You are on page 1of 15

Clinica Chimica Acta 436 (2014) 78–92

Contents lists available at ScienceDirect

Clinica Chimica Acta


journal homepage: www.elsevier.com/locate/clinchim

Invited critical review

Targeted anticancer therapy: Overexpressed receptors


and nanotechnology
Mohd Javed Akhtar a,⁎, Maqusood Ahamed a, Hisham A. Alhadlaq a,b, Salman A. Alrokayan c, Sudhir Kumar d
a
King Abdullah Institute for Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia
b
Department of Medical Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
c
Department of Biochemistry, King Saud University, Riyadh 11451, Saudi Arabia
d
Department of Zoology, University of Lucknow, Lucknow 226007, India

a r t i c l e i n f o a b s t r a c t

Article history: Targeted delivery of anticancer drugs to cancer cells and tissues is a promising field due to its potential to spare
Received 12 February 2014 unaffected cells and tissues, but it has been a major challenge to achieve success in these therapeutic approaches.
Received in revised form 7 May 2014 Several innovative approaches to targeted drug delivery have been devised based on available knowledge in can-
Accepted 10 May 2014
cer biology and on technological advancements. To achieve the desired selectivity of drug delivery, nanotechnol-
Available online 15 May 2014
ogy has enabled researchers to design nanoparticles (NPs) to incorporate anticancer drugs and act as
Keywords:
nanocarriers. Recently, many receptor molecules known to be overexpressed in cancer have been explored as
Nanoparticles docking sites for the targeting of anticancer drugs. In principle, anticancer drugs can be concentrated specifically
Overexpressed receptors in cancer cells and tissues by conjugating drug-containing nanocarriers with ligands against these receptors. Sev-
Ligands eral mechanisms can be employed to induce triggered drug release in response to either endogenous trigger or
Anticancer drugs exogenous trigger so that the anticancer drug is only released upon reaching and preferentially accumulating in
Tumor microenvironment the tumor tissue. This review focuses on overexpressed receptors exploited in targeting drugs to cancerous tis-
sues and the tumor microenvironment. We briefly evaluate the structure and function of these receptor mole-
cules, emphasizing the elegant mechanisms by which certain characteristics of cancer can be exploited in
cancer treatment. After this discussion of receptors, we review their respective ligands and then the anticancer
drugs delivered by nanotechnology in preclinical models of cancer. Ligand-functionalized nanocarriers have de-
livered significantly higher amounts of anticancer drugs in many in vitro and in vivo models of cancer compared
to cancer models lacking such receptors or drug carrying nanocarriers devoid of ligand. This increased concentra-
tion of anticancer drug in the tumor site enabled by nanotechnology could have a major impact on the efficiency
of cancer treatment while reducing systemic side effects.
© 2014 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
2. Overexpressed receptors, their ligands, and drug delivery to cancer using nanotechnology . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
2.1. G protein-coupled receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 80
2.1.1. Bombesin receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
2.1.2. Somatostatin receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
2.1.3. Endothelin receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
2.2. Integrin receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
2.2.1. Integrin αvβ3 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
2.2.2. Integrin α-3 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84

Abbreviations: Bn, bombesin; BnR, bombesin receptor; BR, biotin receptor; c(RGD-K), cyclic {Arginine-Glycine-Aspartic acid (RGD)} containing peptide; CA, cholic acid; DOX, doxoru-
bicin; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; ETRB, endothelin receptor B; FA, folic acid; FGF, fibroblast growth factor; FGFR, fibroblast growth factor re-
ceptor; FR, folate receptor; FSH, follicle stimulating hormone; FSHR, follicle stimulating hormone receptor; NPs, nanoparticles; Nrp-1, neuropilin receptor-1; PEG, poly(ethylene glycol);
PLA, poly(lactic acid); PTMC, poly(trimethylene carbonate); PTX, paclitaxel; QDs, quantum dots; S1R, sigma receptor 1; S2R, sigma receptor 2; SRs, sigma receptors; SSTRs, somatostatin
receptors; Tf, transferrin; TfR, transferrin receptor.
⁎ Corresponding author at: King Abdullah Institute for Nanotechnology (KAIN), King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia. Tel.: +966 559981310 (mobile).
E-mail addresses: mjakhtar@ksu.edu.sa, mohd.j.akhtar@gmail.com (M.J. Akhtar).

http://dx.doi.org/10.1016/j.cca.2014.05.004
0009-8981/© 2014 Elsevier B.V. All rights reserved.
M.J. Akhtar et al. / Clinica Chimica Acta 436 (2014) 78–92 79

2.3. Folate receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84


2.4. Transferrin receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
2.5. Epidermal growth factor receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
2.6. Fibroblast growth factor receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
2.7. Sigma receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
2.8. Other overexpressed receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
3. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
Acknowledgment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89

1. Introduction slightly lower than that in normal tissue, and this difference has been
exploited to achieve pH-triggered drug release in tumor tissue [6]. A
More than 10 million patients are diagnosed with new cases of can- diverse group of functional materials has been designed to accomplish
cer every year, and approximately 27 million new cases of cancer will triggered drug release in response to other stimuli such as temperature
have been recorded by 2030 [1,2]. While cytotoxic chemotherapeutic [7,8], redox potential [9], ultrasound [10], and light [11]. These mecha-
agents such as paclitaxel (PTX) and doxorubicin (DOX) effectively kill nisms of endogenously or exogenously triggered drug release [12] can
cancer cells, they cannot distinguish cancer cells from normal cells. only be applied once the anticancer drug reaches and accumulates at
This lack of selectivity leads to undesirable systemic toxicity when pa- the desired site, i.e., tumor tissues. Molecular markers of cancer include
tients are exposed to the high dosages of cytotoxic agents required to the differential expression of proteins residing in the cytosol, organelles
eradicate the tumor. Improving the selectivity of anticancer drug deliv- or membrane. The group of differentially expressed proteins includes
ery to cancer cells and the tumor microenvironment while sparing nor- receptors that are specifically expressed or overexpressed in cancer
mal cells and tissues is a major challenge in the effective treatment of cells compared to normal cells. These overexpressed receptors have
cancers of various tissues and organs. Marked differences are found in provided important endogenous tools for exploitation in the active
cancer cells and tissues in terms of biochemical, molecular and physio- targeting of drugs to cancer cells. Targeting nanocarriers to a particular
logical features when compared with normal cells and tissues such as organ or tissue through the blood or lymph circulation is referred to
differences in redox status, pH levels, expression of certain cell mem- as primary targeting, while the accumulation around a cancer cell
brane receptors, the leakiness of tumor tissues and the tumor vascula- is named secondary targeting and manipulating the uptake of
ture. Therefore, cancer stands out as a disease likely to benefit from nanocarriers/drugs by cells and cellular compartments is known as ter-
targeted drug delivery approaches exploiting these differences. These tiary targeting [13]. This review focuses on overexpressed receptors
differences between healthy and cancerous cells and tissues, or hall- exploited for targeting drugs to cancer and the tumor microenviron-
marks of cancer, have recently been reviewed [3]. ment (Table 1). We briefly evaluate the structure and function of
Differences in normal and cancer biology are found at the level of these receptor molecules, emphasizing the elegance of exploiting char-
anatomy, biochemistry and molecular biology. The characteristic ana- acteristics of cancer in cancer treatment. After discussing these recep-
tomic features of tumor biology include the leakiness of blood vessels tors, their respective ligands are mentioned, followed by a review of
and poor lymphatic drainage in tumor tissues. The morphology and the anticancer drugs that have been delivered by nanotechnology in
shape of blood endothelial cells are different in cancer vasculature due preclinical models of cancer (Table 2).
to the presence of fenestrae between adjacent cells, and thus the lack
of contact inhibition. Most solid tumors, upon reaching a certain level 2. Overexpressed receptors, their ligands, and drug delivery to
of growth, exhibit enhanced vascular permeability, ensuring a sufficient cancer using nanotechnology
supply of nutrients and oxygen to tumor tissues and outpacing the
growth of surrounding tissues. Blood vessels in tumors are often dilated Receptors overexpressed or specifically expressed in cancers of var-
and convoluted, and compared with normal tissues, exhibit branching ious tissues and cells are listed in Table 1. These receptors provide
patterns that feature excessive loops and arteriolar–venous shunts [4]. unique opportunities to understand cancer biology and its treatment.
All these features enhance the permeability of blood vessels in tumors In attempts to treat cancer, overexpressed receptors are directly modu-
compared with the vasculature in normal tissues, enabling the delivery lated/inhibited by agents such as antibodies or antibody fragments, and
and accumulation of molecules and other substances in tumor tissues also by other small chemicals that directly bind these receptors and
that are generally not able to enter the vasculature in normal tissues. block their activities. These treatment strategies thus block the consis-
This is called the enhanced permeability and retention (EPR) effect, a tent unwanted stimulus for uncontrolled cell division, thus blocking
phenomenon associated with the tumor vasculature. The net result of cancer progression. Other approaches to cancer therapy do not inten-
the EPR effect for circulating molecules depends on their properties tionally interfere with receptor function, but rather exploit receptor
including size, shape, charge, and polarity. The principal difference be- overexpression for the targeted delivery of effective anticancer drugs
tween EPR and passive localization lies in the characteristics of retention that do not discriminate between cancer and normal cells. These
and tissue clearance rather than uptake. Small drug molecules rapidly drugs can be guided by linking them to suitable ligands against such
penetrate into the tumor interstitial space, but in the absence of specific overexpressed receptors. The field of nanotechnology has developed
binding to cellular proteins, drug is not retained and may be free to dif- the ability to synthesize an enormous variety of NPs, providing a plat-
fuse out of the tissue back into the blood pool or the lymphatic system. form for guiding and carrying drugs to cancer cells and tumor tissues.
In contrast, macromolecules have smaller diffusion constants, reducing These NPs can be loaded with anticancer drugs and conjugated with li-
the initial rate of tumor uptake but also tending to increase the half-life gands. Nanosized liposomes, dendrimers, micelles, metals, alloys, mix-
of blood-pool circulation by enhancing tissue retention and decreasing tures of inorganics and organics, and other materials have been
the rate of clearance [5]. Therefore, receptor–ligand interactions are synthesized by nanotechnology for use in diverse applications such as
the most important aspect of active targeting of nanocarriers (or catalysis, sensing and medicine. NPs loaded with drug molecules and
nanoconjugates or macromolecules) and could be further potentiated conjugated with receptor ligands have been variously termed as nano-
by EPR (Fig. 1). The biochemical conditions of acidity (lower pH value) conjugates, nano-formulations, nano-carriers, etc. Table 2 summarizes
and hypoxia (lower concentration of oxygen) generally prevail in can- some recent outcomes of the nanotechnology-based interventions in
cer cells and tumor tissues. The extracellular pH in tumor tissue is the targeting of anticancer drugs in preclinical in vitro and in vivo
80 M.J. Akhtar et al. / Clinica Chimica Acta 436 (2014) 78–92

Endothelial cells

Normal vasculature should allow very little Leaky tumour vasculature should allow
transport, if any, of nanoconjugatesfrom nanoconjugate transportation in either direction
circulation to tissues. with higher rate due to EPR effect.

No ligand -receptor Ligand-receptor interaction causing


interaction accumulation and retention of drug
within tumor microenvironment

Normal Unaffected Cancer Demise of


cell normal cell cell cancer cell

Fig. 1. Different biological responses mediated by receptor ligands and the resulting cellular fate in cancer vs. normal cells. Ligand decorated nanoconjugates would in principle gradually
concentrate within tumor tissues due to the higher leakage mediated by the EPR effect and the consequent entrapment caused by ligand binding with overexpressed receptors in cancer
cells.

models of cancer exploring overexpressed receptors and their respec- derived from. Researchers have recently begun to exploit this difference
tive ligands. in expression in the targeting of chemotherapeutics, radiodiagnostics
and radiotherapeutics to tumors [14]. A variety of external stimuli in-
2.1. G protein-coupled receptors cluding neurotransmitters, hormones, and phospholipids can activate
G-protein coupled receptors (GPCRs), transducing signals from the ex-
Many tumor cells specifically express or overexpress G protein- tracellular environment to the cytoplasm. GPCRs share the common
coupled receptors (GPCRs) compared with the original tissue they are structure of a single polypeptide with seven membrane-spanning

Table 1
Description of overexpressed receptors widely used for selective drug delivery using receptor–ligand pairs in preclinical models of cancer.

Major receptor type Specific receptor(s) Overexpression in cancer cell types

G protein coupled receptors Bombesin receptor (BnR) Lung, prostate, breast, pancreatic, head/neck, colon, uterine, ovarian,
(GPCRs) This family consists of three closely related proteins, based on their renal cell, glioblastomas, neuroblastomas, gastrointestinal carcinoids,
amino acid sequence homology — i) the gastrin-releasing peptide intestinal carcinoids, and bronchial carcinoids.
receptor (GRP receptor); ii) the neuromedin B receptor (NMB receptor)
and iii) the orphan receptor, BRS-3 with unknown ligands.
Somatostatins receptors (SSTRs) Small cell lung, neuroendocrine tumor, prostate cancer, breast cancer,
Five subtypes of SSTRs have been described so far termed as SSTR 1–5. colorectal carcinoma, gastric cancer, hepatocellular carcinoma
SSTR-2 is being widely used.
Endothelin receptors (ETRs) Melanoma tissues
Two highly homologous receptors — i) ETRA and ii) ETRB bind, but with
different affinities, to three closely related ligands, endothelins (ET;
ET-1, ET-2 and ET-3) each consisted of 21 amino acids.
Integrins Among several subtypes formed by various combinations of α and β Activated endothelial cells and tumor cells (such as U87MG
subunits, ανβ3 is of particular interest in selective drug targeting. glioblastoma cells), ovarian cancer cells.
Folate receptors (FRs) FRα, FRβ and FRγ. The best studied, and widely used in selective drug Most tissues including breast cancer cells.
targeting, of these receptors is folate receptor-α (FRα), a cell surface
glycosyl phosphatidylinositol-anchored glycoprotein that can internal-
ize bound folates and folate-conjugated compounds via receptor-
mediated endocytosis.
Transferrin receptors (TfRs) Two types of receptors (ubiquitously expressed TfR1 and TfR2 Breast, ovary, and brain cancers such as glioma and glioblastomas.
restricted to hepatocytes) only have been described so far. The
transferrin receptor 2 (TfR2) shares a 45% identity and 66% similarity in
its extracellular domain with TfR1.
Epidermal growth factor EGFR family consists of four members: EGFR (or ErbB1, HER1), ErbB2 Lung, breast, bladder, and ovarian cancers.
receptor (EGFR) (HER2, neu in rodents), ErbB3 (HER3) and ErbB4 (HER4).
Fibroblast growth factors A hallmark of FGFRs is the presence of an acidic, serine-rich sequence in Breast, prostate, bladder, and gastric cancer
(FGFRs) the linker between D1 and D2, termed the acid box.
Sigma receptors (SRs) The two sigma receptors – S1R and S2R – were distinguished classically Non-small cell lung carcinoma, prostate cancer, melanoma, and breast
on the basis of their binding affinity for pentazocine and guanidine- cancer.
DTG. Both bind pentazocine whereas only the latter binds with DTG.
Others Follicle stimulating hormone receptors (FSHRs) Ovarian surface epithelium
Biotin receptors (BRs) Leukemia
C-type lectin receptors (CLRs). Hepatocytes, dendritic cells, macrophages
Asialoglycoprotein receptor (ASGPR)
NRP-1 Human vascular cells
M.J. Akhtar et al. / Clinica Chimica Acta 436 (2014) 78–92 81

Table 2
Some recent outcomes of nanotechnology in the delivery of anticancer drugs in preclinical models of cancer exploiting overexpressed receptors and their respective ligands. Note that
specific names of nanocarriers, for example liposomes, may consist of the same or different building blocks/monomers. Similarly, micelles and other carriers might also differ in size,
shape, charges, composition, and other properties.

