Download as pdf or txt
Download as pdf or txt
You are on page 1of 42

Subscriber access provided by UCL Library Services

Article
Transferrin-Copper Nanocluster-Doxorubicin
Nanoparticles as Targeted Theranostic Cancer Nanodrug
Upashi Goswami, Anushree Dutta, Asif Raza, Raghuram Kandimalla,
Sanjeeb Kalita, Siddhartha Sankar Ghosh, and Arun Chattopadhyay
ACS Appl. Mater. Interfaces, Just Accepted Manuscript • DOI: 10.1021/acsami.7b15165 • Publication Date (Web): 26 Dec 2017
Downloaded from http://pubs.acs.org on December 26, 2017

Just Accepted

“Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted
online prior to technical editing, formatting for publication and author proofing. The American Chemical
Society provides “Just Accepted” as a free service to the research community to expedite the
dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts
appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been
fully peer reviewed, but should not be considered the official version of record. They are accessible to all
readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered
to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published
in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just
Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor
changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers
and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors
or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

ACS Applied Materials & Interfaces is published by the American Chemical Society.
1155 Sixteenth Street N.W., Washington, DC 20036
Published by American Chemical Society. Copyright © American Chemical Society.
However, no copyright claim is made to original U.S. Government works, or works
produced by employees of any Commonwealth realm Crown government in the course
of their duties.
Page 1 of 41 ACS Applied Materials & Interfaces

1
2
3
4
5
6
7 Transferrin-Copper Nanocluster-Doxorubicin Nanoparticles
8
9
10 as Targeted Theranostic Cancer Nanodrug
11
12
13
14
15 Upashi Goswami,a Anushree Dutta,b Asif Raza,c Raghuram Kandimalla,d Sanjeeb Kalita,d
16
17 Siddhartha Sankar Ghosh* ac and Arun Chattopadhyay* ab
18
19
20 a
Centre for nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039.
21
22
23 b
24 Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039.
25
26
c
27 Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati,
28
29 Guwahati 781039.
30
31
32 d
33
Drug discovery Lab,Institute of Advanced Study in Science and Technology, Guwahati-781035,
34
35 Assam, India.
36
37
38 *Email: arun@iitg.ernet.in; sghosh@iitg.ernet.in
39
40
41
42
43
44 KEYWORDS: transferrin copper nanoclusters, Förster resonance energy transfer, nanodrug,
45
46
47
synergistic targeted therapy, cancer
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
1
ACS Applied Materials & Interfaces Page 2 of 41

1
2
3 ABSTRACT:
4
5
6
7 Transferrin (Tf) templated luminescent blue copper nanoclusters (Tf-CuNCs) are synthesized.
8
9
They are further formulated into spherical transferrin copper nanocluster-doxorubicin
10
11
12 nanoparticles (Tf-CuNC-Dox-NPs) based on electrostatic interaction with doxorubicin (Dox).
13
14 The as synthesized Tf-CuNC-Dox-NPs are explored for bioimaging and targeted drug delivery to
15
16 delineate high therapeutic efficacy. Förster resonance energy transfer (FRET) within the Tf-
17
18
19 CuNC-Dox-NPs exhibited striking red luminescence, wherein the blue luminescence of Tf-
20
21 CuNCs (donor) is quenched due to absorption by Dox (acceptor). Interestingly, blue
22
23 luminescence of Tf-CuNCs is restored in the cytoplasm of cancer cells upon internalization of
24
25
26
the NPs through overexpressed transferrin receptor (TfR) present on the cell surface. Finally,
27
28 gradual release of Dox from the NPs leads to the generation of its red luminescence inside the
29
30 nucleus. The biocompatible Tf-CuNC-Dox-NPs displayed superior targeting efficiency on TfR
31
32
overexpressed cells (HeLa and MCF-7) as compared to the less TfR expressed cells (HEK-293
33
34
35 and 3T3-L1). Combination index (CI) revealed synergistic interaction of Tf-CuNCs and Dox
36
37 upon Tf-CuNC-Dox-NPs treatment. In vivo assessment of the NPs on TfR positive Daltons
38
39 Lymphoma Ascites (DLA) bearing mice revealed significant inhibition of tumor growth
40
41
42 rendering prolonged survival of the mice.
43
44
45 Introduction
46
47
48 In parallel to the conventional radiotherapy,1 chemotherapy2 and surgery for cancer
49
50 treatments, contemporarily preferred modes include photo-thermal therapy,3 immunotherapy,4,5
51
52
53
hormone therapy,6 recombinant protein therapy7,8 and stem cell therapy9 which are based on the
54
55 incidence and progression of cancer. Importantly, non-specificity and low solubility with
56
57
58
59
60 ACS Paragon Plus Environment
2
Page 3 of 41 ACS Applied Materials & Interfaces

1
2
3 reduced retention time of majority of the chemotherapeutic drugs lead not only to severe side
4
5
6 effects but also to eventual development of multidrug resistance.10–14 Thus in clinical practice
7
8 along with surgery, combining other therapies with chemotherapy helps curtail such
9
10 inadequacies to some extent with better therapeutic outcome.15–17 On the other hand,
11
12
13
nanotechnology based composite drug delivery systems have been reported to improve loading
14
15 capacity, detection and specific release at the site of the disease, circulation time and
16
17 bioavailability of drugs and thus help reduce untoward side effects.18–21 In this regard, molecule-
18
19
like properties with discrete electronic states and size-dependent photoluminescence (PL) of
20
21
22 metal nanoclusters of size near to the Fermi wavelength of electrons i.e., < 2 nm (for Ag, Au and
23
24 Cu) are useful in the fields of bioimaging, biosensing and drug delivery.22–27 The ability to
25
26 evaluate the treatment by introducing multiple functions into single nano-platform (as a part of
27
28
29 theranostics) can be achieved with nanoclusters to advance the therapeutic paradigm of cancer
30
31 treatment.8,22,23 Additionally, interaction of the designed luminescent nanoclusters with cellular
32
33 species or any external molecule like drug or enzyme, guided by changes in their luminescence
34
35
36
based on available mechanisms such as Förster resonance energy transfer (FRET), photoinduced
37
38 electron transfer (PET) etc. may allow monitoring of intracellular interactions, stimuli responsive
39
40 28–32
behaviour, imaging, drug localization and enhanced release with ease. Although Au and Ag
41
42
nanoclusters are widely reported to have been incorporated into nanocarriers for imaging and
43
44
45 therapy,8,21,23 use of Cu nanoclusters (CuNCs) has been limited due to lack of stability even in
46
47 mild oxidizing environment.26 However, CuNCs in a hydrogel nanocarrier have shown enhanced
48
49 activity of cisplatin and synergy of action.33 Thus, the advantage of photoluminescent drug
50
51
52 carrier lies in real-time monitoring of drug localization and release; however, the application has
53
54 been limited due to non-specificity.7,8,33 Nanoparticle based drugs like Doxil/Lipoplatin
55
56
57
58
59
60 ACS Paragon Plus Environment
3
ACS Applied Materials & Interfaces Page 4 of 41

1
2
3 (PEGylated liposome), Abraxane (paclitaxel albumin-stabilized nanoparticle), DepoCyt
4
5
6 (liposomal cytarabine), Oncaspar (PEG-asparaginase), CAELYXTM (PEGylated liposomal
7
8 doxorubicin), and Genexol-PM (polymeric micellar nanoparticle), NK105 (Paclitaxel loaded
9
10 polymeric micelles) are in clinical trials for cancer therapy,21,24 but options for targeted imaging
11
12
13
and diagnosis are not available in these compositions. Mostly, therapeutic agents are released in
14
15 the perivascular cells of tumor where the nanoparticles tend to accumulate in spleen and liver as
16
17 captured by reticuloendothelial system. However, the tumor targeting could be achieved by
18
19
incorporating specific receptor protein(s) in the carriers.34–37 Transferrin (Tf), a plasma protein,
20
21
22 which plays a critical role in homeostasis of iron, is overexpressed in many cancers due to -
23
24 augmented iron requirements in cell proliferation. Tf easily enters the TfR-overexpressing cells
25
26 through active receptor-mediated endocytosis.38 Thus the use of Tf in a carrier could help
27
28
29 targeted delivery of drugs in cancer tissues.39 Doxorubicin (Dox) is one of the most widely used
30
31 drug for cancer therapy, but its use is associated with side effects, unalterable cardiac toxicity
32
33 and occurrence of drug resistance.10,13-15 Thus minimization of its use, in combination with other
34
35
36
less toxic candidates, would be beneficial for enhanced therapeutic values.
37
38
39 Herein, a formulation of nanodrug containing blue emitting Tf stabilized CuNCs and Dox as
40
41 hydrophobic drug, referred to henceforth as Tf-Cu NC-Dox – nanoparticles (NPs), is reported.
42
43 Following UV light excitation, FRET from the blue emitting Tf-CuNCs as donor to the Dox
44
45
46
acting as acceptor led to the NPs emitting in the red thus acting as a probe. Interestingly, the blue
47
48 emission of Tf-CuNCs could be effectively recovered in the cytoplasm of overexpressed
49
50 transferrin receptors in cancer cells, allowing observation of simultaneous and gradual release of
51
52
Dox into the nucleus following internalization. That, on the other hand, allowed real time
53
54
55 monitoring of intracellular drug release based on FRET. At the same time, synergistic anticancer
56
57
58
59
60 ACS Paragon Plus Environment
4
Page 5 of 41 ACS Applied Materials & Interfaces

1
2
3 activity could also be achieved involving Tf-CuNCs and Dox present in Tf-CuNC-Dox-NPs. To
4
5
6 the best of our knowledge for the first time a nanodrug incorporating Tf-CuNCs and Dox has
7
8 been developed for targeted imaging, and therapeutic action in conjunction with synergy of the
9
10 components. In addition to the in vitro studies, the nanodrug was tested for efficacies by
11
12
13
investigating in vivo using Dalton’s lymphoma ascites (DLA) cells, a T-cell lymphoma first
14
15 identified in thymus of DBA (H2D) mice and later adapted to close inbred strain of the Swiss
16
17 albino mice, which mimics characteristics similar to human lymphoma. It is one of the most
18
19
convenient and widely used model for peritoneal carcinoma to screen the activity of various
20
21
22 anticancer agents within a short period of time.34,40 The schematic depiction of formulation of
23
24 nanodrug for FRET asissted bio-imaging, targeted delivery and synergistic therapeutic activity in
25
26 vitro as well as in vivo has been elucidated in Scheme 1.
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
5
ACS Applied Materials & Interfaces Page 6 of 41

