 1.785.22 1.04 8.87 1.85
MCF-10A 6.771.04 55.93 3.57 46.53 5.2117.17 0.92
HEK293 6.531.12 18.95 3.87 15.14 2.016.33 1.04
Figure 5. Apoptosis of HepG2 cells by using an annexinV/PIassay treatedwith CaP, cisplatin, 1,Pt–HSA, and
Pt–HSA/CaP.
Chem. Eur.J.2015,21,16547 –16554 www.chemeurj.org Ó2015 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim16551
Full Paper
system are highly biocompatible, including PEG. The mild con-
ditions used for the preparation of hybrid nanoparticles, such
as low temperature, short preparation time, and absence of or-
ganic solvents, are highly desirable for protecting the native
protein from denaturation.Thus,the hybrid CaP system could
also be applicable for delivering other native medicinal
proteins.
Conclusion
We designed abiocompatible, dual-responsive, drug-delivery
system by using HSA protein and CaP hybrid nanoparticles to
deliver aprodrug of cisplatin. The hybrid system was stable in
the extracellularenvironment;however,upon cellular uptake
by endocytosis, the CaP nanoparticles decomposed in the
weakly acidic environment of endosomes/lysosomes and the
HSA protein was then released.The prodrug loaded on HSA
could be reduced by cellular reductants, such as AsA, and the
active form of cisplatin be generated. The PtIV prodrug loaded
on HSA reacted with the target DNA only in the presence of
reductant.The formation of HSA/CaP nanoparticle protected
the PtIV prodrug from premature reduction in the extracellular
environment;hence, it was an ideal platform for controlled
drug delivery.Moreover,the Pt–HSA/CaP hybrid inhibited the
proliferation of various cancer cells more efficiently than cispla-
tin. Different cell cycle arrests of Pt–HSA/CaP,relative to Pt–
HSA and cisplatin, suggested adifferent cellular response of
the PtIV prodrug in the CaP nanocarrier.All materials used in
the preparation were highly biocompatible and the mild prep-
aration process led to unaffected protein folding. Therefore,
this methodcould have potential utility for the preparationof
delivery systems for proteins and drugs.
Experimental Section
Materials
MTT,AsA, and herring sperm DNA were purchased from Sangon
Biotech (Shanghai)Co.,Ltd. 1-Ethyl-3-(3-dimethylaminopropyl)car-
bodiimide hydrochloride (EDC·HCl) and N-hydroxysuccinimide
(NHS) were obtained from Aldrich. Ultrapure water (18.2 MW)was
obtained from aMillipore Milli-Q Biocel purification system with
a0.22 mmfilter.All other reagents and solvents were purchased
commercially and used without further purification.
Cell culture
The human cancer cells, including cervical HeLa, breast carcinoma
MDA-MB-231, hepatocellular carcinoma HepG2, breast carcinoma
MCF-7, and lung carcinoma A549 cells, were obtained from the
American Type Culture Collection (ATCC). The normal liver cells
HL7702, normal human breast cells (MCF-10A), and human embry-
onic kidney cells (HEK293) were obtained from the cell bank of the
Chinese Academy of Sciences. The cells were maintained in DMEM
containing 10%FBS (for HeLa, MDA-MB-231, HepG2 cells, MCF-7,
HL7702);RPMI1640 (for A549);DMEM-based medium containing
sodium pyruvate, 2mml-glutamine, and 10%FBS (for HEK293) ;or
DMEM-based medium containing 2.5 mml-glutamine, 20 ng mL¢1
epidermal growth factor,0.1 mgmL¢1cholera toxin, 10 mgmL¢1in-
sulin, 500 ng mL¢1hydrocortisone, and 5% FBS (for MCF-10A) in
ahumidified atmosphere containing 5% CO2at 37 8C.
Synthesis of Pt–HSA and RhB–HSA
The PtIV prodrug of cisplatin, 1,was synthesized according to apro-
cedure reported in the literature.[44] 10 mmPtIV prodrug 1was pre-
pared in HEPES buffer (50 mm)before EDC (50 mm,5equiv) and
NHS (20 mm,2equiv) were added. The mixture was incubated at
room temperature for 30 min before adding HSA (1 mm). After stir-
ring at room temperature for 24 h, the solution was dialyzed
against distilled water before the solution of Pt–HSA was
lyophilized. The product was characterized by MALDI-TOF mass
and ICP-MS.
RhB–HSA was prepared by using the same method as that for
Pt–HSA with RhB instead of 1.
