ArticlePDF Available

Receptor-Targeted Surface Engineered Nanomaterials for Breast Cancer Imaging and Theranostic Applications

Authors:
  • National Institute of Pharmaceutical Education and Research (NIPER) Raebareli
  • Saveetha Institute of Medical and Technical Sciences, Chennai - 602105. Tamil Nadu, India

Abstract

Breast cancer is one of the most frequently diagnosed cancers in women and the major cause of worldwide cancer-related deaths among women. Various treatment strategies including conventional chemotherapy, immunotherapy, gene therapy, gene silencing and deliberately engineered nanomaterials for receptor mediated targeted delivery of anticancer drugs, antibodies, and small-molecule inhibitors, etc are being investigated by scientists to combat breast cancer. Smartly designed/fabricated nanomaterials are being explored to target breast cancer through enhanced permeation and retention effect; and also, being conjugated with suitable ligand for receptor-mediated endocytosis to target breast cancer for diagnostic, and theranostic applications. These receptor-targeted nanomedicines have shown efficacy to target specific tumor tissue/cells abstaining the healthy tissues/cells from cytotoxic effect of anticancer drug molecules. In the last few decades, theranostic nanomedicines have gained much attention among other nanoparticle systems due to their unique ability to deliver chemotherapeutic as well as diagnostic agents, simultaneously. Theranostic nanomaterials are emerging as novel paradigm with ability for concurrent delivery of imaging (with contrasting agents), targeting (with biomarkers), and anticancer therapeutics with one delivery system (as cancer theranostics) and can transpire as promising strategy to overcome various hurdles for effective management of breast cancer including its most aggressive form, triple-negative breast cancer.
Receptor-Targeted Surface-Engineered
Nanomaterials for Breast Cancer Imaging
and Theranostic Applications
Javed Ahmad,a,*,† Md. Rizwanullah,b,† Teeja Suthar,c,† Hassan A. Albarqi,a
Mohammad Zaki Ahmad,a Parameswara Rao Vuddanda,d
Mohammad Ahmed Khan,e & Keerti Jainc,*
aDepartment of Pharmaceutics, College of Pharmacy, Najran University, Najran, Saudi Arabia;
bDepartment of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia
Hamdard, New Delhi, India; cDepartment of Pharmaceutics, National Institute of Pharmaceutical
Education and Research (NIPER) – Raebareli, Lucknow, India; dResearch Centre for Topical
Drug Delivery and Toxicology (TDDT), University of Hertfordshire, Hertfordshire, United
Kingdom; eDepartment of Pharmacology, School of Pharmaceutical Education and Research,
Jamia Hamdard, New Delhi, India
*Address all correspondence to: Dr. Keerti Jain, Department of Pharmaceutics, National Institute of Pharmaceutical
Education and Research (NIPER) – Raebareli, Lucknow, India; Tel.: +91-522-2497903; Fax: +91-522-2497905,
E-mail: keertijain02@gmail.com; keertijain.02@niperraebareli.edu.in; or Dr. Javed Ahmad, Department
of Pharmaceutics, College of Pharmacy, Najran University, Najran, Saudi Arabia; Tel.: +966 17542 8744,
E-mail: jahmad18@gmail.com; jaahmed@nu.edu.sa
†These authors contributed equally to this work.
ABSTRACT: Breast cancer is one of the most frequently diagnosed cancers in women and the
major cause of worldwide cancer-related deaths among women. Various treatment strategies includ-
ing conventional chemotherapy, immunotherapy, gene therapy, gene silencing and deliberately en-
gineered nanomaterials for receptor mediated targeted delivery of anticancer drugs, antibodies, and
small-molecule inhibitors, etc are being investigated by scientists to combat breast cancer. Smartly
designed/fabricated nanomaterials are being explored to target breast cancer through enhanced per-
meation and retention effect; and also, being conjugated with suitable ligand for receptor-mediated
endocytosis to target breast cancer for diagnostic, and theranostic applications. These receptor-tar-
geted nanomedicines have shown efcacy to target specic tumor tissue/cells abstaining the healthy
tissues/cells from cytotoxic effect of anticancer drug molecules. In the last few decades, theranostic
nanomedicines have gained much attention among other nanoparticle systems due to their unique
ability to deliver chemotherapeutic as well as diagnostic agents, simultaneously. Theranostic nano-
materials are emerging as novel paradigm with ability for concurrent delivery of imaging (with con-
trasting agents), targeting (with biomarkers), and anticancer therapeutics with one delivery system (as
cancer theranostics) and can transpire as promising strategy to overcome various hurdles for effective
management of breast cancer including its most aggressive form, triple-negative breast cancer.
KEY WORDS: breast cancer, receptor, ligand, active targeting, imaging, nanoparticles
ABBREVIATIONS: BRBP1, brain metastatic breast cancer cell-(231-BR)-binding peptide; DOX, doxorubi-
cin; EGFR, epidermal growth factor receptor; EPR, enhanced permeability and retention; FA, folic acid; FR, fo-
late receptor; FSiNP, uorescent silica nanoparticles; HA, hyaluronic acid; HER2, human epidermal growth fac-
tor receptor 2; ICG, indocyanine green; IONPs, iron oxide nanoparticles; LHRH, luteinizing hormone-releasing
hormone; MDR, multidrug resistance; MMC, mouse mammary carcinoma; MRI, magnetic resonance imaging;
NIR, near-infrared; NPs, nanoparticles; PAMAM, poly(amidoamine); PA , photoacoustic; PT, photothermal;
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 1 Manila Typesetting Company 06/21/2022 09:12AM
0743-4863/22/$35.00 © 2022 by Begell House, Inc. www.begellhouse.com 1
Critical Reviews™ in Therapeutic Drug Carrier Systems, 39(6):144 (2022)
2 Ahmad et al.
PAI , photoacoustic imaging; PAT, photoacoustic tomography; PEG, polyethylene glycol; PLA, polylactic acid;
PLGA, poly (D, L-lactide-co-glycolide); QDs, quantum dots; RGD, arginine-glycine-aspartic acid; scFv, single-
chain fragment variable; SPIONs, superparamagnetic iron oxide nanoparticles; Tf, transferrin; TfR, transfer-
rin receptors; TNBC, triple-negative breast cancer; TPGS, D-α-tocopheryl polyethylene glycol 1000 succinate;
VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor
I. INTRODUCTION
Breast cancer is one of the most common cancers in females, with an annual incidence
of about 2.3 million in 2020. It has been associated with the largest number of cancer-
related deaths in women and caused approximately 685,000 deaths, accounting for 15%
of all cancer-related mortality in females. It has been recently declared as the world’s
most prevalent cancer with a count of 7.8 million cases in the last ve years by the WHO
in March 2020.1 While the incidence rates of breast cancer are on the rise in almost every
region, the rates are relatively higher in the developed world.2 Recurrence is a major
concern in the treatment of breast cancer and patients with metastatic breast cancer show
a mean ve-year survival rate of 27%.3 The prognosis of breast cancer is dependent
upon the extent of expression of three receptors namely human epidermal growth factor
receptor-2 (HER2/Neu/ErbB2), estrogen and progesterone receptors.4 Heterogeneity in
their molecular expression in different solid tumor tissue makes it difcult to diagnose
and treat breast cancer. Triple-negative breast cancers (TNBC) which do not express any
of these targets, i.e., estrogen, progesterone, or HER2 receptors are the most challenging
to treat as these show a poor response to both HER2 targeted and hormonal therapies.5,6
Breast cancer therapy has advanced over time from monotherapy to a combinational ap-
proach. However, the development of multidrug resistance (MDR) has also emerged as
a huge challenge in the treatment of breast cancer. MDR to breast cancer occurs through
various complex mechanisms mediated by cellular and non-cellular pathways.7,8
Various advanced approaches such as nanotechnology, precision medicine, and three-
dimensional (3D) printing have recently emerged as an important area of interdisciplinary
research that has the potential to offer solutions for a variety of unresolved disease issues.9–15
Further, the advent of novel metallic, lipids, and polymeric nanoparticles (NPs), as
well as consequent smart design and development of targeting ligand-based NPs have
shown the utility of nanotechnology in the last two decades in the treatment of cancer
as well as other diseases including infectious diseases, cardiovascular and brain disor-
ders, etc.16–20 The main objective of nanocarrier-mediated anticancer drug delivery are,
to enhance the drug concentration in the cancerous tissue through active and passive
targeting, containment of drug during transit in the body, reducing its distribution in the
normal tissues through improvement in its pharmacokinetic prole, facilitate intrave-
nous administration by improving drug solubility, provide better stability and prevent
degradation, improve bioavailability and patient compliance, and to enhance cellular
uptake/intracellular delivery of anticancer agent.21–25
Nanotechnology-based carriers have been widely explored to reduce the dose of an-
ticancer therapeutics by preventing its off-target accumulation using active and passive
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 2 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 3
targeting approaches. Engineered NPs can also be developed to vitalize the conven-
tional cancer therapeutic modalities.26,27 Multifunctional NPs have the unique strength
that they can carry multiple payloads including therapeutic and diagnostic agents or
their combination (i.e., theranostic) for the treatment as well as to evaluate the progress
of therapy,28 as depicted in Fig. 1. This parallel multifunctional property opens a new
horizon of opportunities in cancer diagnosis and therapy that have been conceptualized
recently.