1
2
3 Scheme 1: Schematic depiction of formulation of targeted nanodrug for FRET assisted bio-
4
5
6 imaging, targeted delivery and synergistic therapeutic activity in vitro as well as in vivo.
7
8
9 Experimental Section
10
11
12 TEM Analysis: The size and morphology of as-synthesized Tf-Cu NC and Tf-CuNC-Dox-NPs
13
14 were probed using JEOL, JEM 2100 TEM (Peabody, MA, USA), which operates at maximum
15
16
17
accelerating voltage of 200 kV. For analysis, first 1 mL of as-synthesized particle was taken and
18
19 centrifuged at 15,000 rpm for 30 min. The pellet was then redispersed in 1 mL of Milli-Q water
20
21 and from which 200 µL of Tf-CuNC-Dox-NPs were taken and diluted in 1 mL of water. Briefly
22
23 8 μL of the composite from the above dispersion was drop cast onto copper coated TEM grid.
24
25
26 The sample was allowed to dry at room temperature for 6 h before analysis.
27
28
29 Dynamic Light Scattering Based Measurements: Hydrodynamic diameter and net surface
30
31 charge of the composite nanoparticles were measured using Malvern Zeta Size Nano ZS-90
32
33
instrument. The change in DLS hydrodynamic size and surface zeta potential of Tf-CuNCs
34
35
36 before and after drug binding were performed at a temperature of 25 °C. In each case, 50 µL of
37
38 the dispersion was diluted in 950 µL water and then measurements were performed. For each
39
40 sample dispersion, three DLS measurements were made with a fixed run time of 11 s with
41
42
43 scattering angle set at 90°. The Malvern DTS 5.10 software was used for data analysis.
44
45
46 Synthesis of Tf-Cu NCs. In a typical synthesis, 0.2 mL (25 mM) aqueous solution of copper
47
48 sulphate (CuSO4) was added dropwise to 2 mL aqueous solution of transferrin (5 mg/mL) under
49
50 vigorous stirring at 37 °C. After stirring for 10 min, pH was adjusted to ~ 12 by adding freshly
51
52
53 made 1M NaOH solution during which the reaction mixture turned purple from colourless
54
55 solution. After 1 h of stirring, 80 μL N2H4 (80 %), was introduced into the reaction mixture
56
57
58
59
60 ACS Paragon Plus Environment
6
Page 7 of 41 ACS Applied Materials & Interfaces

1
2
3 dropwise, and was further allowed to stir for 10 h at 37 °C. The color of the reaction mixture
4
5
6 gradually changed from purple to light brown ensuring the completion of reaction. The as-
7
8 synthesized Tf-Cu NCs was then further stored at 4 °C for further use.
9
10
11 Formulation of Tf Copper Nanocluster-Doxorubicin Nanoparticle (Tf-CuNC-Dox-NPs).
12
13 0.017 mL (2.5 mM with respect to copper) as synthesized Tf-Cu NCs was added to 0.8 mL
14
15
16 aqueous solution in a reaction vial and temperature was set at 37 °C. Under stirring condition, 50
17
18 μL Dox solution (from 400 μM stock) was added successively after a gap of 15 min each and
19
20 fluorescence spectrum was recorded simultaneously. After final addition of 250 μL of Dox
21
22
23
solution, the reaction mixture attained a pinkish red color. The final resulting mixture, was then
24
25 allowed to age for ~ 2 h at 37 °C. The as-synthesized Tf-CuNC-Dox-NPs was then collected by
26
27 centrifugation at 10000 rpm for 30 min. The pellet so obtained was re-dispersed in 1 mL Milli-Q
28
29
water for further use.
30
31
32
33 Encapsulation Efficiency and Release Study of Doxorubicin. In order to calculate the amount
34
35 of Dox present in the Tf-CuNC-Dox-NPs, the as synthesized Tf-CuNC-Dox-NPs were
36
37 centrifuged at 10,000 rpm for 30 min at 15 °C. The supernatant was then collected and the
38
39
40
emission spectrum was recorded with λex = 480 nm. Similarly, emission spectrum of control Dox
41
42 i.e., the initial concentration used during NP synthesis was recorded (λex = 480 nm). The
43
44 emission intensities of the supernatant of Tf-CuNC-Dox-NPs and Dox only at λem= 590 nm were
45
46 then measured. The encapsulation efficiency (E.E %) was then calculated using the following
47
48
49 equation:
50
51 Doxi  Dox f
52 E. E % 
53 Doxi
54
55
56
57
58
59
60 ACS Paragon Plus Environment
7
ACS Applied Materials & Interfaces Page 8 of 41

1
2
3 where Doxi refers to emission intensity of Dox corresponding to initial concentartion of Dox
4
5
6 used in composite formulation and Doxf refers to emission intensity of Dox in the supernatant of
7
8 the composite after synthesis.
9
10
11 For release study, the drug loaded NPs were incubated in two different buffers (acetate
12
13 buffer pH 5 and PBS pH 7) at 37 °C and release was then monitored for 48 h by UV-vis
14
15
16 spectroscopy. At different time intervals, supernatant was collected after centrifugation at 10,000
17
18 rpm for 15 min and the absorbance was measured at 480 nm for Dox.
19
20
21 In vitro experiments (Mammalian Cell Culture)
22
23
24 Human cervical cancer cell line (HeLa TfR positive), human breast cancer cell line (MCF-7 TfR
25
26
27
positive), human embryonal kidney cell line (HEK-293 TfR partially positive) and mouse
28
29 embryo fibroblast cell line (Balb/3T3, control TfR negative) were procured from National Centre
30
31 for Cell Sciences, Pune. Cells were grown in CO2 incubator with 5% carbon dioxide in
32
33
humidified atmosphere and cultured in Dulbecco’s Modified Eagle’s Medium (DMEM, high
34
35
36 glucose) containing 10% FBS, 10,000 units of Penicillin and 10mg/mL streptomycin.
37
38
39 Cell Viability Assay
40
41
42 To study the effect of Tf-Cu NC- Dox -NPs on different types of cell lines with varied TfR
43
44 expressions, MTT (3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide) assay was
45
46
47 carried out after 48 h of treatment. Briefly, 5× 103 cells were seeded in each well of 96 well plate
48
49 and were left for 24 h for attachment. Further, varying concentrations of Tf-Cu NCs (10 µg/mL),
50
51 Dox (10 µg/mL) and Tf-Cu NC- Dox-NPs (10 µg/mL) were used for treatment for 48 h, in
52
53
54
triplicates. After the completion of the treatment duration, MTT was added in each well and the
55
56 resulting formation of formazon was dissolved in DMSO. The absorbance of formazan, which
57
58
59
60 ACS Paragon Plus Environment
8
Page 9 of 41 ACS Applied Materials & Interfaces

1
2
3 corresponds to the number of living cells was then measured at 550 nm with background
4
5
6 reference measured at 655 nm in Multiplate Reader (Tecan). The normalized percentage cell
7
8 viability was calculated as:
9
10
11
12 ( A550  A655) sample
13 % of cell viability  100
14 ( A550  A655) control
15
16
17 Expression analysis of transferrin receptors
18
19
20 The total RNA was isolated from HeLa, MCF-7, HEK-293 and 3T3-L1 cells using GenElute
21
22 Mammalian Total RNA Miniprep Kit (Sigma-Aldrich). Further, 1 µg of total RNA was used to
23
24
25
generate cDNA pool of the respective cell lines with the help of the Verso cDNA Kit (Thermo
26
27 Scientific, MA, USA). A semi-quantitative polymerase chain reaction (PCR) was performed with
28
29 transferrin receptor primers using cDNA pool of the respective cell lines. β-actin gene was taken
30
31 as endogenous control. The products of PCR were run on a 1.3% agarose gel pre-stained with
32
33
34 ethidium bromide.
35
36
37 Confocal Microscopy
38
39
40 To study the difference in uptake, based on the TfR expression, confocal microscopy studies
41
42 were carried out in 4 different cell lines (HeLa, MCF-7, HEK-293 and 3T3-L1) using Zeiss LSM
43
44
45
880 microscope. 5× 103 cells were grown on a coverslip in 35 mm culture plate for 24 h under
46
47 integral condition of CO2 incubator. The uptake study was carried out after 4 h incubation with
48
49 Tf-Cu NC-Dox- NPs followed by PBS washing and fixing with 4% formaldehyde. Thereafter,
50
51
the coverslips were gently taken with the help of tweezers and were placed upside down on the
52
53
54 glass slide and the sides were sealed. For time dependent uptake the same procedure was
55
56 followed where the cells were fixed at appropriate time interval. The control cells without any
57
58
59
60 ACS Paragon Plus Environment
9
ACS Applied Materials & Interfaces Page 10 of 41

1
2
3 treatment were prepared in the similar manner for all sets of experiments. The prepared samples
4
5
6 were observed under simultaneous mode (with λex 405 nm for Tf-CuNCs and λex 488 nm for
7
8 Dox) under Zeiss microscope LSM 880.
9
10
11 Mechanism of cellular uptake with inhibitors
12
13
14 The uptake study was carried out with fluorescence based assays on both Tecan and CSLM. For
15
16
17
Tecan based assay 5× 103 cells were grown in 96 well plate for 24 h at 37° C and cells were
18
19 grown on coverslip in 35 mm culture for CSLM study. For active and passive transport the cells
20
21 were incubated at 4°C for 4h and with 1mM NaN3 for 30min at 37°C.These pre-treated cells
22
23 were then incubated with composite (Tf-CuNC-Dox-NPs) for 4h.To confirm the mode of uptake
24
25
26 two inhibitor assays were performed with Chlorazole Black E which is transferrin receptors
27
28 inhibitor and chlorpromazine which inhibits clathrin mediated endocytosis. Before the treatment
29
30 with composite for 4h the cells were pre-treated with Chlorazole Black E (50µM for 6h) and
31
32
33 chlorpromazine (5µM for 4h).
34
35
36 In vivo experiments
37
38
39 Adult male Swiss albino mice weighing 22-25 g were procured from Chakraborty enterprise
40
41 (1443/PO/b/11/CPCSEA), Kolkata, India. All mice were housed at Central Animal Facility,
42
43
Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam. The mice
44
45
46 were maintained in polypropylene cages and the room conditions was maintained at 22 ± 2 ºC,
47
48 60–70% relative humidity and 12–12 h light–dark cycle. All mice were fed with standard Rodent
49
50 pellet diet (Provimi Animal Nutrition India Pvt. Ltd., India) and water ad libitum. Mice were
51
52
53 allowed to acclimate for a week and after close monitoring for a week all the experiments were
54
55 conducted. Before commencing the work, the protocols were designed and approved
56
57
58
59
60 ACS Paragon Plus Environment
10
Page 11 of 41 ACS Applied Materials & Interfaces