Synthesis of protein-loaded CaP nanoparticles
The HSA-containing CaP nanoparticles were synthesized by the re-
action of Ca(NO3)2with (NH4)2HPO4in the presence of HSA. HSA
(20 mg) and PEG (4 g; MW =4000) were added to asolution of
Ca(NO3)2(100 mL, 2.5 mm). The pH of this solution was adjusted to
10.0 with asolution of ammonia (1 m). The mixture was stirred at
room temperature for 10 min.
A2.5 mmsolution of (NH4)2HPO4(100 mL) was prepared and the
pH was adjusted to 10.0 with asolution of ammonia, which was
added dropwise to asolution of HSA/Ca(NO3)2at arate of
10 mLmin¢1with stirring. The precipitate was collected by centrifu-
gation at 10 000 rpm for 10 min and washed twice with ultrapure
water.The precipitate of HSA/CaP nanoparticles was dispersed in
2mLofultrapure water or DMEM/FBS medium.
The Pt–HSA/CaP and RhB–HSA/CaP nanoparticles were prepared
by means of the same procedure by replacing HSA with Pt–HSA or
RhB–HSA.
To test the effect of surfactant on the formation of HSA/CaP,the
nanoparticles were prepared with different surfactants, including
CTAB, AOT,ortween-80, instead of PEG.
pH-response study
The pH-response study was conducted by measuring HSA released
from the hybrid nanoparticles in MES buffer at pH 4.5, 5.0 6.0, 6.5,
7.0, or 7.4. Buffer (200 mL) was added to asolution of HSA/CaP
nanoparticles (50 mL), then the mixture was incubated at 37 8C. Su-
pernatants were collected at 1, 4, or 8h by centrifugation at
12000 rpm for 10 min. HSA in the supernatant was quantified by
measuring the fluorescence on aHitachi F-4600 spectrophoto-
meter.
Reaction of Pt–HSAwith DNA
Herring sperm DNA was dissolved in 10 mmphosphate-buffered
saline (PBS) and the DNA concentration was measured by deter-
mining the UV/Vis absorption at l=260 nm with an extinction co-
efficient of 6600m¢1cm¢1.The platinum concentration was mea-
sured by ICP-MS. The reaction was carried out on 0.04 mg mL¢1
DNA and 0.1 mmplatinum complex in 10 mmPBS (pH 7.4) contain-
ing 10 mmAsA and 10 mmNaClO4at 37 8Cinthe dark. Samples at
different reaction times (0, 2, 4, 8, 12, 24, 36, 48 h) were taken for
DNA platination measurements by adding 0.025 mg mL¢1EtBr and
0.4mNaCl. Then the fluorescence spectra were recorded on aHita-
chi F-4600 fluorescence spectrometer by using a1cm path length
cuvette under the following conditions:scan speed 1200 nmmin¢1,
Chem. Eur.J.2015,21,16547 –16554 www.chemeurj.org Ó2015 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim16552
Full Paper
excitation slit width 5nm, and emission slit width 10 nm. The
excitation and emission wavelengths were l=530 and 592 nm,
respectively.
Cell uptake imaging
HeLa cells were seeded in 24-well plates at adensity of 104cells
per well. After 24 hofincubation at 378Cin5%CO
2,the cells
were treated with RhB–HSA/CaP nanoparticles (in 2.6 mgmL¢1RhB).
After further incubation for different times (0, 0.5, 1, 2, 3, or 4h),
the medium was removed and cells were washed three times with
PBS. The fluorescence imaging of cells was analyzed by
fluorescence microscopy.
CLSM imaging was recorded on HepG2 cells treated with RhB–
HSA/CaP nanoparticles for 4h.Then the cells were washed there
times with PBS, and fixed in 4% paraformaldehyde. The nuclei
were stained with DAPI and the cytoskeletons were stained with
Alexa Fluor 488. After staining, the cells were washed three times
with PBS and mounted on microscope slides for imaging by using
aLeica TCS-SP5 spectral confocal laser scanning microscope (Leica
Microsystems, Germany,with two-photon system). Excitation wave-
lengths were l=364, 488, and 560 nm for DAPI, Alexa Fluor 488,
and RhB, respectively.
Cellular uptake measured by flow cytometry
HepG2 cells were seeded in 24-well plates at adensity of 15Õ
104cells per well in DMEM medium (0.5 mL) and incubated in ahu-
midified 5% CO2atmosphere for 24 h. Then the cells were treated
with RhB–HSA/CaP nanoparticles at aseries of concentrations.
After removal of the medium, cells were washed with cold PBS
three times. Then the cells were harvested and the fluorescence of
RhB in the cells was analyzed by means of aFACS Calibur flow
cytometer (BD Biosciences, USA). The results were analyzed with
Flowjo 7.6 software.