Breast cancer is one of the major thrust areas for which various nanotechnological
therapeutic approaches are being explored and some of the NPs have shown promising
results and even reached the clinical stage for therapy of resistant breast cancer cases.
For example, several liposomal formulations of doxorubicin (DOX) have been designed
and developed to improve its therapeutic index without altering its efcacy.29 Thus, nan-
otechnology-based drug targeting approaches can be further explored to resolve long-
standing pitfalls in the treatment of breast cancer for a better prognosis of the therapy.
II. OVERVIEW AND SIGNIFICANCE OF DRUG TARGETING IN BREAST
CANCER
Targeted nanocarriers have special features that make them suitable for targeted delivery
of anticancer drugs by facilitating the accumulation of drugs via leaky vasculature in the
tumor. If the nanocarrier is decorated with a targeting ligand, it can enter the tumor cells
by endocytosis in an efcient manner.30 Intracellularly targeted nanocarriers also protect
the drug particles from substrate-specic efux pumps and prevent their expulsion.31
The drug targeting approach is classied into two major categories, i.e., passive and ac-
tive targeting approach (Fig. 2).
FIG. 1: Diagram of cancer-targeted theranostic nanomedicine
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 3 Manila Typesetting Company 06/21/2022 09:12AM
4 Ahmad et al.
Tumor vasculature has a very high number of proliferating endothelial cells and an
aberrant basement membrane. This leads to an increased vascular permeability and the
presentation of open fenestrations and inammatory processes on the vascular endothe-
lium. Further, tumor tissue also develops intermittent hypoxic areas.32,33 The mentioned
abnormal structural alterations provide an opportunity that is exploited by circulating
nanocarrier systems having a size range of 20–200 nm to extravasate and accumulate in
the tumor microenvironment. Further, due to the absence of lymphatic drainage in the
tumor, the accumulated nanocarriers are detained in tumor tissues leading to their pro-
longed retention at the target tissue, i.e., tumor. This phenomenon is called the “enhanced
permeability and retention (EPR) effect.”8,34–36 Thus, the abnormal vascular structure is
crucial for the EPR effect and drug targeting at the tissue level in cancer. The phenom-
enon of the EPR effect has been characterized in almost all types of cancers. The EPR
effect applies to and is utilized by almost all noncarriers and it is regarded as the gold
standard in designing cancer targeted drug delivery systems.37–39 Various factors affect
the EPR effect in solid tumors, including: (i) unique and abnormal tumor vasculature,
(ii) active angiogenesis and high vascular density, (iii) impaired lymphatic clearance,
and (iv) extensive production of vascular mediators that facilitate extravasation includ-
ing bradykinin, nitric oxide, carbon monoxide, heme oxygenase-1, vascular endothelial
growth factor (VEGF), prostaglandins and inammatory cytokines, collagenase (matrix
metalloproteinase), angiotensin-converting enzyme inhibitors, etc.40–43 The factors af-
fecting optimal EPR effect are schematically presented in Fig. 3.
The optimal EPR effect is obtained in the case of carriers that can evade the
immune system and can remain in circulation for a relatively longer duration. It
can lead to many fold increase in accumulation of the anticancer drug in tumors
FIG. 2: Diagram of passive and active targeting to tumor site depicting passive targeting of NPs
via EPR effect where NPs can pass through leaky vasculature of tumor blood vessels and accu-
mulates in tumor tissue, while in active targeting ligand conjugated NPs bind to receptors present
on tumor cells and enter tumor cell via receptor-mediated endocytosis. Figure reproduced from
Navya et al.26 under a Creative Commons license.
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 4 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 5
compared with normal tissues.44 To achieve this fate a noncarrier needs to have three
ideal properties; rstly, its size should be in the range of 10–100 nm. In any case,
the size for extravasation should not exceed 400 nm. If the size is below 10 nm, it
will be ltered out by kidneys and if is above 100 nm, there is a probability of it
being trapped by the liver. Secondly, the carrier should have a neutral or anionic
charge to prevent its elimination by kidneys. Thirdly, it should bypass the trapping,
opsonization, and phagocytosis by the reticuloendothelial system.45 Opsonization
refers to an immune process, which utilizes proteins called opsonins, for tagging the
foreign nanocarriers which can be recognized and eliminated by phagocytes. Major
opsonins include immunoglubulins and complement components (C3, C4, and C5).
Opsonization is often unfavorable in active-targeting approaches as the adhered
opsonins masks the targeting ligands, resulting in a marked reduction in specic-
ity. The effective interaction between the opsonins and nanocarriers depends on the
size, surface charges and composition of the nanocarriers. Generally, hydropho-
bic and charged particles tend to be opsonized more easily in the bloodstream.46,47
Different properties of NPs, including size, surface properties, composition, etc
which affect their in vitro and in vivo performance including targeting ability in the
treatment of cancer are shown in Fig. 4.
The degree of tumor vascularization and angiogenesis may limit the passive target-
ing efciency of NPs and therefore, the extravasation will vary depending upon ana-
tomical location and type of tumor. Sometimes, solid tumors exhibit high uid pressure
in the interstitium which affects the uptake and distribution of nanocarriers.39,48 Thus,
a combination of high interstitial uid pressure and poor lymphatic drainage can be
FIG. 3: Schematic representation of factors affecting EPR effect
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 5 Manila Typesetting Company 06/21/2022 09:12AM
6 Ahmad et al.
correlated as the relation between EPR effect and size, i.e., there is more retention of
larger and long-circulating carriers than the smaller molecules.49,50
Active targeting involves the attachment of a ligand to the nanocarrier which can bind
to the receptors overexpressed by tumor cells or vasculature but not or less expressed
by normal cells of the body.51,52 Such targeted receptors should have a homogenous
distribution in the targeted tumor tissue. These targeting ligands could be non-antibody
peptides, antibody fragments, monoclonal antibodies, or endogenous ligands such as
folate, estrone, etc. Tumor penetration is affected by the binding afnity of the targeting
ligand to the binding site and higher afnity binding is observed in tumor vasculature
due to easy accessibility to the cells owing to the dynamic environment of systemic
circulation.37,53–55 The understanding of the difference in tumor microenvironment com-
pared with that of normal tissue has been utilized by researchers in designing better an-
ticancer therapies. Here, the differential expression of targeted receptors in normal and
cancerous cells has particular signicance in designing tumor-targeted nanocarriers for
delivery of anticancer drugs in breast cancer.56–58 The mechanism of receptor-mediated
targeting is depicted in Fig. 5.
III. RECEPTOR-MEDIATED TARGETING IN BREAST CANCER IMAGING
Imaging agents are crucial in the management of cancer attributed to their critical role
in screening and diagnosis of cancer which helps in therapy monitoring and planning
FIG. 4: Tunable physicochemical characteristics of cancer-targeted nanomedicine inuencing in
vitro/in vivo performance of loaded therapeutics/imaging agents to advance the therapy outcome
in breast cancer
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 6 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 7
of current and future treatment strategies.59,60 In breast cancer, the degree of hormone-
receptor expression is directly related to the benecial effects offered by hormonal ther-
apy. For example, in a patient with overexpression of HER2 receptors, there are more
binding sites available for the monoclonal antibody (trastuzumab) to work.61,62 Although
immune-histochemistry has been the conventional method used to determine the degree
of hormone-receptor expression, its use in quantifying receptor expression is not wide-
spread.63 In addition, an attempt to detect different types of receptor proteins at once on
a single tumor specimen requires several steps.61,64 Receptor-targeted nanomedicines, on
the other hand, can quantify and prole several biomarkers more accurately and sensi-
tively, thus offering clear advantages over immune-histochemistry.65,66
Nanomedicines are being engineered as imaging and contrast agents for breast im-
aging techniques including magnetic resonance imaging (MRI),67–70 uorescence imag-
ing,71–74 ultrasound imaging,75–78 and photoacoustic tomography (PAT).79–82 In addition,
FIG. 5: Schematic representation of the mechanism of receptor-mediated targeted delivery
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 7 Manila Typesetting Company 06/21/2022 09:12AM
8 Ahmad et al.
nanoparticulate systems that can serve as multimodal imaging contrast agents are also
being developed and investigated.83–85 Various receptors which are overexpressed in
breast cancer are presented diagrammatically in Fig. 6.
