Curcumin in combination with anti-cancer drugs: A nanomedicine review

Curcumin in combination with anti-cancer drugs: A nanomedicine review

Accepted Manuscript Title: Curcumin in Combination with Anti-Cancer Drugs: A Nanomedicine Review Authors: Harshul Batra, Shrikant Pawar, Dherya Bahl P...

2MB Sizes 0 Downloads 29 Views

Accepted Manuscript Title: Curcumin in Combination with Anti-Cancer Drugs: A Nanomedicine Review Authors: Harshul Batra, Shrikant Pawar, Dherya Bahl PII: DOI: Reference:

S1043-6618(18)30436-5 https://doi.org/10.1016/j.phrs.2018.11.005 YPHRS 4054

To appear in:

Pharmacological Research

Received date: Revised date: Accepted date:

28 March 2018 31 October 2018 4 November 2018

Please cite this article as: Batra H, Pawar S, Bahl D, Curcumin in Combination with Anti-Cancer Drugs: A Nanomedicine Review, Pharmacological Research (2018), https://doi.org/10.1016/j.phrs.2018.11.005 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Curcumin in Combination with Anti-Cancer Drugs: A Nanomedicine Review

1

2

Neuroscience Institute & Center for Behavioral Neuroscience, Georgia State University, 789 Petit Science Center, Atlanta, GA, 30303

Department of Computer Science, Georgia State University, 34 Peachtree Street, Atlanta, GA, 30303. 3

Department of Biology, Georgia State University, 34 Peachtree Street, Atlanta, GA, 30303.

.

U

Division of Pharmaceutics and Translational Therapeutics, University of Iowa, Iowa City, Iowa 52242

N

4

SC RI PT

Harshul Batra1, Shrikant Pawar2, 3, and Dherya Bahl4

A

Article Type: Review.

M

*Correspondence author:

D

Dr. Harshul. Batra, Neuroscience Institute & Center for Behavioral Neuroscience, Georgia State University, 789 Petit Science Center Atlanta, GA, 30303.

A

CC

EP

Graphical abstract

TE

Email address: [email protected]

1

Abstract

2

D

TE

EP

CC

A

SC RI PT

U

N

A

M

A huge surge of research is being conducted on combination therapy with anticancer compounds formulated in the form of nanoparticles (NPs). Numerous advantages like dose minimalization and synergism, reversal of multi drug resistance (MDRs), enhanced efficacy have emerged with

SC RI PT

nanoencapsulation of chemotherapeutic agents with chemo-sensitizing agent like curcumin. Within last couple of years various nano-sized formulations have been designed and tested both in vitro with cell lines for different types of cancers and in vivo with cancer types and drug resistance models. Despite the combinatorial models being advanced, translation to human trials has not been as smooth as one would have hoped, with as few as twenty ongoing clinical trials

U

with curcumin combination, with less than 1/10th being nano-particulate formulations. Mass

N

production of nano-formulation based on their physico-chemical and pharmacokinetics deficits

A

poses as major hurdle up the ladder. Combination of these nano-sized dosage with poorly

M

bioavailable drugs, unspecific target binding ability and naturally unstable curcumin further complicates the formulation aspects. Emphasis is now therefore being laid on altering natural

D

forms of curcumin and usage of formulations like prodrug or coating of curcumin to overcome

TE

stability issues and focus more on enhancing the pharmaceutical and therapeutic ability of the

EP

nano-composites. Current studies and futuristic outlook in this direction are discussed in the review, which can serve as the basis for upcoming research which could boost commercial

CC

translational of improved nano-sized curcumin combination chemotherapy.

A

Abbreviations

MDR, multi drug resistance; NPs, nanoparticles; PTX, paclitaxel, Dox, doxorubicin; P-gp, pglycoprotein; PEG, polyethylene glycol; PLGA, poly(lactic-co-glycolic acid); PLA, polylactic acid; BTZ, Bortezomib; PLACL, poly L-lactic acid-co-ε-caprolactone; DDP, cisplatin Keywords: Curcumin, Nano-formulations, combination therapy, Cancer, Drug Delivery

3

1. Introduction One-fifth of the deaths worldwide annually are caused by various type of cancers [1]. Cancer is a

SC RI PT

result of successive genetic and epigenetic alterations resulting in apoptosis, uncontrolled cell proliferation, metastasis, and angiogenesis [2, 3]. Several strategies have been employed in recent years to combat cancer evasion, including surgery, hormonal therapy, chemotherapy, and radiation therapy. Chemotherapy which involves usage of cytotoxic antineoplastic drugs (alkylating agents and antimetabolites), although effective in most cases, has immense side-

U

effects and lead to drug resistance (intrinsic and acquired). Acquired resistance is considered

N

more severe and is caused by mutations induced by over-expression of therapeutic targets or

A

stimulation of cancer-promoting pathway. Also, tumors contain a high degree of molecular

M

heterogeneity leading to enhanced drug resistance [4].

D

Curcumin is a natural phenolic antioxidant [5]. At high concentrations, it functions as

TE

cardioprotective, neuroprotective and antidiabetic agent [6-8]. It has shown pharmacological effects against diseases like type II diabetes, Alzheimer's disease, atherosclerosis and human

EP

immunodeficiency virus (HIV) replication [6-8]. Anti-cancer activity of curcumin has been

CC

extensively investigated recently, and significant improvements in gastrointestinal, melanoma, genito-urinary, breast, and lung cancers have been seen [9-12] (Insert Figure 1 here). It has

A

pleiotropic properties, due to which it is seen to be most effective against single pathway targeted cancers [13, 14] (Insert Figure 2 here). It has also shown anti-inflammatory activity with a tolerance dose of at 12 g/day [15, 16]. The significance of curcumin to be considered as a possible drug lies in its free passage through cellular membranes due to its high lipophilicity. However, it does have low aqueous solubility making it susceptible to alkaline degradation. This 4

could be a probable reason for its low bioavailability and an increased dose for reaching therapeutic optimal blood concentrations [17-20]. Almost half a century ago, nanomedicine emerged as a specific niche for drug delivery, within

SC RI PT

the multidisciplinary field of nanotechnology. A principal focus for the last 20 years has been on the development of nano-formulation driven drug delivery of nanocarriers [21, 22]. Various nanocarriers that have been investigated for drug delivery [21, 22] include polymeric micelles, liposomes,

magnetic

nanoparticles,

conjugates,

and

peptide

carriers.

Integration

of

Nanotechnology within cancer research has proved to be advantageous in several ways,

U

including (a) cancer treatment and detection (diagnostics and imaging agents); (b) biomarker

N

identification for disease prediction; and (c) mechanism of cancer progression [23-25]. A narrow

A

therapeutic window of drugs however is one of the major challenges with anticancer drugs and is

M

the reason for serious side effects due to non-specific drug uptake by healthy cells. The efficacy

D

of cancer drugs is also restrained due to multidrug resistance (MDR), often associated with

TE

chemotherapy. To overcome these issues the primary strategy of combination chemotherapy along with drug delivery systems using nanoparticles (NPs) is being actively explored [26].

EP

Curcumin could be a potential elementary candidate to be used for combination chemotherapy as

CC

it could overcome the issues associated with anti-cancer therapeutics. This article tries to summarize current research on combination of curcumin with several anticancer compounds

A

with different mechanisms, as a possible target for treating various cancers. We provide a global overview of the role of nanoparticles (NPs) in oncology and different NPs currently used in cancer clinically. We briefly describe advantages of combination therapy in nanomedicine and summarize the current data on curcumin nano-formulations with an emphasis on its effect in invitro and in-vivo studies. Finally, we discuss futuristic clinical outlook, challenges, and 5

perspectives of developing nanoparticle-based curcumin combination therapy in cancer. This work envisions to serve as an up to date comprehensive literature basis, which could be utilized to build upon any future studies for better understanding of mechanism and ADME of curcumin

SC RI PT

along with a selection of proper combinational drug and fabrication of appropriate delivery technology. This review could be used as the go-to reference, to begin with in-vitro and in-vivo studies for successful development and in-clinic adaptation of nano-sized curcumin combination therapy.

U

2. Nanoparticles in oncology

N

Nanomedicine today is an entire field of research and development, with oncology as one of its

A

major application. Lack of specificity is a primary drawback of cancer therapeutics for which

M

nano-strategies based designing of personalized cancer treatment could be a solution. Recent decades have noticed emergence and approval of several nanoparticulate dosage forms by U.S.

D

Food and Drug Administration (FDA). They overpower traditional delivery techniques with

TE

improved drug delivery, reduced toxicity, better safety profile, targeted therapy and extended

EP

product life-cycles [27].

CC

2.1. Type of Nanoparticles in Cancer therapy There are two categories of therapeutic and diagnostic nanoparticles: (a) organic (e.g., polymeric,

A

liposomes, micelles, etc. (b) inorganic (e.g., gold, silica, iron oxide, etc.). Natural or synthetic organic molecules are the template for the formation of organic NPs. Organic and inorganic NPs differ in their technique of fabrication. For organic NPs, the encapsulation technique of biodegradable materials used are relatively simple and require several self-organizing or chemical binding organic molecules. Whereas, inorganic NPs involve precipitation of inorganic 6

salts in which atoms are often linked by covalent/metallic/magnetic bonding leading to formation of a three-dimensional array. Broad applications involving organic NPs in the clinic include formulations for hemostasis, vaccination, long-lasting depot delivery systems, and topical agents

SC RI PT

for systemic delivery through the skin [28-30]. Whereas inorganic nanoparticles are typically used in developing formulations for thermal ablation of tumors, intraoperative sentinel lymph node imaging, imaging applications, and anemia treatment [31, 32]. Intravenous organic NP formulations are characterized into two categories: (a) gene therapy applications [33, 34], or (b) small molecule drug delivery for cancer treatment (e.g., head and neck, melanoma, breast,

U

metastatic, etc.) [35, 36]. Nanoparticle technologies defeat free drug counterparts as a drug-

N

delivery system with an extended circulation of a drug, improved targeting to disease tissues,

A

enhanced drug protection, and controlled release [37, 38]. These advantages make the

M

nanoparticulate delivery system a potential model to dramatically change clinical care by

D

improving current therapies or avail new treatment options (Insert Figure 3 here).

TE

3. Combination chemotherapy in cancer

EP

The interconnected pathways in cancer physiology reduce effectivity of monotherapy strategy. Drug resistance and chances of tumor recurrence due to pathway overlapping [39], cross-talk

CC

[40] and neutralizing response [41, 42] could be various complications hindering full potential of independent drugs. The easiest approach to overcome this issue could involve utilization of

A

combination chemotherapy strategy, which has shown to be successful in preliminary clinical trials. Combination chemotherapy design involves an understanding of several principles like non-overlapping toxicity, non-cross resistance, and enhanced tumor cell killing efficacy [43]. Synergistically acting drugs in vitro are governed by molar ratios, vs. Maximal Tolerated Dose

7

(MTD) govern drug ratios in-vivo [44, 45]. Nanocarriers like liposomes and polymeric micelles could further help to overcome mono-therapeutic complications. A previous generation of cancer combination chemotherapy comprised of traditional drug combinations including anthracycline,

SC RI PT

methotrexate, and paclitaxel (PTX)-based combinations [46]. A study on one such combination displayed an effective reversal of chemotherapeutic drug resistance with Dox and rapamycin codelivery. This combination leads to complete tumor remission, as compared to dox and rapamycin alone [47]. In another such study, in vivo effects of curcumin combination with antitumor drug was studied and displayed effectively. Docetaxel (DTX) and Curcumin (CUR)

U

co-encapsulated lipid nanoparticles (LPN’s) were evaluated on PC3 tumor xenografts in mice

N

(human prostate cancer-bearing Balb/c nude mice model). These potent nanoparticles inhibited

A

tumor volume growth significantly, when compared to other groups, with no visible side effects.

M

It was concluded that this combination could prove to be an effective prostate cancer treatment [166] (insert Figure 4 here). Additionally, nanoparticulate delivery for combination

D

chemotherapy could benefit hydrophobic and hydrophilic drug codelivery [48], spatiotemporal

TE

control release behavior, and ratio-metric drug loading adjustments [49, 50]. The focus of this

EP

therapy lies in exploring new combinations to provide better insight into molecular and cellular

CC

mechanisms of cancer treatment.

One of the most challenging approaches towards treating cancers is overcoming MDR. The

A

major mechanism of resistance of cancer cells to drugs includes altered apoptosis pathway and overexpression of multidrug transporters [50, 51]. A powerful approach to disrupt MDR and to overcome the negative impact of the anticancer drugs is by co-delivery of chemo-sensitizing and chemotherapeutic agents. One of the major player involved in MDR is efflux of a variety of anticancer drugs by efflux transporter p-glycoprotein (P-gp), which is the adenosine triphosphate 8

(ATP)-binding cassette transporters, encoded by the MDR-1 gene [52]. In liver, pancreatic, gastrointestinal, and ovarian cancer, overexpression of P-gp leads to the reduction in the efficacy of drugs such as vinblastine, doxorubicin, vinblastine, paclitaxel, etc. [53].

In apoptotic

of nuclear factor kappa B (NF-kB) or BCL-2 [54].

SC RI PT

pathway-dependent MDR, the apoptotic response to anticancer drugs dampens by deregulation To combat MDR in cancer, several

nanoparticle formulations are designed to include the combination of drugs with chemosensitizers, MDR cytotoxic, and modulating agents. A Doxorubicin (DOX)-curcumin composite nanoparticle formulation called NanoDoxCurc (NDC) for overcoming DOX

U

resistance was developed by Pramanik et al. [168]. Its effectivity in the form of inhibited MDR

N

phenotype, is tested in athymic nude mice (DOX resistant) for several models of DOX resistant

A

cancers (multiple myeloma, acute leukemia, prostate and ovarian cancers). Additionally, reduced

M

cardiac toxicity and bone marrow suppression was further indicated with this course of

D

treatment, which was subjected to reduction in DOX induced oxidative stress (Figure 5).