1
2
3 (IASST/IAEC/2016–17/2301) by the Institutional Animal Ethics Committee (IAEC) of IASST
4
5
6 and implemented as per guidelines of Committee for the Purpose of Control and Supervision of
7
8 Experiments on Animals (CPCSEA), Government of India.
9
10
11 Development of in vivo tumor (Dalton’s Ascites Lymphoma Cells)
12
13
14 Dalton’s ascites lymphoma (DLA) cells were acquired from IASST Central Animal Facility, and
15
16
17
1×106 viable tumour cells/mice were transplanted through intraperitoneal (I.P.) route. The
18
19 growth of tumor was ascertained by abnormal belly swelling and increased body weight, which
20
21 were visible in 8–10 days, post induction.
22
23
24 Animal Grouping and in vivo Experimental design
25
26
27 Based on the acute toxicity studies on Tf-Cu NC-Dox-NPs and from previous literature data
28
29
30 Dox doses were selected for in vivo experiments. For the tumor progression, DLA cells (0.2 mL
31
32 of 1 × 106 cells/mice) were injected intraperitoneally on day 0 leaving aside the normal group.
33
34 The drug treatments were started from day 8 of post tumour transplantation in the interval of 24
35
36
37
h for 8 days. The work involved 46 mice, which were taken and alienated in 5 groups with 10
38
39 mice in each group besides group-I having 6 mice.
40
41
42 Group-I: Mice with no DLA tumour + No drug treatment
43
44
45 Group-II: DLA bearing mice + 0.2 ml PBS (I.P) for 8 days
46
47
48 Group-III: DLA bearing mice + Tf- Cu NC 2 mg/kg (I.P) for 8 days
49
50
51 Group-IV: DLA bearing mice + Dox 2 mg/kg (I.P) for 8 days
52
53
54
55
Group-V: DLA bearing mice + Tf- Cu NC-Dox NP 2 mg/kg (I.P) for 8 days
56
57
58
59
60 ACS Paragon Plus Environment
11
ACS Applied Materials & Interfaces Page 12 of 41

1
2
3 Effect of treatment on tumor Growth
4
5
6
During the experimental period the body weight changes were measured daily and cells from all
7
8
9 the groups were collected at the end of the drug treatment period (16 days) to determine the cell
10
11 viability and morphological changes (FESEM analysis). At day 17, five mice from each group
12
13 were sacrificed by decapitation to collect the blood, liver and kidney for biochemical and
14
15
16 histopathological analysis. Remaining 5 mice were observed up to 50 days to estimate the mean
17
18 survival time (MST) and % increase in life span (% ILS).
19
20
21 Results and Discussions
22
23
24 The nanodrug formulation was achieved using a blue emitting Tf-CuNCs and well known
25
26
27
hydrophobic drug – Dox - with the availability of FRET-based energy transfer between the
28
29 components. Blue emitting Tf-CuNCs was synthesized by an aqueous method with hydrazine
30
31 hydrate as the reducing agent at pH=12 (Refer experimental section for details). Here Tf acted as
32
33
a stabilizer by forming strong metal-ligand bond between Cu ions and cysteine residues along
34
35
36 with contribution from N-atom of histidine and O-atom of tyrosyl residues 41 apart from acting as
37
38 functional ligand for targeting TfR overexpressed in cancer cells. The 26 tyrosine residues of Tf
39
40 (at pH > 10) along with N2H4 imparted the reducing environment, whereas some of the 38
41
42
43 cysteine sulphur residues might have acted as stabilizing ligands to the copper core in the
44
45 cluster.39,42 The as-synthesized Tf-CuNCs exhibited strong blue emission band at 460 nm (λex
46
47 =375 nm) as shown in Figure 1a. Additionally, the absence of any surface plasmon resonance
48
49
50
(SPR) peak in the visible region discounted the presence of large sized copper nanoparticles
51
52 (Figure S1, Supporting Information). The PL quantum yield of the as synthesized Tf-CuNCs was
53
54 quantified to be 7.5 %, using quinine sulphate as the standard. This is analogous to the reported
55
56
57
58
59
60 ACS Paragon Plus Environment
12
Page 13 of 41 ACS Applied Materials & Interfaces

1
2
3 emission efficiency of CuNCs and therefore supported their suitability to be used as bio-imaging
4
5
6 probe.26,33 The formation of Tf-CuNCs was further validated from TEM image as shown in
7
8 (Figure 1b) with average particle size distribution of 2.08 nm ± 0.84 (Figure 1c). The average
9
10 size was calculated from about 100 particles in TEM images recorded with different sets of
11
12
13
experiments and with the help of Image J software. The characterization of the as-synthesized
14
15 Tf-CuNCs was further pursued by both matrix assisted laser desorption/ionisation - time of flight
16
17 (MALDI-TOF) based mass spectrometric measurements (using sinapinic acid as the matrix) and
18
19
X-ray photoelectron spectroscopy (XPS), which signified the number of atoms responsible for
20
21
22 cluster formation and the oxidation state of the concerned atoms, respectively. MALDI TOF
23
24 mass spectrum showed a base peak due to singly charged Tf at m/z = 80027, whereas mass
25
26 spectrum of Tf-CuNCs - as recorded - showed a weak peak at m/z = 80207 and two prominent
27
28
29 peaks at m/z = 80312 and m/z = 80496 indicating the presence of 3, 5 and 7 Cu atoms in the as-
30
31 synthesized Tf-CuNCs (Figure 1d). XPS spectrum showing two peaks at 932.2 eV and 952.2 eV,
32
33 corresponding to Cu 2p3/2 and Cu 2p1/2, respectively (Figure 1e), indicated the Cu(0) state of
34
35
36
copper in the as - synthesized Tf-CuNCs along with Cu(I), which however, differs from Cu(0)
37
38 binding energy by 0.1 eV. Thus both Cu(0) and Cu(I) species might have been present in the
39
40 clusters. Additionally, absence of peak at 942 eV ruled out the possibility of Cu(II) in the system.
41
42
Further, circular dichroism (CD) measurements were carried out to probe the targeting ability of
43
44
45 the protein by evaluating the degree of conformational change in transferrin following the
46
47 synthesis of Tf-CuNCs. It was found that after Tf-CuNC synthesis there was a slender decrease
48
49
in the -helix conformation from 28.6 % to 26.2 % and -sheet from 21.3 % to 20.3%,
50
51
52 respectively (Figure 1f). This on the other hand indicated that the targeting efficiency of Tf might
53
54 have remained unaffected after Tf-CuNCs synthesis.
55
56
57
58
59
60 ACS Paragon Plus Environment
13
ACS Applied Materials & Interfaces Page 14 of 41

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23 Figure 1. (a) Photoluminescence intensity of as synthesized Tf-CuNCs (blue curve, λex =375nm) and
24
25 transferrin (cyan-blue curve) with the inset showing digital image of Tf-Cu NCs under UV trans-
26
illuminator, (b) TEM image of Tf-CuNCs (scale bar 50 nm), (c) histogram showing size distribution of
27
28 Tf-Cu NCs (as obtained from Figure 1b and else), (d) MALDI-TOF spectra of Tf (red) and Tf-CuNCs
29
30 (blue) due to singly charged species. Peaks assigned to respective number of Cu atoms are circled in
31 green, (e) XPS spectrum of as synthesized Tf-CuNCs showing two peak corresponding to Cu2p electrons,
32
33 (f) CD spectra of Tf (red) and Tf-CuNCs (blue).
34
35 For the formulation of organic-inorganic nanodrug composite, freshly prepared Dox was
36
37
38
added to a dilute aqueous dispersion of the as-synthesized Tf-CuNCs, under constant stirring at
39
40 ambient temperature, which was followed by aging for 2 h. Details of synthesis have been
41
42 described in the Experimental Section. The Tf-CuNC-Dox-NPs were then collected after
43
44
centrifugation at 10,000 rpm for 30 min. The formulation of organic-inorganic nanodrug was
45
46
47 monitored using steady state fluorescence spectroscopy. The inorganic moiety of Tf-CuNCs
48
49 showed a characteristic emission at 460 nm (λex = 375 nm), whereas Dox employed as organic
50
51 drug has characteristic absorption at 480 nm, which emits at 590 nm. Thus, prominent spectral
52
53
54 overlap between Tf-CuNCs (donor, λex=375 nm and λem= 460 nm) and Dox (acceptor, λex= 480
55
56 nm and λem= 590 nm) allowed us to realize the existence of a FRET mechanism between them
57
58
59
60 ACS Paragon Plus Environment
14
Page 15 of 41 ACS Applied Materials & Interfaces

1
2
3 (Figure S2a, Supporting Information). Additionally, the excitation spectra recorded for Dox only
4
5
6 (acceptor) and composite NPs at λem= 590 nm provided further evidence of the energy transfer
7
8 process from donor Tf-CuNCs (Figure S2b, Supporting Information). A good spectral overlap
9
10 between the excitation spectrum of composite NPs (Figure S2b(ii), Supporting Information) and
11
12
13
emission spectrum of Tf-CuNCs can be observed in Figure S2b(iii) Supporting Information,
14
15 whereas Figure S2b(iv), Supporting Information shows the emission spectrum of final composite
16
17 NPs (nanodrug). To understand the FRET process, emission characteristic change of Tf-CuNCs
18
19
was monitored by addition of varying concentration of Dox at fixed intervals of time
20
21
22 (λex=375m). Briefly, fixed concentration of Tf-CuNCs (Refer Experimental Section) was taken
23
24 and Dox was added under stirring condition after an interval of 15 min each. The fluorescence
25
26 spectrum of the composite dispersion was then recorded after each successive addition of 50 µL
27
28
29 Dox (from 400 µM stock). As is evident in Figure 2a, the emission intensity at 460 nm due to Tf-
30
31 Cu NCs showed gradual drop with concomitant rise in emission intensity at 590 nm due to Dox
32
33 (when excited by 375 nm light). The results indicated the energy transfer from the Tf-CuNCs to
34
35
36
Dox based on FRET mechanism. At a final Dox concentration of 93.7 µM—in the composite
37
38 (which is equal to 50.92 µg of dox), the blue fluorescence of Tf-CuNCs was completely
39
40 quenched showing maximum emission at 590 nm due to Dox thereby reaching a saturation and
41
42
hence finally giving rise to pink color dispersion that exhibited red emission. To get better
43
44
45 insight into the occurrence of FRET between Tf-CuNCs and Dox, time-resolved
46
47 photoluminescence (TRPL) spectroscopic measurements were carried out. Figure 2b shows the
48
49 PL decay profile of Tf–Cu NCs and Tf-CuNC-Dox–NPs with λex=375 nm. The drop in average
50
51
52 PL lifetime of Tf-CuNCs from 3.3 ns to 1.2 ns in the Tf-CuNCs-Dox-NPs (Figure 2b) gave
53
54 evidence of the steady state emission quenching of the donor in the presence of the acceptor
55
56
57
58
59
60 ACS Paragon Plus Environment
15
ACS Applied Materials & Interfaces Page 16 of 41