A. MRI
MRI, a radiological technique, is widely used to analyze tissues. In the case of MRI,
rstly, hydrogen atoms present in tissues are excited by the application of the magnetic
eld of appropriate resonance frequency, then these excited hydrogen atoms return to
their equilibrium state while emitting a radiofrequency signal which is used to form the
image. The contrast difference among different tissues is attributed to the different rates
of relaxation of hydrogen atoms of different tissues. MRI offers high spatial resolution
without any ionizing radiation. Superparamagnetic iron oxide nanoparticles (SPIONs)
are the majorly used MRI contrast agents among various paramagnetic and superpara-
magnetic metals.86–89
Various nanocarriers apart from SPIONs has also been designed and investigated
by scientists for targeted imaging of breast tumors. Luteinizing hormone-releasing hor-
mone (LHRH) templated ferrosoferric oxide (Fe3O4) NPs has been investigated as MRI
agent for targeted diagnosis and treatment of breast tumors. The designed LHRH-Fe3O4
NPs showed promising results as a contrast agent to be used in the T2 eld as observed
in MRI experiments performed in female Balb/c mice.79 G3 decorated ankyrin repeat
protein and uorescein-5-maleimide multifunctional SPIONs (G3-5MF-SPIONs) have
been designed by Li and coworkers for MRI of breast tumors. The G3-5MF-SPIONs
showed selective binding (even in the presence of trastuzumab), long retention time,
signicant accumulation, and biodistribution in SK-BR-3 tumor-bearing female Balb/c
FIG. 6: Diagrammatic illustration of commonly overexpressed receptors in breast cancer. HER2,
human epidermal growth factor receptor 2; EGFR, epidermal growth factor receptor (HER1);
ER, estrogen receptor; VEGFR, vascular endothelial growth factor receptor; FR, folate receptor.
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 8 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 9
nude mice. Additionally, G3-5MF-SPIONs showed the selectivity to image HER2-
positive breast tumors with remarkable T2 signal reduction after 24 h. These ndings
supported the promising credential of targeted nanomedicine in imaging breast cancer
by MRI.90
Ding et al. developed anti-HER2 single-chain antibody-conjugated iron oxide
nanoparticles (IONPs) as MRI agents. The NPs showed higher afnity toward HER2
overexpressing cells (NCI-N87), compared with HER2 lower expressing cells (SUIT2)
in in vitro cell lines studies. Further, these results were supported by in vivo studies with
NCI-N87 and SUIT2 tumor-bearing mice where a signicantly higher reduction in MRI
signals was observed with NCI-N87 tumors (19.3%) than SUIT2 tumors (6.2%) attrib-
uted to overexpression of HER2 receptors on NCI-N87 cells.91 In another study, Lee et
al. observed that hyaluronic acid (HA)-modied manganese ferrite (MnFe2O4) nanocrys-
tals enhanced the detection ability of MRI in CD44 expressing breast carcinoma cells
(MDA-MB-23 and MCF-7).92 Lim et al. also observed that hyaluronan-modied mag-
netic nanoclusters benet in MRI technique for the detection of CD44-overexpressed
breast cancer cells in MCF-7 and MDA-MB-231 cell lines as well as in MDA-MB-231
bearing female Balb/c nude mice. It was found that the developed nanoparticulate sys-
tem exhibited superior ability for targeted diagnosis of CD44-overexpressed breast can-
cer with excellent sensitivity for MRI.68
Meier et al. developed folic acid (FA) conjugated ultra-small SPIONs which
showed specic retention in imaging in various breast carcinoma cell lines including
MDA-MB-468, ZR-75-1, MDA-MB-435, MCF-7, SK-BR-3, and MDA-MB-231 as
well as in vivo imaging in folate receptors (FR) overexpressing cancer xenograft mice
model. The retention of FR-specic SPIONs led to a signicant negative enhancement
of FR expressed breast tumors on delayed magnetic resonance images at 24 hours post
injection.67 Similarly, Meng et al. found that LHRH-conjugated SPIONs exhibited much
greater efciency than that of unconjugated SPIONs by MRI technique in the detection
of LHRH overexpressed human breast cancer (Hs 578T) as well as breast cancer xeno-
grafts model in addition to metastases in the lungs of athymic nude mice.93 From the re-
search discussed above, it is clear that nanoconjugates systems investigated by scientists
for MRI of breast cancer have shown promising results and are encouraging enough for
scientists to further explore possible clinical applications of these nanomedicine-based
MRI contrast agents for detection of breast cancer to achieve better prognosis in cancer
management.
B. Fluorescence Imaging
Fluorescence spectroscopy is an important technique being used for years for imag-
ing in various elds related to biological and medical sciences such as biochemistry,
biotechnology, molecular biology, pharmaceutical technology, nanotechnology, and
nanomedicine, etc. Fluorescence spectroscopy can provide molecular specicity as
many biomolecules such as proteins, lipids, amino acids, drug molecules such as DOX
can inherently uoresce on excitation with UV-visible light and hence can be used as
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 9 Manila Typesetting Company 06/21/2022 09:12AM
10 Ahmad et al.
uorescent probes to label target molecules in uorescent imaging.94,95 In this spectros-
copy, the image is formed by photons emitted either from biomolecules having inherent
uorescence ability or from external uorescent active probes.96,97 It offers an inexpen-
sive imaging technique with suitable spatial resolution and is similarly sensitive to radio-
isotopes generally used in single-photon emission computed tomography (SPECT) and
positron emission tomography (PET).98–100 Limited tissue penetration with signicantly
high noise, interference from water molecules, light absorption by proteins, tissue scat-
tering, and autouorescence are the few limitations of uorescence microscopy. Some
of these challenges such as tissue scattering and autouorescence have been reduced to
some extent with tissue penetration to many centimeters by use of near-infrared (NIR)
light during in vivo imaging.101–103
Gold nanoclusters encapsulating protein are being explored for bioimaging and
biosensing applications in cancer due to their biocompatibility, relatively safe prole,
and signicant uorescing ability. Scientists have designed mannose conjugated bovine
serum albumin (BSA) encapsulated gold (Man-BSA-Au) nanoclusters for detection of
concanavalin A and imaging of breast tumor cells using uorescence imaging technique
(Fig. 7A). Man-BSA-Au nanoclusters showed bright red uorescence after incubation
with mannose receptor-expressing MDA-MB-231 cells for 1 h attributed to targeting
of nanoclusters to cancer cells mediated by mannose receptors, whereas cells incubated
with phosphate buffer and BSA encapsulated gold (BSA-Au) nanoclusters showed no
noticeable uorescence (Fig. 7B).73
Scientists have fabricated platinum nanocluster bio-nanoprobes with red emission
using polyamidoamine (PAMAM) dendrimer combined with Protein A (adapter), which
exhibited small size, low cytotoxicity and exceptional photostability for uorescence
imaging of HER2 in SK-BR-3 cells. These bio-nanoprobes exhibit minimal nonspecic
binding that allows highly sensitive optical imaging with visualization of deep anatomy
and increase in tissue penetration due to long-wavelength emission.74 Yamaguchi et al.
observed that technetium-99m (99mTc) and indocyanine green (ICG) labeled anti-HER2
antibody decorated PAMAM-coated silica NPs showed higher imaging efciency
with stronger NIR uorescent signals for HER2 positive SK-BR-3 cells than that in
HER2 negative MDA-MB-231 cells. The in vivo imaging results with SK-BR-3 and
MDA-MB-231 xenograft tumor mice model were also found in accordance with the in
vitro results.104
Quantum dots (QDs) have been exploited by scientists for spectrum analysis and
immunouorescence detection of epidermal growth factor receptors (EGFR). The out-
comes of this study showed the importance of EGFR in the prognosis of lymph node-
positive and HER2-positive invasive breast cancer via immunouorescent detection.63
Li et al. developed FA-functionalized two-photon absorbing 1,2-Distearoyl-sn-glycero-
3-phosphoethanolamine (DSPE)- polyethylene glycol (PEG) NPs with aggregation-
induced emission for specic targeting and imaging in MCF-7 cancer cells using a
two-photon uorescence microscope.105 Wu et al. described the targeted imaging potential
of arginine-glycine-aspartic acid (RGD) peptide-doped uorescent silica NPs (FSiNPs)
in athymic nude mice bearing the MDA-MB-231 tumor on intravenous injection. It
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 10 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 11
exhibited a high and specic αvβ3 integrin expression level in the MDA-MB-231 tumor
due to the specic targeting potential of RGD peptide-doped FSiNPs and tumor uores-
cence reached maximum intensity after 1 h of injection.72
FIG. 7: (A) Schematic presentation of preparation of Man-BSA-Au nanoclusters for uores-
cence imaging of human breast cancer cells. (B) Confocal uorescence images of MDA-MB-231
cells after incubation with (a) PBS, (b) BSA-Au nanoclusters, and (c) Man-BSA-Au nanoclusters
for 1 h (excitation wavelength: 488 nm; scale bar: 20 μm) (reprinted from Sha et al.73 with per-
mission from Elsevier, copyright 2020).
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 11 Manila Typesetting Company 06/21/2022 09:12AM
12 Ahmad et al.
Different nanomaterials including lipidic NPs, polymeric NPs, metallic NPs, QDs,
and dendrimers, etc have been engineered smartly by researchers for uorescent imag-
ing of breast cancer with promising results in preclinical studies in animals.