TE

In the last decade, NPs with multiple chemotherapeutic agents, each with a different mechanism of action forms a basis of combination drug therapy. This approach helps in reducing unwanted

EP

side effects. Specific interactions with a target sites for a different type of cancer is the main

CC

advantage of nanomedicine combination therapy. Previously, most combination NPs contained drugs with similar water solubility. Polymeric nanoparticles in their hydrophobic core carry poor

A

water-soluble drugs and liposomes contain hydrophilic drugs. Whereas in recent times development of more sophisticated NP drug delivery system has made it possible to deliver hydrophobic and hydrophilic drugs in one system. Several challenges in delivering drugs with different physiochemical attributes remain to be addressed warranting future studies.

9

3.1. Advantages of combination therapy For a rational design to achieve optimal efficacy, a proper understanding of specific mechanisms of parent drug and combination strategies used are essential. Combination of two drugs can

SC RI PT

generate a combinatorial effect with lower, equal or higher than the total effects of the individual partner drugs. Combination effects could be synergistic, antagonistic or potentiation. The synergistic combinatorial effect is when the effect produced is larger than the summed effect of the drugs individually, with no cross-reactivity and different acting target sites or pathways. When the effect is greater than or equal to the summed effect of the individual drugs with a final

U

target as same site or pathway, the effect is additive. In potentiation effects, alteration of

N

activity/effect of one drug by the other is seen. With enhanced activity, a reduction in side effect

A

via regulation of ADME properties is also seen [46, 55, 56]. A nanoparticulate combination has

M

explored all these effects to achieve a variety of favorable outcomes, including reduction of

D

unwanted side effects, drug-resistance prevention, a decrease in dose of individual drugs, and

TE

enhanced efficacy (Insert Figure 6 here)

EP

3.2. Formulas used for determining combinatorial effects

CC

Combination Index (CI) = D1/Dm1 + D2/Dm2

A

Where D1 and D2 represent the dose of a drug in combined administration, Dm1 and Dm2 represent the dose required to produce the same effect when used alone. When CI < 1, synergism is indicated; CI = 1, additive nature is indicated; CI > 1, mean antagonism [57].

Reversal fold (RF) =

IC50 anticancer drug alone 10

IC50 anticancer drug + modulator

RF > 1 indicates a synergistic effect, RF = 1 indicates no effect, and RF < 1 indicates an

SC RI PT

antagonistic effect [58]. (IC50 anticancer drug alone in MDR cells - IC50 anticancer drug + modulator in MDR cells) Relative Reversal Rate

(IC50 anticancer drug alone in MDR cells - IC50 anticancer (RRR %) [59]

=

* 100

U

drug alone in parental cells)

N

In a recent study by Gwade et al [167], synergistic anticancer activity of paclitaxel (PTX) and

A

curcumin (CUR) combination was noted against ovarian and cervical cancers. The authors

M

performed encapsulation of BSA (bovine serum albumin) nanoparticles which encapsulated PTX and CUR along with a folate receptor ligand conjugation on the nanoparticles. This formulation

D

is reported to possess a synergistic cell killing effect and combination effectivity compared to

TE

control groups, when studied in HeLa and SKOV3 Cell lines. The CI values were reported to be

EP

less than 0.5 indicating an average synergism, as indicated in Figure 7. High potency of the drugs and different anticancer targeting pathways, make these two drugs apt candidates for

CC

combination, which then results in lowered dose levels and synergism in their combined

A

therapeutics. Other such studies have also been indicated elsewhere [169]

4. Curcumin combination nanomedicine Curcumin has shown to be successful in several types of cancer lines, mainly because of its ubiquitous action on different modulator of anti-cancer effects (Insert Figure 8 here). Curcumin

11

alone or in combination is nontoxic and is proposed to accentuate the therapeutic efficiency of chemotherapeutics by inhibiting ABC efflux transporter. Curcumin inhibits tumor growth by arresting cell cycle progression, inducing apoptosis, inhibiting the expression of antiapoptotic

SC RI PT

proteins, inhibiting multiple cell survival signaling pathways and their cross-communication, and modulating immune responses [4, 60, 61]. All these properties make curcumin a promising drug for mono or combination therapy. Table 1 summarizes a few examples of curcumin monotherapy. Although its pharmacokinetic profile is not as appealing as pharmacological profile, nano-based formulations could be a savior and balance this out. Its modulating effects on

U

P-gp efflux transporters, which are a major cause of MDR in the first place, makes it a perfect

N

adjunct for a combination [62]. Curcumin combination nanoparticulate therapeutic doses tested

A

in-vitro and in-vivo for several types of cancers are listed in Table 1.

M

Table 1: Selected examples of Curcumin nano-particulate delivery system’s monotherapy in

Nanoparticle platform Hyaluronic Acid linked Nanomicelles

2.

PEG-PLGA NPs

Cancer type

TE

Serial no. 1.

D

cancers

Pancreatic cancer

89.5±2.8

Breast cancer

70-300 nm

EP

CC

Size

Polymeric micelles

Colon cancer

27.6± 0.7nm

4.

pH-chitosan

Glioblastoma

Not reported

A

3.

mesoporous

12

Status of investigation In vitro

Observations

Ref

Improved cell killing and internalization in MiaPaca-2 (IC50= 140±2.42 nM ) and AsPc-1(160±2.96 nM) cell line.

[63]

In vitro

Improved cell growth inhibition in MCF-7 (17.86±1.08 µM) cell lines with the formulation.

[64]

In vitro/in vivo

The cytotoxic activity of the nanoformulation was higher than free curcumin. (5.50μg/mL). 70% inhibition of tumor growth in mice inoculated with MCF-7 cells. MTT assay for the formulation showed a higher IC50 value of 5.21 µg/mL for

[65]

In vitro

[66]

silica nanoparticles

the formulation as compared to non-pH and free drug group.

PLGA-PEGFe3O4 NPs

Lung cancer

Not reported

In vitro

Enhanced cytotoxicity observed in A549 with the formulation (IC50=7.3 µM). hTERT downregulated.

[67]

6.

folate-BSA-CDF NPs

Ovarian cancer

279 nm

In vitro

High cell viability (IC50=0.25µM) and cellular uptake compared to free drug and non-targeted nanoformulation.

[68]

7.

Cholesterol-PLA polymeric micelles

Breast cancer

Nearly 1.87 fold higher tumor growth inhibition in B16F10-xenografted tumorbearing mice with the polymeric micelles formulation (627.72 ± 0.9 mm3) as compared to free CUR (1174.68 ± 1.64 mm3).

[69]

8.

Nanoemulsions

Prostate cancer

The results showed that the cell update and cytotoxicity considerably increased with Cur nanoemulsions compared to free Cur. Cur nanoemulsions exhibited a significantly prolonged biological activity and demonstrated better therapeutic efficacy than free Cur.

[70]

A

N

U

In vitro/in vivo

SC RI PT

5.

In vitro

CC

EP

TE

D

M

34.54±2. 2 nm

4.1. Liver cancer

A

Liver cancer is the second most common cause of cancer death among men and the sixth leading cause of cancer death among women. Globally, Hepatocellular Carcinoma (HCC) accounts for 75-80 % of the cancer deaths. Due to metastasis and recurrence, a 5-year survival rate of patients with HCC is at 15 % [71]. Hu et al. investigated the combined effect of nanoparticulate delivery of curcumin (Cur) with a kinase inhibitor, sorafenib, in the treatment of HCC. The combination 13

down-regulated expression of MMP9 via NF-κB/p65 signaling pathway. Furthermore, the population of cancer-initiating cells CD133-positive significantly decreased in both MHCCLM3 (4.82 ± 1.22%) and Huh7 (5.46 ± 0.68%) cells with this combination therapy. In vivo outcomes

SC RI PT

demonstrated lung metastatic tumors to be significantly reduced compared with control treatment (16.7% vs. 100%; P = 0.015) [72]. Combining chemotherapeutic agent and chemosensitizer in nanocarriers to overcome MDR in HCC was a focus of Zhao et al. research. Synthesized DOX/Cur-NPs increased Caspase-3 and Bax/Bcl-2 ratio, and decreased C-myc, PCNA and VEGF as seen in liver tissue. With DOX treatment there was a 46.44-fold increase in Resistance

U

Index (RI) of BEL 7402/5-FU cells in comparison with the BEL 7402 cells, significantly

N

decreased by DOX/Cur-NPs (2.07). This indicates a modulatory effect of DOX/Cur-NPs on

A

MDR. The modulation effect calculated by the reversal fold (RF) value in which IC50 of DOX-

M

NPs in BEL 7402/5-FU cells divided by that of DOX/Cur-NPs. The DOX/Cur-NPs showed an

D

RF value of 1.94, indicative of a synergistic effect [73].

TE

Yan et al. developed glycyrrhetinic acid (GA)-modified chitosan-cystamine-poly(є-caprolactone) copolymer (PCL -SS-CTS-GA) micelle for co-delivery of DOX and curcumin to HCC. GA aided

EP

in target specificity to hepatocytes due to its affinity to the liver membrane [74, 75]. In HepG2

CC

and HUVEC cell lines, PCL-SS-CTS-GA/DOX/CCM at a DOX-CCM molar ratio of 1:1, 2:1, and 3:1 were used in-vitro. CI value of less than 1 indicated a strong synergistic effect at all

A

molar ratios [76]. A PH sensitive prodrug conjugate was produced by conjugation mechanism, with self-assembling property in water at pH 7.4 into nanoparticles (PEG-DOX NPs). Cur being hydrophobic encapsulates into the core through hydrophobic interaction (PEG-DOX-Cur NPs). After internalization of these NPs in tumor cells due to an acidic environment (PH 5.4) inside the tumors, PEG and DOX holding Schiff base breaks open and lead to releasing of both parent 14

drugs CUR and DOX into the tumor cells. HeG 2 and Hela cells used for in vitro cytotoxicity studies. The studies showed higher cytotoxicity values compared to individual drugs. NP formulation for the PEG-DOX-Cur NPs in both the cell lines reported an IC50 value of 1.700

SC RI PT

µg/mL (DOX) and 0.680 µg/mL (Cur) in HEG 2 cells and 1.741 µg/mL (DOX) and 0.697 µg/mL (Cur) in Hela cells respectively. The primary reason for higher toxicity observed was due to internalization and rapid release of drug in tumor cells. Moreover in vivo studies using xenograft mice with hepatic tumors also showed a significant reduction in the tumor volume with the PEG-DOX-Cur NPs [77].

U

Recently, Peng et al. designed novel PEGylated lipid/PAA/CaCO3 ternary system encapsulating

N

Cur and Dox (LPCCD) with a goal of increasing cancer cell toxicity and reduction of cardiotoxic

A

side effect. An in vitro cytotoxicity study in HepG2 cells demonstrated better IC50 with LPCCD

M

as compared to free Dox & Cur formulation. Also, cardiotoxicity which is a major side effect of

D

DOX treatment was also evaluated in mice cardiac sections by histology. LPCCD group reported

TE

no inflammatory and hypertrophic cardiac cells [78]. Zhang et al. developed pH-sensitive NPs for co-delivery of DOX and CUR. MTT assay in SMMC 7721 cell line established an increase in

EP

cell growth inhibition with D+C/NP formulation. Additionally, as noted by flow cytometry, the

CC

apoptotic rate for the formulation was nearly 1.99-fold higher than that of the free drug treatment. To investigate the anti-tumor efficacy and potential toxicity of Dox & Cur co-loaded

A

NPs, in vivo studies were conducted. A significant increase in tumor weight inhibition by formulation compared to a combined free drug (73.37 % and 32.6% and 73.37%, respectively) was observed [79].

15

4.2. Colon cancer As the third-most common malignant tumor, colon cancer accounts for more than 1.4 million new cases and over half a million deaths worldwide annually [71]. Surgery, chemotherapy, and

SC RI PT

radiotherapy are the go-to therapeutic approaches against colon cancer with chemotherapy as the first line of treatment [80]. MDR as well as the heterogeneous nature of cancer cells leads to resistance and increase in adverse effects and subsequently results in an increase of effective doses [81]. Like other cancers, combination therapy with NPs drug delivery system helps to overcome issues associated with monotherapy in colon cancer [82]. Xiao et al. developed

U

cationic polymeric NPs for co-delivery of camptothecin (CPT) and curcumin. Combination

N

index (CI) value of 0.46 indicated a strong synergism with the CPT/CUR at a molar ratio of 4: 1

A

after 24 h of incubation [83].

M

Overexpression of CD44, a single-chain glycoprotein, has been reported in a variety of colon

D

cancer [84-86]. Hyaluronic Acid (HA) is shown to have a high affinity for CD-44 receptors [87].

TE

So, the addition of HA provided a colon cancer-targeting capability due to more efficient internalization of HA-functionalized NPs in cells through an HA receptor-mediated endocytosis

EP

pathway by interacting with CD44 receptors [85]. Xia et al. capitalized this property for

CC

formulating HA-CPT/CUR-NPs. Ex vivo studies in AOM/DSS-induced colon cancer mouse model with the developed NPs showed higher permeation and accumulation of HA-

A

functionalized NPs in colon tumor region. Strong synergistic effects observed between CPT and Cur in this formulation, depicted by an IC50 value of 0.7 μM in Colon-26 cells at both 24 and 48 hours [88]. Chrysin (Chr), a naturally occurring flavone, is found in plant extracts. It is one of the most broadly used herbal medicine. Chr has been reported to have several effects such as anti16

inflammatory, antioxidant, antidiabetic, anti-allergic, antibacterial, antiestrogenic, and anticancer [89, 90]. As seen in in-vitro and in-vivo studies, anticancer effect of Chr is mainly due to arresting of the cell cycle, induction of apoptosis, inhibition of angiogenesis, and metastasis with

SC RI PT

no toxicity on normal cells [91]. Lotfi-Attari et al. formulated PLGA/PEG NPs, encapsulating a combination of Cur and Chr for the first time. In vitro studies in Caco-2 cell lines with Cur–Chr– PLGA/PEG NPs demonstrated significant growth arrest of cancer cells and high intracellular concentrations. The CIs value for the formulation was <1 which is indicative of a synergistic effect of the combination [92].