Platinum content in cells
HepG2 cells were seeded in 6-well plates and incubated overnight
in ahumidified atmosphere containing 5% CO2at 37 8C. Cells were
treated with drugs (cisplatin, Pt–HSA, or Pt–HSA/CaP at aconcen-
tration of 25 mmPt). After 2h of incubation, cells were washed
three times with PBS buffer and digested with trypsinization. The
harvested cells were digested by nitric acid before ICP-MS
measurements.
Cytotoxicity assay
The cancer cells were seeded at adensity of 103cells per well in
a96-well plate in medium (100 mL) and incubated for 24 hbefore
the addition of drug. Then the cells were incubated with different
concentrations of drugs for 72 h. Subsequently,the medium was
changed and asolution of MTT (25 mL; 5mgmL¢1in PBS) was
added to each well. After incubation for 4hto allow viable cells to
reduce the yellow tetrazolium salt (MTT) into dark blue formazan
crystals, the culture medium was removed. DMSO (150 mL) was
added to each well and the cells were shaken for 20 min. The ab-
sorbance was measured at l=570 nm by using aBio-Rad 680 mi-
croplate reader.The IC50 values were calculated by using GraphPad
Prism software (version 5.01) based on data from three parallel
experiments.
Cell cycle analyses
HepG2 cells cultured in 12-well plates were treated with cisplatin,
1,Pt–HSA, of Pt–HSA/CaP at aPtconcentration of 10 mmfor 48 or
72 h. Cells were then harvested and the DNA content was analyzed
by using aCycleTEST PLUS DNA Reagent Kit (BD Biosciences, San
Jose, USA) according to the manufacturer’s instructions. Cell cycle
distributions and DNA contents were determined by using aBD
FACS Calibur flow cytometer.The results were analyzed by using
FlowJo 7.6 software. Percentages of cells in G1(resting), S(DNA
synthesis), and G2/M (mitotic cells) phases, as well as of those
undergoing apoptosis (sub-G1), were recorded.
Acknowledgements
This work was supported by the National Basic Research
ProgramofChina (973 Program, 2012CB932502), the National
ScienceFoundationofChina (U1332210, 21171156,21573213),
and the Collaborative Innovation Center of Suzhou Nano
Scienceand Technology.
Keywords: cancer ·drug delivery ·nanoparticles ·platinum ·
proteins
[1] L. Kelland, Nat. Rev.Cancer 2007,7,573 –584.
[2] D. Wang, S. J. Lippard, Nat. Rev.Drug Discovery 2005,4,307 –320.
[3] J. M. Perez, V. M. Gonzalez, M. A. Fuertes, C. Alonso, Mol. Pharmacol.
2001,59,657 –663.
[4] D. J. Stewart, Crit. Rev.Oncol. Hematol. 2007,63,12–31.
[5] D. M. Liu, C. B. He, A. Z. Wang,W.B.Lin, Int. J. Nanomed. 2013,8,3309
3319.
[6] K. J. Haxton, H. M. Burt, J. Pharm. Sci. 2009,98,2299 –2316.
[7] Y. Z. Min, C. Q. Mao, D. C. Xu, J. Wang, Y. Z. Liu, Chem. Commun. 2010,
46,8424 –8426.
[8] Y. Z. Min, C. Q. Mao, S. M. Chen, G. L. Ma, J. Wang, Y. Z. Liu, Angew.
Chem. Int. Ed. 2012,51,6
742–6747;Angew.C
hem. 2012,124,6
846
6851.
[9] B. S. Wong, S. L. Yo ong, A. Jagusiak, T. Panczyk, H. K. Ho, W. H. Ang, G.
Pastorin, Adv.Drug Delivery Rev. 2013,65,1964 –2015.
[10] W. B. Lin, Abstr.Pap.Am. Chem.Soc. 2014,248.
[11] H. S. Oberoi, N. V. Nukolova,A.V.Kabanov,T.K.Bronich, Adv.Drug Deliv-
ery Rev. 2013,65,1667 –1685.
[12] F. Kratz, J. Controlled Release 2008,132,171 –183.
[13] E. Neumann, E. Frei, D. Funk, M. D. Becker,H.-H. Schrenk,U.Mìller-
Ladner, C. Fiehn, ExpertOpin. Drug Delivery 2010,7,915 –925.
[14] Y. Noguchi, J. Wu, R. Duncan,J.Strohalm, K. Ulbrich,T.Akaike, H.
Maeda, Cancer Sci. 1998,89,307 –314Zeitschrift wurdeerst 2003 gerìn-
det! .
[15] Y. R. Zheng, K. Suntharalingam,T.C.Johnstone, H. Yoo, W. Lin, J. G.