C. Ultrasound Imaging
Ultrasound imaging is widely used as a tomography technique for the clinical diagno-
sis of breast cancer. It can show minute internal details of organs and tissues attributed
to the use of sound waves of a shorter wavelength having frequencies of 2 MHz or
greater. Transmitting and receiving transducers are set to capture ultrasound wave in-
teractions.106 Ultrasound imaging is commonly used to diagnose various diseases via
imaging different internal organs, muscles, and tendons, and gives information related
to the structure, size, or presence of any abrasion, injury, or lesions in these areas. High
resolution with low cost, non-invasive procedure, and precise targetability of ultrasound
energy deposition are some of the advantages of ultrasound imaging.86,107
Xu et al. developed dual-targeted gold nanoshell poly (D,L-lactide-co-glycolide)
(PLGA) nanocapsules decorated with vascular endothelial growth factor receptor
(VEGFR)-2 and p53 antibodies for specic ultrasound molecular imaging of breast
cancer, which showed excellent biocompatibility. The results of in vivo studies in
the MCF-7 orthotopic mice model indicated efcient binding to cells overexpressing
VEGFR2 or p53 protein with no signs of organ damage.77 Polylactic acid (PLA) NPs
have been designed and evaluated for comparison of targeted ultrasound imaging of
HER2-overexpressing and HER2-negative breast cancer cells by scientists. In results,
HER2-overexpressing cells showed more staining after incubation with NPs/antibody
conjugates, whereas minimal staining was seen in HER2-negative cells, indicating di-
rect receptor binding of targeted NPs. The average gray matter of HER2-positive cells
was signicantly higher in NP-treated cells compared with the control shown in high-
resolution ultrasound images.76
In another study, Sakamoto et al. carried out the molecular analysis of breast cancer
using herceptin conjugated IONPs-based ultrasound imaging agents. The ultrasound im-
aging results supported the ability of herceptin decorated IONPs to differentiate between
HER2/neu positive SK-BR-3 cells as compared with the non-tagged NPs.75 These stud-
ies validated that conjugated NPs may have promising applications in receptor-targeted
non-invasive imaging of breast cancer.
D. Photoacoustic Imaging/Tomography
Photoacoustic imaging (PAI) or photoacoustic tomography (PAT) is an emerging non-
ionizing imaging tool combining high optical contrast and high ultrasound resolution.108
PAT makes use of pulsed laser light to generate light-induced acoustic signals.109 PAT
with nite element reconstruction has been used to image small nanometer-sized par-
ticles containing objects with high resolution.110 Photoacoustic provides in vivo morpho-
logical and functional information regarding the tumors within the surrounding tissue.
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 12 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 13
Photoacoustic (PA) with the use of targeted contrast agents is also capable of performing
in vivo molecular imaging, thus enabling further molecular and cellular characterization
of cancer.59,111,112
In vivo PAI studies using VEGF targeted, anti-VEGFR antibody-conjugated iron
platinum alloy NPs showed that with these NPs the PAI depth could be extended to more
than 5 cm in chicken breast tissues with the use of a laser energy density within the safe
limit. Further, it was observed that NPs were removed from the site of the tumor and
metabolized mainly by the liver supporting the safe use of this nanomedicine in early
detection and theragnosis of breast cancer with whole-body PAI.82 Balasundaram et al.
demonstrated that folate decorated NPs showed ~ 4-fold enhancement in the PA signal
in the MCF-7 breast cancer xenograft model compared with untargeted NPs, thus pro-
viding a better delineation from the surrounding tissue.113 Furthermore, Kanazaki et al.
developed a single-chain fragment variable (scFv) anti-HER2 moiety conjugated IONPs
for PAI studies. Their results demonstrated that anti-HER2 scFv-conjugated IONPs
showed high afnity and specic binding to HER2-expressing N87 tumor-bearing fe-
male Balb/c-nude mice and SUIT2 cells compared with untargeted NPs.114
Scientists have investigated breast cancer imaging techniques by combining high-
resolution NIR induced PAT using NIR dye-labeled with amino-terminal fragments of
urokinase plasminogen activator receptor-targeted magnetic IONPs in orthotopic mouse
mammary tumor model (mouse bearing mammary carcinoma cell line 4T1). Their re-
sults showed 4–10 fold amplication in PA signals in the tumor with receptor-targeted
NPs as compared with non-targeted ones.115 Wang et al., 2014 designed FR-targeted
ICG-loaded PLGA (ICG-PLGA) lipid NPs. It exhibited high targeting efciency for FR
and remarkable optical absorption in NIR wavelengths resulting in excellent PA signals
in in vitro as well as in vivo studies in MCF-7 bearing female Balb/c nude mice over
non-targeted ICG-PLGA lipid NPs.11 6 Furthermore, a strategically designed study using
HA decorated gold NPs for PAI showed that PA/photothermal (PT) signals from targeted
NPs in MDA-MB-231 cancer cells were 10- to 50-fold higher than untargeted NPs, and
the rate of ashing PA signals signicantly increased in Balb/c nude mice model.79
The use of receptor-targeted nanomedicines such as antibody-conjugated NPs, fo-
late, HA conjugated nanomaterials, etc have shown that the use of these ligand conju-
gated nanomedicines targeted toward a particular receptor overexpressed on tumor cells
can promisingly improve in vivo imaging efciency of PAI along with may provide
further details regarding the cellular and molecular characterization of tumor.
E. Multimodal Imaging
The intrinsic merits and demerits of each imaging technique are signicant and hardly
overcome by the improvement of imaging instrumentation alone. Multimodal imaging
includes the combined usage of various imaging tools such as MRI/uorescence imag-
ing and it has gathered signicant attention to advance the currently used techniques for
diagnosis of breast cancer. It is a powerful technique that provides more consistent and
accurate recognition of disease sites.117,118
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 13 Manila Typesetting Company 06/21/2022 09:12AM
14 Ahmad et al.
Du et al., 2020 developed brain metastatic breast cancer cell-231-BR-binding pep-
tide (BRBP1)-functionalized ultra-small IONPs containing DiR uorescent dye for tar-
geted MR/NIR uorescent dual-modal detection of breast cancer brain metastasis. Their
results showed that the NPs exhibited a size of 10 nm with core-shell structure, high
relaxivity values, and efcient photon emission in vitro. Moreover, the NPs presented a
T2 contrast imaging effect and NIR uorescence signal enhancement. The MR/NIR u-
orescence signal of BRBP1-modied NPs in tumor tissue was signicantly enhanced as
compared with the control. The results showed that BRBP1-IONPs-mPEG NPs loaded
with DiR can be employed as an attractive targeting strategy for the detection of breast
cancer brain metastasis.85 Similarly, Han et al. developed CD44 monoclonal antibodies
coupled magnetic-uorescent iron oxide carbon hybrid nanomaterials which demon-
strated strong excitation wavelength-dependent uorescence in the blue-red region with
a quantum yield of 58.4%, and they displayed higher stability and T2 relaxivity than
simple IONPs. Moreover, there was signicant preferential uptake of the conjugates by
4T1 breast cancer cells as seen in biological transmission electron microscope imaging.
In addition, the developed CD44-hybrid nanomaterials were able to distinguish 4T1
cells from normal cells by virtue of its high binding afnity and specicity of the CD44
antibodies to the CD44 receptors on the surface of cancer cells.119
Herceptin-decorated paclitaxel-loaded ultrasmall superparamagnetic iron ox-
ide nanobubbles exhibited signicantly enhanced cytotoxic effects against HER2-
overexpressing SK-BR-3 breast carcinoma cells and signicantly lower cytotoxicity
against HER2-negative MDA-MB-231 breast carcinoma cells. In addition, the devel-
oped magnetic nanobubbles showed ability to enhance ultrasound, magnetic resonance,
and photoacoustics trimodal imaging.120 Hyaluronan-modied SPIONs were designed
for multimodal imaging of breast cancer which showed signicantly enhanced spe-
cic cellular uptake of HA-SPIONs in HA receptor-overexpressing MDA-MB-231
cells which was conrmed by uorescence imaging. In addition, HA-SPIONs demon-
strated signicant negative contrast enhancement on T2-weighted MR images of HA
receptor-overexpressing MDA-MB-231 breast tumor-bearing balb/c nude mice com-
pared with untargeted SPIONs.121 Kievit et al. developed herceptin decorated uores-
cent dye loaded multifunctional SPIONs that exhibited a 4-fold improvement in cellular
uptake and specicity to target the neu/HER2-overexpressing mouse mammary carci-
noma cells. It showed signicant contrast enhancement in magnetic resonance images
of breast tumors.83
These studies demonstrated that nanomedicine may advance the diagnosis of vari-
ous breast cancers by utilizing receptor-mediated targeting of imaging agents. The po-
tential of various receptor-targeted nanomedicines explored by different researchers/
investigators for the detection or imaging of breast cancer are summarized in Table 1.
IV. RECEPTOR-MEDIATED THERANOSTICS IN BREAST CANCER TREATMENT
NPs can be designed with some inherent properties to offer unique imaging and sur-
face functionalization ability. Also, NPs have optimum size, high surface area to volume
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 14 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 15
TABLE 1: Receptor-targeted nanomedicines for detection/imaging of breast cancer
Imaging
technique
Types of
nanomedicine
Ligand Receptor targeted Breast cancer
model
Outcome Ref.
Magnetic
resonance
imaging
Fe3O4 NPs LHRH LHRH Human MCF-7
breast cancer cells
LHRH-Fe3O4 effectively accumulated in
the tumor region due to magnetic targeting.
It showed a good negative enhancement
effect that signicantly reduced the
intensity of T2 weighted images, allowing
it to be used as a contrast agent
70
Fe3O4-Au NPs Thiolated
AS1411
aptamer
Urokinase-type
plasminogen
activator (uPA)
4T1, mouse
mammary
carcinoma, and
HFFF-PI6, human
foreskin broblast
cell lines
–Developed nanoprobe produced strongly
darkened T2-weighted MR images with
4T1 cells, whereas brightened images with
HFFF-PI6 cells
–Specic targeting of 4T1 cells
overexpressing nucleolin on the cell surface
–Nanoprobe binds specically to breast
cancer cells and has very low accumulation
in normal cells.