U

Anirudhan et al. designed a transdermal drug delivery system (TDDS) to encapsulate 5-

N

fluorouracil (5-FU) and curcumin (CUR). Polymer β-Cyclodextrin and aminated nano dextran

A

used to entrap CUR and 5-FU respectively which provides flexible kinetics for the release of

M

drugs after treatment with specific solvents. Polysaccharides ALG and CS were used to coat the

D

surfaces of NP's with opposite charges in the final product. In vitro studies on human carcinoma

TE

cell lines, HCT-116 depicted a reduction in cell viability from 71.1 to 48.7 % at increasing

EP

concentrations, when treated with solvent dipped final formulation [93]. Bagheri et al. showed the synergistic effect of co-delivery of Chrysin and CUR nanoparticulate

CC

delivery in SW480 colon cancer cell line. MTT assay favored the combination depicted by twice the decrease in cell growth inhibition rate as compared to free CUR and chrysin. These effects

A

were attributed to downregulation of hTERT by the combinatorial therapy [94].

4.3. Breast cancer Breast cancer is one of the most common types of cancer with more than 508,000 deaths till 2011. In 2012, nearly 1.7 million women diagnosed with breast cancer [71]. In the US alone, 17

246,660 cases of breast cancer are expected to register in 2018 [95]. Doxorubicin is the first line of therapy for several types of cancers including breast cancer [96]. Doxorubicin functions by DNA intercalation leading to disruption of DNA damage repair, causing the release of reactive

SC RI PT

oxygen species and eventually programmed cell death [97]. Like other anti-cancer drugs, drug resistance is a major issue associated with doxorubicin. To overcome MDR associated resistance with DOX, Wang et al. fabricated polymeric micelles and incorporated DOX with CUR to function as P-gp and apoptosis inhibitor. In the MCF-7/Adr (DOX-resistant) breast cancer cell lines, the formulation (DOX/Cur)-PMs compared with DOX-treated cells lead to the 18.29-fold

U

increase of apoptotic cells and enhanced invitro toxicity. In vivo studies in 4T1 bearing Balb/c

N

mice model for breast cancer showed a significant reduction in tumor weight as compared to

A

control and free drugs [98].

M

Ruttala et al. designed CUR and albumin-PTX hybrid encapsulation liposomes (CL-APN) and

D

performed in vitro cytotoxicity assay in MCF-7 and B16F10 carcinoma cell lines. 1:1 molar ratio

TE

of the PTX and CUR in formulation showed enhanced cytotoxicity. In cell migration assay CL-

drugs [99].

EP

APN demonstrated the highest inhibition in both cell lines compared to control and individual

CC

To target breast cancer, Cui et al. co-delivered CUR and DOX in a pH-sensitive prodrug using transferrin Tf-PEG-CUR. Several malignant tumors overexpress Tf. This property makes it a

A

perfect candidate for target-specific delivery to these type of cancers [100-102]. Both drugs being hydrophobic were entrapped in the core of Tf-PEG-CUR/DOX NPs. Mildly acidic pH release of CUR was significantly increased from 33.6% at pH 7.4 to 72.4 % at pH 5. A similar trendas observed for DOX release. In vitro cytotoxicity assay in MCF-7 cell lines showed the highest inhibition by the Tf-PEG-CUR/DOX NPs with IC50 of 2.5 μM. The concentration of 18

DOX and CUR in tumor, lung, and liver, major target areas in breast cancer, was the highest. The preparation also presented fewer adverse effects. Its concentration in heart and kidney was lower than others in the group. This formulation showed a high inhibition rate of tumor growth

SC RI PT

(IRT) (83.5%) [103].

Using curcumin as a theranostic agent (diagnostic + therapeutic) was studied by Nguyen et al. by fabricating Cur-PLA-TPGS NPs (curcumin + paclitaxel) co-loaded with PLA-TPGS NPs (Cur+PTX)- PLA-TPGS NPs. Fluorescence of CUR in MCF7 cell and MCF7 spheroid monitored by confocal fluorescence microscopy. This concluded that the monitoring of the

U

delivery and bio-distribution of the drug delivery system, curcumin acted as a potential

A

N

fluorescent probe [104].

M

Sahu et al. utilized nanosuspensions technique to co-deliver CUR and DOX. In vitro cytotoxicity in MCF7 cell line with CUR-DOX co-loaded nanosuspension showed the

D

highest inhibition rate, compared to individual drug nanosuspensions of 83.31% and 81.65% at

TE

0.5 and 1.0μg/mL respectively. Moreover, in vivo studies showed a highest tumor inhibition

EP

ratio (TIR) of 70% compared to groups treated with CUR nanosuspension (34%) and DOX

CC

nanosuspension (15%) [105]. In vivo stability and increased drug loading capacity are a few advantages of coating super-

A

magnetic iron oxide nanoparticles (SPIOs) with albumin. However, synthesis of SPIOs system is complicated and time-consuming. To overcome this limitation, Chen et al. used a one-step synthesis strategy of in situ co-precipitation, to synthesize albumin-stabilized SPIOs (BSASPIOs). Further to test for the effectiveness of this system in-vitro and in vivo, they encapsulated two cytotoxic drugs CUR and Sunitinib in BSA-SPIOs. Values for the free drug Sun and Cur in 19

MCF-7 were 6.15 μM and 23.8 μM respectively. The IC50 values of BSA-SPIOs enclosed CUR and Sunitinib were enhanced to 2.43 μM (for Sun) and 6.79 μM (for Cur). Moreover, an increase in inhibition noted with the application of external magnetic field with IC50 values of 2.03 μM

SC RI PT

(for Sun) and 5.67 μM (for CUR), respectively. In vivo studies showed the highest inhibition of tumor growth with nearly 2.3-fold tumor weight reduction by the BSA-SPIOs SunCUR combination, compared to free SunCUR therapy [106].

Increased sensitivity of MDR tumor cells by addition of small-molecule compounds known as chemosensitizers, which lead to an improvement of the efficacy of chemotherapeutic drugs [107-

U

109]. As a novel chemo/photo- co-therapeutic nanomaterial, mesoporous carbon along with a

N

chemosensitizer (CUR) can be used to develop mesoporous carbon nanoparticles (MCNs). Some

A

advantage of MCNs comprises high pore volume ratio, larger surface area, and efficient drug

M

loading capability [110]. The PCDA backbone, which is made up of disulfide bond linked mono-

D

curcumin, degrades at a significantly higher concentration of glutathione (GSH) in the cytoplasm

TE

than in the extracellular environment, leading to a GSH-responsive release of curcumin (CUR) [111]. Li et al. designed PEG-PCDA coated MCN (chemo/photo chemotherapy activity) to

EP

deliver DOX to drug-resistant MCF-7/ADR cells. In vitro studies on MCF-7/Adr cell lines

CC

showed the maximum inhibition (IC50= 17.41 mg/mL) with the formulated PEGPCDA/DOX@MCNs. Additionally, the IC50 value reduced to 11.16 mg/mL after NIR laser

A

irradiation [112]. Results of this study are illustrated in Figure 6. (Insert Figure 9 here)

Another approach against MDR resistant MCF-7/Adr cells is through Folate conjugation on the surface of nanoparticles. It could become a useful approach to develop target-specific NPs due to its affinity to folate receptors which are overexpressed in several cancers. Baek et al. fabricated

20

2-hydroxypropyl-β-cyclodextrin (HPCD) conjugated with folate for co-delivery of curcumin with paclitaxel (FPCHN-30). The formulation showed enhanced absorption of paclitaxel as compared to that of free drug, which was due to P-gp inhibition of curcumin. In vitro studies in

SC RI PT

MCF-7/Adr cells revealed that FPCHN-30 with folic acid showed the highest cytotoxicity and cellular uptake [113].

Medel et al. tested the efficiency of a biodegradable and biocompatible block copolymer (PEGb-PLA) in co-delivery of the curcumin-bortezomib system to treat breast cancer. NP formulation in MCF-7 and MDAMB-231 breast cancer cells caused significant cytotoxicity with an IC50

A

N

values of 18.8 nM and 122.4 nM respectively [114].

U

value of 7.5 nM and 59.2 nM respectively as compared to free Cur-BTZ combination with IC50

M

HA can interact with the CD44 receptor. Tumor-specific nano-formulations involve HA conjugation in the final design. Yang et al. designed nanoparticles to directly target breast cancer

D

stem cells (bCSCs). They prepared and evaluated targeting co-delivery system of Hyaluronic

TE

Acid (HA-Hybrid NPs) to the surface of hydrophobic PLGA NPs to co-deliver PTX and CUR. In

EP

the MCF-7 cells, the formulation exhibited the lowest IC50 values of 0.751×10-3 μg/mL and 0.4318 μg/mL for PTX and CUR respectively. Compared to free PTX+CUR CI (0.77), the CI of

CC

HA-Hybrid NPs/PTX+CUR was significantly lower at 0.32. In vivo studies pointed out improved tumor growth inhibition (TGI) at 67.5% as compared to other groups. Additionally,

A

average tumor volume for HA-Hybrid NP/PTX+CUR group was found to be 2.5 times smaller than control. [115]. Lin et al. used silica-based materials, mesoporous silica with the PEG (PLMSNs) for co-delivery of Tax and Cur. The in-vitro activity in canine breast cancer line showed enhanced IC50 values for the formulation [116].

21

Sudakaran et al. exploited a new synthetic biodegradable/ biocompatible copolymer poly L-lactic acid-co-ε-caprolactone (PLACL) with Aloe Vera (AV) MGO for co-delivery of CUR/β-CD in breast cancer cell lines. In vitro studies on MCF-7 cells showed 52.19% cell growth inhibition by

SC RI PT

PLACL/AV/MgO/CUR/β-CD compared to PLACL/AV/MgO [117].

Yuan et al. designed and fabricated pH-sensitive NPs to co-deliver DOX and CUR to MDR resistance breast cancer cells. In-vitro cytotoxicity assay on MCF-7/ADR cells showed 2.2 and 7.2 times lower IC50 value for CURDOX-NPs as compared to DOX-NPs and DOX solution respectively. In vivo studies revealed the slowest tumor growth and maximum growth inhibition

U

rate by CURDOX-NPs. Moreover, the formulation demonstrated a lower percentage of cancer

A

N

stem cells as compared to CUR-NPs formulation alone (15.0% vs. 6.82%) [118].

M

4.4. Brain cancer

The most common and aggressive brain tumors in human are malignant glioma with a 5-year

D

survival rate of only 5% [119]. Blood-Brain Barrier (BBB) is the principal obstruction for

TE

delivery for limited glioma chemotherapy (e.g., temozolomide, carmustine). Cui et al. fabricated

EP

T7-modified magnetic PLGA nanoparticulate system (MNP/T7-PLGA NPs), consisting of hydrophobic magnetic nanoparticles (MNPs), to co-deliver PTX and CUR. In vitro cytotoxicity

CC

effects were examined in U87 cells. IC50 for the MNP/T7-PLGA NPs loaded with PTX+CUR was reported to significantly improve at 3.06 μg/mL, compared to other groups. In vivo tumor

A

studies also revealed superior anti-glioma efficacy for the formulation [120]. Due to a continuous requirement of glucose supply in cancer cells, GLUT-1 is significantly overexpressed [121]. BBB in glioblastoma contains significantly overexpressed GLUT-1 [122, 123]. This could be a good target for the anticancer drug delivery to glioblastoma cells. 22

Sarisozen et al. developed polymeric micelles coated with a single chain fragment variable (scFv) against GLUT-1 to co-deliver CUR and DOX. In vitro cytotoxicity assay was tested in U87MG glioblastoma cells. The CI for polymeric micelles at 0.63 further reduced to 0.46 with

SC RI PT

GLUT1 targeting strategy. Moreover, the formulation leads to a 3-fold increase in nuclear localization of DOX in U87MG cells, which was the apparent reason for enhanced cytotoxicity [124].

4.5. Skin cancer

Every year nearly two million new cases of skin cancer are diagnosed in the United States alone

U

[95]. 5-fluorouracil, imiquimod, bleomycin, and interferon alpha are the most widely used

N

chemotherapeutic agents against skin cancer [125]. Recently, small interference (siRNA) has

A

shown effectivity in preclinical and clinical trials against cancer [126]. Jose et al. fabricated and

M

evaluated liposomes for co-delivery of curcumin and STAT3 siRNA, combined with anodal

D

iontophoresis technique. Iontophoresis application enhances skin penetration of positively

TE

charged lipoplexes (liposome-siRNA complexes) by electro-repulsion and electro-osmosis technique [127]. In vitro cytotoxicity assay in A431 cells with the formulation, demonstrated

EP

higher inhibition of cell growth (72.9±2.3%), compared to individual treatments. Anodal

CC

iontophoresis applied to reach the target depth of 100 μm inside the skin and for better penetration of the compounds in the ski[128].

A

Effect of curcumin in combination with the paclitaxel in folic acid conjugated PLGA-PEG NPs was studied by Kumar et al. In in vivo studies, CUR-PLGA-PEG-FA formulation demonstrated enhanced bioavailability and retention time of curcumin with no effect on toxicity profile. Further, in combination with Paclitaxel on cervical cancer xenograft model revealed enhanced

23

tumor growth inhibition. A nearly 2-fold decrease in tumor volume compared to individual drug treatment groups [129].

Jose et al. exploited the effect of Iontophoresis on a liposomal formulation of CUR and STAT

SC RI PT

siRNA on a melanoma tumor mouse model. In vitro cytotoxicity studies with B16F10 cell lines showed higher growth inhibition (76.3 ± 4.0%) compared to curcumin-loaded cationic liposomes (46.0 ± 3.4%) and cationic liposome-STAT3 siRNA group (58.4 ± 3.0%). In vivo studies on melanoma tumor model also supported the in vitro results with maximum nearly three-fold increase in inhibition on tumor growth and volume by the curcumin-loaded liposome-STAT3

N

U

siRNA complex compared to other groups [130].