Brooks, S. J. Lippard, J. Am. Chem. Soc. 2014,136,8790 –8798.
[16] S. S.-Y.Lee, J. Li, J. N. Tai, T. L. Ratliff, K. Park,J.-X. Cheng, ACS Nano
2015,9,2420 –2432.
[17] E. Miele, G. P. Spinelli, E. Miele, F. To mao, S. To mao, Int. J. Nanomed.
2009,4,99–105.
[18] M. J. Hawkins, P. Soon-Shiong, N. Desai, Adv.Drug Delivery Rev. 2008,60,
876–885.
[19] G. Stehle, H. Sinn,A.Wunder,H.H.Schrenk,S.Schutt, W. MaierBorst,
D. L. Heene, Anti-Cancer Drugs 1997,8,677 –685.
[20] N. Desai, V. Trieu, Z. Yao, Clin.Cancer Res. 2006,12,1317.
[21] R. Hong,G.Han, J. M. Fernandez, B. J. Kim, N. S. Forbes, V. M. Rotello, J.
Am. Chem. Soc. 2006,128,1078 –1079.
[22] R. Haag, Angew.Chem.Int. Ed. 2004,43,278 –282; Angew.Chem. 2004,
116 ,280–284.
[23] G. Ilangovan, H. Q. Li, J. L. Zweier,P.Kuppusamy, Mol. Cell.Biochem.
2002,234,393 –398.
Chem. Eur.J.2015,21,16547 –16554 www.chemeurj.org Ó2015 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim16553
Full Paper
[24] I. J. Stratford, G. E. Adams, J. C. M. Bremner,S.Cole, H. S. Edwards, N.
Robertson, P. J. Wood, Int. J. Radiat. Biol. 1994,65,85–94.
[25] N. Oh, J. H. Park, Int. J. Nanomed. 2014,9,51–63.
[26] V. Brabec, O. Vrana, O. Novakova, V. Kleinwachter,F.P.Intini, M. Coluccia,
G. Natile, NucleicAcids Res. 1996,24,336 –341.
[27] E. Wexselblatt, D. Gibson, J. Inorg. Biochem. 2012,117 ,220 –229.
[28] S. W. Fesik, Nat. Rev.Cancer 2005,5,876 –885.
[29] I. Baran, Biophys. J. 1996,70,1198 –1213.
[30] M. J. Berridge, P. Lipp, M. D. Bootman, Nat. Rev.Mol. Cell Biol. 2000,1,
11 –21.
[31] M. Upreti, A. Jyoti,P.Sethi, Transl CancerRes. 2013,2,309 –319.
[32] J. Liu, Y. Huang, A. Kumar,A.Tan, S. Jin, A. Mozhi, X. J. Liang, Biotechnol.
Adv. 2014,32,693 –710.
[33] S. Mura, J. Nicolas, P. Couvreur, Nat. Mater. 2013,12,991 –1003.
[34] Y. Cai, R. Tang, J. Mater.Chem. 2008,18,3775 –3787.
[35] H. P. Rim, K. H. Min, H. J. Lee, S. Y. Jeong,S.C.Lee, Angew.Chem.Int. Ed.
2011,50,8853 –8857; Angew.Chem. 2011,123,9015 –9019.
[36] W. Chen,Y.Xiao, X. Y. Liu, Y. H. Chen,J.J.Zhang, X. R. Xu, R. K. Tang,
Chem. Commun. 2013,49,4932–4934.
[37] M. Epple, K. Ganesan, R. Heumann,J.Klesing, A. Kovtun, S. Neumann,V.
Sokolova, J. Mater. Chem. 2010,20,18–23.
[38] C. Fei Chin, D. Y. Wong, R. Jothibasu, W. H. Ang, Curr.Top. Med.Chem.
2011,11,2602 –2612.
[39] V. Pichler,J.Mayr,P.Heffeter,O.Domotor,E.A.Enyedy, G. Hermann,D.
Groza, G. Kollensperger,M.Galanksi, W. Berger,B.K.Keppler,C.R.
Kowol, Chem. Commun. 2013,49,2249–2251.
[40] E. Frei, Diabetes Metab. Syndr.Obes. 2011,3.
[41] J. Xu, J. Wang,J.C.Luft, S. Tian,G.Owens, Jr., A. A. Pandya, P. Berglund,
P. Pohlhaus,B.W.Maynor, J. Smith, B. Hubby,M.E.Napier,J.M.DeSi-
mone, J. Am. Chem. Soc. 2012,134,8774 –8777.
[42] X. Y. Zhao, Y. J. Zhu, F. Chen,J.Wu, Chem. Asian J. 2012,7,1610 –1615.