1
2
3 (Dox) and its active participation in the FRET process. Based on this, the energy transfer
4
5
6 efficiency from Tf-CuNCs to Dox in the NPs was calculated to be 63.6 % following equation S3,
7
8 Supporting Information. The degree of energy transfer was further determined from spectral
9
10 overlap integral value calculated following equation S4, Supporting Information which was
11
12
13
found to be 3.71 × 1014 M-1cm-1nm4. This contributed to a critical transfer distance called Förster
14
15 radius (R0) equal to 2.83 nm (using equation S5, Supporting Information), which was found to be
16
17 in the desired range (1-10 nm) of an ideal FRET pair. Therefore, using the value obtained for R0
18
19
and E, the actual distance between the FRET pair (using equation S6, Supporting Information)
20
21
22 was calculated to be 2.55 nm. (Refer Section 3, Supporting Information) for FRET parameters
23
24 calculation details).
25
26
27 Further, to shed light into the mechanism of NP formation, DLS based zeta potential
28
29
measurements were performed. The zeta potential data revealed a negative charge of -13.2 ±
30
31
32 5.02 mV for the as-synthesized Tf-CuNCs. The increase in zeta potential of the Tf-CuNCs in the
33
34 final composite to a value of 2.74 ± 1.59 mV, upon addition of positively charged doxorubicin
35
36 suggested the electrostatic mode of interaction between the two species. However, H-bond
37
38
39 interaction as well as π-π stacking interaction between Dox and hydrophilic and hydrophobic
40
41 sites in protein could also be the basis of interaction.32 Thus, after the formation of NPs, the net
42
43 surface charge of the composite NPs was found to be 2.74 ± 1.59 mV, indicating the propensity
44
45
46
of electrostatic and hydrogen bond interaction towards strong association between as synthesized
47
48 Tf-CuNCs and Dox (Figure S3, Supporting Information). The resulting Tf-CuNC-Dox- NPs
49
50 dispersion (pH = 6.5) was centrifuged at 10000 rpm at 15 °C and supernatant was collected. The
51
52
drug encapsulation efficiency was then obtained by measuring the emission intensity (λex= 480
53
54
55 nm) for Dox present in supernatant, following centrifugation of Tf-CuNC-Dox-NPs, with respect
56
57
58
59
60 ACS Paragon Plus Environment
16
Page 17 of 41 ACS Applied Materials & Interfaces

1
2
3 to the emission intensity of Dox used for synthesis (Figure S4, Supporting Information). Further
4
5
6 detail of the procedure is available in Experimental Section. Based on this, the encapsulation
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
Figure 2. (a) Change in PL emission intensity of Tf -CuNCs recorded at varying concentrations of Dox
41 (400 μM stock) as mentioned in the legends (λex=375 nm), (b) time-resolved photoluminescence decay
42
43 curve of as-synthesised Tf-CuNCs (blue) and Tf-CuNC-Dox-NPs (red) monitored at λem=460 nm (and
44 λex=375 nm),(c) TEM image of Tf-CuNC-Dox-NPs and (d) corresponding particle size distribution of as -
45
46 synthesized Tf-CuNC-Dox-NPs.
47
48 efficiency was found to be ~ 89% in the final composite collected after centrifugation, which was
49
50
51 equivalent to 44.5 µg/mL of Dox in the dispersion. For successful drug delivery through passive
52
53 targeting of tumor cells using enhanced permeability and retention (EPR) effect, fulfilment of
54
55 size criterion remains an essential feature. Studies revealed that particles with diameter < 200 nm
56
57
58
59
60 ACS Paragon Plus Environment
17
ACS Applied Materials & Interfaces Page 18 of 41

1
2
3 are effective for easy extravasation through leaky tumour cells via EPR effect. 24 Therefore, the
4
5
6 as-synthesized NPs were characterized by transmission electron microscopy (TEM), field
7
8 emission scanning electron microscopy (FESEM) and dynamic light scattering (DLS) based
9
10 studies. TEM study revealed spherical nature of the as-synthesized Tf-CuNC-Dox-NPs (Figure
11
12
13
2c) with an average particle size 42.14 nm ± 16.83 nm, which represents the thermodynamically
14
15 most stable structure and hence shape under the given reaction condition. In addition, kinetic
16
17 stability of the particles during the observation period may also have contributed to such
18
19
particular size range. Additionally, the histogram plot of the as-synthesized Tf-CuNC-Dox-NPs
20
21
22 is shown in Figure 2d. The CuNCs of size < 2 nm in Tf-CuNC-Dox-NPs are noticeable in the
23
24 TEM image (Figure S5c Supporting Information), while no characteristic SAED pattern for Cu
25
26 metal (Figure S5d, Supporting Information) was observed. The FESEM image (Figure S6a,
27
28
29 Supporting Information) of Tf-Cu NCs-Dox-NPs revealed an average particle size of 47.79 ±
30
31 8.53, which matched closely with the TEM results. However, DLS based measurement of the
32
33 hydrodynamic size of the Tf-CuNCs-Dox-NPs revealed size of 117.35 ± 10.15 nm, which may
34
35
36
be due to the protein in the liquid medium and the hydration layer surrounding the NC core
37
38 (Figure S5e, Supporting Information). Thus, the present composite NP system fulfils the size
39
40 criterion required for passive targeting based on EPR effect. To further explore the cellular
41
42
internalization and real time imaging of cancer cells, confocal microscopy imaging experiments
43
44
45 were carried out on TfR overexpressed cells. Confocal microscopy image along with depth
46
47 projection image of HeLa cells after 4 h of treatment with Tf-Cu NCs revealed their successful
48
49 internalization in the cells. The cells exhibited bright blue luminescence in the cytoplasm
50
51
52 (Figure S7a, Supporting Information) signifying their uptake, which was corroborated by Z-stack
53
54 image as shown in (Figure S7d, Supporting Information). However, similar phenomenon was not
55
56
57
58
59
60 ACS Paragon Plus Environment
18
Page 19 of 41 ACS Applied Materials & Interfaces

1
2
3 observed in 3T3-L1 cells, which have low expression of TfR (Figure S8b, Supporting
4
5
6 Information).
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
Figure 3. (a) Agarose gel electrophoresis results (image) showing different expressions of TfR on HeLa,
45 MCF-7, HEK-293 and 3T3-LI cells, and -actin, which was taken as endogenous control, (b) FACS
46
47 analysis confirming the uptake of the composite by cells without Tf inhibitor as observed by shift in
48
fluorescence intensity profile in PE-H channel than the cells treated with inhibitor, (c) confocal
49
50 fluorescence microscopic images of HeLa cells treated with Tf-Cu NC-Dox-NPs for 4 h, and then with
51
52 inhibitor for 6 h, only Dox and cells treated with Tf-Cu NC-Dox-NPs without inhibitor. First column
53 represents the bright-field images; in the second and third columns fluorescence images were collected in
54
55 emission window 580−700 and 380−490 nm at excitation wavelength 405 and 488 nm, respectively.
56
57
58
59
60 ACS Paragon Plus Environment
19
ACS Applied Materials & Interfaces Page 20 of 41

1
2
3 Fourth column is the merged image. The scale bar is 5 µm for first 3 columns, whereas image
4
5 representing the control has a scale bar of 10 µm.
6
7 It has been reported that transferrin receptors are overexpressed in HeLa and MCF-7 cells,
8
9
10 whereas HEK-293 and 3T3-L1 cells have low expression of them as shown by semi-quantitative
11
12 polymerase chain reaction (PCR) using TfR gene primers (Figure 3a). Relative expression level
13
14 of the TfR was measured by extracting the RNA of the respective cells followed by
15
16
17
transformation into cDNA by reverse transcription polymerase chain reaction (RT-PCR). Semi
18
19 quantitative PCR analysis and agarose gel electrophoresis of cDNA gave an insight into the
20
21 expression level of TfR in all the cells. To confirm the receptor mediated active endocytosis,
22
23
Fluorescence-activated cell sorting (FACS) based experiments were performed with transferrin
24
25
26 inhibitor chlorazole black E (50 µM), which was added to the cells 6 h prior to the treatment (in
27
28 order to block the TfR). The overgrown HeLa cells on treatment with transferrin inhibitor
29
30 blocked the uptake of Tf-Cu NC-Dox-NPs. The same amount of Tf-CuNC-Dox-NPs without the
31
32
33 inhibitor showed higher shift in fluorescence intensity plot than the cells treated with inhibitor
34
35 after 4 h of treatment (Figure 3b) as analysed by FACS. It is to be stated here that without
36
37 inhibitor, the uptake of Tf-CuNC-Dox-NPs was more facile compared to that of control Dox
38
39
40
used for the treatment (Figure S9, Supporting Information). It is apparent from the FACS data
41
42 that there was an observable shift of fluorescence intensity plot of HeLa cells after 4 h of
43
44 treatment with Tf-Cu NC-Dox-NPs than only Dox as depicted in (Figure S9, Supporting
45
46
Information). To evaluate whether the uptake was an active or passive process, the cells were
47
48
49 pre-incubated (i) at 4°C for 4 h and (ii) in ATP-depleted environment (using 10mM NaN3 for
50
51 30min). This is followed by incubation with (Tf-CuNC-Dox-NPs) for 4h. The control was also
52
53 maintained at 37°C following treatment for 4h. These exposures resulted in prominent reduction
54
55
56 of fluorescence intensity by 67.45% and 58.96%, respectively (Figure S10, Supporting
57
58
59
60 ACS Paragon Plus Environment
20
Page 21 of 41 ACS Applied Materials & Interfaces