A

Combination of chemotherapy with phototherapy for the treatment of skin cancer exploited by

M

Singh et al. They designed gold coated Hydrogenated Soya Phosphatidyl Choline (HSPC) liposomes encapsulating CUR. In vitro studies on B16F10 cell lines showed the 9-fold increase

D

in cell growth inhibition by Au-Lipos Cur NPs compare with free curcumin after laser irradiation

EP

TE

and ~8 fold compared to non-gold coated/non-irradiated formulation of lipos-Cur NPs [131].

4.6. Pancreatic cancer

CC

Pancreatic cancer is the second most common cause of cancer-related deaths. It is estimated to account for 55,440 new cases and 44,330 deaths in 2018 in the US alone [95]. New therapeutic

A

avenues are essential to overcome an abysmal 5-year survival rate of pancreatic cancerare essential. Kim et al. utilized nanoparticle albumin-bound (NabTM) technology using highpressure homogenization, to co-deliver PTX and Cur. Efficient internalization into MiaPaca-2 cells observed with PTX/CCM Alb-NPs. Moreover in vitro cytotoxicity studies revealed 71%

24

increase in IC50 with this formulation in MiaPaca-2 cells [132]. Toxicological study of the formulation containing chitosan-SLNs (c-SLNs) in combination with sulforaphane (ACS combination) on co-delivery of Aspirin and Cur studied by Thakkar et al. for pancreatic cancer.

SC RI PT

This formulation has been previously shown to be effective against pancreatic cancer in in-vitro studies, but the toxicity profile of the c-SLNs and ACS is not studied before. The formulation was found to be safe for long-term oral administration against pancreatic cancer with no signs of toxicity in acute, subacute, and sub-chronic studies. Future direction points in the utilization of cSLNs and ACS technique for formulating other anti-cancer drug combinations [133].

U

4.7. Osteosarcoma

N

In kids and young adults, Osteosarcoma (OS) accounts for 60% of primary bone cancers [134].

A

Currently approved treatments for OS include surgery and chemotherapy [95]. Combination of

M

surgery with chemotherapy increases the cure rate of patients with OS from 20 % to 70 % [135].

D

The commonly used chemotherapeutics to treat OS include DOX, cisplatin, and methotrexate.

TE

Even with the combination of chemotherapy and surgery, failure in nearly 30% of stage IV patients due to drug resistance is observed [136]. Polymeric nanoparticles coated with lipid-PEG

EP

or (LPNs) were constructed to overcome the resistance issues and to inhibit the rapid clearance

CC

effect of the reticuloendothelial system. Constructed LPNs utilized to co-deliver DOX, and CUR (DOX+CUR LPNs) and tested the formulation for in-vitro and in-vivo effects in OS. In vitro

A

cytotoxicity assay was performed in Human osteosarcoma cell lines (KHOS cells). The IC50 of DOX+CUR LPNs was found to be lowest at 0.6 μM as compared to other groups. In-vivo efficacy was observed in BALB/c mice injected with KHOS cell model. On 21-day posttreatment, OX+CUR LPNs group treated mice, the tumor volume was lowest at about 182 mm3,

25

compared to the saline-treated group (973 mm3). Moreover, with DOX+CUR LPNs, the Tumor Growth Inhibition (TGI) was significantly higher at 81.3% compared to other groups [137].

4.8. Gastrointestinal cancer

SC RI PT

Gastrointestinal cancers are the third leading cause of cancer-related deaths worldwide [138]. Intravenous (IV) administration is the most common route of chemotherapeutic delivery in GI cancer patients. IV route has several disadvantages like discomfort, stress to patients, multiple hospitalizations trip, high cost, and high risk of hospital-acquired infections [139, 140]. To overcome this problem, Bar-Zeev et al. developed a novel oral delivery nano-delivery system

U

using self-assembling β-casein (CN) enclosing hydrophobic chemotherapeutic drugs (PTX) and

N

MDR (TQD) chemosensitizers. In vitro cytotoxicity assays were performed in EPG85-257RDB

A

(P-gp overexpressing) and EPG85-257P gastric carcinoma cell lines. The β-CM formulation with

M

combination showed significantly lower IC50 value with MDR resistance EPG85-257RDB cell

TE

4.9. Prostate cancer

D

line (111 nM) compared to EPG85-257P cell line (2185 nM) [141].

EP

Prostate cancer is the second most common cause of cancer-related deaths. In 2018, nearly 164, 690 new case and 29, 430 deaths are estimated in the US alone [95]. Taxane drugs and platinum

CC

compounds first line for chemotherapy against prostate cancer. Although due to severe adverse effects due to MDR, they are not as effective with monotherapy [142]. To overcome this adverse

A

effect, Saralkar et al. used hydrophobic phytochemicals Curcumin and resveratrol and formulated calcium alginate NPs to encapsulate them. The drug-loaded nanoparticles had a cytotoxic effect in DU145 prostate cancer cells with no hemolytic effects [143].

26

4.10. Cervical cancer Cervical cancer is one of the most common causes of cancer-related deaths in females. The first line of therapy for cervical cancer is Cisplatin (DDP) as monotherapy, or it’s combination with

SC RI PT

paclitaxel [144, 145]. Serious adverse effects related to DPP are nephrotoxicity, hepatotoxicity, and drug resistance [146-148], which could be overcome by nano-carrier-based delivery of DDP to the tumor sites and combination chemotherapy [149-151]. Li et al. combined both techniques and constructed lipid-polymer hybrid nanoparticles (LPNs) for DDP and Cur co-delivery. Moreover, they compared the LPN formulation with the most common NPs delivery formulation

U

such as Polymeric Nanoparticles (PNPs). HeLa cells and HUVEC cells used for in-vitro

N

cytotoxicity assay. Highest in vitro tumor activity was exhibited by DDP/CUR/LPNs as

A

indicated by lowest IC50 value among the groups. CI of DPP/CUR/LPNs was significant at 0.61

M

as compared to DPP/CUR/PNPs, and DPP/CUR solution with CI value of 0.96 and 1.40, respectively. Investigated the in vivo antitumor efficacy of LPNs in cervical cancer-bearing

D

BALB/c mice model DPP/CUR/LPNs treated group showed the most significant reduction in

EP

4.11. Leukemia

TE

tumor size compared to other groups [152].

CC

According to the American cancer society report, in 2018 nearly 60,300 new cases and 24,370 deaths are expected due to leukemia [95]. Silver nanoparticles (AgNPs) have antimicrobial

A

activity and anti-cancer effect [153]. The generation of reactive oxygen species due to the release of silver ions is the reason behind AgNPs mediated cytotoxicity [154]. Petrov et al. utilized this property of AgNPs and evaluated the effect of a combination of AgNPs and curcumin in a multilayer polymeric micelle of poly (ethylene oxide)-b-poly (n-butyl acrylate)-b-poly (acrylic acid) (PEO-b-PnBA-b-PAA) triblock terpolymer micelles, in acute myeloid leukemia. This 27

formulation specifically presented a pronounced cytotoxic effect on acute myeloid leukemia (HL-60), and it's multidrug-resistant subline HL-60/DOX, as compared to an individually loaded carrier [155].

SC RI PT

4.12. Lung cancer

Lung cancer has one of the highest incidence among all forms of cancer. Suboptimal therapy due to lack of targeted effects and severe side effects are some disadvantages of conventional therapy against lung cancer. The advent of using siRNA for cancer therapy has attracted more attention, as RNA interference is more specific to the target gene and can knock down some oncogene

U

expression for cancer therapy [156]. Zhang et al. demonstrated the effect of epidermal growth

N

factor (EGF) modified monomethoxy (polyethylene glycol)-poly (D, L-lactide-co-glycolide)-

A

poly(L-lysine) (mPEG-PLGA-PLL, PEAL) NPs (EGF-PEAL), loaded with Dox and Bcl-2-

M

siRNA NPs on lung cancer. The blank EGF-PEAL NPs showed minimal cytotoxicity in the

D

H1299 cell lines, indicating a useful drug delivery carrier. The co-delivery of siRNA and Dox in

TE

EGF-PEAL also reported higher cytotoxicity. Annexin V/PI staining assay showed maximum apoptosis in H1299 cells was induced by the combination of early and late apoptosis rates of

EP

61.56% and 11.37 % respectively. In vivo studies demonstrated tumor volume to be the smallest

CC

(2-3 fold) with the co-delivery of Dox and Bcl-2-siRNA group [157].

Tang et al. designed SLNs for the co-delivery of CUR and piperine in paclitaxel-resistant human

A

ovarian carcinoma cell line A2780/Taxol. The purpose of the study was to evaluate whether the presence of Cur could reverse MDR associate resistance in this cell line. The in vitro cytotoxicity studies showed significantly higher cell growth inhibition with (Cur + Pip)-SLN (59.90±8.40%), as compared to Cur (25.85±5.74%) and Pip (6.87±4.52%) alone [158].

28

Boron Neutron Capture Therapy (BNCT) radiotherapy combines a boron-containing compound with low energy neutron irradiation to target cancer cells.10B captures the neutrons and disintegrates into alpha particles and lithium nuclei, which induce site-specific damage to tumor

SC RI PT

cells after accumulation [159]. Diego et al. exploited this target for anticancer effects by formulating Rubro-curcumin (stable adduct of boric acid, curcumin, and oxalic acid) [160] in PLGA NPS. Prepared folate specific PLGA NPs, to provide tumor-specific accumulationlate specific PLGA NPs were prepared. In vitro cytotoxicity assays showed enhanced inhibition rate

U

with the formulation compared to individual drugs [161].

N

5. Clinical trials involving curcumin

A

Curcumin both in its free form and as nano-particulate formulations have been under

M

investigation in human clinical trials for many years. It has shown clinical benefits in patients with colorectal cancer, pancreatic cancer, breast cancer and multiple myeloma. Searching for

EP

TE

summarized in Table 2 [162].

D

Curcumin as a treatment for cancer in www.clincaltrials.gov shows 22 recent trials which are

Table 2: Recent clinical trials involving curcumin NCT number

CC

Serial No. 1.

Phase

Trial Status Active, not recruiting

Cancer type Advanced colorectal cancer

Description

Phase 1

2.

NCT01333917

Phase 1

Completed

Colorectal cancer

Curcumin Biomarkers

2016

3.

NCT01294072

Phase 1

Active, not recruiting

Colorectal cancer

Study Investigating the Ability of Plant Exosomes to Deliver

2020

A

NCT01859858

29

Effect of Cur on Dose Limiting Toxicity and Pharmacokinetics of Irinotecan in Patients with Solid Tumors

Completion date 2018

Curcumin to Normal and Colon Cancer Tissue Phase 1/2

Active, not recruiting

Colorectal cancer

Combining Curcumin with FOLFOX Chemotherapy in Patients with Inoperable Colorectal Cancer

2019

5.

NCT02598726

Phase 1

Recruiting

Malignant Neoplasm

Curcumin and Piperine in Reducing Inflammation for Ureteral StentInduced Symptoms in Patients with Cancer

2021

6.

NCT02138955

Phase1/ 2

Active, not recruiting

Advanced Cancer/Faile d standard therapy

A Phase IB Dose Escalation Study of Lipocurc in Patients with Cancer

2017

7.

NCT02336087

Phase 1

Recruiting

Pancreatic cancer

Gemcitabine Hydrochloride, Paclitaxel AlbuminStabilized Nanoparticle Formulation, Metformin Hydrochloride, and a Standardized Dietary Supplement in Treating Patients with Pancreatic Cancer That Cannot be Removed by Surgery

2018

8.

NCT02439385

N

A

M D TE EP

Enrolling by invitation

Colorectal cancer

Avastin/FOLFIRI in Combination with Curcumin in Colorectal Cancer Patients with Unresectable Metastasis

2019

Phase 2

Recruiting

Breast cancer

"Curcumin" in Combination with Chemotherapy in Advanced Breast Cancer

2018

CC

Phase 2

A 9.

NCT03072992

SC RI PT

NCT01490996

U

4.

30

NCT00745134

Phase 2

Active, not recruiting

Rectal cancer

Curcumin with Preoperative Capecitabine and Radiation Therapy Followed by Surgery for Rectal Cancer

2019

11.

NCT02724618

Phase 2

Active, not recruiting

Prostate cancer

Nanocurcumin for Prostate Cancer Patients Undergoing Radiotherapy (RT)

2022

12.

NCT02095717

Phase 2

Active, not recruiting

Prostate cancer

2018

13.

NCT00852332

Phase 2

Recruiting

N

Multicenter Study Comparing Taxotere Plus Curcumin Versus Taxotere Plus Placebo Combination in First-line Treatment of Prostate Cancer Metastatic Castration-Resistant (CURTAXEL)

Breast cancer

Docetaxel with or Without a Phytochemical in Treating Patients with Breast Cancer

2017

14.

NCT02944578

Phase 2

Recruiting

Cervical cancer

Topical Curcumin for Precancer Cervical Lesions

2021

15.

NCT02138955

16.

NCT02556632

TE

D

M

A

U

SC RI PT

10.

EP

Phase 2

A Phase IB Dose Escalation Study of Lipo-cur in Patients with Cancer Completed

Breast cancer

Prophylactic Topical Agents in Reducing Radiation-Induced Dermatitis in Patients with NonInflammatory Breast Cancer

2016

Phase 2

Recruiting

Cervical, Endometrial , and Uterine cancer

Study of Pembrolizumab, Radiation and Immune Modulatory Cocktail in Cervical/Uterine

2022

A

CC

Phase 2

17.

NCT03192059

31

Cancer Phase 2

Completed

Endometrial cancer

Effect of Curcumin Addition to Standard Treatment on Tumour-induced Inflammation in Endometrial Carcinoma

2016

19.

NCT02782949

Phase 2

Recruiting

Gastric cancer

Curcumin in Preventing Gastric Cancer in Patients with Chronic Atrophic Gastritis or Gastric Intestinal Metaplasia

2019

20.

NCT00641147

Phase 2

Completed

Familial Adenomato us Polyposis

Curcumin in Treating Patients with Familial Adenomatous Polyposis

2016

21.

NCT03061591

Phase 2

Not yet recruiting

Familial Adenomato us Polyposis

Turmeric Supplementation on Polyp Number and Size in Patients with Familial Adenomatous Polyposis.