[43] G. J. Ding, Y. J. Zhu, C. Qi, T. W. Sun, J. Wu, F. Chen, Chem. Eur.J.2015,
21,9868 –9876.
[44] S. Dhar,W.L.Daniel, D. A. Giljohann,C.A.Mirkin, S. J. Lippard, J. Am.
Chem. Soc. 2010,132,17335 –17335.
Received: July 14, 2015
Publishedonline on September 25, 2015
Chem. Eur.J.2015,21,16547 –16554 www.chemeurj.org Ó2015 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim16554
Full Paper
... Redox/pH dual-responsive Pt(IV) prodrugs. Schematic illustration of complex 47 was reproduced with permission from ref. (Shi et al., 2015). Copyright 2015 John Wiley. ...
... A Pt(IV) prodrug was attached to Human serum albumin (HSA) that protected it from premature reduction before being uptaken by cells and improved cancer targeting efficacy (Desai et al., 2006;Neumann et al., 2010;Zheng et al., 2014;Lee et al., 2015). Pt(IV)-HSA was further conjugated with calcium phosphate (CaP) nanoparticles, obtaining a hybrid Pt(IV)-HAS-CaP 47 (Figure 12) (Shi et al., 2015). 47 was decomposed in an acidic environment (pH < 6.0) to release the Pt-HSA complex, which was reduced by reducing agents inside cells to generate active cisplatin, thus exerting anti-tumor effects. ...
Article
Full-text available
Lung cancer is the most common cause of cancer-related deaths worldwide. More efficient treatments are desperately needed. For decades, the success of platinum-based anticancer drugs has promoted the exploration of metal-based agents. Four ruthenium-based complexes have also entered clinical trials as candidates of anticancer metallodrugs. However, systemic toxicity, severe side effects and drug-resistance impeded their applications and efficacy. Stimuli-responsiveness of Pt- and Ru-based complexes provide a great chance to weaken the side effects and strengthen the clinical efficacy in drug design. This review provides an overview on the stimuli-responsive Pt- and Ru-based metallic anticancer drugs for lung cancer. They are categorized as endo-stimuli-responsive, exo-stimuli-responsive, and dual-stimuli-responsive prodrugs based on the nature of stimuli. We describe various representative examples of structure, response mechanism, and potential medical applications in lung cancer. In the end, we discuss the future opportunities and challenges in this field.
... In recent years, HSA-based nanoparticles achieved more attention due to their high biocompatibility and biodegradability in physiologic conditions, surface modification, enhanced permeation and retention in tumors, specific release in tumor tissue, and lower immune response. Thus, these nanoparticles were applied to deliver various cancer drugs such as noscapine, docetaxel, and paclitaxel [4,10,11]. ...
Article
Full-text available
Background: Human Serum Albumin (HSA) is one of the most prominent proteins in human blood. Trimethoprim (TMP) is an efficient antibiotic drug for treating pneumocystis pneumonia. Patients with HIV/AIDS and cancer are highly affected by this disease due to immune system deficiency. Objective: This study aims to evaluate the Molecular Dynamics (MD) simulation of HSA with TMP for drug delivery systems. Methods: In the first step, the 3D structure of HSA and TMP were determined by PDB (Protein Data Bank) and PubChem, respectively. Then, the molecular docking was done via AutoDock Vina software, and the best complex was selected based on the lowest binding energy. Finally, the structural characteristics of the above complex were evaluated. Results: The results showed that TMP binds to the HSA molecule with a binding energy of-7.3 kcal/mol, and this binding causes changes in the third and second structures of the HSA. Thus, Root-Mean-Square Deviation (RMSD) and radius of gyration results proved the third structural change, and the results obtained from DSSP (Database of Secondary Structure assignment for all Protein entries) confirmed the second structural modification. The TMP-HSA complex formation is accompanied by hydrophobic interaction between residues of Tyr150, Ala291, His288, Leu238, Leu219, Lys199, Lys195, Glu153, and TMP. The TMP molecule had two hydrogen bonds with Arg222 residue and three with Ser192. Furthermore, the final PDB file of the MD simulation process showed that the TMP molecule reacted with HSA (IIA chain). Conclusion: Because of the extensive application of TMP in infectious diseases and appropriate interaction with HSA, the complex could be used for the purposeful transport of nanoparticles in the future.
... However, such a one-component, bio-derived system would be more practical and might also offer better biocompatibility than synthetic drug carriers. Albumin particles are a very promising candidate for this multi-faceted problem, but also those carriers come with limitations: this particulate system is only stable in a pH range from 4 to 9 [21,22]; thus, when applied orally, the stability of albumin-based systems in the harsh acidic environment of the stomach is low [23,24]. ...