69
MnFe2O4
nanocrystals
HA HA receptor In vitro/
MDA-MB-231 and
MCF-7 cells
Excellent MR imaging sensitivity to
diagnose CD44-overexpressing cancer cells
92
Magnetic
nanoclusters
Hyaluronan HA receptor In vitro/
MDA-MB-231
and MCF-7 breast
carcinoma cells
In vivo/Balb/c-Slc
nude mice bearing
MDA-MB-231
cancer cells
Nanoclusters showed excellent ability for
targeted detection of CD44-positive breast
carcinoma cells by showing excellent
sensitivity as MR imaging agents
122
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 15 Manila Typesetting Company 06/21/2022 09:12AM
16 Ahmad et al.
TABLE 1: (continued)
Imaging
technique
Types of
nanomedicine
Ligand
Receptor targeted
Breast cancer
model
Outcome
Ref.
SPIONs FA FA receptor In vitro/
MDA-MB-231
In vivo/breast
cancer xenograft
in female athymic
Harlan
rats
–Specic retention in FR-positive
MDA-MB-231 cell line and in an FR-
positive human breast carcinoma xenograft
model in vivo
–Signicant negative enhancement of
FR-positive breast tumors on delayed MR
images at 24 hours post injection
67
SPIONs LHRH LHRH receptor In vitro- Hs 578T
cells
In vivo-breast
cancer xenografts
and lung
metastases of
athymic nude mice
–LHRH conjugated SPIONs exhibited
signicantly higher uptake in the cells than
that of the unconjugated SPIONs in both in
vitro and in vivo studies
–The enhanced uptake of intracellularly
accumulated LHRH conjugated SPIONs
provided T2 contrast enhancement leading
to improved spatial resolution in MRI by
classical T2 imaging
93
Fluorescence
imaging
Red-emitting
Platinum
nanoclusters
Protein A and
anti-HER2
antibody
HER2 SK-BR-3 breast
cancer cells
Platinum bio-nanoprobe binds to HER2-
overexpressing SK-BR-3 cells with a
higher afnity and selectivity
74
Mannose
functionalized
BSA Gold
Nanoclusters
Mannose
Concanavalin A on
Mannose receptors
Human
breast cancer
MDA-MB-231
cells,
HUVEC cells
Increased sensitivity and selectivity
of Man-BSA-Au nanoclusters for
determination of Concanavalin A on
mannose receptor
73
99mTc-
radiolabeled
nanosilica system
Trastuzumab HER2 Female Balb/c
nude mice
SK-BR-3 breast
cancer cells
–Radiolabeled system exhibited an
enhanced active targeting to the tumor,
compared with EPR-based passive
diffusion
–Signicantly enhanced accumulation of
SiNPs within the HER2 overexpressing
SK-BR-3 cells
123
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 16 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 17
TABLE 1: (continued)
DSPE-PEG NPs FA FA receptor
In vitro
/MCF-7
breast cancer cells
Two-photon uorescence images obtained
from the uorescence signal from
cytoplasm after incubation with developed
NPs clearly distinguished the MCF-7 breast
cancer cells from others
105
FSiNPs RGD peptide Integrin receptor In vitro/
MDA-MB-231 cell
Ex vivo/athymic
nude mice bearing
the MDA-MB-231
tumors
RGD peptide-conjugated FSiNPs were
able to image the specic αvβ3 integrin
expression level in the MDA-MB-231 cells
due to its special receptor-based targeting
effects
72
Ultrasound
imaging
Gold-Nanoshells
PLGA
Magnetic Hybrid
NPs
Anti-HER2
antibodies
HER2 Human breast
cancer SK-BR-3
cells
Conjugation with anti-HER2 antibodies led
to specic and sensitive binding with HER2
overexpressing SK-BR-3 breast cancer
cells.
Excellent contrast enhancement in vitro
78
Gold nano-
shelled PLGA
nanocapsules
Anti-VEGFR2
and anti-p53
antibodies
(DNCs)
Vascular endothelial
growth factor
receptor type 2
HUVECs, 4T1,
MCF-7, and
MDA-MB-231
breast cancer cells
MCF-7 cell bearing
Female Balb/c
nude mice
–DNCs exhibited high target specicities in
vitro in VEGFR2- and p53-overexpressing
cells compared with control cells
–Ultrasound imaging studies using novel
nano-sized, dual-targeted PLGA-Au
nanocapsules ultrasound contrast agents
allowed highly precise and reliable
detection of breast cancer in MCF-7
orthotopic mice model
77
Anti-EGFR
antibody-
conjugated gold
nanorods
Anti-HER2
antibody
EGFR EGFR-negative
MCF-7 cells
EGFR-positive
MDA-MB-231
cells
Mouse model of
human TNBC
Ultrasound-guided PAI using anti-
EGFR decorated gold nanorods showed
a very high sensitivity for the selective
visualization of EGFR-positive primary
breast tumors as well as Lymph Nodes
micrometastases
124
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 17 Manila Typesetting Company 06/21/2022 09:12AM
18 Ahmad et al.
TABLE 1: (continued)
Imaging
technique
Types of
nanomedicine
Ligand
Receptor targeted
Breast cancer
model
Outcome
Ref.
PLA-NPs Anti-HER2
antibody
HER2 receptor In vitro/SK-BR-3
and MDA-MB-231
cell line
In high-resolution ultrasound B-mode
images, the average gray scale of the
HER2-positive cells (SK-BR-3 cell) was
consistently and signicantly higher
76
IONPs Herceptin HER2 receptor In vitro/SK-BR-3
human breast
cancer cells
Ultrasound imaging exhibited excellent
sensitivity and specicity in the detection
of HER2 overexpressed breast cancer cells
75
Photoacoustic
(PA) imaging
Iron-platinum NPs anti-VEGFR VEGF 4T1 breast cancer
cells
Accumulation in the tumor region,
Targeted delivery of NPs by EPR effect
82
Irradiated
nanodiamonds
Anti-HER2
peptide
HER2
4T1.2 breast cancer
cells
Female Balb/c
mice
–PA images demonstrated that
nanodiamonds signicantly accumulated in
breast tumors and traced the entire tumor in
less than 10 hours
–HER2 conjugation signicantly enhanced
the imaging of HER2-positive tumors
–The conjugation of PEGylated
nanodiamonds with anti-HER2 peptide
led to enhanced internalization of INDs
by HER2 positive tumor cells (4T1.2-neu)
and longer residence time in the region of
HER2 positive tumor
81
ICG conjugated
PLGA lipid NPs
FA FR In vitro/MCF-7
cells
In vivo/Female
Balb/c nude mice
bearing human
breast cancer
(MCF-7) cells
Developed NPs showed excellent optical
properties with reduced toxicity, enhanced
tumor-targeting capability, prolonged
circulation time, and signicantly improved
PA contrast for preclinical imaging
applications
116
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 18 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 19
TABLE 1: (continued)
Gold nanorods Bombesin
(BBN)
Gastrin releasing
peptide (GRP)
receptor
Breast tumor-
bearing Balb/c
mice
T47D cells
In vitro studies showed a very selective
binding of PEG-BBN gold nanorods
towards breast cancer cells.
Biodistribution studies showed that after
the injection of PEG-BBN gold nanorods
conjugate (high uptake) and PEG gold
nanorods conjugate (low uptake) approved
the targeting ability of PEG-BBN gold
nanorods conjugate to breast cancer cells.
PEG-BBN gold nanorods is a reliable
vector for targeting GRP receptors over-
expressed in various cancers
125
IONPs NIR830 Urokinase
plasminogen
activator receptor
In vivo/Balb/c mice
bearing Mouse
mammary tumor
4T1 cells
In vivo studies in mice showed 4- and 10-
fold enhancement in PA signals of IONPs
compared with non-targeted IONP or
control Balb/c mice
115
Gold carbon
nanotubes
CD44
receptor-
specic
antibody
CD44 receptor
In vitro
/
MDA-MB-231
cells
In vivo/nude mice
–PA/PT signals from targeted
MDA-MB-231 cells were 10 to 50-fold
higher than those from unbound nanotubes
distributed randomly in suspension between
cells at relatively high laser uences of
300–500 mJ/cm2
–After subsequent injections of CD44-
decorated gold carbon nanotubes, the rate
of ashing PA signals increased to 46.7 ±
6.8 cells/min in nude mice
79
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 19 Manila Typesetting Company 06/21/2022 09:12AM
20 Ahmad et al.
TABLE 1: (continued)
Imaging
technique
Types of
nanomedicine
Ligand
Receptor targeted
Breast cancer
model
Outcome
Ref.