Specific Ligands/ligands analogues Nanoparticles/nanocarriers Anticancer drugs/agents Major outcome References


receptor(s)

BnR Peptide fragment called BN Liposome DOX Comparatively greater amount of DOX was delivered [27]
(7–14) to PC-3 cancer cells and xenografts expressing higher
BnR.
BnR BN (6–14) No nanocarrier Mitochondria-disrupting peptide, Greater cytotoxicity to various human cancer cells [31]
B28 (DU145, PC-3, MCF-7, HSF, MRC-5, PrSC)-expressing
BnR.
SSTR OCT Liposome DOX OCT enhanced DOX uptake in SMMC-7721 cells, and [45]
tumor of pancreas and melanoma of B16 tumor
bearing BALB/c mice.
SSTR OCT Liposome DOX Sterically stabilized liposomes (SSL) increased [46]
intracellular delivery of DOX in SSTR2-positive cells
through a mechanism of receptor-mediated endocy-
tosis.
SSTR OCT Micelles DOX OCT caused higher cytotoxicity to MCF-7 cells with [47]
high expression of SSTR. Nude mice bearing MCF-7
cancer xenografts exhibited enhanced anti-tumor
efficacy and decreased systemic toxicity.
ETRB mAB against N-terminal tail Antibody–drug conjugate Monomethylauristatin E (MMAE) ADC demonstrated remarkable efficacy against [56]
of ETRB (ADC) human melanoma cell lines and xenograft tumor
models in correlation with the levels of receptor
expression.
αvβ3 c(RGDfK) Liposome DOX This delivery method resulted in a 15-fold improve- [67]
ment in tumor and anti-metastatic activity in human
umbilical vein endothelial cells (HUVECs) expressing
high levels of αvβ3 when compared with free drug.
αvβ3 c(RGDyK) Iron oxide NPs coated with PEG Internalization potential testing Integrin-specific association between the ligand [68]
c(RGDyK)-iron oxide nanoparticle adducts and
U87MG glioblastoma cells.
αvβ3 c(RGDyK) Micelles PTX Presence of c(RGDyK) enhanced cytotoxicity by 2.5 [69]
folds in U87MG glioblastoma cells. Median survival
time was prolonged in mice bearing intracranial
U87MG tumor xenografts.
αvβ3 c(RGDyK) Micelles PTX and taxol Cellular uptake and, thus, cytotoxicity of c(RGDyK)- [70]
NP/PTX was found to be significantly higher than that
of NPs/PTX due to the integrin protein-mediated
endocytosis effect. Better bioavailability of PTX than
Taxol.
α-3 Peptide OA02 Micelles PTX Dramatically enhanced the uptake efficiency of these [72]
OA02-PEG-CA NPs in SKOV-3 and ES-2 ovarian cancer
cells via receptor-mediated endocytosis, but not in
α-3 integrin-negative K562 leukemia cells.
FR FA Liposome DOX Folate receptor targeted-liposomes loaded with [91]
doxorubicin (FA-L-DOX) inhibited proliferation of
nonfunctioning pituitary adenoma (NFPA) cells
FR FA Copolymeric NPs PTX NPs formulation has great advantages over the [92]
pristine drug, PTX, in achieving better therapeutic
effect in MCF-7 breast cancer cells.
FR FA Surface functionalized QDs – Established folate targeting of QDs in FR expressing [93]
MCF-7 cells.
FR FA Liposome DOX FR mediated endocytosis induced higher cytotoxicity [94]
against the 4T1 mouse mammary carcinoma cell line.
FR FA Liposome DOX Higher cytotoxicity in KB human carcinoma cells; [95]
greater tumor growth inhibition and almost a 50%
increase in life span of mice compared with mice
taking drug in absence of targeting.
FR FA Hybrid polymeric NPs PTX Folate significantly promoted drug-loaded NP's [96]
cellular uptake through folate receptor-mediated
endocytosis in FR overexpressing HeLa and glioma C6
cells as compared to no significant difference in FR-
mediated drug uptake in NIH 3T3 cells with normally
expressed folate receptors.
FR FA Liposome DOX More effective drug uptake in the KB and KB-V [97]
xenograft models, and in the J6456 model irrespec-
tive of administered routes.
TfR Transferrin Iron oxide NPs MRI Retention of Tf-SPIONs in cytoplasm of tumor glioma [116]
cells achieved
TfR Single-chain antibody Liposome p53-gemcitabine The combination treatment prolonged median [118]
fragment (TfRscFv) survival when compared with single drug treatment.
Also decreased tumor burden.
EGFR Peptide ligand (D4: Leu-Ala- Liposome Tested EGFR targeting potential D4 peptide conjugated liposomes efficiently entered [128]
Arg-Leu-Leu-Thr) in EGFR expressing cancer cells (H1299) and specifi-
cally accumulated in EGFR-expressing xenografts
tumor tissues.

(continued on next page)


82 M.J. Akhtar et al. / Clinica Chimica Acta 436 (2014) 78–92

Table 2 (continued)
Specific Ligands/ligands analogues Nanoparticles/nanocarriers Anticancer drugs/agents Major outcome References
receptor(s)

HER2 mAB trastuzumab (TMAB) Human serum albumin (HSA) Methotrexate (MTX) The cytotoxicity of TMAB–MTX–HSA NPs on HER2 [129]
NPs positive cells was found to be significantly higher
than that for non-targeted MTX–HSA NPs and free
MTX.
EGFR Heptapeptide Liposome DOX Peptide-conjugated PLGA-PEG NPs loaded with DOX [131]
showed three-fold higher uptake than by peptide-
free PLGA-PEG NPs in a SKOV3 cell line with high
expression of epidermal growth factor receptor
EGFR Pentapeptide Lipid nanocarriers Tested EGFR targeting potential Higher internalization of pentapeptide ligand [132]
conjugated NPs in high EGFR expressing H1299 and
K562 cells.
FGFR2 Truncated (t) fibroblast Liposome DOX An enhanced antiproliferative activity was achieved [141]
growth factor (tbFGF) by co-delivering DOX and plasmid DNA with impor-
peptide tant a mutant gene simultaneously to the Lewis lung
carcinoma cells (LLC).
FGFR tbFGF Liposome PTX tbFGF-LPs-PTX significantly accumulated in tumor [142]
and prolonged the retention time in tumor-bearing
mice.
FGFR tbFGF Liposome PTX tbFGF, significantly enhanced cytotoxicity of PTX in [143]
FGFR expressing LL/2 cancer cells.
FGFR tbFGF Liposome PTX Higher accumulation of PTX loaded with PEGylated- [144]
tFGF nanoconjugates in tumor tissues expressing
FGFRs.
FGFR1 FGF1 Gold nanoparticles Irradiation FGF1 variant-AuNP conjugates was specifically inter- [145]
nalized only in cells expressing FGFRs. Cell viability
reduced significantly after irradiation with near-
infrared light whereas the proliferation potential of
cells lacking FGFRs is not affected.
S2R SV119 Liposome DOX DOX-loaded SV119 liposomes showed significantly [175]
higher cytotoxicity to DU-145 cells compared to the
DOX-liposomes without SV119.
S2R SV119 Gold nano-cages Tested targeting potential of SV119-ligand might be a promising tool in targeting [176]
SV119 certain tumor.
FSHR FSH33–53 peptide Liposome PTX FSH33–NP–PTX displayed stronger anti-proliferation [181]
and antitumor effects compared with free PTX or
NP-PTX both in vitro and in vivo.
BR Biotin Liposome PTX Results showed that the biotin conjugated NPs could [182]
improve the selective delivery of PTX into cancer cells
with overexpressed biotin receptors.
BR Biotin Taxoid (SB-T-1214) Excellent drug uptake and cytotoxicity in L1210FR [183]
leukemia cells.
BR Biotin Gold NNPs PTX Gold NPs surface-functionalized with PTX and biotin [184]
played a significant role in the diagnosis and therapy
of the cancer cells.
CLR GalNAc residue Liposome Targeting asialoglycoprotein Glycolipid conjugated liposome internalized [194]
receptor. significantly into hepatocytes by ASGPR-mediated
endocytosis.
CLRs Lectin Magnetic NPs (MNPs) PTX Five times higher uptake of PTX-MNPs conjugated [195]
with lectins in K562 leukemia cells as compared with
lectin receptors negative HEK 293 cell line.
NRP-1 CRGDK peptide Gold NPs p-12 (a peptide) CRGDK peptide increased intracellular uptake of gold [198]
NPs carrying therapeutic P12 peptide in NRP-1
receptor overexpressing MDA-MB-321 cells.

domains. Binding of specific agonists (or ligands) to these receptors head/neck cancer, colon cancer, uterine cancer, ovarian cancer, renal
leads to the subsequent activation of heterotrimeric G-proteins (Gs, cell cancers, glioblastomas, neuroblastomas, gastrointestinal carcinoids,
Gi/Go, Gq/G11 and G12/G13), which in turn regulate the activity of intestinal carcinoids, and bronchial carcinoids. Thus, there is a great deal
one or several effectors such as second-messenger-producing enzymes of interest in developing BnR-mediated agents to treat these tumors [17,
or ion channels [15,16]. 18]. The mammalian BnR family consists of three closely related G pro-
Here, we focus only on those GPCRs reported to be overexpressed in tein coupled receptors (GPCRs): the gastrin-releasing peptide receptor
various tumor cells, making them of interest to the field of cancer ther- (GRP receptor), whose native ligand is the 27 amino acid peptide GRP;
apy. A diverse array of ligands have been developed for drug targeting the neuromedin B receptor (NMB receptor), which mediates the action
against GPCRs. Bombesin and octreotide are among the major ligands of the 10 amino acid peptide NMB, and the orphan receptor BRS-3,
for a certain class of GPCRs used in the targeted delivery of anticancer which shares 47–51% amino acid sequence homology with the GRP/
drugs by nanotechnology. NMB receptors but still has an unknown ligand. Each of these receptors
and their ligands are widely distributed in both the central nervous sys-
2.1.1. Bombesin receptors tem and the peripheral tissues. Moreover, studies in animals suggest
Bombesin receptors (BnRs), also known as gastrin-releasing peptide that these receptors are involved in a broad range of physiological and
(GRP) receptors belonging to the GPCR superfamily, have been found to pathophysiological processes [19–22].
be overexpressed in cell lines derived from several human tumor types, Numerous radiolabeled bombesin ligands (Bn) or their analogues
including lung cancer, prostate cancer, breast cancer, pancreatic cancer, are currently undergoing investigation for applications in tumor
M.J. Akhtar et al. / Clinica Chimica Acta 436 (2014) 78–92 83