2020

22.

NCT02064673

Phase 3

Prostate cancer

Adjuvant Curcumin to Assess Recurrence-Free Survival in Patients Who Have Had a Radical Prostatectomy

2020

SC RI PT

NCT02017353

Recruiting

CC

EP

TE

D

M

A

N

U

18.

A

6. Challenges with curcumin combination therapy Effective combination therapy for anticancer treatment requires dealing with an investigation of multiple hurdles. Initial studies demand precise tuning of an optimal mass ratio (ratio-metric dosing) of each drug in a combination, based on individual drug efficacy and pharmacokinetic profile [163]. Due to low bioavailability and poor absorption, the formulation of combination 32

becomes more challenging with curcumin [4]. One of the major challenges of non-site-specific combination nanoparticulate formulation is off-site effects due to action on healthy cells. Although, recently several chemotherapeutic nano-formulations combine folate or other specific

SC RI PT

antigens, to enhance site specificity for the delivery of cytotoxic drugs to tumor cells. Delivery system could be further extended into combination chemotherapy for achieving maximum treatment efficacy as described in Figure 7 via a potential delivery system of curcumin combination nanoparticles with paclitaxel. (Insert Figure 10 here). Also, some of the materials used for fabricating nano-formulations are toxic, so each formulation should be thoroughly tested

U

for short and long-term toxicity. As the intravenous route is the most popular route of

N

administration of nano-formulation, toxicity related to the interaction between endothelial cells

A

of blood vessels and nanocarrier should be considered [164]. For nanocarrier-based combination

M

therapy, other critical aspects to be considered are Quality by design parameters, rationally based

D

on size and surface physicochemical property of materials to be fabricated. [165].

TE

7. Conclusion and future directions

EP

Delivery of nano-encapsulated anticancer drugs with curcumin as an adjunct, is emerging

CC

globally as an improvement to monotherapy. Along with the chemo-sensitizing synergism and tissue specificity imparted by this approach, the combinatorial therapy is now being

A

acknowledged for fighting back multidrug resistance and providing effective treatment strategies. The present in-vitro and in-vivo work pose a promising picture, but a lot must be done before transition of this therapy could be made from lab-scale to safe human dosage forms. The biggest challenge from pharmaceutical point is alteration of curcumin from its natural form to a formulation relevant derivative, which is more stable and bio-pharmaceutically possess more 33

“drug like properties”. In one such attempt, poly (curcumin-dithiodipropionic acid) PCDA coating on nanoparticles was used. This coating was made up of disulfide bond linked monocurcumin, which degrades at a significantly higher concentration of glutathione (GSH) in the

SC RI PT

cytoplasm than in the extracellular environment, leading to GSH-responsive release of curcumin; thus, altering curcumin’s original form to a more practically viable form for formulation. Other intense fabrications like prodrug approach, polymer coating, complexation, co-crystallization of curcumin etc. would require time and cost inputs, both of which are general offsets for pharmaceutical industries. More research thus needs to be put into incorporation of curcumin in a

U

delivery system, which could be optimized to its best possible usage, counterbalancing the

N

monetary input. Other major barriers to human usage of this approach lie in lack of its target

A

specific properties. Methods like ligand attachment, PEGylation of NPs, tissue specific coating,

M

or upcoming external means like electrophoresis and iontophoresis etc. could be possible solutions to such hindrances. Human dose optimization for curcumin alone or with the

D

combination drugs needs to be further investigated. With no commercial drugs in the market so

TE

far, dosing would be a major rate limiting step here. In vitro dose-response studies and possible

EP

FDA approval with preclinical or clinical data backups must be set in place to even start those conversations. Further, a complete pharmacokinetic (PK) profiling and in vitro-in vivo

CC

correlation (IVIVC) for the drug combinations, needs to be drawn to further move up the ladder. Also, upon dosage designing, antitumor activity of the combination NPs needs to be tested in

A

actual cancer settings. Effectivity of these formulations is often determined by altered vasculature, characteristic to cancer masses. Toxicity studies for many developmental doses discussed in the work above have been performed, but extensive toxicologic profiling and

34

percentage damage to healthy tissue is required to weight in the risk to benefit ratio of this chemotherapeutic approach and justify their incorporation. Switching the course of present chemotherapy to nano-particulate curcumin combination therapy

SC RI PT

is a translational attempt which requires work from basic molecular aspects to pharmaceutical and then to preclinical and clinical trials. Extensive efforts are required from current prospects that curcumin combination NPs possess, to ultimately make this approach a potential future treatment improvement in cancer chemotherapy

U

Funding

N

This research did not receive any specific grant from funding agencies in the public, commercial,

M

A

or not-for-profit sectors.

D

Conflict of interest

References

Research

UK,

Cancer

statistics

reports

for

the

UK.

CC

[1] Cancer

EP

TE

The authors declare that there are no conflicts of interest.

http://www.cancerresearchuk.org/aboutcancer/statistics/cancerstatsreport/, 2012 (accessed 1

A

March 2012).

[2] D. Hanahan, R.A. Weinberg, Hallmarks of cancer: the next generation, cell 144(5) (2011) 646-674. [3] K. Nakano, K.H. Vousden, PUMA, a novel proapoptotic gene, is induced by p53, Molecular cell 7(3) (2001) 683-694. 35

[4] G. Sa, T. Das, Anti cancer effects of curcumin: cycle of life and death, Cell division 3(1) (2008) 14. [5] B.B. Aggarwal, I.D. Bhatt, H. Ichikawa, K.S. Ahn, G. Sethi, S.K. Sandur, C. Natarajan, N.

SC RI PT

Seeram, S. Shishodia, 10 Curcumin—Biological and Medicinal Properties, (2006). [6] Y. Liu, X. Hong, Effect of three different curcumin pigmens on the prdiferation of vascular smooth muscle cells by ox-LDL and the expression of LDL-R, Zhongguo Zhong yao za zhi= Zhongguo zhongyao zazhi= China journal of Chinese materia medica 31(6) (2006) 500-503.

[7] D.S. Kim, S.-Y. Park, J.-Y. Kim, Curcuminoids from Curcuma longa L.(Zingiberaceae) that

N

(1–42) insult, Neuroscience Letters 303(1) (2001) 57-61.

U

protect PC12 rat pheochromocytoma and normal human umbilical vein endothelial cells from βA

A

[8] T. Nishiyama, T. Mae, H. Kishida, M. Tsukagawa, Y. Mimaki, M. Kuroda, Y. Sashida, K.

M

Takahashi, T. Kawada, K. Nakagawa, Curcuminoids and sesquiterpenoids in turmeric (Curcuma longa L.) suppress an increase in blood glucose level in type 2 diabetic KK-Ay mice, Journal of

D

Agricultural and food Chemistry 53(4) (2005) 959-963.

TE

[9] A. Duvoix, R. Blasius, S. Delhalle, M. Schnekenburger, F. Morceau, E. Henry, M. Dicato, M.

181-190.

EP

Diederich, Chemopreventive and therapeutic effects of curcumin, Cancer letters 223(2) (2005)

CC

[10] P. Anand, C. Sundaram, S. Jhurani, A.B. Kunnumakkara, B.B. Aggarwal, Curcumin and cancer: an “old-age” disease with an “age-old” solution, Cancer letters 267(1) (2008) 133-164.

A

[11] G. Bar-Sela, R. Epelbaum, M. Schaffer, Curcumin as an anti-cancer agent: review of the gap between basic and clinical applications, Current medicinal chemistry 17(3) (2010) 190-197. [12] J. Ravindran, S. Prasad, B.B. Aggarwal, Curcumin and cancer cells: how many ways can curry kill tumor cells selectively?, The AAPS journal 11(3) (2009) 495-510.

36

[13] B.B. Aggarwal, H. Ichikawa, P. Garodia, P. Weerasinghe, G. Sethi, I.D. Bhatt, M.K. Pandey, S. Shishodia, M.G. Nair, From traditional Ayurvedic medicine to modern medicine: identification of therapeutic targets for suppression of inflammation and cancer, Expert opinion

SC RI PT

on therapeutic targets 10(1) (2006) 87-118. [14] A.M. Kamat, G. Sethi, B.B. Aggarwal, Curcumin potentiates the apoptotic effects of chemotherapeutic agents and cytokines through down-regulation of nuclear factor-κB and nuclear factor-κB–regulated gene products in IFN-α–sensitive and IFN-α–resistant human bladder cancer cells, Molecular cancer therapeutics 6(3) (2007) 1022-1030.

U

[15] H. Hatcher, R. Planalp, J. Cho, F. Torti, S. Torti, Curcumin: from ancient medicine to

N

current clinical trials, Cellular and Molecular Life Sciences 65(11) (2008) 1631-1652.

A

[16] S. Balaji, B. Chempakam, Toxicity prediction of compounds from turmeric (Curcuma longa

M

L), Food and Chemical Toxicology 48(10) (2010) 2951-2959. [17] H.H. Tønnesen, M. Másson, T. Loftsson, Studies of curcumin and curcuminoids. XXVII.

D

Cyclodextrin complexation: solubility, chemical and photochemical stability, International

TE

Journal of Pharmaceutics 244(1-2) (2002) 127-135.

EP

[18] Y.-J. Wang, M.-H. Pan, A.-L. Cheng, L.-I. Lin, Y.-S. Ho, C.-Y. Hsieh, J.-K. Lin, Stability of curcumin in buffer solutions and characterization of its degradation products, Journal of

CC

pharmaceutical and biomedical analysis 15(12) (1997) 1867-1876. [19] P. Anand, A.B. Kunnumakkara, R.A. Newman, B.B. Aggarwal, Bioavailability of

A

curcumin: problems and promises, Molecular pharmaceutics 4(6) (2007) 807-818. [20] H. Maeda, The enhanced permeability and retention (EPR) effect in tumor vasculature: the key role of tumor-selective macromolecular drug targeting, Advances in enzyme regulation 41(1) (2001) 189-207.

37

[21] J. Fang, H. Nakamura, H. Maeda, The EPR effect: unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect, Advanced drug delivery reviews 63(3) (2011) 136-151.

SC RI PT

[22] V. Torchilin, Tumor delivery of macromolecular drugs based on the EPR effect, Advanced drug delivery reviews 63(3) (2011) 131-135.

[23] K.Y. Kim, Nanotechnology platforms and physiological challenges for cancer therapeutics, Nanomedicine: Nanotechnology, Biology and Medicine 3(2) (2007) 103-110.

[24] I.A. Siddiqui, V.M. Adhami, J. Christopher, Impact of nanotechnology in cancer: emphasis

U

on nanochemoprevention, International journal of nanomedicine 7 (2012) 591.

N

[25] R. Misra, S. Acharya, S.K. Sahoo, Cancer nanotechnology: application of nanotechnology

A

in cancer therapy, Drug discovery today 15(19-20) (2010) 842-850.

M

[26] C. Holohan, S. Van Schaeybroeck, D.B. Longley, P.G. Johnston, Cancer drug resistance: an evolving paradigm, Nature Reviews Cancer 13(10) (2013) 714.

D

[27] Y. Zhang, H.F. Chan, K.W. Leong, Advanced materials and processing for drug delivery:

TE

the past and the future, Advanced drug delivery reviews 65(1) (2013) 104-120.

EP

[28] A.C. Anselmo, S. Mitragotri, An overview of clinical and commercial impact of drug delivery systems, Journal of Controlled Release 190 (2014) 15-28.

CC

[29] V.P. Torchilin, Multifunctional, stimuli-sensitive nanoparticulate systems for drug delivery, Nature reviews Drug discovery 13(11) (2014) 813.

A

[30] Y. Min, J.M. Caster, M.J. Eblan, A.Z. Wang, Clinical translation of nanomedicine, Chemical reviews 115(19) (2015) 11147-11190. [31] A.C. Anselmo, S. Mitragotri, A review of clinical translation of inorganic nanoparticles, The AAPS journal 17(5) (2015) 1041-1054.

38

[32] H.-C. Huang, S. Barua, G. Sharma, S.K. Dey, K. Rege, Inorganic nanoparticles for cancer imaging and therapy, Journal of controlled Release 155(3) (2011) 344-357. [33] R. Kanasty, J.R. Dorkin, A. Vegas, D. Anderson, Delivery materials for siRNA

SC RI PT

therapeutics, Nature materials 12(11) (2013) 967. [34] K.A. Whitehead, R. Langer, D.G. Anderson, Knocking down barriers: advances in siRNA delivery, Nature reviews Drug discovery 8(2) (2009) 129.

[35] R.A. Petros, J.M. DeSimone, Strategies in the design of nanoparticles for therapeutic applications, Nature reviews Drug discovery 9(8) (2010) 615.

U

[36] D. Peer, J.M. Karp, S. Hong, O.C. Farokhzad, R. Margalit, R. Langer, Nanocarriers as an

N

emerging platform for cancer therapy, Nature nanotechnology 2(12) (2007) 751.

M

review of medicine 63 (2012) 185-198.

A

[37] A.Z. Wang, R. Langer, O.C. Farokhzad, Nanoparticle delivery of cancer drugs, Annual

materials 12(11) (2013) 991.

D

[38] S. Mura, J. Nicolas, P. Couvreur, Stimuli-responsive nanocarriers for drug delivery, Nature

TE

[39] Y. Pilpel, P. Sudarsanam, G.M. Church, Identifying regulatory networks by combinatorial

EP

analysis of promoter elements, Nature genetics 29(2) (2001) 153. [40] R. Müller, Crosstalk of oncogenic and prostanoid signaling pathways, Journal of cancer

CC

research and clinical oncology 130(8) (2004) 429-444. [41] N.V. Sergina, M. Rausch, D. Wang, J. Blair, B. Hann, K.M. Shokat, M.M. Moasser, Escape

A

from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3, Nature 445(7126) (2007) 437. [42] W. Kassouf, C.P. Dinney, G. Brown, D.J. McConkey, A.J. Diehl, M. Bar-Eli, L. Adam, Uncoupling between epidermal growth factor receptor and downstream signals defines resistance

39

to the antiproliferative effect of Gefitinib in bladder cancer cells, Cancer research 65(22) (2005) 10524-10535. [43] L.D. Mayer, A.S. Janoff, Optimizing combination chemotherapy by controlling drug ratios,

SC RI PT

Molecular interventions 7(4) (2007) 216. [44] D.S. Couto, L. Perez-Breva, P. Saraiva, C.L. Cooney, Lessons from innovation in drugdevice combination products, Advanced drug delivery reviews 64(1) (2012) 69-77.