Multimodal
imaging
Ultra-small IONPs BRBP1
peptide
EGFR receptor Mice bearing 231-
BR xenografts
–Ultra-small IONPs offered a T2 contrast
imaging effect and enhanced NIR
uorescent signal
–The MR/NIR uorescence imaging signal
of BRBP1-modied NPs in tumor tissue
was signicantly enhanced as compared
with control NPs, which shows the
targeting ability of BRBP1 peptide
85
Natural magnetic
NPs with Gold
nanorods
Folate FA receptor In vitro
MDA-MB-231 cell
line
Mouse model
injected with breast
cancer cells
–Magnetotactic bacteria, natural magnetic
NPs, and natural magnetic NPs-gold
nanorods can be used for scanning as well
as in PA and PT imaging
–Signicant enhancement in PA signals
from natural magnetic NPs and natural
magnetic NPs-gold nanorods both in vitro
and in vivo
–Natural magnetic NPs-gold nanorods were
able to provide for real-time counting of
labeled cells, magnetic trapping of labeled
cells (e.g., CTCs), and killing of diseased
circulating cells
84
SPIONs Herceptin HER2 receptor In vitro/
MDA-MB-231
cells
In vivo/transgenic
breast cancer mice
–SPIONs showed up to 4-fold improved
cellular uptake and specic ability to
target neu/HER2 overexpressing mouse
mammary carcinoma (MMC) cells in vitro
and in vivo
–Signicant enhancement in the contrast
of SPIONs MR images in primary breast
tumors
83
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 20 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 21
ratio, higher loading capacity for contrast agents, targeting ligands, and therapeutic mol-
ecules.126 Furthermore, many NPs have inherent imaging properties, which can further be
functionalized to become nanotheranostics. Theranostics refers to the integrated applica-
tions of diagnostic/ imaging agents and therapeutic moiety in one system. Theranostic
nanomedicines or nanotheranostics are multifunctional nanosystems designed for pre-
cise and personalized disease management.127 They consist of macromolecular materials/
polymers in which imaging and therapeutic agents can be absorbed, adsorbed, encapsu-
lated, entrapped, or conjugated for simultaneous imaging and therapy at the cellular and
molecular level.128 Ideal theranostic nanomedicines should have the following properties:
(i) safe for administration in humans, (ii) shows rapid and selective accumulation at the
target site, (iii) recognizes biochemical and morphologic characteristics of a disease, (iv)
able to deliver sufcient drug on-demand without damaging healthy organs, and (v) rap-
idly cleared from the body within hours or be biodegraded into nontoxic byproducts.129
Generally, theranostic nanomedicines can be fabricated in several ways: (i) thera-
peutic agents (e.g., anticancer drugs and photosensitizers) may be adsorbed, conjugated,
entrapped, or loaded, to existing NPs having intrinsic imaging abilities such as QDs,
IONPs, and gold nanocages; (ii) attaching the imaging agents, such as uorescent dyes,
optical or magnetic NPs, and various radioisotopes, to existing therapeutic NPs; (iii)
encapsulating both diagnosing and therapeutic agents together in different nanocarriers
such as polymeric NPs, porous silica NPs and ferritin nanocages; and (iv) some unique
NPs (e.g., porphysomes, 64Cu-CuS, and gold nanoshells or cages) with dual inherent im-
aging and therapeutic properties can also be engineered for theranostic applications.130,131
The surface of these nanocarriers can be modied with PEG and various target-
ing ligands to prolong the blood circulation half-life and to provide the active tumor-
targeting capability. Targeting ligands can be exploited to identify and selectively bind
to cell surface receptors overexpressed on tumor cells or surfaces. Various targeting
ligands including, antibodies, small peptides, aptamers, lectins, some proteins, or pro-
tein fragments can be used depending on the physicochemical properties of theranostic
nanocarrier or tumor of interest. In the last few decades, substantial efforts have been
made by researchers to design and evaluate different receptor-targeted nanomedicines
for simultaneous diagnosis and treatment of breast cancer with certain promising results;
however, some challenges have also been faced by scientists while designing targeted
theranostic nanomedicines. The following subsection of the article discusses different
receptor-targeted nanomedicines for theranostic applications in the breast cancer treat-
ment particularly in the invasive variants such as TNBC and deals with the challenges
encountered while designing and developing these theranostic nanomedicines.
A. HER2 Targeted Theranostics
HER2 receptors are overexpressed on breast cancer and have been explored by scientists
for targeted theranostic applications. Mechanistic approaches for exploring HER2 recep-
tors for targeted breast cancer theranostics are shown diagrammatically in Fig. 8. Recently,
Khaniabadi et al. investigated the theranostic potential of superparamagnetic Fe3O4 NPs
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 21 Manila Typesetting Company 06/21/2022 09:12AM
22 Ahmad et al.
decorated with protoporphyrin and trastuzumab for imaging and treatment of EGFR2-
overexpressing breast cancer. The results showed no signicant cytotoxicity after incubat-
ing the MCF-7 breast cancer cells under various Fe concentrations of NPs and theranostic
agents, whereas in vitro photothermal ablation of targeted NPs showed a reduction of 74%
in MCF-7 cells after 10 min at the highest Fe concentration.132 In another study, Choi et
al. developed IONPs and DOX-loaded multifunctional nanocarriers with an ability of in-
tegrated cancer-targeting via a HER2 monoclonal antibody for controlled delivery of an-
tineoplastic drug (DOX) as well as imaging agent (IONPs) for MRI and NIR uorescence
imaging. The results of in vitro studies demonstrated up to 5-fold higher cellular uptake and
signicantly higher cytotoxicity to HER2 overexpressing SK-BR-3 cells than HER2 nega-
tive MCF-7 cells, signifying HER2 receptor-based cancer targeting. In in vivo tumor xe-
nograft model, antibody-targeted nanocarriers exhibited signicantly higher uptake in the
cancerous cells and the size of the tumor signicantly reduced than the non-targeted ones.133
Parhi and Sahoo developed trastuzumab-conjugated rapamycin, and QDs-loaded
theranostic NPs which exhibited signicantly higher uptake in SK-BR-3 cells and
MDA-MB-231 cells compared with untargeted NPs. Targeted NPs showed ~ 3 and ~
1.5-fold higher toxicity to SK-BR-3 cells and MDA-MB-231 cells, respectively, com-
pared with untargeted NPs. The cellular uptake studies were performed using couma-
rin-6 dye and the results of cell uptake studies showed that confocal images indicated
a higher uorescence intensity in tumor spheroids treated with targeted NPs than that
treated with untargeted NPs (Fig. 9). Further, the therapeutic benets of targeted NPs
were explored at the molecular level and the results showed augmented downregulation
of mTOR signaling pathway thereby inducing further cell death.134
Kievit et al. developed multifunctional SPIONs for diagnosis and treatment of meta-
static breast cancer and evaluated in a FVB/N transgenic mouse model. They fabricated
uorescent dye-loaded NPs using a copolymer of chitosan and PEG for optical detection
FIG. 8: Diagrammatic representation of theranostic applications achieved with HER2 recep-
tor targeting, where QD: Quantum dots; Tmab-QD-rapa-NPs: Trastuzumab-conjugated rapamy-
cin and QDs-loaded theranostic NPs (reprinted from Parhi and Sahoo134 with permission from
Elsevier, copyright 2015)
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 22 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 23
and labeled them with trastuzumab against the HER2 receptor. The results of cellular
uptake studies showed that trastuzumab-doped NPs exhibited a 4-fold higher cellular
uptake and could specically target HER2 overexpressing mouse breast cancer cells.
Also, the dye-coated NPs provided signicant enhancement in contrast of MR images
of primary breast tumors that were detected with MRI. Moreover, these NPs were able
FIG. 9: Qualitative intracellular uptake of coumarin-6, coumarin-6-NPs, and Tmab-coumarin-
6-NPs (50 ng/mL) in SKBR 3 cells and MDA-MB-231 cells by confocal microscopy after 2
hrs of incubation (reprinted from Parhi and Sahoo134 with permission from Elsevier, copyright
2015)
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 23 Manila Typesetting Company 06/21/2022 09:12AM
24 Ahmad et al.
to detect micrometastases by recognizing and tagging spontaneous micrometastases in
the lungs, livers, and bone marrow of these mice.83
In another study, Day et al. developed NIR resonant anti-HER2 antibody conjugated
gold-gold sulde NPs as theranostic agents for management of breast cancer via mul-
tiphoton microscopy and higher intensity photoablation. The study demonstrated that
anti-HER2 antibody-doped NPs could efciently bind to SK-BR-3 breast cancer cells.
Antibody conjugated NPs emitted luminescence upon excitation with a pulsed laser
resulted from a two-photon absorption process. At 1 mW laser power, Anti-HER2 anti-
body conjugated gold-gold sulde NPs could specically visualize SK-BR-3 cells and
cell death was induced upon increasing laser power to 50 mW followed by membrane
blebbing.135 Similarly, Yang et al. developed anti-HER2 antibody conjugated DOX-
loaded magnetic PLGA NPs for imaging and treatment of breast cancer. The in vitro
studies of anti-HER2 antibody conjugated DOX NPs using HER2/neu-overexpressing
broblast NIH3T6.7 cells and HER2-negative SK-BR-3 and MDA-MB-231 cell lines
demonstrated 87.4 times higher uorescence intensity than that of other cells. The mul-
timodal NPs demonstrated remarkable cancer cell afnity. Also, the NPs followed a
sustained release of loaded anticancer drugs for 3 weeks.136
HER2 receptors and HER2 targeting antibody, trastuzumab have been investigated
for targeted theranostic applications in cancer and have shown efcacy to be explored as
promising theranostic nanomedicines for treatment and imaging of breast cancer.