1
2
3 Information). The results were further supported by CLSM studies (Figure S11, Supporting
4
5
6 Information) hence suggesting the prevalence of active transport mechanism. Further, to validate
7
8 the pathway involved in endocytosis, the cells were pre-incubated with chlorpromazine (5µM for
9
10 4h), which is a known clathrin mediated endocytosis inhibitor. Experimental results indicated
11
12
13
drastic reduction of fluorescence intensity (76.67%) compared with that of control (Figure S10,
14
15 Supporting Information). The reduced luminescence as seen in CLSM images (Figure S11,
16
17 Supporting Information) also revealed less uptake of Tf-CuNC-Dox-NPs in chlorpromazine
18
19
treated cells thus confirming the clathrin mediated endocytosis pathway.
20
21
22
23
Further, the confocal microscopy studies also supported the inhibitory action of the chlorazole
24
25 black E, as no activation of luminescence was observed on HeLa cells treated with chlorazole
26
27 black E for 6 h followed by 4 h treatment with Tf-Cu NC-Dox-NPs (Figure 3c). This observation
28
29
was possibly due to blocking of TfR receptor mediated endocytosis pathway by the inhibitor.
30
31
32 Further, the internalisation of Tf-Cu NCs-Dox-NPs after 4 h of treatment was clearly observed in
33
34 case of HeLa and MCF-7 cells as opposed to HEK-293 and 3T3-L1 cells, which have low
35
36 expression levels of transferrin receptors (Figure S12, Supporting Information). To get a better
37
38
39 insight into the manifestation of FRET in the as-synthesized NPs vis-à-vis being inside cells,
40
41 time-dependent confocal microscopy studies were carried out in HeLa cells.
42
43
44 Interestingly, receptor mediated endocytosis of Tf-CuNC-Dox-NPs in TfR overexpressed
45
46 cells led to gradual activation of blue luminescence of Tf-CuNCs in the cytoplasm as is clear
47
48
49 from Figure 4. The time dependent confocal laser scanning microscopic studies revealed no
50
51 luminescence at 0 h of treatment by the as-synthesized NPs (Figure 4). However, after 2 h of
52
53 treatment, slender blue luminescence was observed in the cytoplasm, which was gradually
54
55
56
57
58
59
60 ACS Paragon Plus Environment
21
ACS Applied Materials & Interfaces Page 22 of 41

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36 Figure 4. Fluorescence confocal microscopy images of HeLa cells at different time intervals – 0 h, 2 h, 4
37
38
h and 6 h of treatment, indicating the gradual uptake of Tf-CuNC-Dox-NPs. The excitation wavelength
39 was 405 and 488 nm. First column is the bright-field image, for the second and third columns the
40
41 fluorescence images were collected in the range of 580−700 and 380−490 nm, respectively. Fourth
42 column is the merged image. The scale bar is 10 µm.
43
44
45 enhanced over the time possibly due to gradual disruption of donor-acceptor interaction between
46
47 Tf- CuNCs and Dox following internalisation. This led to gradual recovery of blue fluorescence
48
49
50
in cytoplasm and hence simultaneous release of Dox in the nucleus. Figure 4 clearly revealed the
51
52 blue luminescence observed in cytoplasm after 4 h of treatment with red fluorescence of the Dox
53
54 in the nucleus, intensity of both of which was augmented at the end of 6 h of study. In contrast,
55
56
57
58
59
60 ACS Paragon Plus Environment
22
Page 23 of 41 ACS Applied Materials & Interfaces

1
2
3 no significant recovery of blue luminescence was observed in 3T3-L1 cells - which have
4
5
6 relatively low TfR expression - when treated with the Dox-loaded composite (Figure S13,
7
8 Supporting Information).
9
10
11 To evaluate the intracellular drug release and its therapeutic activity, MTT based cell viability
12
13
14
assay was carried out. The HeLa cells were incubated at different concentrations of Tf-CuNCs,
15
16 free Dox and Tf-Cu NCs-Dox-NPs for 48 h at 37 °C. The final concentrations of copper and Dox
17
18 in the Tf-Cu NCs-Dox-NPs was 1.82 µg/mL and 44.5 µg/mL, respectively. The copper
19
20
concentration in the Tf-Cu NCs-Dox-NPs was determined by AAS. Further, 75% of HeLa cells
21
22
23 were found to be viable when treated with Tf-CuNCs only at a concentration of 0.44 µg/mL
24
25 (with respect to copper) as shown in (Figure S14a, Supporting Information). Further, MTT assay
26
27 carried out in HeLa cells at Dox concentration in the range of 0.2-1.33 µg/mL showed viability
28
29
30 of around 50 % at 1.23 µg/mL as shown in Figure 5a. Interestingly, the same concentration of
31
32 Dox on cells treated with Tf-CuNCs-Dox-NPs displayed viability of only 22%. The IC50 values,
33
34 calculated by nonlinear regression curve fit (using Graphpad Prism Software), were found to be
35
36
37
0.65 µg/mL for Dox and 0.32 µg/mL for copper (Figure 5a), suggesting the occurrence of
38
39 synergistic effect. To establish the synergy of Cu and Dox in Tf-CuNC-Dox-NP, the
40
41 isobologram and combination index (CI) were calculated based on the median effect principle of
42
43
Chou and Talalay, 43 using the CalcuSyn software (Biosoft, Version 2.1). The line of additivity of
44
45
46 isobologram plot can be interpreted in terms of synergy (CI < 1) placed below the line, additivity
47
48 (CI=1) on the line and antagonism (CI > 1) above the line. The isobologram plot of MTT data
49
50 shown in Figure 5b depicts that all the data points were below the line of additivity, which
51
52
53 represented drug-drug interactions i.e., synergy. The details of the CI values are given in (Table
54
55 S1, Supporting Information). Similar dose dependent cytotoxicity of nanodrug was also observed
56
57
58
59
60 ACS Paragon Plus Environment
23
ACS Applied Materials & Interfaces Page 24 of 41

1
2
3 in MCF-7 cells (Figure S14b, Supporting Information), where cell viability was decreased to
4
5
6 20% at 1.33 µg/mL of Dox. The IC50 value was found to be at 0.6 µg/mL of Dox and 0.24
7
8 µg/mL of copper. However, in case of HEK-293 and 3T3-L1 cells, around 80 to 90% of cells
9
10 were viable at the IC50 dose of HeLa cells (Figure S14 c and d). In fact, at the highest
11
12
13
concentration of Dox (1.33µg/mL) of our experiment, the viability of cells was more than 70% in
14
15 case of 3T3-L1 cells. This is essentially because of the selective targeting of the Tf-Cu NCs-
16
17 Dox-NPs, based on TfR expressions that promoted increased uptake by HeLa and MCF-7 cells
18
19
as opposed to HEK-293 and 3T3-L1 cells. Thus, the significant decrease of cell viability with
20
21
22 nanodrug (composite NPs) as opposed to free drug molecule evidenced synergistic therapeutic
23
24 efficacy herein. The anti-cell proliferative effect of Dox was remarkably enhanced in HeLa cells
25
26 when incorporated in Tf-Cu NCs. Hence, the subsequent intracellular release of anticancer drug -
27
28
29 with synergistic therapeutic activity - was observed in case of overexpressed TfR cells (HeLa
30
31 and MCF-7). To support the cytotoxicity of Tf-CuNC-Dox-NPs, in vitro release profile of Dox
32
33 was pursued at pH 4.5 (acetate buffer) and pH 7.0 (phosphate buffer), which resemble the
34
35
36
physiological conditions of tumour cells and normal cells, respectively. The time dependent
37
38 release profile shown in Figure 5c indicates that at pH 4.5 nearly 65% of Dox was released after
39
40 48 h, whereas around 40% of drug was released at pH 7.0. This outcome was attributed to
41
42
enhanced disintegration of the inorganic-organic composite NP system at acidic pH.
43
44
45
46 To further examine the cytotoxicity of Tf-CuNC-Dox-NPs, analyses of live and dead cells
47
48 were performed using Calcein AM and EtBr. For this HeLa cells were treated with IC 50 dose of
49
50 Tf-CuNC-Dox-NPs with respective Tf-CuNC and Dox concentrations as mentioned above.
51
52
53 Observation under epi-fluorescence microscope revealed heightened Et-Br-stained apoptotic
54
55 cells, which signified the preferential uptake of Tf-CuNC-Dox-NPs as compared with Dox by the
56
57
58
59
60 ACS Paragon Plus Environment
24
Page 25 of 41 ACS Applied Materials & Interfaces

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
Figure 5. (a) Cell viability based on MTT assay after 48 h of treatment with varying concentrations of
28
29 Dox and Tf-CuNC-Dox-NPs in HeLa cells. The experiments were carried out in triplicates and are
30
31 represented as the mean ± SD. The ANOVA test revealed the statistical significance between Dox and Tf-
32 CuNC-Dox-NPs which is denoted by ‘*’ (p < 0.05), ‘**’ (p < 0.005), and ‘***’ (p < 0.001), (b)
33
34 Isobologram analysis showing all points below additive line indicating synergistic effect. Combination
35
index (CI) values calculated using Calcusyn software, (c) Time-dependent in-vitro release profile (in %)
36
37 of Dox from Tf-CuNC-Dox-NPs in the PBS (pH 7.0) and acetate buffer (pH 4.5). Experiments were
38
39 carried out at 37°C and are represented as the mean ± SD from three individual experiments , (d) FACS
40 analysis results of the generation of reactive oxygen species (ROS) in HeLa cells investigated in DCF
41
42 channel (FITC) corresponds to the green emission of the DCF. Cells treated with Tf-CuNC-Dox-NPs
43 exhibited prominent shift as compared to the control cells.
44
45
46 treated cells (Figure S15, Supporting Information). Additionally, the onset of apoptosis was
47
48 documented in FESEM images, after 48 h of treatment with Dox and Tf-CuNC-Dox-NPs.
49
50
51 Initiation of apoptosis was illustrated by gradual indentation and irregularity in the cell
52
53 morphology treated with Dox (Figure S16b, Supporting Information) than control (Figure S16a,
54
55 Supporting Information). On the other hand, more blebbing and distortion were observed in case
56
57
58
59
60 ACS Paragon Plus Environment
25
ACS Applied Materials & Interfaces Page 26 of 41