[45] J.H. Lee, A. Nan, Combination drug delivery approaches in metastatic breast cancer, Journal of drug delivery 2012 (2012).

U

[46] J. Jia, F. Zhu, X. Ma, Z.W. Cao, Y.X. Li, Y.Z. Chen, Mechanisms of drug combinations:

N

interaction and network perspectives, Nature reviews Drug discovery 8(2) (2009) 111.

A

[47] H.-G. Wendel, E. de Stanchina, J.S. Fridman, A. Malina, S. Ray, S. Kogan, C. Cordon-

M

Cardo, J. Pelletier, S.W. Lowe, Survival signalling by Akt and eIF4E in oncogenesis and cancer therapy, Nature 428(6980) (2004) 332.

D

[48] N. Kolishetti, S. Dhar, P.M. Valencia, L.Q. Lin, R. Karnik, S.J. Lippard, R. Langer, O.C.

TE

Farokhzad, Engineering of self-assembled nanoparticle platform for precisely controlled

EP

combination drug therapy, Proceedings of the National Academy of Sciences 107(42) (2010) 17939-17944.

CC

[49] S. Aryal, C.-M.J. Hu, L. Zhang, Polymeric nanoparticles with precise ratiometric control over drug loading for combination therapy, Molecular pharmaceutics 8(4) (2011) 1401-1407.

A

[50] T. Lammers, V. Subr, K. Ulbrich, P. Peschke, P.E. Huber, W.E. Hennink, G. Storm, Simultaneous delivery of doxorubicin and gemcitabine to tumors in vivo using prototypic polymeric drug carriers, Biomaterials 30(20) (2009) 3466-3475.

40

[51] Z. Gao, L. Zhang, Y. Sun, Nanotechnology applied to overcome tumor drug resistance, Journal of controlled release 162(1) (2012) 45-55. [52] Y. Yan, M. Björnmalm, F. Caruso, Particle carriers for combating multidrug-resistant

SC RI PT

cancer, ACS nano 7(11) (2013) 9512-9517. [53] M. Koval, K. Preiter, C. Adles, P.D. Stahl, T.H. Steinberg, Size of IgG-opsonized particles determines macrophage response during internalization, Experimental cell research 242(1) (1998) 265-273.

[54] Z.S. Guo, Z. Liu, D.L. Bartlett, D. Tang, M.T. Lotze, Life after death: targeting high

U

mobility group box 1 in emergent cancer therapies, American journal of cancer research 3(1)

N

(2013) 1.

A

[55] T.-C. Chou, Theoretical basis, experimental design, and computerized simulation of

M

synergism and antagonism in drug combination studies, Pharmacological reviews 58(3) (2006) 621-681.

D

[56] R.B. Mokhtari, T.S. Homayouni, N. Baluch, E. Morgatskaya, S. Kumar, B. Das, H. Yeger,

TE

Combination therapy in combating cancer, Oncotarget 8(23) (2017) 38022.

EP

[57] T.-C. Chou, P. Talalay, Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors, Advances in enzyme regulation 22 (1984) 27-55.

CC

[58] J. Jin, F.-P. Wang, H. Wei, G. Liu, Reversal of multidrug resistance of cancer through inhibition of P-glycoprotein by 5-bromotetrandrine, Cancer chemotherapy and pharmacology

A

55(2) (2005) 179-188. [59] F. Gao, F. Wang, J. Wu, X. Le, Q. Zhang, Screening effective sequences of small interfering RNAs targeting MDR1 gene in human gastric cancer SGC7901/VCR cells, Zhonghua zhong liu za zhi [Chinese journal of oncology] 28(3) (2006) 178-182.

41

[60] T. Choudhuri, S. Pal, T. Das, G. Sa, Curcumin selectively induces apoptosis in deregulated cyclin D1-expressed cells at G2 phase of cell cycle in a p53-dependent manner, Journal of Biological Chemistry 280(20) (2005) 20059-20068.

SC RI PT

[61] S. Bhattacharyya, D. Mandal, G.S. Sen, S. Pal, S. Banerjee, L. Lahiry, J.H. Finke, C.S. Tannenbaum, T. Das, G. Sa, Tumor-Induced Oxidative Stress Perturbs Nuclear Factor-κB Activity-Augmenting Tumor Necrosis Factor-α–Mediated T-Cell Death: Protection by Curcumin, Cancer research 67(1) (2007) 362-370.

[62] V. Lopes-Rodrigues, E. Sousa, M.H. Vasconcelos, Curcumin as a modulator of P-

U

glycoprotein in cancer: challenges and perspectives, Pharmaceuticals 9(4) (2016) 71.

N

[63] P. Kesharwani, S. Banerjee, S. Padhye, F.H. Sarkar, A.K. Iyer, Hyaluronic acid engineered

A

nanomicelles loaded with 3, 4-difluorobenzylidene curcumin for targeted killing of CD44+ stem-

M

like pancreatic cancer cells, Biomacromolecules 16(9) (2015) 3042-3053. [64] F.S. Tabatabaei Mirakabad, A. Akbarzadeh, M. Milani, N. Zarghami, M. Taheri-Anganeh,

D

V. Zeighamian, F. Badrzadeh, M. Rahmati-Yamchi, A Comparison between the cytotoxic effects

TE

of pure curcumin and curcumin-loaded PLGA-PEG nanoparticles on the MCF-7 human breast

EP

cancer cell line, Artificial cells, nanomedicine, and biotechnology 44(1) (2016) 423-430. [65] X. Yang, Z. Li, N. Wang, L. Li, L. Song, T. He, L. Sun, Z. Wang, Q. Wu, N. Luo,

CC

Curcumin-encapsulated polymeric micelles suppress the development of colon cancer in vitro and in vivo, Scientific reports 5 (2015) 10322.

A

[66] N. Ahmadi Nasab, H. Hassani Kumleh, M. Beygzadeh, S. Teimourian, M. Kazemzad, Delivery of curcumin by a pH-responsive chitosan mesoporous silica nanoparticles for cancer treatment, Artificial cells, nanomedicine, and biotechnology 46(1) (2018) 75-81.

42

[67] H. Sadeghzadeh, Y. Pilehvar-Soltanahmadi, A. Akbarzadeh, H. Dariushnejad, F. Sanjarian, N. Zarghami, The effects of nanoencapsulated curcumin-Fe3O4 on proliferation and hTERT gene expression in lung cancer cells, Anti-Cancer Agents in Medicinal Chemistry (Formerly

SC RI PT

Current Medicinal Chemistry-Anti-Cancer Agents) 17(10) (2017) 1363-1373. [68] K.A. Gawde, P. Kesharwani, S. Sau, F.H. Sarkar, S. Padhye, S.K. Kashaw, A.K. Iyer, Synthesis and characterization of folate decorated albumin bio-conjugate nanoparticles loaded with a synthetic curcumin difluorinated analogue, Journal of colloid and interface science 496 (2017) 290-299.

U

[69] P. Kumari, O.S. Muddineti, S.V.K. Rompicharla, P. Ghanta, A.K. BBN, B. Ghosh, S.

N

Biswas, Cholesterol-conjugated poly (D, L-lactide)-based micelles as a nanocarrier system for

A

effective delivery of curcumin in cancer therapy, Drug delivery 24(1) (2017) 209-223.

M

[70] Y.-b. Guan, S.-y. Zhou, Y.-q. Zhang, Y.-d. Tian, Y.-y. Jia, Y.-j. Sun, Therapeutic effects of curcumin nanoemulsions on prostate cancer, Journal of Huazhong University of Science and

D

Technology [Medical Sciences] 37(3) (2017) 371-378.

TE

[71] L.A. Torre, F. Bray, R.L. Siegel, J. Ferlay, J. Lortet‐ Tieulent, A. Jemal, Global cancer

EP

statistics, 2012, CA: a cancer journal for clinicians 65(2) (2015) 87-108. [72] B. Hu, D. Sun, C. Sun, Y.-F. Sun, H.-X. Sun, Q.-F. Zhu, X.-R. Yang, Y.-B. Gao, W.-G.

CC

Tang, J. Fan, A polymeric nanoparticle formulation of curcumin in combination with sorafenib synergistically inhibits tumor growth and metastasis in an orthotopic model of human

A

hepatocellular carcinoma, Biochemical and biophysical research communications 468(4) (2015) 525-532.

43

[73] X. Zhao, Q. Chen, Y. Li, H. Tang, W. Liu, X. Yang, Doxorubicin and curcumin co-delivery by lipid nanoparticles for enhanced treatment of diethylnitrosamine-induced hepatocellular carcinoma in mice, European Journal of Pharmaceutics and Biopharmaceutics 93 (2015) 27-36.

SC RI PT

[74] W. Huang, W. Wang, P. Wang, Q. Tian, C. Zhang, C. Wang, Z. Yuan, M. Liu, H. Wan, H. Tang, Glycyrrhetinic acid-modified poly (ethylene glycol)–b-poly (γ-benzyl l-glutamate) micelles for liver targeting therapy, Acta biomaterialia 6(10) (2010) 3927-3935.

[75] Z.Y. He, X. Zheng, X.H. Wu, X.R. Song, G. He, W.F. Wu, S. Yu, S.J. Mao, Y.Q. Wei, Development of glycyrrhetinic acid-modified stealth cationic liposomes for gene delivery,

U

International journal of pharmaceutics 397(1-2) (2010) 147-154.

N

[76] T. Yan, D. Li, J. Li, F. Cheng, J. Cheng, Y. Huang, J. He, Effective co-delivery of

A

doxorubicin and curcumin using a glycyrrhetinic acid-modified chitosan-cystamine-poly (ε-

B: Biointerfaces 145 (2016) 526-538.

M

caprolactone) copolymer micelle for combination cancer chemotherapy, Colloids and Surfaces

D

[77] Y. Zhang, C. Yang, W. Wang, J. Liu, Q. Liu, F. Huang, L. Chu, H. Gao, C. Li, D. Kong,

TE

Co-delivery of doxorubicin and curcumin by pH-sensitive prodrug nanoparticle for combination

EP

therapy of cancer, Scientific reports 6 (2016) 21225. [78] J. Peng, S. Fumoto, H. Miyamoto, Y. Chen, N. Kuroda, K. Nishida, One-step formation of

CC

lipid-polyacrylic acid-calcium carbonate nanoparticles for co-delivery of doxorubicin and curcumin, Journal of drug targeting 25(8) (2017) 704-714.

A

[79] J. Zhang, J. Li, Z. Shi, Y. Yang, X. Xie, S.M. Lee, Y. Wang, K.W. Leong, M. Chen, pHsensitive polymeric nanoparticles for co-delivery of doxorubicin and curcumin to treat cancer via enhanced pro-apoptotic and anti-angiogenic activities, Acta biomaterialia 58 (2017) 349-364.

44

[80] C. Liu, G. Zhao, J. Liu, N. Ma, P. Chivukula, L. Perelman, K. Okada, Z. Chen, D. Gough, L. Yu, Novel biodegradable lipid nano complex for siRNA delivery significantly improving the chemosensitivity of human colon cancer stem cells to paclitaxel, Journal of controlled release

SC RI PT

140(3) (2009) 277-283. [81] S.J. Dylla, L. Beviglia, I.-K. Park, C. Chartier, J. Raval, L. Ngan, K. Pickell, J. Aguilar, S. Lazetic, S. Smith-Berdan, Colorectal cancer stem cells are enriched in xenogeneic tumors following chemotherapy, PloS one 3(6) (2008) e2428.

[82] W. Tian, J. Liu, Y. Guo, Y. Shen, D. Zhou, S. Guo, Self-assembled micelles of amphiphilic

U

PEGylated rapamycin for loading paclitaxel and resisting multidrug resistant cancer cells,

N

Journal of Materials Chemistry B 3(7) (2015) 1204-1207.

A

[83] B. Xiao, X. Si, M.K. Han, E. Viennois, M. Zhang, D. Merlin, Co-delivery of camptothecin

M

and curcumin by cationic polymeric nanoparticles for synergistic colon cancer combination chemotherapy, Journal of Materials Chemistry B 3(39) (2015) 7724-7733.

D

[84] D. Naor, S.B. Wallach-Dayan, M.A. Zahalka, R.V. Sionov, Involvement of CD44, a

TE

molecule with a thousand faces, in cancer dissemination, Hyaluronan in Cancer Biology,

EP

Elsevier2009, pp. 127-146.

[85] M. Yu, S. Jambhrunkar, P. Thorn, J. Chen, W. Gu, C. Yu, Hyaluronic acid modified

CC

mesoporous silica nanoparticles for targeted drug delivery to CD44-overexpressing cancer cells, Nanoscale 5(1) (2013) 178-183.

A

[86] S.-D. Li, S.B. Howell, CD44-targeted microparticles for delivery of cisplatin to peritoneal metastases, Molecular pharmaceutics 7(1) (2009) 280-290.

45

[87] P.V. Almeida, M.-A. Shahbazi, E. Mäkilä, M. Kaasalainen, J. Salonen, J. Hirvonen, H.A. Santos, Amine-modified hyaluronic acid-functionalized porous silicon nanoparticles for targeting breast cancer tumors, Nanoscale 6(17) (2014) 10377-10387.

SC RI PT

[88] B. Xiao, M.K. Han, E. Viennois, L. Wang, M. Zhang, X. Si, D. Merlin, Hyaluronic acidfunctionalized polymeric nanoparticles for colon cancer-targeted combination chemotherapy, Nanoscale 7(42) (2015) 17745-17755.