B. Folate-Targeted Theranostics
FR are highly overexpressed on different types of cancer cells including breast cancer
and have been exploited by scientists for targeted delivery of antineoplastic drugs, con-
trast agents as well as theranostic applications in cancer. Soleymani et al. fabricated fo-
late-targeted IONPs which showed a signicant reduction in the T2-weighted MR signal
intensity of breast tumors indicating the accumulation and retention of NPs in the tumor
tissue. Moreover, a signicant reduction in tumor progression was seen in the mice.137
In another study, Alibolandi et al. developed folate-decorated QDs and DOX-loaded
theranostic polymersomes for the imaging and treatment of breast cancer. The results
of uorescence microscopic and cytotoxicity studies showed that the folate-conjugated
DOX-QD NPs displayed signicantly higher cellular uptake and cytotoxicity in 4T1
and MCF-7 cells compared with untargeted NPs and pure drug solution. Fluorescence
imaging studies in Balb/c mice bearing 4T1 breast adenocarcinoma demonstrated that
the targeted NPs showed signicant accumulation at the tumor site after 6 h post intra-
venous injection. Also, acute toxicity studies showed no evidence of long-term harmful
histopathological and physiological changes in the treated animals. Moreover, the tar-
geted NPs demonstrated much better therapeutic efcacy in vivo compared with untar-
geted NPs and free drugs.138
FA and cisplatin prodrug conjugated gold nanoclusters showed that FA-conjugation
led to a signicant increase in the cellular uptake and cytotoxicity of cisplatin gold
nanoclusters in murine 4T1 breast cancer cells. Fluorescence imaging studies in 4T1
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 24 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 25
tumor-bearing nude mouse showed that FA-cisplatin gold nanoclusters selectively ac-
cumulated in the 4T1 tumor cells and produced intense uorescence signals due to the
tumor-targeting effect of FA. In addition, FA-cisplatin gold nanoclusters showed sig-
nicant inhibition in the growth and lung metastasis of the orthotopically implanted
4T1 breast tumors.139 In another study, Heidari Majd et al. developed tamoxifen-loaded
FA-armed PEGylated MnFe2O4 NPs for imaging and treatment of the FR-positive breast
cancer cells. Fluorescence imaging and ow cytometry analyses revealed substan-
tial interaction of developed NPs with FR-overexpressing MCF-7 breast cancer cells.
Cytotoxicity studies demonstrated signicant inhibition in the growth of MCF-7 cells
treated with developed NPs.140 Muthu et al. also developed FR-targeted D-α-tocopheryl
polyethylene glycol 1000 succinate (TPGS) coated theranostic liposomes containing
docetaxel and QDs for diagnosis and therapy of breast cancer. Confocal laser micros-
copy study showed that folate-targeted liposomes exhibited a signicantly higher red
uorescence intensity and higher cell uptake in MCF-7 cells compared with untargeted
liposomes. Also, they observed a reduction in IC50 values of docetaxel-loaded TPGS
coated targeted and non-targeted liposomes by 97.58% and 83.64% compared with
Taxotere® after 24 h incubation with MCF-7 cells.141
FA targeting has shown promising outcomes in the treatment of breast cancer and
could be utilized for its potential theranostic applications in cancer.
C. CD44-Targeted Theranostics
CD44 receptors are overexpressed in cancer cells including breast cancer. HA is a prin-
cipal ligand for CD44 receptors and scientists have explored HA conjugated nanomed-
icines to achieve targeted delivery of therapeutic, diagnostic and theranostic agents.
Scientists have investigated HA-decorated, red emission cationic BSA protected gold
nanoclusters loaded with ICG and paclitaxel for chemo-photothermal therapy and nitric
oxide to modulate the tumor microenvironment and enhance the delivery of drug to the
cancer cells. The developed nanoclusters presented size-reducible ability in presence
of hyaluronidase and signicantly higher accumulation in breast cancer cells with the
homogenous intra-tumor distribution. Furthermore, their study showed a suppression of
95.3% in in situ tumor growth and 88.4% inhibition of lung metastasis growth.142
Similarly, Wang et al. developed pH-responsive HA prodrug micelles with aggre-
gation-induced emission properties by a chemical graft of biocompatible phosphoryl-
choline, DOX, and aggregation-induced emission uorogen tetraphenylene to the HA
backbone. The intracellular distribution of HA was observed by uorescence microscopy.
Flow cytometry and uorescent microscopy indicated that HA prodrug micelles were ef-
fectively internalized by MDA-MB-231 carcinoma cells due to HA-mediated endocyto-
sis. Also, the cytotoxicity results indicated that the prodrug micelles showed signicant
inhibition in the proliferation of cancerous cells. HA backbone could greatly enhance
the cellular internalization ability, furthermore, they observed a signicant inhibition
in the proliferation of cancer cells and higher cytotoxicity in MDA-MB-231 cells.143
Receptor-targeted theranostic nanomedicines based on CD44 receptor overexpression
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 25 Manila Typesetting Company 06/21/2022 09:12AM
26 Ahmad et al.
on cancer cells have shown the ability for simultaneous targeted delivery of antineoplas-
tic drugs and imaging agents to cancer cells.
D. EGFR-Targeted Theranostics
EGFR receptors are overexpressed on breast cancer cells and scientists have explored
anti-EGFR antibodies and aptamers for targeted theranostic applications in breast can-
cer. Kim et al. designed aptamer-loaded lipid-based nanocarriers containing QDs and
siRNAs for imaging and treatment of TNBC. The hydrophobic QDs were incorporated
in the lipid bilayer of liposomes, and then liposomes containing QDs were complexed
with therapeutic siRNAs and functionalized with anti-EGFR aptamer for targeting
TNBC. The results of uorescence imaging studies showed enhanced delivery of de-
veloped nanocarriers to EGFR overexpressing cancerous cells and thus, more effective
gene silencing and enhanced tumor imaging compared with non-targeted nanocarriers.
Moreover, combinatorial therapy with Bcl-2 and PKC-ι siRNAs loaded into the anti-
EGFR QD lipid nanocarriers in tumor-bearing female Balb/c nude mice caused remark-
able inhibition in tumor growth and metastasis.144
In another study, Wang et al. developed anti-EGFR nanobody-decorated QDs based
theranostic micelles loaded with an antineoplastic drug, aminoavone for TNBC ther-
apy. The results of in vitro studies in EGFR overexpressing MDA-MB-468 cells showed
that anti-EGFR nanobody 7D12 conjugation resulted in an enhanced cellular uptake and
cytotoxicity of the QD-based micelles. Furthermore, there was a higher concentration of
targeted micelles in tumors as compared with non-targeted ones, leading to more effec-
tive tumor regression in an orthotopic TNBC xenograft mouse model. Also, they did not
observe any systemic toxicity with the treatments.145 From these studies, it could be con-
cluded that EGFR targeted nanomedicine could be explored promisingly for theranostic
applications in breast cancer.
E. Transferrin-Targeted Theranostics
Transferrin (Tf) functionalized nanomedicines have signicantly been explored for
targeted delivery of anticancer therapeutic agents. Scientists are also exploring Tf for
targeted theranostic applications in breast cancer. Tf-functionalized DOX-loaded lumi-
nescent blue copper (Tf-Cu) nanoclusters have been fabricated by Goswami et al. for
breast cancer theranostic applications. The results of Förster resonance energy transfer
within the DOX-loaded Tf-Cu nanoclusters showed striking red luminescence, wherein
the blue luminescence of Tf-Cu nanoclusters (donor) was quenched due to absorption
by DOX (acceptor). The blue luminescence of Tf-Cu nanoclusters was restored in the
cytoplasm of cancer cells upon Tf mediated internalization. Finally, the gradual release
of DOX from the nanoclusters led to the generation of red luminescence inside the
nucleus. Moreover, a confocal microscopy image of HeLa cells revealed successful in-
ternalization of Tf-Cu nanoclusters in the cells. The results of in vivo studies showed
superior targeting efciency of DOX-loaded Tf-Cu nanoclusters on transferrin receptor
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 26 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 27
(TfR)–positive HeLa and MCF-7 cells compared with TfR-negative HEK-293 and 3T3-
L1 cells. Furthermore, the Combination index showed synergistic interaction of Tf-Cu
nanocluster and DOX upon treatment with DOX-loaded Tf-Cu nanoclusters. In vivo as-
sessment of the nanoclusters on TfR overexpressing Dalton’s lymphoma ascites-bearing
mice model showed signicant inhibition of tumor growth rendering prolonged survival
of the mice.146
In another study, Muthu et al. designed docetaxel and ultra-bright gold clusters
loaded Tf-tagged TPGS based theranostic micelles. Their results indicated that the mi-
celles showed signicantly higher cytotoxicity and cellular uptake along with the tar-
geted co-delivery, and enhanced theranostic activity compared with the non-targeted
micelles. Also, they observed a reduction in IC50 values of docetaxel-loaded TPGS based
non-targeted and targeted micelles by 15.31- and 71.73-fold as compared with that of
Taxotere® after 24 h incubation with MDA-MB-231-luc breast cancer cells, respectively.