1
2
3 of cells treated with Tf-CuNC-Dox-NPs (Figure S16c, Supporting Information). Moreover,
4
5
6 presence of apoptosis was also confirmed using propidium iodide (PI) assays after 48 h of
7
8 treatment at IC50 dose of Tf- CuNC-Dox-NPs along with the control Tf-Cu NCs and Dox. The
9
10 results suggested upsurge of dead cells population to 51.12%, 10.38% and 35.93%, for cells
11
12
13
treated with composite NPs, Tf-Cu NCs and Dox, respectively (Figure S17, Supporting
14
15 Information), which corroborated the cell viability results.
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47 Figure 6. (a) Propidium iodide based cell cycle analysis using FACS where 1 represents that of the
48
49 control and 2, 3, 4 are the cells treated with Tf-CuNCs, Dox and Tf-CuNC-Dox-NPs, respectively, (b)
50 Flow cytometric analyses via annexin-V-7-AAD staining in order to find out the apoptotic and necrotic
51
52 populations (%). In the dot plots the second, and third quadrants denotes early apoptotic (E.A.), and late
53 apoptotic (L.A.) population of control cells and the cells treated with Tf-CuNCs, Dox and Tf-CuNC-Dox-
54
55 NPs respectively.
56
57
58
59
60 ACS Paragon Plus Environment
26
Page 27 of 41 ACS Applied Materials & Interfaces

1
2
3 To elucidate the mode of cell death exerted by the nanodrug (Tf-CuNCs-Dox NPs),
4
5
6 fluorescence-activated cell sorting (FACS) based assays were carried out. Firstly, reactive
7
8 oxygen species (ROS) generation was investigated using the 2’,7’–dichlorofluorescin diacetate
9
10 (DCFDA) staining method where DCFH is to be oxidized to green fluorescent 2’, 7’ –
11
12
13
dichlorofluorescein (DCF) in the presence of ROS. It can be inferred from Figure 5d, that there
14
15 was shift in fluorescence intensity in cells treated with nanodrug (Tf-CuNCs-Dox NPs), which
16
17 correlated with the amount of ROS production inside the cells. Tf-Cu NCs and Dox on the other
18
19
hand induced less oxidative stress than Tf-CuNC-Dox-NPs. Cell cycle analysis revealed the
20
21
22 arrest of G2/M phase in cells treated with Dox, whereas S-phase arrest was observed in case of
23
24 cells treated with Tf-CuNCs-Dox-NPs (Figure 6a). Additionally, FACS based Annexin V-7AAD
25
26 assay confirmed apoptosis mode of cell death, where significantly greater percentage of
27
28
29 apoptotic cells (17.47%) was observed for Tf-CuNC-Dox-NPs treated cells than control (2.59%),
30
31 Tf-CuNCs (4.56%) and Dox (10.15%) treatment.
32
33
34 Finally, in vivo efficacy of Tf-CuNC-Dox-NPs was examined on tumour bearing Dalton’s
35
36
37
lymphoma ascites (DLA) mice. Adult male Swiss albino mice were used to induce DLA by
38
39 subcutaneous inoculation of DLA cells in group G-II to V for respective experiments and
40
41 subsequently size of tumor growth was monitored. The DLA cells collected from the mice
42
43
harboring tumour showed adequate expression of transferrin receptor (Figure S18a, Supporting
44
45
46 Information) and hence, the transferrin carrying NPs could be employed on DLA mice model for
47
48 selective targeting in vivo. The uptake of Tf-CuNC-Dox-NPs on DLA cells was confirmed by
49
50 confocal microscopy (Figure S18b, Supporting Information). The toxicity studies of Tf-CuNC-
51
52
53 Dox-NPs suggested that Tf-CuNC-Dox-NPs at 20 mg/kg did not produce any deleterious effects
54
55 on animal health and no mortality was seen up to 14 days of observation period.
56
57
58
59
60 ACS Paragon Plus Environment
27
ACS Applied Materials & Interfaces Page 28 of 41

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27 Figure 7. (a) Experimental scheme representing the effect of Tf-Cu NC-Dox-NPs in tumour bearing
28
29 Swiss albino mice monitored for 30 days, (b) Change in the body weight of tumor bearing mice after
30 treatment with Tf-Cu NCs, Dox and Tf-CuNC-Dox-NPs, respectively, till 16 days of treatment, (c)
31
32 Survival probability of DLA induced mice monitored up to 50 days after treatment with Tf-Cu NCs, Dox
33 and Tf-CuNC-Dox-NPs, as represented by Kaplan–Meir curve.
34
35
36 Therefore, 1/10th of the No Observed Adverse Effect Level (NOAEL) i.e., 2 mg/kg was selected
37
38 for cytotoxic evaluation and tumour suppression activities of Tf-CuNC-Dox-NPs, Tf-CuNCs and
39
40
41 free Dox. The experiments were conducted in Swiss albino mice bearing DLA tumourogenesis.
42
43 The mice were divided into four groups (n = 10) where the 1st group was assigned as control
44
45 followed by groups treated with Tf-CuNCs (2 mg/kg), free Dox (2 mg/kg) and 2 mg/kg of Tf-
46
47
48
CuNC-Dox-NPs. After 8 days of tumour growth (with the belly swollen due to accumulation of
49
50 ascitic fluid) the non-lethal dose of Tf-CuNC-Dox-NPs (2 mg/kg) was injected into the
51
52 peritoneal cavity of the mice at scheduled intervals for 8 days as illustrated in the experimental
53
54
Figure 7a. After 8 days of drug treatment period, the cells from all mice were tested for cell
55
56
57
58
59
60 ACS Paragon Plus Environment
28
Page 29 of 41 ACS Applied Materials & Interfaces

1
2
3 viability. Results of trypan blue (Table S2, Supporting Information) assay revealed that Tf-
4
5
6 CuNC-Dox-NPs treatment significantly reduced the percentage of viable DLA cells compared to
7
8 the other treatment groups (Free Dox & Tf- CuNCs).It can be inferred from Figure 7b that
9
10 reduction in tumour volume was observed in both free Dox and Tf-CuNC-Dox-NPs treated mice,
11
12
13
which were confirmed by body weight reduction compared to control and Tf-CuNC treated
14
15 groups. Among all the treatments, the Tf-CuNC-Dox-NPs treatment showed the highest potential
16
17 in tumour volume reduction. At the end of the treatment (16th day), mice treated with Dox and
18
19
Tf-CuNC-Dox-NPs showed a lesser increment in body weight (4.32 ± 0.86* g and 1.55 ± 0.42*
20
21
22 g, respectively) compared to control and Tf-CuNCs treated mice (11.28 ± 1.62 g and 10.14 ±
23
24 1.14 g, respectively) to that of initial body weight (Figure 7b). The changes in body weight, cell
25
26 viability and tumor volume in various treatment groups are illustrated in (Figure S19, Supporting
27
28
29 Information) and (Table S2, Supporting Information). The superior activity of Tf-CuNC-Dox-
30
31 NPs compared to free Dox can be attributed to the targeted internalization and longer circulation
32
33 time of Tf-Cu NC-Dox-NPs resulting in enhanced EPR effect. This superior therapeutic efficacy
34
35
36
of Tf-Cu NC-Dox-NPs has a direct impact on the increased life span of DLA mice as compared
37
38 to other treatment groups. In control and Tf-CuNCs treated groups, all mice died by 18 days of
39
40 post tumor development as shown in Figure 7c. Most importantly, though free Dox treatment
41
42
showed promising therapeutic effect on tumour size reduction; due to systemic toxicity issues the
43
44
45 life span of DLA mice were reduced as compared to that of Tf-CuNC-Dox-NPs treated mice.
46
47 The free DOX treated mice showed significant increase in mean survival time compared to
48
49 control and Tf-CuNC group, but periodical death of mice was observed by day 28 (Figure 7c).
50
51
52 Selective targeting efficacy of Tf-CuNC-Dox-NPs led to survival of mice till 50 days of study,
53
54 where systemic toxicity of Dox was greatly reduced as can be viewed from Figure 7c. Further,
55
56
57
58
59
60 ACS Paragon Plus Environment
29
ACS Applied Materials & Interfaces Page 30 of 41

1
2
3 morphological observation of DLA cells from various treatment groups confirmed that Tf-
4
5
6 CuNC-Dox-NPs induced maximal destruction of DLA cells over treatment with other group viz,
7
8 free Dox or Tf-CuNCs (Figure 8).
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39 Figure 8. FESEM images of (a) control DLA cells without treatment, and DLA cells after 16 days of
40
41 treatment with (b) Tf-CuNCs, (c) Dox and (d) Tf-CuNC-Dox- NPs.
42
43
44 In addition, DLA transplanted untreated mice showed increased levels of WBC and decline
45
46 of Hb and RBC counts (Table S3, Supporting Information). Tf-CuNCs administration showed
47
48
49
almost similar counts of those found in DLA transplanted untreated mice whereas free Dox
50
51 treatment showed an increase in RBC and Hb count towards the normal, and in parallel decrease
52
53 in WBC count. Interestingly, treatment with Tf-CuNC-Dox-NPs showed superior results and
54
55
displayed counts almost near to normal levels of WBC, RBC and Hb as shown in (Table S3,
56
57
58
59
60 ACS Paragon Plus Environment
30
Page 31 of 41 ACS Applied Materials & Interfaces

1
2
3 Supporting Information). Further, liver functions tests (LFT) were performed to assess the
4
5
6 hepatotoxicity in DLA mice under different treatment conditions. DLA induction in mice
7
8 significantly raises all the liver function marker enzymes (SGOT, SGPT & ALP) compared to
9
10 normal mice (Table S4, Supporting Information). Tf-CuNC administration did not show any
11
12
13
appreciable variation in these levels. However, free Dox treatment reduced these enzyme levels
14
15 compared to DLA mice, while the Tf-CuNC-Dox-NPs treatment showed their significant
16
17 reduction, which were close to normalcy.
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37 Figure 9. Histopathological examination images of main organs kidney (a-e) and liver (f-j), stained with
38 Hematoxylin and Eosin. The organs were collected on 17th day after treatment with Tf-CuNCs, Dox and
39
40 Tf-CuNC-Dox-NPs, respectively.
41
42
43 Histopathological examination of kidney of untreated DLA mice (Figure 9b) showed
44
45 glomerular atrophy, tubular congestion, infiltration of cells and deformed epithelial cells. Tf-
46
47 CuNC administration (Figure 9c) failed to improve the condition and showed pathological
48
49
50
observations similar to that of DLA mice. Free-Dox treatment (Figure 9d) demonstrated
51
52 significant improvement in the condition with decreased occurrence of tubular, glomerular and
53
54 blood vessel congestion. However, Tf-CuNC-Dox-NPs treated mice (Figure 9e) exhibited
55
56
57
58
59
60 ACS Paragon Plus Environment
31
ACS Applied Materials & Interfaces Page 32 of 41