[89] F. Mohammadian, Y. Pilehvar-Soltanahmadi, M. Mofarrah, M. Dastani-Habashi, N. Zarghami, Down regulation of miR-18a, miR-21 and miR-221 genes in gastric cancer cell line

U

by chrysin-loaded PLGA-PEG nanoparticles, Artificial cells, nanomedicine, and biotechnology

N

44(8) (2016) 1972-1978.

A

[90] S. Mohammadinejad, A. Akbarzadeh, M. Rahmati-Yamchi, S. Hatam, S. Kachalaki, S.

M

Zohreh, N. Zarghami, Preparation and evaluation of chrysin encapsulated in PLGA-PEG nanoparticles in the T47-D breast cancer cell line, Asian Pac J Cancer Prev 16(9) (2015) 3753-

D

3758.

TE

[91] B.Y. Khoo, S.L. Chua, P. Balaram, Apoptotic effects of chrysin in human cancer cell lines,

EP

International journal of molecular sciences 11(5) (2010) 2188-2199. [92] J. Lotfi-Attari, Y. Pilehvar-Soltanahmadi, M. Dadashpour, S. Alipour, R. Farajzadeh, S.

CC

Javidfar, N. Zarghami, Co-Delivery of Curcumin and Chrysin by Polymeric Nanoparticles Inhibit Synergistically Growth and hTERT Gene Expression in Human Colorectal Cancer Cells,

A

Nutrition and cancer 69(8) (2017) 1290-1299. [93] T. Anirudhan, A.S. Nair, S.J. Bino, Nanoparticle assisted solvent selective transdermal combination therapy of curcumin and 5-flurouracil for efficient cancer treatment, Carbohydrate polymers 173 (2017) 131-142.

46

[94] R. Bagheri, Z. Sanaat, N. Zarghami, Synergistic Effect of Free and Nano-encapsulated Chrysin-Curcumin on Inhibition of hTERT Gene Expression in SW480 Colorectal Cancer Cell Line, Drug research (2018).

SC RI PT

[95] R.L. Siegel, K.D. Miller, A. Jemal, Cancer statistics, 2018, CA Cancer J Clin 68(1) (2018) 7-30.

[96] C.F. Thorn, C. Oshiro, S. Marsh, T. Hernandez-Boussard, H. McLeod, T.E. Klein, R.B. Altman, Doxorubicin pathways: pharmacodynamics and adverse effects, Pharmacogenetics and genomics 21(7) (2011) 440.

U

[97] K. Razavi-Azarkhiavi, M. Iranshahy, A. Sahebkar, K. Shirani, G. Karimi, The protective

N

role of phenolic compounds against doxorubicin-induced cardiotoxicity: a comprehensive

A

review, Nutrition and cancer 68(6) (2016) 892-917.

M

[98] J. Wang, W. Ma, P. Tu, Synergistically Improved Anti‐ tumor Efficacy by Co‐ delivery Doxorubicin and Curcumin Polymeric Micelles, Macromolecular bioscience 15(9) (2015) 1252-

D

1261.

TE

[99] H.B. Ruttala, Y.T. Ko, Liposomal co-delivery of curcumin and albumin/paclitaxel

EP

nanoparticle for enhanced synergistic antitumor efficacy, Colloids and Surfaces B: Biointerfaces 128 (2015) 419-426.

CC

[100] Z.M. Qian, H. Li, H. Sun, K. Ho, Targeted drug delivery via the transferrin receptormediated endocytosis pathway, Pharmacological reviews 54(4) (2002) 561-587.

A

[101] R.S. Mulik, J. Mönkkönen, R.O. Juvonen, K.R. Mahadik, A.R. Paradkar, Transferrin mediated solid lipid nanoparticles containing curcumin: enhanced in vitro anticancer activity by induction of apoptosis, International journal of pharmaceutics 398(1-2) (2010) 190-203.

47

[102] S. Chen, K. Yang, R.G. Tuguntaev, A. Mozhi, J. Zhang, P.C. Wang, X.-J. Liang, Targeting tumor

microenvironment

with

PEG-based

amphiphilic

nanoparticles

to

overcome

chemoresistance, Nanomedicine: Nanotechnology, Biology and Medicine 12(2) (2016) 269-286.

SC RI PT

[103] T. Cui, S. Zhang, H. Sun, Co-delivery of doxorubicin and pH-sensitive curcumin prodrug by transferrin-targeted nanoparticles for breast cancer treatment, Oncology reports 37(2) (2017) 1253-1260.

[104] H.N. Nguyen, P.T. Ha, A. Sao Nguyen, D.T. Nguyen, H.D. Do, Q.N. Thi, M.N.H. Thi, Curcumin as fluorescent probe for directly monitoring in vitro uptake of curcumin combined

U

paclitaxel loaded PLA-TPGS nanoparticles, Advances in Natural Sciences: Nanoscience and

N

Nanotechnology 7(2) (2016) 025001.

A

[105] B.P. Sahu, H. Hazarika, R. Bharadwaj, P. Loying, R. Baishya, S. Dash, M.K. Das,

M

Curcumin-docetaxel co-loaded nanosuspension for enhanced anti-breast cancer activity, Expert opinion on drug delivery 13(8) (2016) 1065-1074.

D

[106] S. Chen, Q. Liang, E. Liu, Z. Yu, L. Sun, J. Ye, M.-C. Shin, J. Wang, H. He,

TE

Curcumin/sunitinib co-loaded BSA-stabilized SPIOs for synergistic combination therapy for

EP

breast cancer, Journal of Materials Chemistry B 5(22) (2017) 4060-4072. [107] M.M. Gottesman, Mechanisms of cancer drug resistance, Annual review of medicine 53(1)

CC

(2002) 615-627.

[108] R. Li, R.a. Wu, L. Zhao, M. Wu, L. Yang, H. Zou, P-glycoprotein antibody functionalized

A

carbon nanotube overcomes the multidrug resistance of human leukemia cells, ACS nano 4(3) (2010) 1399-1408.

48

[109] M. Qin, Y.E.K. Lee, A. Ray, R. Kopelman, Overcoming cancer multidrug resistance by codelivery of doxorubicin and verapamil with hydrogel nanoparticles, Macromolecular bioscience 14(8) (2014) 1106-1115.

SC RI PT

[110] L. Zhang, Y. Li, Z. Jin, K.M. Chan, C.Y. Jimmy, Mesoporous carbon/CuS nanocomposites for pH-dependent drug delivery and near-infrared chemo-photothermal therapy, RSC Advances 5(113) (2015) 93226-93233.

[111] L. Lv, Y. Guo, Y. Shen, J. Liu, W. Zhang, D. Zhou, S. Guo, Intracellularly Degradable, Self‐ Assembled Amphiphilic Block Copolycurcumin Nanoparticles for Efficient In Vivo

U

Cancer Chemotherapy, Advanced healthcare materials 4(10) (2015) 1496-1501.

N

[112] F. Li, Y. Wang, Z. Zhang, Y. Shen, S. Guo, A chemo/photo-co-therapeutic system for

A

enhanced multidrug resistant cancer treatment using multifunctional mesoporous carbon

M

nanoparticles coated with poly (curcumin-dithiodipropionic acid), Carbon 122 (2017) 524-537. [113] J.-S. Baek, C.-W. Cho, A multifunctional lipid nanoparticle for co-delivery of paclitaxel

D

and curcumin for targeted delivery and enhanced cytotoxicity in multidrug resistant breast cancer

TE

cells, Oncotarget 8(18) (2017) 30369.

EP

[114] S. Medel, Z. Syrova, L. Kovacik, J. Hrdy, M. Hornacek, E. Jager, M. Hruby, R. Lund, D. Cmarko, P. Stepanek, Curcumin-bortezomib loaded polymeric nanoparticles for synergistic

CC

cancer therapy, European Polymer Journal 93 (2017) 116-131. [115] Z. Yang, N. Sun, R. Cheng, C. Zhao, J. Liu, Z. Tian, Hybrid nanoparticles coated with

A

hyaluronic acid lipoid for targeted co-delivery of paclitaxel and curcumin to synergistically eliminate breast cancer stem cells, Journal of Materials Chemistry B 5(33) (2017) 6762-6775. [116] J. Lin, Q. Cai, Y. Tang, Y. Xu, Q. Wang, T. Li, H. Xu, S. Wang, K. Fan, Z. Liu, PEGylated Lipid bilayer coated mesoporous silica nanoparticles for co-delivery of paclitaxel and

49

curcumin: Design, characterization and its cytotoxic effect, International journal of pharmaceutics 536(1) (2018) 272-282. [117] S.V. Sudakaran, J.R. Venugopal, G.P. Vijayakumar, S. Abisegapriyan, A.N. Grace, S.

SC RI PT

Ramakrishna, Sequel of MgO nanoparticles in PLACL nanofibers for anti-cancer therapy in synergy with curcumin/β-cyclodextrin, Materials Science and Engineering: C 71 (2017) 620-628. [118] J.-D. Yuan, D.-L. ZhuGe, M.-Q. Tong, M.-T. Lin, X.-F. Xu, X. Tang, Y.-Z. Zhao, H.-L. Xu, pH-sensitive polymeric nanoparticles of mPEG-PLGA-PGlu with hybrid core for simultaneous encapsulation of curcumin and doxorubicin to kill the heterogeneous tumour cells

U

in breast cancer, Artificial cells, nanomedicine, and biotechnology (2018) 1-12.

N

[119] O. Gallego, Nonsurgical treatment of recurrent glioblastoma, Current oncology 22(4)

A

(2015) e273.

M

[120] Y. Cui, M. Zhang, F. Zeng, H. Jin, Q. Xu, Y. Huang, Dual-targeting magnetic PLGA nanoparticles for codelivery of paclitaxel and curcumin for brain tumor therapy, ACS applied

D

materials & interfaces 8(47) (2016) 32159-32169.

TE

[121] L. Szablewski, Expression of glucose transporters in cancers, Biochimica et Biophysica

EP

Acta (BBA)-Reviews on Cancer 1835(2) (2013) 164-169. [122] W.-L. Yeh, C.-J. Lin, W.-M. Fu, Enhancement of glucose transporter expression of brain

CC

endothelial cells by vascular endothelial growth factor derived from glioma exposed to hypoxia, Molecular pharmacology 73(1) (2008) 170-177.

A

[123] R.J. Boado, K.L. Black, W.M. Pardridge, Gene expression of GLUT3 and GLUT1 glucose transporters in human brain tumors, Molecular brain research 27(1) (1994) 51-57. [124] C. Sarisozen, S. Dhokai, E.G. Tsikudo, E. Luther, I.M. Rachman, V.P. Torchilin, Nanomedicine based curcumin and doxorubicin combination treatment of glioblastoma with

50

scFv-targeted micelles: in vitro evaluation on 2D and 3D tumor models, European Journal of Pharmaceutics and Biopharmaceutics 108 (2016) 54-67. [125] J.S. Kirby, C.J. Miller, Intralesional chemotherapy for nonmelanoma skin cancer: a

SC RI PT

practical review, Journal of the American Academy of Dermatology 63(4) (2010) 689-702. [126] J.E. Zuckerman, M.E. Davis, Clinical experiences with systemically administered siRNAbased therapeutics in cancer, Nature reviews Drug discovery 14(12) (2015) 843.

[127] X.-M. Wu, H. Todo, K. Sugibayashi, Enhancement of skin permeation of high molecular compounds by a combination of microneedle pretreatment and iontophoresis, Journal of

U

controlled release 118(2) (2007) 189-195.

N

[128] A. Jose, S. Labala, V.V.K. Venuganti, Co-delivery of curcumin and STAT3 siRNA using

A

deformable cationic liposomes to treat skin cancer, Journal of drug targeting 25(4) (2017) 330-

M

341.

[129] A.K.T. Thulasidasan, A.P. Retnakumari, M. Shankar, V. Vijayakurup, S. Anwar, S.

and

chemosensitizing

TE

bioavailability

D

Thankachan, K.S. Pillai, J.J. Pillai, C.D. Nandan, V.V. Alex, Folic acid conjugation improves the efficacy

of

curcumin-encapsulated

PLGA-PEG

EP

nanoparticles towards paclitaxel chemotherapy, Oncotarget 8(64) (2017) 107374. [130] A. Jose, S. Labala, K.M. Ninave, S.K. Gade, V.V.K. Venuganti, Effective Skin Cancer

CC

Treatment by Topical Co-delivery of Curcumin and STAT3 siRNA Using Cationic Liposomes, AAPS PharmSciTech 19(1) (2018) 166-175.

A

[131] S.P. Singh, S.B. Alvi, D.B. Pemmaraju, A.D. Singh, S.V. Manda, R. Srivastava, A.K. Rengan, NIR triggered liposome gold nanoparticles entrapping curcumin as in situ adjuvant for photothermal treatment of skin cancer, International journal of biological macromolecules (2017).

51

[132] B. Kim, C. Lee, E.S. Lee, B.S. Shin, Y.S. Youn, Paclitaxel and curcumin co-bound albumin nanoparticles having antitumor potential to pancreatic cancer, asian journal of pharmaceutical sciences 11(6) (2016) 708-714.

SC RI PT

[133] A. Thakkar, S. Chenreddy, A. Thio, W. Khamas, J. Wang, S. Prabhu, Preclinical systemic toxicity evaluation of chitosan-solid lipid nanoparticle-encapsulated aspirin and curcumin in combination with free sulforaphane in BALB/c mice, International journal of nanomedicine 11 (2016) 3265.

[134] A. Longhi, C. Errani, M. De Paolis, M. Mercuri, G. Bacci, Primary bone osteosarcoma in

U

the pediatric age: state of the art, Cancer treatment reviews 32(6) (2006) 423-436.

N

[135] C.L. Schwartz, R. Gorlick, L. Teot, M. Krailo, Z. Chen, A. Goorin, H.E. Grier, M.L.

A

Bernstein, P. Meyers, Multiple drug resistance in osteogenic sarcoma: INT0133 from the

M

Children's Oncology Group, Journal of clinical oncology 25(15) (2007) 2057-2062. [136] S. Li, W. Sun, H. Wang, D. Zuo, Y. Hua, Z. Cai, Research progress on the multidrug

D

resistance mechanisms of osteosarcoma chemotherapy and reversal, Tumor Biology 36(3) (2015)

TE

1329-1338.