Article
Biopolymer-based drug carriers are commonly used for the development of safe delivery systems. However, biopolymer-based systems are often highly sensitive to the acidic pH levels in the stomach and release most of their cargo before they have reached their point of destination. Such premature drug release combined with the resulting high dose requirements is not cost-efficient and comes with the risk of unwanted side effects on non-target tissues/organs. This problem can be mitigated by the mucin-based drug carriers developed here, which exhibit good stability at acidic pH levels as proven by dynamic light scattering and enzymatic degradation tests with pepsin. In addition, the mucin-based particles can deliver hydrophobic and hydrophilic drugs simultaneously, which is demonstrated both with experiments performed under in vitro sink conditions and with drug transport tests involving eukaryotic cells as targets. As photo-induced cross-links covalently stabilize those particles, they can release their payload over time in a sustained manner. The drug carrier system introduced here combines good stability with high drug encapsulation efficiency and very good biocompatibility and thus may be valuable for a broad spectrum of applications in biological settings.
... The pH responsive study showed that, at normal pH the hybrid is stable and at pH less than or equal to 6, complete release of drug was observed. According to in vitro cytotoxicity analysis, calcium phosphate enhanced the cytotoxicity of hybrid [232]. Fig. 8 represent the schematic illustration of bioceramic nanoparticle used for drug delivery. ...
Article
Drug delivery can be defined as the group of approaches a drug or pharmacologically active agent can be transported to the target cell to treat disease or health issue. Conventional routes for drug delivery are oral, buccal, rectal, subcutaneous, intranasal, intramuscular, intravenous, pulmonary and transdermal. These are the commonly used methods for treatment of various medical problems but have certain limitations such as instability, risk of displacement, uncontrolled release, side effect such as irritation and pain, slow absorption, enzymatic deterioration and many others. Incorporation of drug into nanocarrier is one of the efficient methods for targeted and sustained delivery of drug. Applications of nanocarriers such as solid nanoparticles, liposomes, dendrimers, polymeric nanoparticles, polymeric micelles, virus like nanoparticles, carbon nanotube and mesoporous silica nanoparticles are discussed in this review. To overcome drawbacks of drug delivery, innovative delivery systems are designed usually termed as smart drug delivery systems which include nucleic acid-based drug delivery system, cell-based drug delivery system, self-nano emulsifying drug delivery system, self-micro emulsifying drug delivery system, chemical and physical stimuli-based drug delivery system, nanoneedles, patches, ultrasound drug delivery and microchip technology. This article focuses of the basic mechanism of drug delivery, pharmacokinetic study, recent innovations and future trends of the drug delivery system.
Article
Full-text available
Platinum-based anticancer drugs play a crucial role in the clinical treatment of various cancers. However, the application of platinum-based drugs is heavily restricted by their severe toxicity and drug resistance/cross resistance. Various drug delivery systems have been developed to overcome these limitations of platinum-based chemotherapy. Stimuli-responsive nanocarrier drug delivery systems as one of the most promising strategies attract more attention. And huge progress in stimuli-responsive nanocarrier delivery systems of platinum-based drugs has been made. In these systems, a variety of triggers including endogenous and extracorporeal stimuli have been employed. Endogenous stimuli mainly include pH-, thermo-, enzyme- and redox-responsive nanocarriers. Extracorporeal stimuli include light-, magnetic field- and ultrasound responsive nanocarriers. In this review, we present the recent advances in stimuli-responsive drug delivery systems with different nanocarriers for improving the efficacy and reducing the side effects of platinum-based anticancer drugs.
Article
Introduction: Human serum albumin is the most abundant transport protein in plasma, which has recently been extensively utilized to form nanoparticles for drug delivery in cancer. The primary reason for selecting albumin protein as drug delivery cargo is its excellent biocompatibility, biodegradability, and non-immunogenicity. Moreover, the albumin structure containing three homologous domains constituted of a single polypeptide (585 amino acid) incorporates various hydrophobic drugs by non-covalent interactions. Albumin shows active tumor targeting via their interaction with gp60 and SPARC proteins abundant in the tumor-associated endothelial cells and the tumor microenvironment. Areas covered: The review discusses the importance of albumin as a drug-carrier system, general procedures to prepare albumin NPs, and the current trends in using albumin-based nanomedicines to deliver various chemotherapeutic agents. The various applications of albumin in the nanomedicines, such as NPs surface modifier and fabrication of hybrid/active-tumor targeted NPs, are delineated based on current trends. Expert opinion: Nanomedicines have the potential to revolutionize cancer treatment. However, clinical translation is limited majorly due to the lack of suitable nanomaterials offering systemic stability, optimum drug encapsulation, tumor-targeted delivery, sustained drug release, and biocompatibility. The potential of albumin could be explored in nanomedicines fabrication for superior treatment outcomes in cancer.