imaging and radiotherapy. A few non-radiolabeled ligand–drug com- anticancer drug delivery. Liposomes containing the anticancer drug
plexes constructed by the conjugation of Bn with chemotherapeutic DOX with OCT ligand noticeably increased the uptake of DOX in
agents such as camptothecin, DOX, and PTX have successfully increased human hepatocellular carcinoma (SMMC-7721) cells and led to greater
the selectivity and efficacy of these drugs in preclinical studies [23–25]. cytotoxicity than liposomes without targeting ligands [45]. In contrast,
Recently, several studies have used nanotechnology to exploit the spe- no significant difference in cytotoxicity occurred between drugs encap-
cific overexpression of BnR to target anticancer drugs loaded with NPs sulated in liposomes with or without OCT in CHO cells not expressing
conjugated with Bn and/or Bn analogues. Accardo A et al. [26] have suc- SSTRs. Moreover, the study of the ex vivo fluorescence imaging of
cessfully delivered more of the anticancer drug DOX to PC-3 cancer cells BALB/c mouse tissues and the in vivo tissue distribution of B16 tumor-
using drug-containing liposomes conjugated with the BnR ligand bearing mice indicated that OCT caused a remarkable accumulation of
(7–14) peptide fragment called BN(7–14). The same study reported DOX in SSTRs expressing melanoma and pancreatic tumors compared
significant cytotoxicity in PC-3 cells expressing high BnR as well as with the DOX-liposomal formulation without ligand [45]. Using the
greater inhibition of tumor growth in PC-3 xenograft-bearing mice same drug and ligand conjugated with novel sterically stabilized lipo-
compared with liposome–DOX complexes without ligand. Previously, somes (SSLs) composed of polyethylene glycol (OCT-SSL-DOX) led to
radiolabeled liposomes containing the BN(7–14) peptide fragment increased intracellular delivery of DOX in SSTR2-positive cells through
demonstrated that the peptide exposed on the liposome surface main- a mechanism of receptor-mediated endocytosis [46]. Compared to SSL,
tains the ability to selectively target aggregates of GRPR-expressing xe- OCT modification of SSL exhibited little effect on the physicochemical
nografts [27]. Many studies have demonstrated that the BN(7–14) properties of SSL. In summary, OCT-modified SSL might be a promising
fragment modified on its N-terminus with radiometal complexes for system for the targeting of anticancer drugs in the treatment of SSTR2-
diagnostic or therapeutic nuclear medicine applications preserves its af- overexpressing cancers. Another study described efficient DOX
finity for these receptors [28–30]. Yang et al. [31] again proved the abil- targeting and thus significantly higher cytotoxicity in MCF-7 human
ity of BnR to facilitate targeted drug delivery to several tumor cells such breast cancer cells expressing SSTRs compared with normal human
as human prostate cancer cells (DU145 and PC-3), human breast cancer lung fetal (WI-38) cells with no significant SSTR expression. In this
cells (MCF-7), human skin fibroblast cells (HSF), human lung fibroblast study, investigators constructed micelles of N-deoxycholic acid-O, N-
cells (MRC-5), and human prostate stromal cells (PrSC) expressing BnR. hydroxyethyl chitosan (DAHC) with good DOX loading capacity
They have used the mitochondria-disrupting peptide B28 as an antitu- (named DAHC-DOX). The in vivo administration of micelles to nude
mor agent together with the bombesin analogue BN(6–14), which con- mice bearing MCF-7 cancer xenografts confirmed that OPD (OCT–
tains a bombesin receptor-binding motif for the targeting of B28 to PEG–deoxycholic acid)-DAHC micelles possessed a much higher
these tumor cells. B28 conjugated to BN(6–14) was far more cytotoxic tumor-targeting capacity than the DAHC control and exhibited en-
to tumor cells than unconjugated B28. hanced anti-tumor efficacy and decreased systemic toxicity. These re-
sults suggest that OPD-DAHC micelles might be a promising anti-
2.1.2. Somatostatin receptors cancer drug delivery carrier for targeted cancer therapy [47].
Somatostatin receptors (SSTRs), members of the superfamily of G- Therapy with valproic acid and the SSTR2-targeting cytotoxic conju-
protein coupled receptors (GPCRs), are widely expressed in a variety gate CPT–SST led to greater suppression of cervical cancer compared
of tumors and cancer cell lines, including small cell lung cancer [32], with each agent alone. These findings provide a promising clinical op-
neuroendocrine tumors [33], prostate cancer [34], breast cancer [35], portunity for enhanced cancer therapy using combinations of Notch1-
colorectal carcinoma [36], gastric cancer [37] and hepatocellular carci- activating agents (valproic acid) and SSTR2-targeting agents [48]. We
noma [38]. Not surprisingly, therefore, SSTRs have recently gained at- should expect more research in this area.
tention due to the potential for targeting anticancer drugs to cancer
cells overexpressing SSTRs via suitable ligands against these receptors. 2.1.3. Endothelin receptors
Physiologically, these receptors are involved in mediating signaling to The endothelin (ET) family of molecules (acting as ligands) is com-
regulate the basic processes of secretion, cell division, proliferation posed of three polypeptides, ET-1, ET-2 and ET-3, each consisting of 21
and apoptosis. The ligand for these receptors is the neuropeptide hor- amino acids that bind to two highly homologous G-coupled protein re-
mone somatostatin (SST) [39,40]. Five subtypes of SSTRs have been de- ceptors (GCPRs), endothelin receptor A (ETRA) and endothelin receptor
scribed so far, termed SSTR 1–5 [41]. SSTRs, especially SSTR subtype 2, B (ETRB or EDNRB), triggering a variety of signals according to cell type.
are found to be expressed at higher levels in many tumor cells and in tu- The three ETs are potent vasoconstricting peptides involved in the path-
moral blood vessels relative to normal tissues. The binding of somato- ophysiology of various malignancies. ET-1, ET-2 and ET-3 are character-
statin (SST) to endogenous SSTRs is followed by the internalization of ized by a single α-helix and two disulfide bridges, although the three
SST, and several reports have shown that most hormone-secreting tis- peptides are encoded by distinct genes. ETRB binds with all three ETs
sue tumors have a high density of SSTRs [42]. with equal affinity, while ETRA binds ET-1 and ET-2 with a two-fold
Almost a decade ago, Huang C.M. et al. [43] reported the use of SSTRs higher affinity than the ET-3 [49–52]. ETRB has been reported to be
to target anticancer drugs to tumor cells by conjugating the anticancer overexpressed in the vast majority of human melanoma tissue speci-
drug taxol with the SSTR ligand octreotide (OCT). SSTR-mediated mens examined [53,54]. Differential endothelin receptor expression
delivery of OCT-taxol conjugates triggered apoptosis and was exclusive- and function has also been reported in rat myometrial and leiomyoma
ly toxic to SSTR-expressing human breast carcinoma (MCF-7) cells. ELT3 cells [55].
OCT-conjugated taxol was less toxic to cells expressing low levels of An antibody–drug conjugate demonstrated preclinical efficacy, even
SSTR compared with free taxol. These results suggested that OCT- in a model with a low level of ETRB expression [56]. The receptor ETRB is
conjugated taxol demonstrated cell selectivity and might be used as a particularly attractive as a target for antibody/ligand–drug conjugate
targeting agent for cancer therapy. Thereafter, Shen et al. [44] evaluated therapy due to its minimal expression on normal tissue, its cell surface
the anti-tumor effects of the anticancer drug PTX conjugated with the localization and its rapid endocytosis. However, we are not familiar
SSTR ligand OCT in human lung epithelial adenocarcinoma (A549) with any reports exploiting endothelin receptors in drug targeting
cells and human non-small-cell lung cancer (NSCLC) cells xenografted nanocarriers.
into nude mice. The PTX–OCT conjugate, made up of two molecules of
PTX with one molecule of OCT, could enhance tumor growth inhibition 2.2. Integrin receptors
and reduce toxicity when compared with unconjugated PTX. The two
reports mentioned above did not use NPs to carry ligand–drug conju- Among various integrins, αv (or αvβn) integrin receptors are found
gates, but nanotechnology has since been applied to SSTR-mediated to be highly expressed in activated endothelial cells and tumor cells
84 M.J. Akhtar et al. / Clinica Chimica Acta 436 (2014) 78–92

(such as U87MG glioblastoma cells), but not in resting endothelial cells was observed in a subcutaneous U87MG glioblastoma xenograft
or most normal organ systems. Therefore, these integrins represent po- model by both magnetic resonance imaging (MRI) and NIRF imaging.
tential targets for tumor imaging and therapy [57]. Researchers are The investigators achieved excellent tumor integrin targeting efficiency
currently investigating which integrins act as neoplastic markers. Up- and specificity. Another study also used the same ligand–receptor pair
regulation of ανβ3 has been found to be tightly associated with a [i.e., c(RGDyK)–αvβ3] in U87MG glioblastoma cells to deliver the anti-
wide range of cancer types, making it a broad-spectrum tumor marker cancer drug PTX, which was encapsulated in poly(ethylene glycol)-
[58]. Similarly, another integrin, α-3, is also overexpressed in several block poly(lactic acid) micelles, resulting in a nanoscale formulation of
types of cancers, especially in ovarian cancer, breast cancer, and mela- (c(RGDyK)–PEG–PLA–PTX). In vitro cytotoxicity studies proved that
noma [59]. Before discussing the roles of ανβ3- and α-3 integrin recep- the presence of c(RGDyK) led to a 2.5-fold enhancement in the anti-
tors in the targeted delivery of various anticancer drugs to cancer cells glioblastoma cytotoxic efficacy. In an in vivo model, the c(RGDyK)–
and exploring their respective ligands, it is imperative to briefly review PEG–PLA micelle accumulated in the subcutaneous and intracranial
the structure and function of integrins in normal biology. Integrins are tumor tissues, and the PTX-loaded micelle (c(RGDyK)–PEG–PLA–PTX)
transmembrane receptors that bind extracellular matrix proteins or exhibited the strongest tumor growth inhibition among the PTX formu-
other adhesion receptors on neighboring cells. Heterodimeric pairing lations studied here [69]. Other investigators prepared polymeric
of integrin α and β subunits confers specificity of binding to one or micellar-like NPs (MNP) based on PEGylated poly(trimethylene carbon-
more substrates [60]. In particular, the αv subunit pairs with β1, β3, ate) (PEG-PTMC) conjugated with c(RGDyK) for active targeting to
β5, β6, and β8. While some pairings preferentially bind a single ligand integrin-rich U87MG cancer cells. A pharmacokinetic study in rats dem-
(αvβ5 for vitronectin), others recognize a number of ligands (αvβ3 onstrated that the polymeric micellar NPs significantly enhanced the
binds vitronectin, fibronectin, vWF, tenascin, osteopontin, fibrillin, bioavailability of PTX compared with Taxol. In vivo multispectral fluo-
fibrinogen, and thrombospondin). At least 24 distinct integrin heterodi- rescent imaging indicated that c(RGDyK)-MNP/PTX exhibited high
mers are formed by the combination of 18 α subunits and 8 β subunits. specificity and efficiency in active tumor targeting [70].
Because the integrins expressed on the surface of a cell will determine
whether it can adhere to and survive in a particular microenvironment, 2.2.2. Integrin α-3
the matching of integrins and ligands plays a key role in the regulation Similarly, α-3 integrin is overexpressed in several types of cancers,
of the sprouting ability of endothelial cells during angiogenesis, the especially in ovarian cancer, breast cancer, and melanoma [59]. The
recruitment of inflammatory cells to sites in need of repair, and the in- high affinity α-3 integrin-targeting peptide OA02 has been shown to
vasive potential of tumor cells [61,62]. The role of integrins in cell migra- bind strongly to α-3 integrin-overexpressing ovarian cancer cells and
tion and invasion is one of their functions that has received the most specifically target ovarian cancer xenografts (ES-2) in nude mice
study in tumor biology, and this has been reviewed elsewhere [63,64]. when conjugated to near-infrared fluorescence dyes [71]. The overex-
Antagonists to some integrins such as cilengitide have shown promising pression of α-3 integrin on these ovarian cancer cells has been exploited
results in various phases of clinical trials, including for cancer therapy [72] as a promising pharmacologic target for selective drug delivery in
[65]. Our focus is to review the utility of these well studied integrin re- the treatment of these cancers. Conjugation of micellar NPs (PEG5k-
ceptors for targeted anticancer drug delivery in nanomedicine. Integrins CA8 NPs) composed of polyethylene glycol (PEG) block dendritic cholic
expressed by epithelial cells (including α6β4, α6β1, αvβ5, α2β1 and acid (CA) copolymers with the OA02 peptide dramatically enhanced the
α3β1) are generally retained in the tumor but at different levels. uptake and efficiency of these nanocarriers in SKOV-3 and ES-2 ovarian
Integrin expression also varies considerably between normal and cancer cells via receptor-mediated endocytosis, but uptake was not in-
tumor tissues. Most notably, integrins αvβ3, α5β1 and αvβ6 are usually creased in α-3 integrin negative K562 leukemia cells. Furthermore,
expressed at low or undetectable levels in most adult epithelia, but can the in vivo biodistribution study reported that the OA02 peptide greatly
be highly upregulated in some tumors. However, the expression levels facilitated tumor localization and intracellular uptake of PEG5k-CA8 NPs
of some integrins decrease in tumor cells, potentially increasing tumor into ovarian cancer cells as validated in SKOV3-luc tumor-bearing mice
cell dissemination. In fact, re-expression of α2β1 in breast cancer cells [72].
reversed some of the malignant properties of those cells, suggesting
that α2β1 could function as a tumor suppressor. Several integrins that 2.3. Folate receptors
might have important roles in cancer progression have now emerged
with great potential in targeted drug delivery. As expression of the Among cellular surface targets potentially suitable for use in drug
integrins αvβ3, αvβ5, α5β1, α6β4, α4β1 and αvβ6 is correlated with targeting, folate receptors (FRs) stand out as one of the most promising
disease progression in various tumor types, these integrins are the and most investigated epithelial cancer markers. The FR constitutes a
most frequently studied in cancer [66]. useful target for tumor-specific drug delivery primarily because it is
up-regulated in many human carcinomas, including malignancies of
2.2.1. Integrin αvβ3 the ovary, brain, kidney, breast, colon, myeloid cells, and lung, while it
Recent studies showed that the delivery of targeted NPs loaded with is expressed at low levels in normal cells and tissues [73–76]. FRs
DOX to the integrin αvβ3-positive tumor vasculature inhibited the (FRα, FRβ and FRγ) are cysteine-rich cell-surface glycoproteins that
growth of metastases while eliminating the toxicity and weight loss as- bind folate with high affinity to mediate the cellular uptake of folate. Al-
sociated with systemic administration of this drug [67]. This delivery though expressed at very low levels in most tissues, FRs, especially FR α,
method resulted in a 15-fold improvement in tumor and anti- are expressed at high levels in numerous cancers to meet the folate de-
metastatic activity when compared with administration of the free mand of rapidly dividing cells under low folate conditions [77,78]. The
drug. The preferential activity of these NPs on metastases suggests folate dependency of many tumors has been therapeutically and diag-
that growing metastatic tumors may have a greater dependence on an- nostically exploited by the administration of anti-FRα antibodies,
giogenic vessels, making them more susceptible to integrin αvβ3- high-affinity anti-folates [79,80], folate-based imaging agents and
targeted therapy. Chen et al. [68] coated iron oxide NPs with a folate-conjugated drugs and toxins [81–83]. The best studied isoform
PEGylated amphiphilic triblock copolymer and then conjugated the of these receptors is FRα, a cell surface glycosyl phosphatidylinositol-
near-infrared fluorescent (NIRF) dye IRDye800 and a cyclic Arginine- anchored glycoprotein that can internalize bound folates and folate-
Glycine-Aspartic acid (RGD) containing peptide c(RGDyK) for integrin conjugated compounds via receptor-mediated endocytosis [84]. Very
αvβ3 targeting. In vitro binding assays confirmed the integrin-specific recently, its structural basis for the molecular recognition of folic acid
association between the ligand c(RGDyK)-iron oxide nanoparticle ad- was reported [85]. FR α has a globular structure stabilized by eight di-
ducts and U87MG glioblastoma cells. Successful tumor homing in vivo sulfide bonds and contains a deep open folate-binding pocket
M.J. Akhtar et al. / Clinica Chimica Acta 436 (2014) 78–92 85