1
2
3 significant reduction of tubular cast and congestion, epithelial desquamation, hyperaemia of
4
5
6 medullary part with normal glomerular structures, which resemble the normal renal physiology
7
8 of mice. Similarly, liver tissue sections of DLA mice (Figure 9f) showed abnormal hepatocytes
9
10 with polymorphic neutrophil infiltration, hepatic fibrillation, haemorrhage and perinuclear
11
12
13
clumping of cytoplasm. These histological features were also observed in Tf-CuNC treated mice
14
15 (Figure 9g). On the contrary, free Dox (Figure 9h) and Tf-CuNC-Dox-NPs treatment (Figure 9i)
16
17 significantly reduced the damages with cellular ultra-structures, which were comparable to
18
19
normal mice. Most importantly, hepatic physiology of DLA mice treated with Tf-CuNC-Dox-
20
21
22 NPs was comparable with normal mice.
23
24
25 Conclusion
26
27
28 In brief, a green aqueous method of synthesis of a novel nanodrug system in the form of Tf-
29
30
31 CuNC-Dox-NPs has been reported. The as-synthesized composite NPs was found to be
32
33 efficacious in targeted delivery of anticancer drug Dox to TfR receptor expressing cancer cells,
34
35 where uptake and release of Dox from the nanoparticulate system was monitored by FRET
36
37
38
assisted bioimaging. Additionally, inhibitor based experiments helped understand the cellular
39
40 uptake of the formulated nanodrug specifically in TfR overexpressed HeLa and MCF-7 cells
41
42 than the normal healthy cells, thereby elucidating their targeted imaging and therapeutic
43
44
potential. The work also highlighted the significance of synergistic activity of Tf-CuNC and Dox
45
46
47 in the formulated NPs and their important therapeutic efficacy on TfR positive DLA mice model.
48
49 To the best of our knowledge, this is the first report with Cu NCs, where high drug-loading
50
51 capacity, excellent tumour targeting specificity and monitoring of drug release based on FRET
52
53
54 assisted strategy have been combined for potential clinical applications in cancer theranostics.
55
56
57
58
59
60 ACS Paragon Plus Environment
32
Page 33 of 41 ACS Applied Materials & Interfaces

1
2
3 Associated Content:
4
5
6
7 Supporting Information: This material is available free of charge on ACS publications website
8
9 or from the authors. Characterisation details including UV-Vis, Photoluminescence (PL)
10
11 spectrum, Zeta Potential, TEM, FESEM. Cellular and animal study details like FACS, inhibition
12
13
14
assay, MTT and confocal studies.
15
16
17 Corresponding Author
18
19 Email: *arun@iitg.ernet.in,
20
21
22 Email: *sghosh@iitg.ernet.in
23
24 Present Addresses
25
26
27 Same as above
28
29
30 Author Contributions
31
32
AC conceived the idea, guided in execution of experiments and wrote the manuscript. SSG
33
34
35 contributed ideas and helped in writing the manuscript. UG performed the experiments,
36
37 contributed ideas and wrote the manuscript. AD contributed ideas and helped in synthesis and
38
39 characterisations and in writing the manuscript. AR performed the PCR and expression
40
41
42 experiments. RK and SK performed all the animal experiments and helped in writing the in vivo
43
44 experimental section of the manuscript.
45
46
47 Notes
48
49 The authors declare no competing financial interest.
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
33
ACS Applied Materials & Interfaces Page 34 of 41

1
2
3 ACKNOWLEDGMENT
4
5
6
Authors greatly acknowledge financial supports from the Department of Electronics and
7
8
9 Information Technology, Government of India (No. 5(9)/2012-NANO (Vol. II)) and the DBT
10
11 Programme Support (BT/PR13560/COE/34/44/2015) project. Authors also acknowledge the
12
13 instrumental support of the Centre for Nanotechnology, Centre for Environment (IIT Guwahati)
14
15
16 for atomic absorption spectroscopy, and the Central Instruments Facility at Indian Institute of
17
18 Technology Guwahati. We are thankful to Namami Goswami for helping us in taking FESEM
19
20 images and Mitradip Bhattacharjee for TEM images. We like to acknowledge Anil Bidkar and
21
22
23
Archita Ghoshal for their valuable suggestions. We also thank Prof. Jibon Kotoky for help in
24
25 animal experiments.
26
27
28 REFERENCES
29
30
(1) Kunjachan, S.; Detappe, A.; Kumar, R.; Ireland, T.; Cameron, L.; Biancur, D. E.; Motto-
31
32
33 Ros, V.; Sancey, L.; Sridhar, S.; Makrigiorgos, G. M.; Berbeco, R. I. Nanoparticle
34
35 Mediated Tumor Vascular Disruption: A Novel Strategy in Radiation Therapy. Nano Lett.
36
37 2015, 15 (11), 7488–7496.
38
39
40
41 (2) DeVita, V. T.; Young, R. C.; Canellos, G. P. Combination versus Single Agent
42
43 Chemotherapy: A Review of the Basis for Selection of Drug Treatment of Cancer. Cancer
44
45 1975, 35, 98–110.
46
47
48
49
(3) Zou, L.; Wang, H.; He, B.; Zeng, L.; Tan, T.; Cao, H.; He, X.; Zhang, Z.; Guo, S.; Li, Y.
50
51 Current Approaches of Photothermal Therapy in Treating Cancer Metastasis with
52
53 Nanotherapeutics. Theranostics 2016, 6 (6), 762–772.
54
55
56
57
58
59
60 ACS Paragon Plus Environment
34
Page 35 of 41 ACS Applied Materials & Interfaces

1
2
3 (4) Shao, K.; Singha, S.; Clemente-casares, X.; Tsai, S.; Yang, Y. Nanoparticle-Based
4
5
6 Immunotherapy. ACS Nano 2015, 9, 16–30.
7
8
9 (5) Vanneman, M.; Dranoff, G. Combining Immunotherapy and Targeted Therapies in Cancer
10
11 Treatment. Nat Rev Cancer 2012, 12 (4), 237–251.
12
13
14
15
(6) Zhang, F. L.; Song, M. R.; Yuan, G. K.; Ye, H. N.; Tian, Y.; Huang, M. D.; Xue, J. P.;
16
17 Zhang, Z. H.; Liu, J. Y. A Molecular Combination of Zinc(II) Phthalocyanine and
18
19 Tamoxifen Derivative for Dual Targeting Photodynamic Therapy and Hormone Therapy.
20
21
J. Med. Chem. 2017, 60 (15), 6693–6703.
22
23
24
25 (7) Ghoshal, A.; Goswami, U.; Sahoo, A. K.; Chattopadhyay, A.; Ghosh, S. S. Targeting Wnt
26
27 Canonical Signaling by Recombinant sFRP1 Bound Luminescent Au-Nanocluster
28
29 Embedded Nanoparticles in Cancer Theranostics. ACS Biomater. Sci. Eng. 2015, 1 (12).
30
31
32 1256-1266.
33
34
35 (8) Ghoshal, A.; Goswami, U.; Raza, A.; Chattopadhyay, A.; Ghosh, S. S. Recombinant
36
37 sFRP4 Bound Chitosan-Alginate Composite Nanoparticles Embedded with Silver
38
39
40
Nanoclusters for Wnt/β-Catenin Targeting in Cancer Theranostics. RSC Adv. 2016, 6 (89),
41
42 85763-85772.
43
44
45 (9) Rao, W.; Wang, H.; Han, J.; Zhao, S.; Dumbleton, J.; Agarwal, P.; Zhang, W.; Zhao, G.;
46
47
Yu, J.; Zynger, D. L.; Lu, X.; He, X. Chitosan-Decorated Doxorubicin-Encapsulated
48
49
50 Nanoparticle Targets and Eliminates Tumor Reinitiating Cancer Stem-like Cells. ACS
51
52 Nano 2015, 9 (6), 5725–5740.
53
54
55 (10) Kamimura, M.; Furukawa, T.; Akiyama, S.; Nagasaki, Y. Enhanced Intracellular Drug
56
57
58
59
60 ACS Paragon Plus Environment
35
ACS Applied Materials & Interfaces Page 36 of 41

1
2
3 Delivery of pH-Sensitive Doxorubicin/poly(ethylene Glycol)-Block-poly(4-
4
5
6 Vinylbenzylphosphonate) Nanoparticles in Multi-Drug Resistant Human Epidermoid KB
7
8 Carcinoma Cells. Biomaterials Science 2013, 1 (4), 361-367.
9
10
11 (11) Wang, F.; Wang, Y.; Dou, S.; Xiong, M.; Sun, T.; Wang, J.; Al, W. E. T. Doxorubicin-
12
13
14
Tethered Responsive Gold Nanoparticles Facilitate Intracellular Drug Delivery for
15
16 Overcoming Multidrug Resistance in Cancer Cells. ACS Nano 2011, 5, 3679–3692.
17
18
19 (12) Kong, F.; Zhang, X.; Zhang, H.; Qu, X.; Chen, D.; Servos, M.; Mäkilä, E.; Salonen, J.;
20
21
Santos, H. A.; Hai, M.; Weitz, D. A. Inhibition of Multidrug Resistance of Cancer Cells
22
23
24 by CO-Delivery of DNA Nanostructures and Drugs Using Porous Silicon
25
26 Nanoparticles@giant Liposomes. Adv. Funct. Mater. 2015, 25 (22), 3330–3340.
27
28
29 (13) Li, W. Q.; Wang, Z.; Hao, S.; He, H.; Wan, Y.; Zhu, C.; Sun, L. P.; Cheng, G.; Zheng, S.
30
31
32 Y. Mitochondria-Targeting Polydopamine Nanoparticles to Deliver Doxorubicin for
33
34 Overcoming Drug Resistance. ACS Appl. Mater. Interfaces 2017, 9 (20), 16793–16802.
35
36
37 (14) Yu, H.; Cui, Z.; Yu, P.; Guo, C.; Feng, B.; Jiang, T.; Wang, S.; Yin, Q.; Zhong, D.; Yang,
38
39
40
X.; Zhang, Z.; Li, Y. PH- and NIR Light-Responsive Micelles with Hyperthermia-
41
42 Triggered Tumor Penetration and Cytoplasm Drug Release to Reverse Doxorubicin
43
44 Resistance in Breast Cancer. Adv. Funct. Mater. 2015, 25 (17), 2489–2500.
45
46
47
(15) Luo, W.; Wen, G.; Yang, L.; Tang, J.; Wang, J.; Wang, J.; Zhang, S.; Zhang, L.; Ma, F.;
48
49
50 Xiao, L.; Wang, Y.; Li, Y. Dual-Targeted and pH-Sensitive Doxorubicin Prodrug-
51
52 Microbubble Complex with Ultrasound for Tumor Treatment. Theranostics 2017, 7 (2),
53
54 452–465.
55
56
57
58
59
60 ACS Paragon Plus Environment
36
Page 37 of 41 ACS Applied Materials & Interfaces