Article
Full-text available
The efficacy of platinum drugs is limited by severe side effects, drug resistance, and poor pharmacokinetic properties. Utilizing long‐lasting blood components as drug carriers is a promising strategy to improve the circulation half‐lives and tumor accumulation of platinum drugs. Non‐immunogenic blood cells such as erythrocytes and blood proteins such as albumins, which have long lifespans, are suitable for the delivery of platinum drugs. In this concept, we briefly summarize the strategies of applying blood components as promising carriers to deliver small‐molecule platinum drugs for cancer treatment. Examples of platinum drugs that are encapsulated, non‐covalently attached, and covalently bound to erythrocytes and plasma proteins such as albumin and apoferritin are introduced. The potential methods to increase the stability of platinum‐based thiol–maleimide conjugates involved in these delivery systems are also discussed. This concept may enlighten researchers with more ideas on the future development of novel platinum drugs that have excellent pharmacokinetic properties and antitumor performance in vivo.
Article
Directly encapsulating cisplatin in calcium phosphate nanoparticles (CPNPs) is a challenge because of the low water-solubility and weak binding affinity with calcium phosphate (CaP) of the cisplatin. In this work, we present a unique strategy using a polymer (poly(Pt(IV) prodrug)) bearing numerous carboxyl side groups and incorporating redox-sensitive cisplatin Pt(IV) prodrugs in its backbone as the payload for encapsulation in CPNPs. The poly(Pt(IV) prodrug) was efficiently encapsulated in CPNPs (>90%), attributing to its improved solubility in alkaline water and strong binding affinity with CaP deriving from the plenty of carboxyl side groups. The CPNPs were stable and almost entirely inhibited the premature release of platinum drugs in the medium mimicking the pH condition of the bloodstream, whether there were reduction agents or not. While in an acidic condition with reduction agents, they released platinum drugs rapidly due to simultaneous reduction and hydrolysis of the poly(Pt(IV) prodrug). Further bioactivity experiments demonstrated the poly(Pt(IV) prodrug) encapsulated CPNPs were of higher efficacy against cancerous cells than free cisplatin and poly(Pt(IV) prodrug) owing to the enhanced platinum drugs uptake by cancerous cells via the CPNPs.
Article
Full-text available
Undesirable side effects remain a significant challenge in cancer chemotherapy. Here we report a strategy for cancer-selective chemotherapy by blocking acyl-CoA cholesterol acyltransferase-1 (ACAT-1)-mediated cholesterol esterification. To efficiently block cholesterol esterification in cancer in vivo, we developed a systemically injectable nanoformulation of avasimibe (a potent ACAT-1 inhibitor), called avasimin. In cell lines of human prostate, pancreatic, lung, and colon cancer, avasimin significantly reduced cholesteryl ester storage in lipid droplets and elevated intracellular free cholesterol levels, which led to apoptosis and suppression of proliferation. In xenograft models of prostate cancer and colon cancer, intravenous administration of avasimin caused the concentration of avasimibe in tumors to be 4-fold higher than the IC50 value. Systemic treatment of avasimin notably suppressed tumor growth in mice and extended the length of survival time. No adverse effects of avasimin to normal cells and organs were observed. Together, this study provides an effective approach for selective cancer chemotherapy by targeting altered cholesterol metabolism of cancer cells.
Article
Full-text available
Spurred by recent progress in materials chemistry and drug delivery, stimuli-responsive devices that deliver a drug in spatial-, temporal- and dosage-controlled fashions have become possible. Implementation of such devices requires the use of biocompatible materials that are susceptible to a specific physical incitement or that, in response to a specific stimulus, undergo a protonation, a hydrolytic cleavage or a (supra)molecular conformational change. In this Review, we discuss recent advances in the design of nanoscale stimuli-responsive systems that are able to control drug biodistribution in response to specific stimuli, either exogenous (variations in temperature, magnetic field, ultrasound intensity, light or electric pulses) or endogenous (changes in pH, enzyme concentration or redox gradients).