comprised of residues that are conserved in all receptor subtypes. The found to be greater than that of the DOX-loaded micelles without FA
folate pteroate moiety is buried inside the receptor, whereas its gluta- due to folate-receptor mediated endocytosis in the 4T1 mouse mamma-
mate moiety is solvent-exposed and protrudes out of the pocket en- ry carcinoma cell line. Further, in vitro degradation studies revealed that
trance, allowing for drug conjugation without adversely affecting FR α the H40–PLA-b-MPEG/PEG–FA block copolymer hydrolytically degrad-
binding affinity. The extensive interactions between the receptor and li- ed into polymer fragments within six weeks. Another study tested the
gand readily explain the high folate-binding affinity of folate receptors anticancer efficacy of free DOX and DOX bound with folate targeted li-
and provide a template for the design of more specific drugs targeting posome. DOX-liposome was superior to free DOX in two human
the folate receptor system [85]. Sarcomas, lymphomas, and cancers of tumor models (KB, KB-V) and in one mouse ascitic tumor model (FR-
the pancreas, testicles, bladder, prostate, and liver often do not show expressing J6456). The therapeutic effect was dose-dependent in the
elevated levels of folate receptors. When expressed in normal tissue, fo- KB model receiving DOX-liposome through the i.p. intra-cavitary
late receptors are restricted to the lungs, kidneys, placenta, and choroid route and was schedule-dependent in the J6456 mouse model receiving
plexus; in these tissues, the receptors are limited to the apical surface of DOX-liposome via the intra-cavitary route. In some experiments,
polarized epithelia [86]. Folate or folic acid (FA), the FR ligand is a small however, toxic deaths aggravated by the folate-depleted diet were a
molecule (44 Da) stable over a broad range of temperatures and pH major confounding factor. While promising, this study indicates that
values, inexpensive, and non-immunogenic, and it retains its ability to more sophistication is needed in selecting cancer type associated recep-
bind to the folate receptor after conjugation with drugs or diagnostic tors and ligands [95]. Another in vitro study [96] demonstrated signifi-
markers [87]. The overexpression of folate receptor in cancers of several cant FR-mediated endocytosis of PTX-loaded micellar NPs in FR-
histologies relative to normal tissues, the low cost of folic acid (FA), and overexpressing HeLa and glioma C6 cells, compared with no significant
the vast library of conjugation reactions available make it one of the drug uptake in NIH 3T3 cells with lower folate receptor expression. In
most used ligands for tumor-targeted drug delivery and tumor imaging their study, Riviere et al. [97] investigated the antitumor activity
(reviewed in [88]). Folate is internalized into cells via a low-affinity re- of FA-linked liposome-DOX injected intravenously in mice bearing
duced folate carrier protein or via high-affinity folate receptors. KB tumor xenografts. In conclusion, systemically administered FA-
Due to the elevated FR α expression in cancer cells, FR α has become conjugated DOX-liposomes have demonstrated the potential to en-
one of the most intensively investigated cellular surface antigens for the hance the delivery of anticancer drugs in in vivo models.
targeted delivery of a variety of molecules, including imaging agents,
chemotherapeutic agents, oligodeoxynucleotides, and macromolecules. 2.4. Transferrin receptors
Various types of drug carriers have been conjugated to folate such as
liposomes, lipid NPs, polymeric NPs, polymers, and micelles filled with The efficient cellular uptake of transferrin (Tf) makes this pathway a
the active molecule [89]. Thus, solid tumors that are currently among potential route for the delivery of anticancer drugs, proteins, and thera-
the most difficult to treat by classical therapeutic modalities may be peutic genes primarily into proliferating malignant cells that overex-
readily targeted with FA-linked therapeutics [73,90]. Folate receptor press transferrin receptors (TfRs) [98–100]. Transferrin (Tf) is a single-
targeted-liposomes loaded with DOX (FA-L-DOX) inhibited the prolifer- chain glycoprotein containing approximately 700 amino acids and is
ation of human non-functioning pituitary adenoma (NFPA) cells and one of the most widely used tumor targeting ligands in tumor cells ex-
promoted apoptosis through the activation of caspase 8, caspase 9, pressing transferrin receptors (TfRs) more than other cells [101–103].
and caspase 3/7 more effectively than L-DOX (liposomes without FR TfRs are divided into two subtypes, TfR1 and TfR2. TfR1, also known as
targeted but with DOX). Furthermore, FA-L-DOX also exerted greater CD71, is found to be ubiquitously expressed at low levels in most nor-
anti-invasive ability in NFPA cells than L-DOX through the suppres- mal human tissues, while TfR2, which is homologous to TfR1, is largely
sion of the secretion of matrix metalloproteinase-2 and matrix restricted to hepatocytes [104,113]. Despite its ubiquitous expression,
metalloproteinase-9. Addition of 1 mM free folic acid (to block FR- TfR1 is expressed on malignant cells at levels several fold higher than
mediated endocytosis of liposomes) significantly reduced the those in normal cells, and its expression can be correlated with tumor
pleotropic effects of FA-L-DOX in NFPA cells, suggesting that FRα stage or cancer progression [105–109]. TfR1 is found to be
plays a critical role in mediating the antitumor effect of FA-L-DOX overexpressed in several human carcinomas including breast, ovary,
[91]. Another study using nanotechnology showed that a two- and brain cancers such as glioma and glioblastomas [110–112]. This
component copolymeric NP formulation has great advantages over high expression of the receptor on malignant cells, its ability to internal-
the pristine drug PTX, achieving a better therapeutic effect in MCF-7 ize, and the necessity of iron for cancer cell proliferation make this re-
breast cancer cells. These results further showed that the folate decora- ceptor a widely accessible portal for the delivery of drugs into
tion could promote significantly higher delivery of the drug into the cor- malignant cells, and thus an attractive docking site for targeting antican-
responding cancer cells, enhancing therapeutic effect [92]. Later, the cer drugs in cancer therapy. TfR1, a type-II transmembrane protein, is a
same group prepared folate-decorated quantum dot NPs to improve transmembrane homodimer that can bind up to two molecules of Tf
the imaging effects and reduce their side effects with surface function- through its extracellular carboxyl end binding sites. Transferrin receptor
alized poly(lactide)–vitamin E–D-alpha-tocopheryl polyethylene glycol 2 (TfR2) shares a 45% identity and 66% similarity in its extracellular do-
succinate (PLA-TPGS) copolymer and vitamin E TPGS–carboxyl (TPGS- main with TfR1 [113]. These findings make the transferrin family of re-
COOH) copolymer designed to conjugate folate-NH2. This system pre- ceptors potentially valuable cancer biomarkers as well. Moreover,
sents the advantage of making the targeting effect adjustable. Much transferrin receptor and the receptor for insulin are highly expressed
higher internalization of the folate-decorated QD-loaded PLA-TPGS/ by the endothelial cells forming the blood–brain barrier (BBB), provid-
TPGS-COOH NPs was achieved in MCF-7 cells overexpressing folate re- ing a way for drugs to enter the nervous system [114,115]. As the ex-
ceptors than in mouse NIH 3T3 fibroblast cells with low levels of folate pression of TfR2 is restricted to hepatocytes, TfR1 will be treated as
receptor expression. Compared with free QDs, the QDs conjugated with equivalent to TfR in the remainder of this review for the sake of clarity.
PLA-TPGS/TPGS-COOH NPs showed lower in vitro cytotoxicity in both Jiang et al. [116] reported that transferrin-conjugated super-
MCF-7 and NIH 3T3 cells [93]. Prabaharan et al. [94] encapsulated paramagnetic iron oxide NPs (Tf-SPIONs) acted as promising con-
DOX into folate-conjugated amphiphilic hyperbranched block copoly- trast agents in targeted magnetic resonance imaging (MRI) for the
meric micelles {a hydrophobic poly(L-lactide) inner shell and a hydro- detection of glioma in the brains of rats. MRI showed obvious contrast
philic methoxy poly(ethylene glycol) outer surface}. The DOX-loaded enhancement of brain glioma that could still be clearly observed even
micelles provided an initial burst release (up to 4 h) followed by a 48 h post injection due to the retention of Tf-SPIONs in the cytoplasm
sustained release of the entrapped DOX over a period of approximately of tumor cells, as proven by Prussian blue staining. A second factor
40 h. Cellular uptake of the DOX-loaded micelles linked with FA was supporting TfR as an appropriate target in pancreatic cancer is that the
86 M.J. Akhtar et al. / Clinica Chimica Acta 436 (2014) 78–92

TfR is recycled during internalization in rapidly dividing cancer cells, In an initial study using EGFR to deliver a potential anticancer drug in
thus improving the uptake of Tf-conjugated vectors [117]. Camp et al. nanocarriers conjugated with a novel EGFR ligand called D4 peptide
[118] have delivered human wild type p53 protein (SGT53) (in combi- (D4: Leu-Ala-Arg-Leu-Leu-Thr), liposomes accumulated in EGFR-
nation with anticancer gemcitabine, probably to enhance the anticancer expressing xenograft tumor tissues up to 80 h after intravenous deliv-
activity) bound with liposomal NPs, targeted to the transferrin receptor ery, a marked contrast compared with the control group [128]. Another
by a single-chain antibody fragment (TfRscFv) in an in vivo metastatic study combined the somewhat less frequently used anticancer drug
pancreatic cancer model. Compared with untreated mice harboring methotrexate (MTX) with human serum albumin (HAS) NPs decorated
metastatic tumors with a median survival of 20 days, SGT-53, with trastuzumab (TMAB) molecules to form a nanoconjugate of
gemcitabine and SGT-53–gemcitabine complex increased the median TMAB–MTX–HAS. TMAB–MTX–HAS achieved significantly higher up-
survival to 29, 30 and 37 days, respectively. The tumor targeting take and cytotoxicity in HER2 positive cells than non-targeted MTX–
liposomal-based SGT-53 nanoparticle is capable of sensitizing pancreat- HSA NPs and free MTX [129]. TMAB is a monoclonal antibody working
ic cancer to conventional chemotherapy in pancreatic cancer models. as ligand against HER2 receptors reported to be overexpressed in
This approach has the potential to be translated into a new, more effec- 20–30% of human breast cancers [130]. Other investigators prepared a
tive therapy for pancreatic cancer. Further optimization is ongoing functional liposome of poly(D,L-lactide-co-glycolide)-poly(ethylene gly-
moving towards a Phase 1B/2 clinical trial [118]. col) (PLGA-PEG) with an amino-active group for conjugation with a
Using transferrin-conjugated NPs, however, Salvati et al. [119] have peptide ligand (a heptapeptide). The peptide-conjugated PLGA-PEG
found that proteins in the media can shield transferrin from binding to NPs loaded with DOX showed three-fold higher uptake than peptide-
both its receptors on cells and soluble transferrin receptors. Although free PLGA-PEG NPs in human ovarian carcinoma (SKOV3) cells with
NPs continue to enter cells, the targeting specificity of transferrin is high levels of EGFR expression [131]. Han et al. [132] found that
lost. Their results suggest that when NPs are placed in a complex biolog- conjugation of a pentapeptide (AEYLR or Ala-Glu-Tyr-Leu-Arg) with
ical environment, interaction with other proteins in the medium and nanostructured lipid carriers caused increased cellular uptake of
the formation of a protein corona can ‘screen’ the targeting molecules nanocarriers in tumor cells expressing EGFR. Analyses with flow cytom-
on the surface of NPs and cause a loss of targeting specificity [119]. etry and an internalization assay using human non-small-cell lung car-
cinoma (NCI-H1299) and human leukemia (K562) cells, with high EGFR
2.5. Epidermal growth factor receptors and no EGFR expression, respectively, indicated that FITC-AEYLR had
high EGFR targeting activity. Biotin-AEYLR specifically bound to
Epidermal growth factor receptors (EGFRs) are important targets for human EGFR, demonstrating high affinity for human non-small-cell
anticancer therapy. Special attention has been given to ligands that lung tumors. In a model of multidrug resistance (MTDR), EGFR binding
interact with EGFRs. EGFRs are overexpressed in a wide variety of peptide (EBP) linked DOX was found to accumulate equally in DOX-
human cancers including cancers of the lung, breast, bladder, and resistant (SW480/DOX) and non-resistant (SW480) cell lines of
ovary. EGFR expression has also been found to associate with various human colon cancer, at higher levels than free, unconjugated DOX in
features of advanced disease with poor prognosis [120]. Various EGFR- DOX-resistant cells [133].
targeting vectors and conjugates have been reported as delivery agents
of cytotoxic drugs, toxins, or radionuclides [121]. Moreover, it is an ‘in- 2.6. Fibroblast growth factor receptors
ternalizing’ receptor, indicating that following ligand–EGFR binding, the
ligand–receptor complex is actively endocytosed. Thus, directing Overexpression of fibroblast growth factor receptors (FGFRs) has
nanocarriers by linking them with ligands specific to EGFRs provides a been reported in tumors of the breast, prostate, bladder, and gastric can-
promising way to achieve receptor-mediated intracellular delivery of cer, and has been associated with tumor progression and poor patient
the carrier with its anticancer cargo. The EGFR family consists of four prognosis [134,135]. There are four FGFR genes (FGFR1–FGFR4) that
members: EGFR (ErbB1, HER1), ErbB2 (HER2, neu in rodents), ErbB3 encode receptors consisting of three extracellular immunoglobulin do-
(HER3) and ErbB4 (HER4). These structurally related receptors are mains (D1–D3), a single-pass transmembrane domain and a cytoplas-
single chain transmembrane glycoproteins consisting of an extracellular mic tyrosine kinase (TK) domain [136]. A hallmark of FGFRs is the
ligand-binding ectodomain, a transmembrane domain, a short presence of an acidic, serine-rich sequence in the linker between D1
juxtamembrane section, a tyrosine kinase domain and a tyrosine- and D2, termed the acid box. The D2–D3 fragment of the FGFR
containing C-terminal tail. Binding of soluble ligand to the ectodomain ectodomain is necessary and sufficient for ligand binding and specificity,
of the receptor promotes homo- and hetero-dimer formation between whereas the D1 domain and the acid box are proposed to have roles in
receptors. Receptor dimerization is essential for activation of the intra- receptor autoinhibition [137]. Depending on the localization and cancer
cellular tyrosine kinase (TK) domain and for the phosphorylation of the type, different types of FGFRs are overexpressed. For example, elevated
C-terminal tail [122]. Phosphotyrosine residues then activate, either levels of FGFR1, FGFR2, and FGFR4 are commonly found in cancers of the
directly or through adaptor proteins, downstream components of breast and prostate, whereas only FGFR2 overexpression is observed in
signaling pathways including Ras/MAPK, PLCg1/PKC, PI(3)kinase/Akt, gastric cancers, and papillary thyroid carcinoma is limited to the overex-
and STAT pathways [123]. EGFR and ErbB4 can be thought of as fully pression of FGFR1 and FGFR3 only [138]. Moreover, a low level of FGFR
functional receptors with the ability to bind ligands as well as expression is found on the surface of non-cancerous cells in the vicinity
autophosphorylate C-terminal tails through functional intracellular ty- of the tumor. Due to their specific expression in various cancer types,
rosine kinase domains. ErbB2, however, is unique in that it has no FGFRs represent potential therapeutic targets [139]. Fibroblast growth
known ligand but is the preferred dimerization partner for other factors (FGFs), ligands of FGFRs, exert their physiological roles through
EGFRs [124,125]. ErbB3 is also unique as it has no intrinsic tyrosine ki- FGFRs, regulating developmental pathways such as mesoderm pattern-
nase activity but can transduce signals through interactions with kinase ing in the early embryo. The mammalian FGF family is composed of 18
active receptors, namely EGFR, ErbB2, and ErbB4. Although all ErbB re- ligands that elicit their actions through four (i.e., FGFR1, FGFR2, FGFR3,
ceptors have been localized to the nucleus, ErbB4 is notable for its abil- and FGFR4) highly conserved transmembrane tyrosine kinase receptors
ity to directly transduce extracellular signals to the nucleus through [140].
liberation of the intracellular domain by a ligand-dependent dual prote- Xiao et al. [141] developed a novel cationic liposomal nanocarrier for
ase cleavage of the receptor [126]. In essence, EGFRs bind ligands, DOX which was further conjugated with truncated human basic fibro-
resulting in receptor homo- or hetero-dimerization followed by recep- blast growth factor (tbFGF) peptide, a modified peptide containing
tor internalization (primarily via clathrin-mediated endocytosis) and binding sites for the FGF2 receptor and part of heparin. This peptide
activation of the cytoplasmic tyrosine kinase domain [127]. could effectively bind FGFR2 on the cell surface without stimulating
M.J. Akhtar et al. / Clinica Chimica Acta 436 (2014) 78–92 87