1
2
3 (16) Gao, H.; Bi, Y.; Chen, J.; Peng, L.; Wen, K.; Ji, P.; Ren, W.; Li, X.; Zhang, N.; Gao, J.;
4
5
6 Chai, Z.; Hu, Y. Near-Infrared Light-Triggered Switchable Nanoparticles for Targeted
7
8 Chemo/Photothermal Cancer Therapy. ACS Appl. Mater. Interfaces 2016, 8 (24), 15103–
9
10 15112.
11
12
13
14
(17) Luo, D.; Carter, K. A.; Miranda, D.; Lovell, J. F. Chemophototherapy: An Emerging
15
16 Treatment Option for Solid Tumors. Adv. Sci. 2017, 4 (1), 1–24.
17
18
19 (18) Yu, M. K.; Park, J.; Jon, S. Targeting Strategies for Multifunctional Nanoparticles in
20
21
Cancer Imaging and Therapy. Theranostics 2012, 2 (1), 3–44.
22
23
24
25 (19) Falagan-Lotsch, P.; Grzincic, E. M.; Murphy, C. J. New Advances in Nanotechnology-
26
27 Based Diagnosis and Therapeutics for Breast Cancer: An Assessment of Active-Targeting
28
29 Inorganic Nanoplatforms. Bioconjugate. Chem. 2016, 28,135-152.
30
31
32
33 (20) Lim, E.-K.; Kim, T.; Paik, S.; Haam, S.; Huh, Y.-M.; Lee, K. Nanomaterials for
34
35 Theranostics: Recent Advances and Future Challenges. Chemical Reviews 2015, 115 (1),
36
37 327–394.
38
39
40
41
(21) Su, Y. L.; Fang, J. H.; Liao, C. Y.; Lin, C. T.; Li, Y. T.; Hu, S. H. Targeted Mesoporous
42
43 Iron Oxide Nanoparticles-Encapsulated Perfluorohexane and a Hydrophobic Drug for
44
45 Deep Tumor Penetration and Therapy. Theranostics 2015, 5 (11), 1233–1248.
46
47
48
(22) Tao, Y.; Li, M.; Ren, J.; Qu, X. Metal Nanoclusters: Novel Probes for Diagnostic and
49
50
51 Therapeutic Applications. Chem. Soc. Rev. 2015, 44, 8636–8663.
52
53
54 (23) Sahoo, A. K.; Goswami, U.; Dutta, D.; Banerjee, S.; Chattopadhyay, A.; Ghosh, S. S.
55
56
57
58
59
60 ACS Paragon Plus Environment
37
ACS Applied Materials & Interfaces Page 38 of 41

1
2
3 Silver Nanocluster Embedded Composite Nanoparticles for Targeted Prodrug Delivery in
4
5
6 Cancer Theranostics. Biomater. Sci. Eng. 2016, 2,1395-1402.
7
8
9 (24) Zhou, F.; Feng, B.; Yu, H.; Wang, D.; Wang, T.; Liu, J.; Meng, Q.; Wang, S.; Zhang, P.;
10
11 Zhang, Z.; Li, Y. Cisplatin Prodrug-Conjugated Gold Nanocluster for Fluorescence
12
13
14
Imaging and Targeted Therapy of the Breast Cancer. Theranostics 2016, 6 (5), 679–687.
15
16
17 (25) Sun, J.; Jin, Y. Fluorescent Au Nanoclusters: Recent Progress and Sensing Applications.
18
19 J. Mater. Chem. C 2014, 2 (38), 8000–8011.
20
21
22
(26) Zhou, M.; Tian, M.; Li, C. Copper-Based Nanomaterials for Cancer Imaging and Therapy.
23
24
25 Bioconjug. Chem. 2016, 27 (5), 1188–1199.
26
27
28 (27) Song, X.-R.; Goswami, N.; Yang, H.-H.; Xie, J. Functionalization of Metal Nanoclusters
29
30 for Biomedical Applications. Analyst 2016, 141 (11), 3126–3140.
31
32
33
34 (28) Chen, N.-T.; Cheng, S.-H.; Liu, C.-P.; Souris, J.; Chen, C.-T.; Mou, C.-Y.; Lo, L.-W.
35
36 Recent Advances in Nanoparticle-Based Förster Resonance Energy Transfer for
37
38 Biosensing, Molecular Imaging and Drug Release Profiling. Int. J. Mol. Sci. 2012, 13
39
40
41
(12), 16598–16623.
42
43
44 (29) Wang, S.; Deng, H.; Huang, P.; Sun, P.; Huang, X.; Su, Y.; Zhu, X.; Shen, J.; Yan, D.
45
46 Real-Time Self-Tracking of an Anticancer Small Molecule Nanodrug Based on Colorful
47
48
Fluorescence Variations. RSC Adv. 2016, 6 (15), 12472–12478.
49
50
51
52 (30) Wang, J.; Gao, P. P.; Yang, X. X.; Wang, T. T.; Wang, J.; Huang, C. Z. Real-Time
53
54 Imaging of Intracellular Drug Release from Mesoporous Silica Nanoparticles Based on
55
56
57
58
59
60 ACS Paragon Plus Environment
38
Page 39 of 41 ACS Applied Materials & Interfaces

1
2
3 Fluorescence Resonance Energy Transfer. J. Mater. Chem. B 2014, 2 (27), 4379-4386.
4
5
6
7 (31) Chattoraj, S.; Amin, M. A.; Jana, B.; Mohapatra, S.; Ghosh, S.; Bhattacharyya, K.
8
9 Selective Killing of Breast Cancer Cells by Doxorubicin Loaded Fluorescent Gold Nano-
10
11 Cluster: Confocal Microscopy and FRET. ChemPhysChem 2016, 17, 253-259.
12
13
14
15
(32) Tang, J.; Kong, B.; Wu, H.; Xu, M.; Wang, Y.; Wang, Y.; Zhao, D.; Zheng, G. Carbon
16
17 Nanodots Featuring Efficient FRET for Real-Time Monitoring of Drug Delivery and
18
19 Two-Photon Imaging. Adv. Mater. 2013, 25 (45), 6569–6574.
20
21
22
(33) Ghosh, R.; Goswami, U.; Ghosh, S. S.; Paul, A.; Chattopadhyay, A. Synergistic
23
24
25 Anticancer Activity of Fluorescent Copper Nanoclusters and Cisplatin Delivered through
26
27 a Hydrogel Nanocarrier. ACS Appl. Mater. Interfaces 2015, 7, 209-222.
28
29
30 (34) Laha, D.; Pramanik, A.; Chattopadhyay, S.; Dash, S. kumar; Roy, S.; Pramanik, P.;
31
32
33 Karmakar, P. Folic Acid Modified Copper Oxide Nanoparticles for Targeted Delivery in
34
35 in Vitro and in Vivo Systems. RSC Adv. 2015, 5 (83), 68169–68178.
36
37
38 (35) Wang, Y.; Chen, J.; Yan, X. Fabrication of Transferrin Functionalized Gold Nanoclusters/
39
40
41
Graphene Oxide Nanocomposite for Turn-On Near-Infrared Fluorescent Bioimaging of
42
43 Cancer Cells and Small Animals. Anal.Chem. 2013, 85, 2529-2535.
44
45
46 (36) Pulakkat, S.; Balaji, S. A.; Rangarajan, A.; Raichur, A. M. Surface Engineered Protein
47
48
Nanoparticles with Hyaluronic Acid Based Multilayers for Targeted Delivery of
49
50
51 Anticancer Agents. ACS Appl. Mater. Interfaces 2016, 8 (36), 23437–23449.
52
53
54 (37) Cheng, W.; Nie, J.; Xu, L.; Liang, C.; Peng, Y.; Liu, G.; Wang, T.; Mei, L.; Huang, L.;
55
56
57
58
59
60 ACS Paragon Plus Environment
39
ACS Applied Materials & Interfaces Page 40 of 41

1
2
3 Zeng, X. A pH-Sensitive Delivery Vehicle Based on Folic Acid-Conjugated
4
5
6 Polydopamine-Modified Mesoporous Silica Nanoparticles for Targeted Cancer Therapy.
7
8 ACS Appl. Mater. Interfaces 2017, 9, 18462-18473.
9
10
11 (38) Choi, C. H. J.; Alabi, C. A.; Webster, P.; Davis, M. E. Mechanism of Active Targeting in
12
13
14
Solid Tumors with Transferrin-Containing Gold Nanoparticles. Proc. Natl. Acad. Sci. U.
15
16 S. A. 2010, 107 (3), 1235–1240.
17
18
19 (39) Zhao, T.; He, X.-W.; Li, W.-Y.; Zhang, Y.-K. Transferrin-Directed Preparation of Red-
20
21
Emitting Copper Nanoclusters for Targeted Imaging of Transferrin Receptor over-
22
23
24 Expressed Cancer Cells. J. Mater. Chem. B 2015, 3 (11), 2388–2394.
25
26
27 (40) Sriram, M. I.; Kanth, S. B. M.; Kalishwaralal, K.; Gurunathan, S. Antitumor Activity of
28
29 Silver Nanoparticles in Dalton’s Lymphoma Ascites Tumor Model. Int. J. Nanomedicine
30
31
32 2010, 5 (1), 753–762.
33
34
35 (41) Aasa, R.; Aisen, P. An Electron Paramagnetic Resonance Study of the Iron and Copper
36
37 Complexes of Transferrin. J.Biol.Chem. 1968, 243 (9), 2399–2404.
38
39
40
(42) Welch, S. Transferrin: The Iron Carrier, CRC Press, Inc.: Florida, 1992, pp 70-72.
41
42 (43) Foucquier, J.; Guedj, M. Analysis of Drug Combinations: Current Methodological
43
44 Landscape. Pharmacol. Res. Perspect. 2015, 3, e00149.
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
40
Page 41 of 41 ACS Applied Materials & Interfaces

1
2
3
4
5
6
7 Table of Contents
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
41

You might also like