Article
Full-text available
The sections in this article are Introduction Nano‐Calcium Phosphates in Hard Tissues Bone Tooth Other Biological Organisms Biological Formation of Calcium Phosphates Characteristic Mechanical Properties Stability of Nano‐Calcium Phosphates Demineralization of Biominerals Dissolution of Pure HAP Nanosize Effects in Biomaterials Synthesis of Nano‐Calcium Phosphates Synthesis of Nano‐Calcium Phosphate Particles Biomimetic Construction using HA Nanoparticles Nano‐ HA –Collagen Composites Nano‐ HA Coating Nano‐Calcium Phosphate in Biomedical Engineering Bone Repair Bone‐Related Cells Enamel Repair Other Applications Summary Acknowledgments
Article
Albumin is the most abundant protein in human serum and drugs that are administered intravenously inevitably interact with it. We present here a series of platinum(IV) prodrugs designed specifically to enhance interaction with human serum albumin (HSA) for drug delivery. This goal is achieved by asymmetrically functionalizing the axial ligands of the prodrug so as to mimic the overall features of a fatty acid. Systematic variation of the length of the aliphatic tail tunes the cellular uptake and, consequently, the cytotoxicity of cis,cis,trans-[Pt(NH3)2Cl2(O2CCH2CH2COOH)(OCONHR)], 4, where R is a linear alkyl group. Investigation of an analogue bearing a fluorophore conjugated to the succinate ligand confirmed that these compounds are reduced by biological reductants with loss of the axial ligands. Intracellular release of cisplatin from 4 was further confirmed by observing the characteristic effects of cisplatin on the cell cycle and morphology following treatment with the prodrug. The most potent member of series 4, for which R is a hexadecyl chain, interacts with HSA in a 1:1 stoichiometry to form the platinum-protein complex 7. The interaction is non-covalent and extraction with octanol completely removes the prodrug from an aqueous solution of HSA. Construct 7 is robust and can be isolated following fast protein liquid chromatography. The nature of the tight interaction was investigated computationally, and these studies suggest that the prodrug is buried below the surface of the protein. Consequently, complexation to HSA is able to reduce the rate of reduction of the prodrug by ascorbate. The lead compound from series 4 also exhibited significant stability in whole human blood, attributed to its interaction with HSA. This favorable redox profile, in conjunction with the established nonimmunogenicity, biocompatibility, and enhanced tumor accumulation of HSA, produces a system that holds significant therapeutic potential.
Article
Recent studies delineate a predominant role for the tumor microenvironment in tumor growth and progression. Improved knowledge of cancer biology and investigation of the complex functional interrelation between the cellular and noncellular compartments of the tumor microenvironment have provided an ideal platform for the evolution of novel cancer nanotherapies. In addition, multifunctional "smart" nanoparticles carrying imaging agents and delivering multiple drugs targeted preferentially to the tumor/tumor microenvironment will lead to early diagnosis and better treatment for patients with cancer. The emerging knowledge of the tumor microenvironment has enabled rational designing of nanoparticles for combinatorial treatment strategies that include radiotherapy, antiangiogenesis and chemotherapy. This multimodality approach is thus expected to achieve therapeutic efficacy and enhance the quality of life of cancer patients. This review highlights the unique characteristics of the tumor microenvironment that are exploited by nanotechnology to develop novel drug delivery systems aimed to target the tumor/tumor microenvironment.
Article
In the realm of drug delivery, carbon nanotubes (CNTs) have gained tremendous attention as promising nanocarriers, owing to their distinct characteristics, such as high surface area, enhanced cellular uptake and the possibility to be easily conjugated with many therapeutics, including both small molecules and biologics, displaying superior efficacy, enhanced specificity and diminished side effects. While most CNT-based drug delivery system (DDS) had been engineered to combat cancers, there are also emerging reports that employ CNTs as either the main carrier or adjunct material for the delivery of various non-anticancer drugs. In this review, the delivery of small molecule drugs is expounded, with special attention paid to the current progress of in vitro and in vivo research involving CNT-based DDSs, before finally concluding with some consideration on inevitable complications that hamper successful disease intervention with CNTs.
Article
The fabrication and evaluation of the analytical performance of integrated microfluidic MEMS modified with electrochemically deposited metal nanoparticles are described in this paper. The microfluidic MEMS is a hydride system that comprise two layers, poly-dimethylsiloxane (PDMS) and glass, which in turn enclose a micro channels network and a set of 15 micro electrodes, respectively. The mechanism of electrokinetic phenomenon and electrochemical detection were applied for performing injection and separation, and sensing the components of a biochemical mixture, respectively. The technique of electrochemical deposition was employed for depositing nanoparticles of gold, palladium and platinum on the surface of integrated microelectrodes. The nanoparticles-modified micro electrodes were characterised electrochemically and using scanning electron microscope (SEM). Importantly, the nanoparticles-based decoupler has significantly enhanced the analytical performance of the microfluidic MEMS, where detection limits in the low nanomolar range were recorded for the targeted analytes.