cell proliferation. As a result, conjugation of tbFGF led to an enhanced ligand regulated membrane protein chaperone of 25 kDa possessing
anti-proliferative activity in mouse Lewis lung carcinoma (LLC) cells one putative transmembrane domain and an endoplasmic reticulum re-
by the synergistic actions of DOX and plasmid DNA (encoding the tention signal, is involved in the ER stress response and inter-organelle
phosphorylation-defective mouse survivin threonine 34 → alanine mu- communication [155–157]. S1R is expressed primarily in the cerebral
tant gene known as Msurvivin T34A plasmid) targeted in the same cells cortex, hippocampus, and cerebellar Purkinje cells [158,159], and has
by the same cationic liposome. The concentration of DOX in the co- been proposed as a target for the treatment of central nervous system
delivery system causing 50% cell killing was nearly 3-fold lower than diseases including amnesia, pain, schizophrenia, clinical depression,
the concentration of free DOX responsible for the same cytotoxicity. Alzheimer's, stroke, and addiction [160,148]. S1R is primarily localized
Furthermore, the co-delivery system suppressed tumor growth more to mitochondria-associated ER membranes [161,162], which are the
efficiently than either DOX or the Msurvivin T34A plasmid alone in sites for mitochondrial bioenergetic regulation through the release of
the Lewis lung carcinoma-bearing C57BL/6 mice. The co-delivery sys- ER calcium [163]. S1R activity is modulated by a number of endogenous
tem also caused a 15 day delay of tumor growth, longer than the other molecules such as N,N dimethyltryptamine, progesterone and sphingo-
treatment groups. In conclusion, this novel cationic liposome is an effi- sine, and exogenous molecules including opiates, antipsychotics,
cient vector for the simultaneous delivery of drugs and DNA to the antidepressants, antihistamines, phencyclidine-like compounds, β-
same cells in vitro and in vivo. Another study compared the pharmacoki- adrenergic receptor ligands, and cocaine [164–166]. Upon binding to
netics and tissue distribution of a novel truncated basic fibroblast these molecules, activated S1R causes the dissociation of ankyrin from
growth factor peptide-mediated cationic liposomal PTX (tbFGF-LPs- the inositol triphosphate receptor (IP3R) [167], resulting in calcium re-
PTX) with free PTX and cationic liposomal PTX (LPs-PTX) in tumor- lease at the ER–mitochondrial interface. This released calcium is effi-
bearing mice. The data indicated that the concentration of tbFGF-LPs- ciently taken up by mitochondria to increase energy production and
PTX significantly increased in the tumor and that the retention time other stress responses [168].
was prolonged [142]. Another group prepared biocompatible and bio- Although the natural ligand for the sigma-2 receptor (S2R) and the
degradable nanocarriers of cholesterol-block-poly(ethylene glycol) detailed structure of S2R itself remain unknown, this receptor has
(Chol-PEG2000-COOH) polymer with PTX linked with truncated bFGF been shown to be overexpressed in a variety of human tumors including
fragments (tbFGF). This nanocarrier significantly enhanced the cytotox- breast, pancreas, neuroblastoma, bladder, and lung. As such, various S2R
icity caused by targeted PTX in FGFR expressing LL/2 (a modified Lewis ligands have been extensively studied for their effectiveness in the
lung carcinoma) cells as measured by an MTT {3-(4,5-dimethyl thiazol- treatment of solid tumors due to their preferential uptake in proliferat-
2-yl)-2,5-diphenyl tetrazolium bromide} assay. Flow cytometry re- ing cells [169]. Equally important is the notion that S2R ligands are rap-
vealed that the cellular uptake of rhodamine B, encapsulated in the idly internalized into cancer cells [170,171], including pancreatic cancer
tbFGF conjugated micelles, was increased by 6.6-fold for HepG2 which expresses the receptor at low levels [172]. Synthetic ligands to
(human liver carcinoma), 6.2-fold for A549 (human lung adenocarcino- this receptor could play an important role in cancer diagnosis, imaging,
ma), 2.9-fold for C26 (mouse colon adenocarcinoma) and 2.7-fold for and targeted drug delivery [173].
LL/2 (mouse Lewis lung carcinoma) cells, respectively, compared with SV119, an S2R ligand, binds to pancreatic cancer cells and induces
micelles without tbFGF. The fluorescence spectroscopy further demon- target cell death in vitro and in vivo. When conjugated to SV119, small
strated that the tbFGF-conjugated micelles specifically bound to tumor molecular modulators known to interfere with intracellular pro-
cells over-expressing FGFRs and then released rhodamine B into the cy- survival pathways retained their ability to induce cell death, and their
toplasm [143]. Biodistribution studies in C57BL/6J mice bearing B16 efficiency was enhanced by the targeting effect of SV119. These findings
melanoma revealed a higher accumulation of tFGF targeted PTX deliv- indicate the utility of S2R ligands to increase the tumor-specific delivery
ered by PEGylated liposomes in tumor tissue and organs containing of small molecules [174]. SV119 targeting of DOX-loaded liposomes to
the mononuclear phagocyte system (liver and spleen), but a consider- S2R-overexpressing cells led to significantly higher uptake of SV119-
able decrease in other organs (heart, lung, and kidney) as compared conjugated liposomes in cancer cells of human breast (MCF-7), human
with PEGylated-liposome-PTX without tFGF and free PTX [144]. prostate (PC-3, DU-145) and human lung (A549, 201T) lineages com-
Szlachcic et al. [145] developed novel FGF1 variant-AuNP conjugates pared to normal human bronchial (Beas-2B) cells. Thus, the SV119 li-
found to be specifically internalized only in cells expressing FGFRs. gand caused significantly higher cytotoxicity in DU-145 cells when
FGF1 coated AuNPs significantly reduced viability in cells expressing compared with the cytotoxicity induced by liposomal DOX without li-
FGFRs after irradiation with near-infrared light (down to 40% of control gand [175]. Similarly, gold nano-cages functionalized with SV119
cell viability), whereas the proliferation potential of cells lacking FGFRs again demonstrated the promising ability of this ligand–receptor sys-
was not affected. tem for cancer cell targeting [176]. These results suggest that the
SV119–S2R system might be an effective tool for targeting certain
2.7. Sigma receptors tumors.

Researchers have recently become interested in exploring sigma re- 2.8. Other overexpressed receptors
ceptor ligands for tumor imaging and targeted therapy. Sigma receptors
are overexpressed in a variety of human tumors including cancers of Some less common receptors recently used for targeting drugs to
non-small cell lung carcinoma (NSCLC), prostate, melanoma, and breast cancer cells are follicle-stimulating hormone receptors, biotin receptors,
[146–148]. Sigma receptor was initially proposed to represent a subtype C-type lectin receptors and neuropilin receptors. Overexpression of
of opioid receptors [149]. These subtypes display different tissue distri- follicle-stimulating hormone receptors (FSHRs) during ovarian cancer
butions and distinct physiologic and pharmacologic profiles in both the makes FSHR a possible target against ovarian cancer. Follicle stimulating
central and peripheral nervous systems. Sigma-1 and -2 receptors were hormone (FSH) is a glycoprotein consisting of α and β chains. Some
classically distinguished on the basis of their binding affinity for [3H]- FSHR-binding domains in FSH have been identified including amino
(+)-pentazocine and [3H]-1,3-di(2-tolyl)guanidine ([3H]-DTG). Both acids 1 to 15, 33 to 53, 51 to 65, and 81 to 95 of the FSH β chain [177,
sigma receptors bind pentazocine, while only the sigma-2 receptor 178]. The ovary is the target organ of FSH, which binds to the FSH recep-
binds guanidine-DTG [150–152]. Although the sigma-1 receptor is a tor (FSHR), a G protein-linked receptor. Overexpression of FSHR was
well-characterized protein, the sigma-2 receptor protein has largely found on the ovarian surface epithelium and in some ovarian cancer
remained elusive [153]. However, the novel sigma-2 receptor ligand cell lines [179,180].
SW43 was reported to stabilize the progression of pancreatic cancer in Zhang et al. [181] developed a ligand of FSHR called FSH33–53 that
combination with gemcitabine [154]. The sigma-1 receptor (S1R), a was derived from 33 to 53 amino acids of the FSH β chain. Later,
88 M.J. Akhtar et al. / Clinica Chimica Acta 436 (2014) 78–92

FSH33–53 was conjugated with NPs constructed from PEG-PLA [poly- Ca2+-dependent galactose/GalNAc recognition [193]. In a more recent
ethylene glycol-poly(lactic acid)] forming FSH33–NP complexes. The study, liposomes incorporated with synthetic glycolipids containing a
anticancer drug PTX was chosen to test the targeting potential of terminal GalNAc residue (acting as a ligand for ASGPR) were shown to
FSH33–NPs against ovarian cancer. The FSH33–NP–PTX nanoconjugates be internalized into hepatocytes by ASGPR-mediated endocytosis [194].
displayed stronger anti-proliferation and antitumor effects compared Singh et al. [195] reported five times higher uptake of PTX-loaded mag-
with free PTX or PTX-loaded NPs without FSH33 both in vitro and netic NPs (PTX-MNPs) conjugated with lectins in K562 leukemia cells
in vivo. In summary, this novel FSH33–NP delivery system showed compared to lectin receptor-negative human embryonic kidney (HEK
very high selectivity and efficacy for FSHR-expressing tumor tissues. 293) cells.
Therefore, it has a strong potential to become a new therapeutic ap- Neuropilin receptors (NRPs) were initially considered to be
proach for patients with ovarian cancer. expressed in neuronal cells but were later found to be expressed in
Kim et al. [182] reported that biotin-conjugated poly(ethylene many cell types such as myofibroblasts, endothelial cells and tumors.
oxide)/poly(ε-caprolactone) (PEG/PCL) amphiphilic block NPs carrying The two NRPs (NRP-1 and NRP-2) expressed in vertebrates are trans-
PTX induced higher cytotoxicity in cancer cells than NPs carrying PTX membrane glycoproteins that show 44% homology at the amino acid
but not conjugated with biotin. These results showed that biotin- level. These receptors contain four distinct extracellular domains that
conjugated NPs could improve the delivery of PTX into cancer cells via mediate ligand binding and a short cytoplasmic domain that lacks cata-
interactions with overexpressed biotin receptors. Chen et al. [183] lytic activity [196]. NRP-1 is highly expressed in diverse tumor cell lines
have shown a good correlation between drug uptake and biotin including human neoplasms, and has potential implications in tumor
receptor expression. These investigators have conjugated second- growth and vascularization [196,197]. CRGDK peptide, a ligand of the
generation taxoid (SB-T-1214) as the cytotoxic agent with biotin and NRP-1 receptor, increased the intracellular uptake of gold NPs carrying
reported excellent drug uptake and cytotoxicity in L1210FR leukemia the therapeutic P12 peptide (p12) in MDA-MB-321 human breast can-
cells overexpressing biotin receptors compared with two biotin recep- cer cells overexpressing the NRP-1 receptor [198].
tor negative cell lines, L1210 leukemia cells and WI38 normal human
lung fibroblast cells. Other investigators have shown that gold NP 3. Conclusion
surface-functionalized with PTX and biotin are promising for applica-
tions in cancer diagnosis and therapy [184]. Targeting drugs to cancer cells and the tumor microenvironment is a
C-type lectin receptors (CLRs) have emerged with great potential major challenge in the field of anticancer therapy. This article has
in cell-specific drug and gene delivery [185–187]. CLRs belong to a discussed strategies for achieving the specific delivery of anticancer
large family of receptors that share a structurally homologous drugs to cancer cells and tissues by exploiting specific or overexpressed
carbohydrate-recognition domain and often bind to glycan structures receptors through the use of nanotechnology. Nanotechnology has en-
in a Ca2+-dependent manner [188], including the collectins, selectins, abled many options in manipulating nanocarriers based on proteins,
lymphocyte lectins, and proteoglycans. Because of their endocytic prop- lipids and metals, which can be loaded with anticancer drugs and direct-
erties, CLRs are suitable targets for cell-specific drug delivery [189]. In ed mostly to cancer cells by the attachment of moieties that recognize
addition to cell-specific drug delivery, CLRs may also be exploited to cancer phenotypes. NPs ranging from lipids to metals are under explo-
modulate the functions of CLR-expressing cells such as endocytosis or ration as vehicles for anticancer drug delivery and as moieties guiding
cell activation. In innate immunity, CLRs are mainly expressed by drugs to tumor cells. Lipid-based NPs include polymers, micelles and li-
antigen presenting cells such as dendritic cells and macrophages. They posomes. Note that lipid-based NPs are composed of the same blocks/
serve as pattern-recognition receptors and bind to pathogen-associated monomers, but their main structural differences lie in their three-
molecular patterns (PAMPs) [190], but may also sense self-antigens dimensional structures. Polymeric NPs are single, linear but folded poly-
released by damaged tissue or dead cells [191]. Hepatic lectin receptors mers made of the same or different blocks, whereas dendrimers are
including asialoglycoprotein receptor (ASGPR) are capable of mediating composed of many macromolecules and are thus tree-like and
the endocytosis of bound ligands. Following recognition, bound ligands hyperbranched. Liposomes are well known vesicular structures com-
are internalized via coated pits and the complex is then released into posed of lipid bilayers enclosing an aqueous core, whereas micelles
endosomal compartments while the lectin receptor is recycled to the are composed of lipid monolayers without internal aqueous compart-
cell surface for another round of endocytosis [192]. Thus, hepatic lectin ments. All are colloidal structures with sizes ordered as polymeric NPs
receptors represent promising targets to be exploited in liver-specific ( 10 nm), dendrimers (2–10 nm), micelles (10–100 nm) and liposomes
drug and gene delivery. The human ASGPR, a type II transmembrane pro- (100–200 nm) [199]. Polymeric NPs may be homopolymers (composed
tein, consists of two subunits, H1 and H2, with the H1 subunit mediating of the same monomer) or heteropolymers (composed of more than two

Size and stability of nanoparticle-ligand complex


determining its circulation time?
NP nanoparticle
Nanoparticle’s drug release efficiency at
the desired site?
Anticancer drug molecules
Ligand immunogenicity?