EP

[137] L. Wang, W. Wang, Z. Rui, D. Zhou, The effective combination therapy against human osteosarcoma:

doxorubicin

plus

curcumin

co-encapsulated

lipid-coated

polymeric

CC

nanoparticulate drug delivery system, Drug delivery 23(9) (2016) 3200-3208. [138] K.D. Miller, R.L. Siegel, C.C. Lin, A.B. Mariotto, J.L. Kramer, J.H. Rowland, K.D. Stein,

A

R. Alteri, A. Jemal, Cancer treatment and survivorship statistics, 2016, CA: a cancer journal for clinicians 66(4) (2016) 271-289. [139] G. Liu, E. Franssen, M.I. Fitch, E. Warner, Patient preferences for oral versus intravenous palliative chemotherapy, Journal of Clinical Oncology 15(1) (1997) 110-115.

52

[140] H.M. Ashour, A. El-Sharif, Microbial spectrum and antibiotic susceptibility profile of gram-positive aerobic bacteria isolated from cancer patients, Journal of Clinical Oncology 25(36) (2007) 5763-5769.

SC RI PT

[141] M. Bar-Zeev, Y.G. Assaraf, Y.D. Livney, β-casein nanovehicles for oral delivery of chemotherapeutic Drug combinations overcoming P-glycoprotein-mediated multidrug resistance in human gastric cancer cells, Oncotarget 7(17) (2016) 23322.

[142] S. Oudard, Progress in emerging therapies for advanced prostate cancer, Cancer treatment reviews 39(3) (2013) 275-289.

U

[143] P. Saralkar, A.K. Dash, Alginate nanoparticles containing curcumin and resveratrol:

N

preparation, characterization, and in vitro evaluation against DU145 prostate cancer cell line,

A

AAPS PharmSciTech 18(7) (2017) 2814-2823.

M

[144] H. Hirte, E. Kennedy, L. Elit, M.F.K. Fung, Systemic therapy for recurrent, persistent, or metastatic cervical cancer: a clinical practice guideline, Current Oncology 22(3) (2015) 211.

D

[145] K. Scatchard, J. Forrest, M. Flubacher, P. Cornes, C. Williams, Chemotherapy for

TE

metastatic and recurrent cervical cancer, Cochrane Database Syst Rev 10 (2007).

EP

[146] Z. Wang, J. Lv, T. Zhang, Combination of IL-24 and cisplatin inhibits angiogenesis and lymphangiogenesis of cervical cancer xenografts in a nude mouse model by inhibiting VEGF,

CC

VEGF-C and PDGF-B, Oncology reports 33(5) (2015) 2468-2476. [147] A. Macciò, C. Madeddu, Cisplatin: an old drug with a newfound efficacy–from

A

mechanisms of action to cytotoxicity, Expert opinion on pharmacotherapy 14(13) (2013) 18391857.

53

[148] S. Ugur, R. Ulu, A. Dogukan, A. Gurel, I.P. Yigit, N. Gozel, B. Aygen, N. Ilhan, The renoprotective effect of curcumin in cisplatin-induced nephrotoxicity, Renal failure 37(2) (2015) 332-336.

SC RI PT

[149] H. Kulhari, D. Pooja, M.K. Singh, A.S. Chauhan, Optimization of carboxylate-terminated poly (amidoamine) dendrimer-mediated cisplatin formulation, Drug development and industrial pharmacy 41(2) (2015) 232-238.

[150] P. Chen, J. Li, H.-G. Jiang, T. Lan, Y.-C. Chen, Curcumin reverses cisplatin resistance in cisplatin-resistant lung caner cells by inhibiting FA/BRCA pathway, Tumor Biology 36(5)

U

(2015) 3591-3599.

N

[151] Y. Wang, P.-C. Hu, F.-F. Gao, J.-W. Lv, S. Xu, C.-C. Kuang, L. Wei, J.-W. Zhang, The

A

protective effect of curcumin on hepatotoxicity and ultrastructural damage induced by cisplatin,

M

Ultrastructural pathology 38(5) (2014) 358-362.

[152] C. Li, X. Ge, L. Wang, Construction and comparison of different nanocarriers for co-

D

delivery of cisplatin and curcumin: a synergistic combination nanotherapy for cervical cancer,

TE

Biomedicine & Pharmacotherapy 86 (2017) 628-636.

EP

[153] D. Guo, L. Zhu, Z. Huang, H. Zhou, Y. Ge, W. Ma, J. Wu, X. Zhang, X. Zhou, Y. Zhang, Anti-leukemia activity of PVP-coated silver nanoparticles via generation of reactive oxygen

CC

species and release of silver ions, Biomaterials 34(32) (2013) 7884-7894. [154]

A.

Sangsuwan,

H.

Kawasaki,

Y.

Iwasaki,

Thiolated-2-methacryloyloxyethyl

A

phosphorylcholine protected silver nanoparticles as novel photo-induced cell-killing agents, Colloids and Surfaces B: Biointerfaces 140 (2016) 128-134.

54

[155] P.D. Petrov, K. Yoncheva, V. Gancheva, S. Konstantinov, B. Trzebicka, Multifunctional block copolymer nanocarriers for co-delivery of silver nanoparticles and curcumin: synthesis and enhanced efficacy against tumor cells, European Polymer Journal 81 (2016) 24-33.

SC RI PT

[156] W.-P. Su, F.-Y. Cheng, D.-B. Shieh, C.-S. Yeh, W.-C. Su, PLGA nanoparticles codeliver paclitaxel and Stat3 siRNA to overcome cellular resistance in lung cancer cells, International journal of nanomedicine 7 (2012) 4269.

[157] X. Zhang, Q. Wang, L. Qin, H. Fu, Y. Fang, B. Han, Y. Duan, EGF-modified mPEGPLGA-PLL nanoparticle for delivering doxorubicin combined with Bcl-2 siRNA as a potential

U

treatment strategy for lung cancer, Drug delivery 23(8) (2016) 2936-2945.

N

[158] J. Tang, H. Ji, J. Ren, M. Li, N. Zheng, L. Wu, Solid lipid nanoparticles with TPGS and

A

Brij 78: A co-delivery vehicle of curcumin and piperine for reversing P-glycoprotein-mediated

M

multidrug resistance in vitro, Oncology letters 13(1) (2017) 389-395. [159] A.H. Soloway, W. Tjarks, B.A. Barnum, F.-G. Rong, R.F. Barth, I.M. Codogni, J.G.

D

Wilson, The chemistry of neutron capture therapy, Chemical reviews 98(4) (1998) 1515-1562.

TE

[160] Z. Sui, R. Salto, J. Li, C. Craik, P.R.O. de Montellano, Inhibition of the HIV-1 and HIV-2

EP

proteases by curcumin and curcumin boron complexes, Bioorganic & medicinal chemistry 1(6) (1993) 415-422.

CC

[161] D. Alberti, N. Protti, M. Franck, R. Stefania, S. Bortolussi, S. Altieri, A. Deagostino, S. Aime, S. Geninatti Crich, Theranostic Nanoparticles Loaded with Imaging Probes and

A

Rubrocurcumin for Combined Cancer Therapy by Folate Receptor Targeting, ChemMedChem 12(7) (2017) 502-509. [162] Registry and Results Database of U.S. National Institutes of Health. Available from: www.clinicaltrials.gov. Accessed March 2018.

55

[163] J. Noh, B. Kwon, E. Han, M. Park, W. Yang, W. Cho, W. Yoo, G. Khang, D. Lee, Amplification of oxidative stress by a dual stimuli-responsive hybrid drug enhances cancer cell death, Nature communications 6 (2015) 6907.

SC RI PT

[164] M.I. Setyawati, C.Y. Tay, D. Docter, R.H. Stauber, D.T. Leong, Understanding and exploiting nanoparticles' intimacy with the blood vessel and blood, Chemical Society Reviews 44(22) (2015) 8174-8199.

[165] J. Shi, A.R. Votruba, O.C. Farokhzad, R. Langer, Nanotechnology in drug delivery and tissue engineering: from discovery to applications, Nano letters 10(9) (2010) 3223-3230.

U

[166] J Yan, Y Wang, X Zhang, S Liu, C Tian, H Wang, Targeted nanomedicine for prostate

N

cancer therapy: docetaxel and curcumin co-encapsulated lipid–polymer hybrid nanoparticles for

A

the enhanced anti-tumor activity in vitro and in vivo. Drug delivery 23(5) (2016) 1757-1762.

M

[167] KA Gawde, S Sau, K Tatiparti, SK Kashaw, M Mehrmohammadi, AS Azmi, AK Iyer Paclitaxel and di-fluorinated curcumin loaded in albumin nanoparticles for targeted synergistic

TE

(2018) 8-19.

D

combination therapy of ovarian and cervical cancers. Colloids and Surfaces B: Biointerfaces. 167

EP

[168] D Pramanik, NR Campbell, S Das, S Gupta, V Chenna, S Bisht, Sysa-Shah, D Bedja, C Karikari, C Steenbergen, KL Gabrielson, A composite polymer nanoparticle overcomes

CC

multidrug resistance and ameliorates doxorubicin-associated cardiomyopathy. Oncotarget 3(6) (2012) 640.

A

[169] S Chen, Q Liang, E Liu, Yu, L Sun, J Ye, MC Shin, J Wang, H He, Curcumin/sunitinib coloaded BSA-stabilized SPIOs for synergistic combination therapy for breast cancer Journal of Materials Chemistry B 5(22) (2017) 4060-4072.

56

Figure legends Figure 1: Different type of cancers that curcumin is found to manage.

SC RI PT

Figure 2: The anti-cancer mechanism of curcumin. It is characterized by decrease in angiogenesis, metastasis and proliferation and increase in apoptosis with associated factors.

Figure 3: Different types of possible curcumin nano-formulation with their major advantages. Figure 4: In vivo antitumor efficacy of LPNs as compared to other groups. Tail injection of

different formulation groups of equivalent doses was administered in mice. Co-administration by

N

reproduced with copyright permissions from [166])

U

DTX and CUR LPN's showed most efficient suppression on tumor growth (p < 0.05). (Figure

A

Figure 5: NanoDoxCurc (NDC) surmounts DOX resistance (a) Tumor sections from Dox

M

xenografts examined by fluorescence microscopy; accumulation of DOX nanoparticles observed (b) NDC significantly inhibits the growth of subcutaneous DOX-resistant cancer xenografts.

D

Excised tumors presented significant reduction in tumor growth with NDC group as compared to

TE

other treatment groups (N=5, *p<0.005). (c) Post treatment levels of MDR1 expression measured

EP

by immunofluorescence and (d) western blot in Dox xenografts. (e) Mice with DOX-resistant ascites with different treatment groups; more than 50% increase in survival was observed in

CC

NDC treated mice compared to others (N=8, *p<0.005). (Figure reproduced from open access unrestricted use article [167])

A

Figure 6: Schematic representation of delivery of nanocarrier combination therapy. (A) An anticancer drug in free form and curcumin as nanocarrier (Free drug + Nano), (B) Separate nanocarrier delivery of both drugs (Nano + Nano), C) Co-encapsulation of both drugs in the same nanocarrier (co-encapsulation)

57

Figure 7: (a-d) Combination Index Plots of Folate-BSA-difluorinated curcumin (T C) and Folate-BSA-Paclitaxel (T P) using MTT assay in SKOV3 Cell line (left) and HeLa cell lines (right). The data demonstrates synergistic cell killing effect (e) depicts the Fa-CI plot

SC RI PT

demonstrating synergistic combination efficiency as compare to fraction effected. Average synergism reported at Fa < 0.5. (f) Graph showing the enhance cell killing compared to paclitaxel. (Figures reproduced with copyright permissions from [168]).

Figure 8: Combination Nanocarrier possible dosing and effects. The in-vitro and then in-vivo fixed molar ratio (synergistic effect) can be translated from in vitro assay to in vivo assay using

U

nanoparticle approach strategy.

N

Figure 9: In-vitro cytotoxicity depicting chemotherapy efficacy (chemo-sensitizing effect) and

A

drug resistance reversal of synthesized doxorubicin with curcumin coated mesoporous carbon

M

nanoparticles (PEG-PCDA/MCNs) was tested by MTT assay against drug resistant of MCF7/ADR cells. (A) DOX dose-dependent in vitro cytotoxicity of different DOX formulations

D

against the drug resistant MCF-7/ADR cells after 48 h incubation; Free DOX shows lowest

TE

inhibition concentrations (IC50) vs significantly enhanced IC50 with the designed nanoparticles,

EP

(B) Viability rate of MCF-7/ADR cells after 48 h incubation with MCNs and PEGPCDA/MCNs; a significantly decreased viability of cells is seen with the drug incorporated

CC

nanoparticles vs the controls, (significant difference, *: p < 0.05; **: p < 0.01). (figure reproduced with permission from Li et al. [112])

A

Figure 10: A mechanism of action display for a possible paclitaxel (PTX) and curcumin (CUR) nanoparticle. Their interaction with Folate receptor and subsequent Endocytosis of the PLGAFA-PTX Lipid Nanocarrier has been depicted.

58

N U SC RI PT

Figures

A

CC E

PT

ED

M

A

Figure 1:

59

A ED

PT

CC E A

M

N U SC RI PT

Figure 2:

60

A ED

PT

CC E A

M

N U SC RI PT

Figure 3:

61

A ED

PT

CC E A

M

N U SC RI PT

Figure 4:

62

A ED

PT

CC E A

M

N U SC RI PT

Figure 5:

63

A ED

PT

CC E A

M

N U SC RI PT

Figure 6:

64

A ED

PT

CC E A

M

N U SC RI PT

Figure 7:

65

A ED

PT

CC E A

M

N U SC RI PT

Figure 8:

66

A ED

PT

CC E A

M

N U SC RI PT

Figure 9:

67

A ED

PT

CC E A

M

N U SC RI PT

Figure 10:

68