Nanoparticle’s loading efficiency with Receptor ligands


anticancer drug?

Effect of bound ligand on its native affinity


with receptor? Receptor

Nanoparticle’s biocompatibility and


systemic toxicity?

Spatial and temporal variability in receptor overexpression?

Fig. 2. Major issues that need to be addressed in multidisciplinary research involving material sciences and life sciences.
M.J. Akhtar et al. / Clinica Chimica Acta 436 (2014) 78–92 89

different monomers). Liposomal NPs are mainly composed of PEG, [20] Gonzalez N, Moody TW, Igarashi H, et al. Bombesin-related peptides and their re-
ceptors: recent advances in their role in physiology and disease states. Curr Opin
whereas micellar NPs are primarily based on PEG with another block Endocrinol Diabetes Obes 2008;15:58–64.
of cholic acid (CA) [72], poly(lactic acid) [69] and poly(trimethylene [21] Weber HC. Regulation and signaling of human bombesin receptors and their bio-
carbonate) (PEG-PTMC) [70]. Instances of metal-based NPs include logical effects. Curr Opin Endocrinol Diabetes Obes 2009;16:66–71.
[22] Majumdar ID, Weber HC. Biology of mammalian bombesin-like peptides and their
iron oxide [116] and gold NPs [145,176,184,198]. Human serum albu- receptors. Curr Opin Endocrinol Diabetes Obes 2011;18:68–74.
min (HSA) NPs [129] and quantum dots are other nanoscale drug car- [23] Engel JB, Keller G, Schally AV, et al. Effective inhibition of experimental human
riers [93]. Lipid-based NPs have been frequently used in anticancer ovarian cancers with a targeted cytotoxic bombesin analogue AN-215. Clin Cancer
Res 2005;11:2408–15.
therapy as carriers of anticancer drugs, especially in targeted delivery [24] Moody TW, Sun LC, Mantey SA, et al. In vitro and in vivo antitumour effects of cy-
facilitated by overexpressed receptors. Physicochemical factors such as totoxic camptothecin–bombesin conjugates are mediated by specific interaction
the dimensions and stability of NPs, the ease of ligand conjugation, the with cellular bombesin receptors. J Pharmacol Exp Ther 2006;318:1265–72.
[25] Safavy A, Raisch KP, Matusiak D, et al. Single-drug multiligand conjugates: synthe-
drug loading ability, and retention times all are likely to affect the suit-
sis and preliminary cytotoxicity evaluation of a paclitaxel-dipeptide “scorpion”
ability of individual nanocarriers to varying degrees. molecule. Bioconjug Chem 2006;17:565–70.
While nanotechnology is a highly promising technique for targeted [26] Accardo A, Salsano G, Morisco A, et al. Peptide-modified liposomes for selective
drug delivery to cancer cells and the tissue microenvironment, this ap- targeting of bombesin receptors overexpressed by cancer cells: a potential
theranostic agent. Int J Nanomedicine 2012;7:2007–17.
proach will not achieve its potential success unless some technological [27] Accardo A, Mansi R, Morisco A, et al. Peptide modified nanocarriers for selective
and toxicological challenges are addressed (Fig. 2). Several mechanisms targeting of bombesin receptors. Mol Biosyst 2010;6 [878-7].
of triggered drug release can only be applied once the anticancer drug [28] Rogers BE, Bigott HM, McCarthy DW, et al. MicroPET imaging of a gastrin-releasing
peptide receptor-positive tumour in a mouse model of human prostate cancer
reaches and accumulates at the tumor site. Major issues in nanocarrier 64
using a Cu-labeled bombesin analogue. Bioconjug Chem 2003;14:756–63.
design include nanoparticle–drug loading efficiency, the overall stability [29] Smith CJ, Volkert WA, Hoffman TJ. Radiolabeled peptide conjugates for targeting of
of nanoconjugates with attached ligands, optimal receptor–ligand inter- the bombesin receptor superfamily subtypes. Nucl Med Biol 2005;32:733–40.
[30] Parry JJ, Kelly TS, Andrews R, et al. In vitro and in vivo evaluation of 64Cu-labeled
actions, and the duration of expression of the targeted receptor. Nano- DOTA-linker-bombesin(7–14) analogues containing different amino acid linker
particle toxicity and ligand immunogenicity are additional critical moieties. Bioconjug Chem 2007;18:1110–7.
factors that require proper attention in the engineering of clinically ef- [31] Yang H, Cai H, Wan L, et al. Bombesin analogue-mediated delivery preferentially
enhances the cytotoxicity of a mitochondria-disrupting peptide in tumour cells.
fective nanocarriers for the targeting of anticancer drugs. PLoS One 2013;8:e57358.
[32] Herlin G, Kolbeck KG, Menzel PL, et al. Quantitative assessment of 99mTc-
depreotide uptake in patients with nonsmall-cell lung cancer: immunohistochem-
Acknowledgment ical correlations. Acta Radiol 2009;50:902–8.
[33] Sclafani F, Carnaghi C, Di Tommaso L, et al. Detection of somatostatin receptor
subtypes 2 and 5 by somatostatin receptor scintigraphy and immunohistochemis-
This work was supported by the Research Chair of King Saud Univer-
try: clinical implications in the diagnostic and therapeutic management of
sity on Drug Targeting and Treatment of Cancer using Nanoparticles. gastroenteropancreatic neuroendocrine tumours. Tumori 2011;97:620–8.
[34] Mazzucchelli R, Morichetti D, Santinelli A, et al. Immunohistochemical expression
and localization of somatostatin receptor subtypes in androgen ablated prostate
References cancer. Anal Cell Pathol (Amst) 2010;33:27–36.
[35] He Y, Yuan XM, Lei P, et al. The antiproliferative effects of somatostatin receptor
[1] Boyle P, Levin B. World Cancer Report. Lyon, France: International Agency for subtype 2 in breast cancer cells. Acta Pharmacol Sin 2009;30:1053–9.
Research on Cancer Press; 2008. [36] He SW, Shen KQ, He YJ, et al. Regulatory effect and mechanism of gastrin and its
[2] Sutcliffe SB. Cancer control: life and death in an unequal world. Curr Oncol antagonists on colorectal carcinoma. World J Gastroenterol 1999;5:408–16.
2012;19:12–5. [37] Hu C, Yi C, Hao Z, et al. The effect of somatostatin and SSTR3 on proliferation and
[3] Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell apoptosis of gastric cancer cells. Cancer Biol Ther 2004;3:726–30.
2011;144:646–74. [38] Ji XQ, Ruan XJ, Chen H, et al. Somatostatin analogues in advanced hepatocellular
[4] Iyer AK, Greish K, Fang J, et al. Exploiting the enhanced permeability and retention carcinoma: an updated systematic review and meta-analysis of randomized con-
effect for tumour targeting. Drug Discov Today 2006;11:812–8. trolled trials. Med Sci Monit 2011;17:RA169–76.
[5] Maeda H. Tumour-selective delivery of macromolecular drugs via the EPR effect: [39] Guillermet-Guibert J, Lahlou H, Cordelier P, et al. Physiology of somatostatin recep-
background and future prospects. Bioconjug Chem 2010;21:797–802. tors. J Endocrinol Invest 2005;28:5–9.
[6] Lee ES, Bae YHJ. Recent progress in tumour pH targeting nanotechnology. J Control [40] Mariniello B, Finco I, Sartorato P, et al. Somatostatin receptor expression in adreno-
Release 2008;132:164–70. cortical tumours and effect of a new somatostatin analog SOM230 on hormone se-
[7] Wei H, Zhang XZ, Chen WQ, et al. Self-assembled thermo sensitive micelles based cretion in vitro and in ex vivo adrenal cells. J Endocrinol Invest 2011;34:e131–8.
on poly(L-lactide-star block-N-isopropyl acrylamide) for drug delivery. J Biomed [41] Klagge A, Krause K, Schierle K, et al. Somatostatin receptor subtype expression in
Mater Res 2007;83:980–9. human thyroid tumours. Horm Metab Res 2010;42:237–40.
[8] Wei H, Zhang X, Cheng C, et al. Self-assembled, thermosensitive micelles of a star [42] Sun LC, Coy DH. Somatostatin receptor-targeted anti-cancer therapy. Curr Drug
block copolymer based on PMMA and PNIPAAm for controlled drug delivery. Bio- Deliv 2011;8:2–10.
materials 2007;28:99–107. [43] Huang CM, Wu YT, Chen ST. Targeting delivery of paclitaxel into tumour cells via
[9] Saito G, Swanson JA, Lee KD. Drug delivery strategy utilizing conjugation via revers- somatostatin receptor endocytosis. Chem Biol 2000;7:453–61.
ible disulfide linkages: role and site of cellular reducing activities. Adv Drug Deliv [44] Shen H, Hu D, Du J, et al. Paclitaxel–octreotide conjugates in tumour growth inhi-
Rev 2003;55:199–215. bition of A549 human non-small cell lung cancer xenografted into nude mice.
[10] Rapoport NY, Christensen DA, Fain HD, et al. Ultrasound-triggered drug targeting of Eur J Pharmacol 2008;601:23–9.
tumours in vitro and in vivo. Ultrasonics 2004;42:943–50. [45] Sun M, Wang Y, Shen J, et al. Octreotide-modification enhances the delivery and
[11] Han G, You CC, Kim BJ, et al. Light regulated release of DNA and its delivery to nu- targeting of DOX-loaded liposomes to somatostatin receptors expressing tumour
clei by means of photolabile gold nanoparticles. Angew Chem Int Ed Engl in vitro and in vivo. Nanotechnology 2010;21:475101.
2006;45:3165–9. [46] Zhang J, Jin W, Wang X, et al. A novel octreotide modified lipid vesicle improved
[12] Mura S, Nicolas J, Couvreur P. Stimuli-responsive nanocarriers for drug delivery. the anticancer efficacy of DOX in somatostatin receptor 2 positive tumour models.
Nat Mater 2013;12:991–1003. Mol Pharm 2010;7:1159–68.
[13] Schroeder A, Heller DA, Winslow MM, et al. Treating metastatic cancer with nano- [47] Huo M, Zou A, Yao C, et al. Somatostatin receptor-mediated tumour-targeting drug
technology. Nat Rev Cancer 2012;12:39–50. delivery using octreotide-PEG-deoxycholic acid conjugate-modified N-deoxycholic
[14] Zwanziger D, Beck-Sickinger AG. Radiometal targeted tumour diagnosis and thera- acid-O, N-hydroxyethylation chitosan micelles. Biomaterials 2012;33:6393–407.
py with peptide hormones. Curr Pharm Des 2008;14:2385–400. [48] Franko-Tobin LG, Mackey LV, Huang W, et al. Notch1-mediated tumour suppres-
[15] Wettschureck N, Offermanns S. Mammalian G proteins and their cell type specific sion in cervical cancer with the involvement of SST signaling and its application
functions. Physiol Rev 2005;85:1159–204. in enhanced SSTR-targeted therapeutics. Oncologist 2012;17:220–32.
[16] Oldham WM, Hamm HE. How do receptors activate G proteins? Adv Protein Chem [49] Bagnato A, Tecce R, Moretti C, et al. Autocrine actions of endothelin 1 as a growth
2007;74:67–93. factor in human ovarian carcinoma cells. Clin Cancer Res 1995;1:1059–66.
[17] Jensen RT, Battey JF, Spindel ER, et al. International Union of Pharmacology. LXVIII. [50] Levin ER. Endothelins. N Engl J Med 1995;333:356–63.
Mammalian bombesin receptors: nomenclature, distribution, pharmacology, sig- [51] Masaki T. The endothelin family: an overview. J Cardiovasc Pharmacol 2000;35:
naling, and functions in normal and disease states. Pharmacol Rev 2008;60:1–42. S3–5.
[18] Sancho V, Di Florio A, Moody TW, et al. Bombesin receptor mediated imaging and [52] Nelson J, Bagnato A, Battistini B, et al. The endothelin axis: emerging role in cancer.
cytotoxicity: review and current status. Curr Drug Deliv 2011;8:79–134. Nat Rev Cancer 2003;3:110–6.
[19] Fathi Z, Corjay MH, Shapira H, et al. BRS-3: novel bombesin receptor subtype selec- [53] Bittner M, Meltzer P, Chen Y, et al. Molecular classification of cutaneous malignant
tively expressed in testis and lung carcinoma cells. J Biol Chem 1993;268:5979–84. melanoma by gene expression profiling. Nature (London) 2000;406:536–40.
90 M.J. Akhtar et al. / Clinica Chimica Acta 436 (2014) 78–92

[54] Ross DT, Scherf U, Eisen MB, et al. Systematic variation in gene expression patterns [89] Zhao X, Li H, Lee RJ. Targeted drug delivery via folate receptors. Expert Opin Drug
in human cancer cell lines. Nat Genet 2000;24:227–35. Deliv 2008;5:309–19.
[55] Raymond MN, Robin P, De Zen F, et al. Differential endothelin receptor expression [90] Mansoori A, Brandenburg K, Shakeri-Zadeth A. A comparative study of two folate-
and function in rat myometrial cells and leiomyoma ELT3 cells. Endocrinology conjugated gold nanoparticles for cancer nanotechnology applications. Cancer
2009;150:4766–76. 2010;2:1911–28.
[56] Asundi J, Reed C, Arca J, et al. An antibody–drug conjugate targeting the endothelin [91] Liu X, Ma S, Dai C, et al. Antiproliferative, antiinvasive, and proapoptotic activity of
B receptor for the treatment of melanoma. Clin Cancer Res 2011;17:965–75. folate receptor α-targeted liposomal DOX in nonfunctional pituitary adenoma cells.
[57] Chen X, Plasencia C, Hou Y, et al. Synthesis and biological evaluation of dimeric RGD Endocrinology 2013;154:1414–23.
peptide–paclitaxel conjugate as a model for integrin-targeted drug delivery. J Med [92] Pan J, Feng SS. Targeted delivery of paclitaxel using folate-decorated poly(lactide)-
Chem 2005;48:1098–106. vitamin E TPGS nanoparticles. Biomaterials 2008;29:2663–72.
[58] Cai W, Niu G, Chen X. Imaging of integrins as biomarkers for tumour angiogenesis. [93] Pan J, Feng SS. Targeting and imaging cancer cells by folate-decorated, quantum
Curr Pharm Des 2008;14:2943–73. dots (QDs)-loaded nanoparticles of biodegradable polymers. Biomaterials
[59] Mizejewski GJ. Role of integrins in cancer: survey of expression patterns. Proc Soc 2009;30:1176–83.
Exp Biol Med 1999;222:124–38. [94] Prabaharan M, Grailer JJ, Pilla S, et al. Folate-conjugated amphiphilic
[60] Humphries JD, Byron A, Humphries MJ. Integrin ligands at a glance. J Cell Sci hyperbranched block copolymers based on Boltorn(r) H40, poly (L-lactide) and
2006;119:3901–3. poly(ethylene glycol) for tumour targeted drug delivery. Biomaterials 2009;
[61] Pytela R, Pierschbacher MD, Ruoslahti E. Identification and isolation of a 140 kd cell 30:3009–19.
surface glycoprotein with properties expected of a fibronectin receptor. Cell [95] Gabizon A, Tzemach D, Gorin J, et al. Improved therapeutic activity of folate-
1985;40:191–8. targeted liposomal DOX in folate receptor-expressing tumour models. Cancer
[62] Berrier AL, Yamada KM. Cell–matrix adhesion. J Cell Physiol 2007;213 [565-3]. Chemother Pharmacol 2010;66:43–52.
[63] Guo W, Giancotti FG. Integrin signalling during tumour progression. Nat Rev Mol [96] Wang J, Liu W, Tu Q, et al. Folate-decorated hybrid polymeric nanoparticles for
Cell Biol 2004;5:816–26. chemically and physically combined paclitaxel loading and targeted delivery.
[64] Mitra SK, Schlaepfer DD. Integrin-regulated FAK-Src signaling in normal and cancer Biomacromolecules 2011;12:228–34.
cells. Curr Opin Cell Biol 2006;18:516–23. [97] Riviere K, Huang Z, Jerger K, et al. Antitumour effect of folate-targeted liposomal
[65] Desgrosellier JS, Cheresh DA. Integrins in cancer: biological implications and thera- DOX in KB tumour-bearing mice after intravenous administration. J Drug Target
peutic opportunities. Nat Rev Cancer 2010;10:9–22. 2011;19:14–24.
[66] Kren A, Baeriswyl V, Lehembre F, et al. Increased tumour cell dissemination and cel- [98] Singh M. Transferrin as a targeting ligand for liposomes and anticancer drugs. Curr
lular senescence in the absence of β1-integrin function. EMBO J 2007;26:2832–42. Pharm Des 1999;5:443–51.
[67] Murphy EA, Majeti BK, Barnes LA, et al. Nanoparticle-mediated drug delivery [99] Vyas SP, Sihorkar V. Endogenous carriers and ligands in non-immunogenic site-
to tumour vasculature suppresses metastasis. Proc Natl Acad Sci U S A specific drug delivery. Adv Drug Deliv Rev 2000;43:101–64.
2008;105:9343–8. [100] Kircheis R, Wightman L, Kursa M, et al. Tumour targeted gene delivery: an attrac-
[68] Chen K, Xie J, Xu H, et al. Triblock copolymer coated iron oxide nanoparticle conju- tive strategy to use highly active effector molecules in cancer treatment. Gene
gate for tumour integrin targeting. Biomaterials 2009;30:6912–9. Ther 2002;9:731–5.
[69] Zhan C, Gu B, Xie C, et al. Cyclic RGD conjugated poly(ethylene glycol)-co- [101] Inoue T, Cavanaugh PG, Steck PA, et al. Differences in transferrin response and
poly(lactic acid) micelle enhances paclitaxel anti-glioblastoma effect. J Control Re- numbers of transferrin receptors in rat and human mammary carcinoma lines of
lease 2010;143:136–42. different metastatic potentials. J Cell Physiol 1993;156:212–7.
[70] Jiang X, Sha X, Xin H, et al. Self-aggregated PEGylated poly(trimethylene carbon- [102] Qian ZM, Li H, Sun H, et al. Targeted drug delivery via the transferrin receptor-
ate) nanoparticles decorated with c(RGDyK) peptide for targeted paclitaxel deliv- mediated endocytosis pathway. Pharmacol Rev 2002;54:561–87.
ery to integrin-rich tumours. Biomaterials 2011;32:9457–69. [103] Yoon DJ, Kwan BH, Chao FC, et al. Intratumoural therapy of glioblastoma
[71] Aina OH, Marik J, Gandour-Edwards R, et al. Near-infrared optical imaging of ovar- multiforme using genetically engineered transferrin for drug delivery. Cancer Res
ian cancer xenografts with novel alpha 3-integrin binding peptide “OA02”. Mol Im- 2010;70:4520–7.
aging 2005;4 [439-7]. [104] Daniels TR, Bernabeu E, Rodríguez JA, et al. The transferrin receptor and the
[72] Xiao K, Li Y, Lee JS, et al. OA02 peptide facilitates the precise targeting of paclitaxel- targeted delivery of therapeutic agents against cancer. Biochim Biophys Acta
loaded micellar nanoparticles to ovarian cancer in vivo. Cancer Res 2012; 1820;2012:291–317.
72:2100–10. [105] Habeshaw JA, Lister TA, Stansfeld AG, et al. Correlation of transferrin receptor ex-
[73] Cummings J, McArdle CS. Studies on the in vivo disposition of adriamycin in human pression with histological class and outcome in non-Hodgkin lymphoma. Lancet
tumours which exhibit different responses to the drug. Br J Cancer 1986;53:835–8. 1983;1:498–501.
[74] Sudimack J, Lee RJ. Targeted drug delivery via the folate receptor. Adv Drug Deliv [106] Seymour GJ, Walsh MD, Lavin MF, et al. Transferrin receptor expression by human
Rev 2000;41:147–62. bladder transitional cell carcinomas. Urol Res 1987;15:341–4.
[75] Yoo H, Park T. Folate-receptor-targeted delivery of DOX nano-aggregates stabilized [107] Prior R, Reifenberger G, Wechsler W. Transferrin receptor expression in tumours of
by DOX–PEG–folate conjugate. J Control Release 2004;100:247–56. the human nervous system: relation to tumour type, grading and tumour growth
[76] Shia J, Klimstra DS, Nitzkorski JR, et al. Immunohistochemical expression of folate fraction. Virchows Arch A Pathol Anat Histopathol 1990;416:491–6.
receptor alpha in colorectal carcinoma: patterns and biological significance. Hum [108] Kondo K, Noguchi M, Mukai K, et al. Transferrin receptor expression in adenocarci-
Pathol 2008;39:498–505. noma of the lung as a histopathologic indicator of prognosis. Chest 1990;97:
[77] Matsue H, Rothberg KG, Takashima A, et al. Folate receptor allows cells to grow in 1367–71.
low concentrations of 5-methyltetrahydrofolate. Proc Natl Acad Sci U S A [109] Yang DC, Wang F, Elliott RL, et al. Expression of transferrin receptor and ferritin H-
1992;89:6006–9. chain mRNA are associated with clinical and histopathological prognostic indica-
[78] Kelemen LE. The role of folate receptor α in cancer development, progression and tors in breast cancer. Anticancer Res 2001;21:541–9.
treatment: cause, consequence or innocent bystander? Int J Cancer [110] Recht L, Torres CO, Smith TW, et al. Transferrin receptor in normal and neoplastic
2006;119:243–50. brain tissue: implications for brain-tumour immunotherapy. J Neurosurg
[79] McGuire JJ. Anticancer antifolates: current status and future directions. Curr Pharm 1990;72:941–5.
Des 2003;9:2593–613. [111] Chirasani SR, Markovic DS, Synowitz M, et al. Transferrin-receptor-mediated iron
[80] Deng Y, Zhou X, Kugel Desmoulin S, et al. Synthesis and biological activity of a accumulation controls proliferation and glutamate release in glioma cells. J Mol
novel series of 6-substituted thieno 2,3d-pyrimidine antifolate inhibitors of purine Med 2009;87:153–67.
biosynthesis with selectivity for high affinity folate receptors over the reduced fo- [112] Candolfi M, Kroeger KM, Xiong W, et al. Targeted toxins for glioblastoma
late carrier and proton-coupled folate transporter for cellular entry. J Med Chem multiforme: pre-clinical studies and clinical implementation. Anticancer Agents
2009;52:2940–51. Med Chem 2011;11:729–38.
[81] Leamon CP, Reddy JA. Folate-targeted chemotherapy. Adv Drug Deliv Rev [113] Dufès C. Al Robaian M, Somani S, Transferrin and the transferrin receptor for the
2004;56:1127–41. targeted delivery of therapeutic agents to the brain and cancer cells. Ther Deliv
[82] Leamon CP, Reddy JA, Vlahov IR, et al. Preclinical antitumour activity of a novel 2013;4:629–40.
folate-targeted dual drug conjugate. Mol Pharm 2007;4:659–67. [114] Ulbrich K, Hekmatara T, Herbert E, et al. Transferrin- and transferrin receptor-
[83] Reddy JA, Dorton R, Westrick E, et al. Preclinical evaluation of EC145, a folate–vinca antibody-modified nanoparticles enable drug delivery across the blood–brain
alkaloid conjugate. Cancer Res 2007;67:4434–42. barrier (BBB). Eur J Pharm Biopharm 2009;71:251–6.
[84] Paulos CM, Reddy JA, Leamon CP, et al. Ligand binding and kinetics of folate recep- [115] Ulbrich K, Knobloch T, Kreuter J. Targeting the insulin receptor: nanoparticles for
tor recycling in vivo: impact on receptor-mediated drug delivery. Mol Pharmacol drug delivery across the blood–brain barrier (BBB). J Drug Target 2011;19:125–32.
2004;66:1406–14. [116] Jiang W, Xie H, Ghoorah D, et al. Conjugation of functionalized SPIONs with trans-
[85] Chen C, Ke J, Zhou XE, et al. Structural basis for molecular recognition of folic acid ferrin for targeting and imaging brain glial tumours in rat model. PLoS One 2012;7:
by folate receptors. Nature 2013;500:486–9. e37376.
[86] Parker N, Turk MJ, Westrick E, et al. Folate receptor expression in carcinomas and [117] Pirollo KF, Zon G, Rait A, et al. Tumour-targeting nanoimmunoliposome complex
normal tissues determined by a quantitative radioligand binding assay. Anal for short interfering RNA delivery. Hum Gene Ther 2006;17:117–24.
Biochem 2005;338:284–93. [118] Camp ER, Wang C, Little EC, et al. Transferrin receptor targeting nanomedicine de-
[87] Muller C, Schibli R. Folic acid conjugates for nuclear imaging of folate receptor- livering wild-type p53 gene sensitizes pancreatic cancer to gemcitabine therapy.
positive cancer. J Nucl Med 2011;52:1–4. Cancer Gene Ther 2013;20:222–8.
[88] Low PS, Henne WA, Doorneweerd DD. Discovery and development of folic-acid- [119] Salvati A, Pitek AS, Monopoli MP, et al. Transferrin-functionalized nanoparticles
based receptor targeting for imaging and therapy of cancer and inflammatory dis- lose their targeting capabilities when a biomolecule corona adsorbs on the surface.
eases. Acc Chem Res 2008;41:120–9. Nat Nanotechnol 2013;8:137–43.
M.J. Akhtar et al. / Clinica Chimica Acta 436 (2014) 78–92 91

[120] Salomon DS, Brandt R, Ciardiello F, et al. Epidermal growth factor-related peptides [155] Hayashi T, Su TP. Regulating ankyrin dynamics: roles of sigma-1 receptors. Proc
and their receptors in human malignancies. Crit Rev Oncol Hematol 1995;19: Natl Acad Sci U S A 2001;98:491–6.
183–232. [156] Hayashi T, Su TP. Sigma-1 receptor chaperones at the ER–mitochondrion interface
[121] Blessing T, Kursa M, Holzhauser R, et al. Different strategies for formation of regulate Ca(2+) signaling and cell survival. Cell 2007;131:596–610.
PEGylated EGF-conjugated PEI/DNA complexes for targeted gene delivery. [157] Pal A, Fontanilla D, Gopalakrishnan A, et al. The sigma-1 receptor protects against
Bioconjug Chem 2001;12:529–37. cellular oxidative stress and activates antioxidant response elements. Eur J
[122] Linggi B, Carpenter G. ErbB receptors: new insights on mechanisms and biology. Pharmacol 2012;682:12–20.
Trends Cell Biol 2006;16:649–56. [158] Weissman AD, Su TP, Hedreen JC, et al. Sigma receptors in postmortem human
[123] Scaltriti M, Baselga J. The epidermal growth factor receptor pathway: a model for brains. J Pharmacol Exp Ther 1988;247:29–33.
targeted therapy. Clin Cancer Res 2006;12:5268–72. [159] Seth P, Ganapathy ME, Conway SJ, et al. Expression pattern of the type 1 sigma re-
[124] Graus-Porta D, Beerli RR, Daly JM, et al. ErbB-2, the preferred heterodimerization ceptor in the brain and identity of critical anionic amino acid residues in the ligand-
partner of all ErbB receptors, is a mediator of lateral signaling. EMBO J binding domain of the receptor. Biochim Biophys Acta 2001;1540:59–67.
1997;16:1647–55. [160] Guitart X, Codony X, Monroy X. Sigma receptors: biology and therapeutic potential.
[125] Citri A, Skaria KB, Yarden Y. The deaf and the dumb: the biology of ErbB-2 and Psychopharmacology (Berl) 2004;174:301–19.
ErbB-3. Exp Cell Res 2003;284:54–65. [161] Walter L, Hajnoczky G. Mitochondria and endoplasmic reticulum: the lethal
[126] Schlessinger J, Lemmon MA. Nuclear signaling by receptor tyrosine kinase kinases: interorganelle cross-talk. J Bioenerg Biomembr 2005;37:191–206.
the first robin of spring. Cell 2006;127:45–8. [162] Csordás G, Renken C, Várnai P, et al. Structural and functional features and signifi-
[127] Master AM, Gupta AS. EGF receptor-targeted nanocarriers for enhanced cancer cance of the physical linkage between ER and mitochondria. J Cell Biol
treatment. Nanomedicine (Lond) 2012;7:1895–906. 2006;174:915–21.
[128] Song S, Liu D, Peng J, et al. Novel peptide ligand directs liposomes toward EGF-R [163] Csordás G, Várnai P, Golenár T, et al. Imaging interorganelle contacts and local cal-
high-expressing cancer cells in vitro and in vivo. FASEB J 2009;23:1396–404. cium dynamics at the ER–mitochondrial interface. Mol Cell 2010;39:121–32.
[129] Taheri A, Dinarvand R, Atyabi F, et al. Trastuzumab decorated methotrexate-human [164] Su TP, London ED, Jaffe JH. Steroid binding at sigma receptors suggests a link be-
serum albumin conjugated nanoparticles for targeted delivery to HER2 positive tu- tween endocrine, nervous, and immune systems. Science 1988;240:219–21.
mour cells. Eur J Pharm Sci 2012;47:331–40. [165] Ramachandran S, Chu UB, Mavlyutov TA, et al. The sigma1 receptor interacts with
[130] Engel R, Kaklaman V. HER2-positive breast cancer: current and future treatment N-alkyl amines and endogenous sphingolipids. Eur J Pharmacol 2009;609:19–26.
strategies. Drugs 2007;67:1329–41. [166] Fontanilla D, Johannessen M, Hajipour AR, et al. The hallucinogen N, N-
[131] Liu CW, Lin WJ. Polymeric nanoparticles conjugate a novel heptapeptide as an epi- dimethyltryptamine (DMT) is an endogenous sigma-1 receptor regulator. Science
dermal growth factor receptor-active targeting ligand for DOX. Int J Nanomedicine 2009;323:934–7.
2012;7:4749–67. [167] Wu Z, Bowen WD. Role of sigma-1 receptor C-terminal segment in inositol 1,4,5-
[132] Han CY, Yue LL, Tai LY, et al. A novel small peptide as an epidermal growth fac- trisphosphate receptor activation: constitutive enhancement of calcium signaling
tor receptor targeting ligand for nanodelivery in vitro. Int J Nanomedicine in MCF-7 tumour cells. J Biol Chem 2008;283:28198–215.
2013;8:1541–9. [168] Ortega-Roldan JL, Ossa F, Schnell JR. Characterization of the human sigma-1 receptor
[133] Ai S, Jia T, Ai W, et al. Targeted delivery of DOX through conjugation with EGF chaperone domain structure and BiP interactions. J Biol Chem 2013;288:21448–57.
receptor-binding peptide overcomes drug resistance in human colon cancer cells. [169] van Waarde A, Rybczynska AA, Ramakrishnan N, et al. Sigma receptors in oncology:
Br J Pharmacol 2013;168:1719–35. therapeutic and diagnostic applications of sigma ligands. Curr Pharm Des
[134] Knights V, Cook SJ. De-regulated FGF receptors as therapeutic targets in cancer. 2010;16:3519–37.
Pharmacol Ther 2010;125:105–17. [170] Hou C, Tu Z, Mach R, et al. Characterization of a novel iodinated sigma-2 receptor
[135] Wesche J, Haglund K, Haugsten EM. Fibroblast growth factors and their receptors in ligand as a cell proliferation marker. Nucl Med Biol 2006;33:203–9.
cancer. Biochem J 2011;437:199–213. [171] Zeng C, Vangveravong S, Xu J, et al. Subcellular localization of sigma-2 receptors in
[136] Mohammadi M, Olsen SK, Ibrahimi OA. Structural basis for fibroblast growth factor breast cancer cells using two-photon and confocal microscopy. Cancer Res
receptor activation. Cytokine Growth Factor Rev 2005;16:107–37. 2007;67:6708–16.
[137] Wang F, Kan M, Yan G, et al. Alternately spliced NH2-terminal immunoglobulin- [172] Kashiwagi H, McDunn JE, Simon Jr PO, et al. Selective sigma-2 ligands preferentially
like loop I in the ectodomain of the fibroblast growth factor (FGF) receptor 1 bind to pancreatic adenocarcinomas: applications in diagnostic imaging and thera-
lowers affinity for both heparin and FGF-1. J Biol Chem 1995;270:10231–5. py. Mol Cancer 2007;6:48.
[138] Haugsten EM, Wiedlocha A, Olsnes S. Roles of fibroblast growth factor receptors in [173] Crawford KW, Bowen WD. Sigma-2 receptor agonists activate a novel apoptotic
carcinogenesis. Mol Cancer Res 2010;8:1439–52. pathway and potentiate antineoplastic drugs in breast tumour cell lines. Cancer
[139] Heinzle C, Sutterlüty H, Grusch M, et al. Targeting fibroblast-growth-factor-receptor- Res 2002;62:313–22.
dependent signaling for cancer therapy. Expert Opin Ther Targets 2011;15:829–46. [174] Spitzer D, Simon Jr PO, Kashiwagi H, et al. Use of multifunctional sigma-2 receptor
[140] Turner N, Grose R. Fibroblast growth factor signalling: from development to cancer. ligand conjugates to trigger cancer-selective cell death signaling. Cancer Res
Nat Rev Cancer 2010;10:116–29. 2012;72:201–9.
[141] Xiao W, Chen X, Yang L, et al. Co-delivery of DOX and plasmid by a novel FGFR- [175] Zhang Y, Huang Y, Zhang P, et al. Incorporation of a selective sigma-2 receptor li-
mediated cationic liposome. Int J Pharm 2010;393:119–26. gand enhances uptake of liposomes by multiple cancer cells. Int J Nanomedicine
[142] Wang X, Deng L, Chen X, et al. Truncated bFGF-mediated cationic liposomal pacli- 2012;7:4473–85.
taxel for tumour-targeted drug delivery: improved pharmacokinetics and [176] Wang Y, Xu J, Xia X, et al. SV119-gold nanocage conjugates: a new platform for
biodistribution in tumour-bearing mice. J Pharm Sci 2011;100:1196–205. targeting cancer cells via sigma-2 receptors. Nanoscale 2012;4:421–4.
[143] Cai L, Qiu N, Li X, et al. A novel truncated basic fibroblast growth factor fragment- [177] Agris PF, Guenther RH, Sierzputowska-Gracz HE, et al. Solution structure of a syn-
conjugated poly(ethylene glycol)-cholesterol amphiphilic polymeric drug delivery thetic peptide corresponding to a receptor binding region of FSH (hFSH-h
system for targeting to the FGFR-overexpressing tumour cells. Int J Pharm 33–53). J Protein Chem 1992;11:495–7.
2011;408:173–82. [178] Konishi I. Gonadotropins and ovarian carcinogenesis: a new era of basic research
[144] Cai L, Wang X, Wang W, et al. Peptide ligand and PEG-mediated long-circulating li- and its clinical implications. Int J Gynecol Cancer 2006;16:16–22.
posome targeted to FGFR overexpressing tumour in vivo. Int J Nanomedicine [179] Minegishi T, Kameda T, Hirakawa T, et al. Expression of gonadotropin and activin
2012;7:4499–510. receptor messenger ribonucleic acid in human ovarian epithelial neoplasms. Clin
[145] Szlachcic A, Pala K, Zakrzewska M, et al. FGF1-gold nanoparticle conjugates Cancer Res 2000;6:2764–70.
targeting FGFR efficiently decrease cell viability upon NIR irradiation. Int J [180] Wang J, Lin L, Parkash V, et al. Quantitative analysis of follicle-stimulating hormone
Nanomedicine 2012;7:5915–27. receptor in ovarian epithelial tumours: a novel approach to explain the field effect
[146] John CS, Vilner BJ, Geyer BC, et al. Targeting sigma receptor-binding benzamides as of ovarian cancer development in secondary Mullerian systems. Int J Cancer
in vivo diagnostic and therapeutic agents for human prostate tumours. Cancer Res 2003;103:328–34.
1999;59:4578–83. [181] Zhang XY, Chen J, Zheng YF, et al. Follicle-stimulating hormone peptide can facili-
[147] Cobos EJ, Entrena JM, Nieto FR, et al. Pharmacology and therapeutic potential of tate paclitaxel nanoparticles to target ovarian carcinoma in vivo. Cancer Res
sigma(1) receptor ligands. Curr Neuropharmacol 2008;6:344–66. 2009;69:6506–14.
[148] Maurice T, Su TP. The pharmacology of sigma-1 receptors. Pharmacol Ther [182] Kim SY, Cho SH, Lee YM, et al. Biotin-conjugated block copolymeric nanoparticles
2009;124:195–206. as tumour-targeted drug delivery systems. Macromol Res 2007;15:646–55.
[149] Martins CP, Brown-Swigart L, Evan GI. Modeling the therapeutic efficacy of p53 res- [183] Chen S, Zhao X, Chen J, et al. Mechanism-based tumour-targeting drug delivery
toration in tumours. Cell 2006;127:1323–34. system validation of efficient vitamin receptor-mediated endocytosis and drug re-
[150] Su TP. Evidence for sigma opioid receptor: binding of [3H] SKF-10047 to etorphine- lease. Bioconjug Chem 2010;21:979–87.
inaccessible sites in guinea-pig brain. J Pharmacol Exp Ther 1982;223:284–90. [184] Heo DN, Yang DH, Moon HJ, et al. Gold nanoparticles surface-functionalized with
[151] Largent BL, Gundlach AL, Snyder SH. Sigma receptors on NCB-20 hybrid paclitaxel drug and biotin receptor as theranostic agents for cancer therapy. Bioma-
neurotumour cells labeled with (+)[3H]SKF 10,047 and (+)[3H]3-PPP. Eur J terials 2012;33:856–66.
Pharmacol 1986;124:183–7. [185] Lepenies B, Yin J, Seeberger PH. Applications of synthetic carbohydrates to chemical
[152] Walker JM, Bowen WD, Walker FO, et al. Sigma receptors: biology and function. biology. Curr Opin Chem Biol 2010;14:404–11.
Pharmacol Rev 1990;42:355–402. [186] Lepenies B, Seeberger PH. The promise of glycomics, glycan arrays and carbohydrate-
[153] Hornick JR, Spitzer D, Goedegebuure P, et al. Therapeutic targeting of pancreatic based vaccines. Immunopharmacol Immunotoxicol 2010;32:196–207.
cancer utilizing sigma-2 ligands. Surgery 2012;152:S152–6. [187] Lepenies B, Lee J, Sonkaria S. Targeting C-type lectin receptors with multivalent car-
[154] Hornick JR, Xu J, Vangveravong S, et al. The novel sigma-2 receptor ligand SW43 bohydrate ligands. Adv Drug Deliv Rev 2013;65:1271–81.
stabilizes pancreas cancer progression in combination with gemcitabine. Mol Can- [188] Kooyk Y, Rabinovich GA. Protein–glycan interactions in the control of innate and
cer 2010;9:298. adaptive immune responses. Nat Immunol 2008;9:593–601.
92 M.J. Akhtar et al. / Clinica Chimica Acta 436 (2014) 78–92

[189] Tang CK, Sheng KC, Apostolopoulos V, et al. Protein/peptide and DNA vaccine deliv- [195] Singh A, Dilnawaz F, Sahoo SK. Long circulating lectin conjugated paclitaxel loaded
ery by targeting C-type lectin receptors. Expert Rev Vaccines 2008;7:1005–18. magnetic nanoparticles: a new theranostic avenue for leukemia therapy. PLoS One
[190] Sancho D, Reis ESC. Signaling by myeloid C-type lectin receptors in immunity and 2011;6:e26803.
homeostasis. Annu Rev Immunol 2012;30:491–529. [196] Bielenberg DR, Pettaway CA, Takashima S, et al. Neuropilins in neoplasms: expres-
[191] Sancho D, Reis ESC. Sensing of cell death by myeloid C-type lectin receptors. Curr sion, regulation, and function. Exp Cell Res 2006;312:584–93.
Opin Immunol 2013;25:46–52. [197] Sugahara KN, Teesalu T, Karmali PP, et al. Tissue-penetrating delivery of com-
[192] Cummings RD, McEver RP. C-type lectins. In: Varki A, Cummings RD, Esko JD, pounds and nanoparticles into tumours. Cancer Cell 2009;16:510–20.
Freeze HH, Stanley P, Bertozzi CR, Hart GW, Etzler ME, editors. Essentials of [198] Kumar A, Ma H, Zhang X, et al. Gold nanoparticles functionalized with therapeutic
glycobiology. NY: Cold Spring Harbor; 2009. and targeted peptides for cancer treatment. Biomaterials 2012;33:1180–9.
[193] Hashida M, Nishikawa M, Yamashita F, et al. Cell-specific delivery of genes with [199] Blanco E, Kessinger CW, Sumer BD, Gao J. Multifunctional micellar nanomedicine
glycosylated carriers. Adv Drug Deliv Rev 2001;52:187–96. for cancer therapy. Exp Biol Med (Maywood) 2009;234:123–31.
[194] Bernardes GJ, Kikkeri R, Maglinao M, et al. Design, synthesis and biological evalua-
tion of carbohydrate functionalized cyclodextrins and liposomes for hepatocyte-
specific targeting. Org Biomol Chem 2010;8:4987–96.

You might also like