Thus, they proposed that the developed micelles could be an attractive system for the
co-delivery of poorly soluble anticancer drugs and imaging agents to TfR overexpress-
ing carcinomas.147 Tf-conjugated micelles, nanoclusters and other nanomedicines have
shown promising potential for targeted delivery of antineoplastic drugs and simultane-
ous imaging of TfR overexpressing carcinoma.
The potential of receptor-targeted theranostic nanomedicines explored for breast
cancer is summarized in Table 2. Systematic research on ligand conjugated nanomedi-
cines for receptor-mediated targeting of breast cancer for theranostic applications may
result in a promising outcome and will surely aid in the clinical management of breast
cancers including its most invasive variant TNBC.
V. CHALLENGES IN DEVELOPMENT OF BREAST CANCER THERANOSTICS
AND FUTURE PERSPECTIVES
Major challenges in developing theranostic nanomedicines include (i) developing
simple, controllable, and reproducible methods for synthesis of nanomedicines; (ii)
insufcient batch-to-batch reproducibility, low yield, and variable physicochemical
characteristics; (iii) understanding the in vivo biochemical mechanisms through which
nanomedicines function (i.e., how nanomedicines target certain tissues and factors that
affect the release of drugs from nanocarriers); (iv) understanding the biodistribution of
NPs in vivo; (v) identifying the transformation and metabolic pathways; (vi) understand-
ing the chronic toxicity of nanotheranostics in vivo; (vii) stringent regulatory and safety
guidelines for timely and effective translation of theranostics to market. Finally, to bring
nanomedicine to the clinic for the effective diagnosis and treatment of human diseases,
we need multidisciplinary knowledge and techniques from pharmaceutical scientists,
nanochemists, nanophysics, nanotoxicologists, clinical doctors, and so forth to construct
safe and effective nanotheranostics.154,155
The high heterogeneity of breast cancers poses a big challenge for its successful
prognosis and treatment, which could be resolved by selectively targeting the theranostic
agents to cancer cells. Although, development of an effective theranostic nanomedicine
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 27 Manila Typesetting Company 06/21/2022 09:12AM
28 Ahmad et al.
Receptor
targeted
Targeting
ligand
Type of
nanomedicines
Type of
imaging
Imaging agent
Therapeutic
agent
Breast cancer
model
Outcome
Ref.
HER2 Trastuzumab SPIONs MRI Protoporphyrin IONPs MCF-7 breast
cancer cell line
In vitro photothermal
ablation of IONP-
protoporphyrin-
trastuzumab revealed
a 74% reduction in
MCF-7 cells after
10 min exposure
at the highest Fe
concentration
132
Anti-HER2
antibodies
Gold nanoshell
PLGA hybrid
nanocapsules
Ultrasound/
MRI
Peruorooctyl
bromide
DOX and
SPIONs
HER2-positive
SK-BR-3 cells
and HER2-
negative
MDA-MB-231
cells
–Signicant
photothermal
cytotoxicity
Signicantly enhanced
anti-tumor effect
148
Herceptin Graphene QDs Fluorescence Graphene QDs DOX HER2-negative
MCF-7 breast
cancer cells,
HER2-positive
BT-474 breast
cancer cells
Signicant
accumulation of
nanocarriers in the
cancer cells,
Internalization into the
cells via endocytosis
149
Trastuzumab
Lipid NPs
Fluorescence
QDs
Rapamycin
SK-BR-3 cells,
MDA-MB-231
cells
signicantly higher
cellular uptake, higher
uorescence intensity
in tumor spheroid,
~ 3 and ~ 1.5-fold
higher toxicity to
SK-BR-3 cells and
MDA-MB-231 cells,
respectively
134
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 28 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 29
TABLE 2: (continued)
Herceptin IONPs MRI and
Fluorescence
IONPs DOX
In vitro
/
SK-BR-3 and
MCF-7 breast
cancer cells
In vivo/
Athymic nude
mice bearing
SK-BR-3 cancer
cells
–5 fold higher cellular
uptake
–Signicantly
enhanced tumor
regression in vitro and
in vivo
133
Herceptin Mesoporous
silica NPs
Ultrasound Mesoporous
silica NPs
Mesoporous
silica NPs
In vitro/
SK-BR-3 and
MDA-MB-231
breast cancer
cells
–Signicantly higher
cellular uptake
–Signicant
enhancement in
ultrasound image
contrast and enhanced
breast tumor-specic
toxicity
150
Trastuzumab SPIONs MRI Fluorescent
dye
SPIONs In vivo/MMC
cells
In vivo/FVB/N
transgenic
mouse model
–4-fold higher cellular
uptake
–Signicant contrast
enhancement in MR
images of primary
breast tumors in vitro
and in vivo
83
Herceptin Magnetic PLGA
NPs
MRI Magnetic
nanocrystals
DOX In vitro/
NIH3T6.7,
SK-BR-3, and
MDA-MB-231
cancer cells
–87.4 times greater
uorescent intensity
–Signicantly higher
cancer cell afnity
–Signicantly higher
cellular uptake
–Excellent tumor
growth retardation both
in vitro and in vivo
136
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 29 Manila Typesetting Company 06/21/2022 09:12AM
30 Ahmad et al.
TABLE 2: (continued)
Receptor
targeted
Targeting
ligand
Type of
nanomedicines
Type of
imaging
Imaging agent
Therapeutic
agent
Breast cancer
model
Outcome
Ref.
Folic acid FA Fe3O4 NPs MRI IONPs IONPs MC4-L2 cells,
Female Balb/c
mice
–The NPs were
signicantly
accumulated and
retained within the
tumor tissues.
–MRI experiments
showed a signicant
decrease in T2-
weighted MR signal
intensity of breast
tumors indicating the
accumulation and
retention of NPs in the
tumor tissue in vivo.
–Reduction in tumor
progression
137
FA Nanocomposite Fluorescence Graphene
oxide
manganese-
doped zinc
sulde QDs
DOX Breast cancer
cell line
MDA-MB-231
(FA receptor
positive) and
NIH-3T3
cell line (FA
receptor
negative)
–FA functionalization
improves the selectivity
for discriminating
positive FR cancer
cells.
–Signicant reduction
in the toxicity of the
nanocomposite
151
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 30 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 31
TABLE 2: (continued)
FA IONPs MRI IONPs DOX Breast cancer
cell lines,
i.e., MCF-7,
BT549, and
MD-MBA-231
Female Balb/c
nude mice
–FA conjugated NPs
showed the strongest
cytotoxicity against
breast cancer cells,
compared with
non-targeted NPs,
and caused cellular
apoptosis.
–Suppression in in vivo
tumor growth in mice,
No signs of toxicity
against healthy organs
152
FA PEGylated
magnetic NPs
MRI Magnetic NPs Tamoxifen In vitro/MCF-7
breast cancer
cell line
–Signicantly enhanced
cellular uptake
–Signicant inhibition in
growth of MCF-7 cells
140
FA TPGS micelles Fluorescence QDs Docetaxel MCF-7 breast
cancer cells
–Signicantly higher
red uorescence
intensity and higher
cellular uptake
–Signicantly improved
cytotoxicity in vitro
141
CD44 HA Cationic BSA
protected gold
nanoclusters
PAI ICG Paclitaxel 4T1, A549, and
RAW 246.7
Cells
Female Balb/c
mice
–Signicantly
enhanced cellular
uptake
–Suppression in in situ
tumor growth by 95.3%
142
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 31 Manila Typesetting Company 06/21/2022 09:12AM
32 Ahmad et al.
TABLE 2: (continued)
Receptor
targeted
Targeting
ligand
Type of
nanomedicines
Type of
imaging
Imaging agent
Therapeutic
agent
Breast cancer
model
Outcome
Ref.
HA HA prodrug
micelles
Fluorescence Aggregation-
induced
emission
uorogen
tetraphenylene
DOX CD44-
overexpressing
MDA-MB-231
cell line CD44
negative
NIH3T3 cell
line
–Flow cytometry and
uorescent microscopy
indicated that HA
prodrug micelles were
effectively internalized
by MDA-MB-231 cells
due to HA-mediated
endocytosis
–Signicant inhibition
of the proliferation of
cancer cells
–Signicantly enhanced
cytotoxicity against
MDA-MB-231 cells
143
EGFR Anti-EGFR
aptamer
Lipid
nanocarriers
Fluorescence QDs siRNAs EGFR positive
MDA-MB-231
and EGFR
negative
MDA-MB-453
cell lines,
Tumor-bearing
female Balb/c
nude mice
–Enhanced delivery to
target cancer cells
–Signicant reduction
in target gene
expression
–Inhibition of tumor
growth and metastasis
144
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 32 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 33
TABLE 2: (continued)
Anti-EGFR
Nanobody
Micelles Fluorescence QDs Aminoavone EGFR
overexpressing
MDA-MB-468
TNBC cells
MDA-MB-468
breast cancer
xenograft mouse
model
–Nb-conjugated
micelles accumulated
in tumors at higher
concentrations, leading
to more effective tumor
regression in the mouse
model
–No systemic toxicity
observed
–Enhanced cellular
uptake and increased
cytotoxicity
145
Transferrin Tf Nanoclusters Fluorescence luminescent
blue copper
DOX TfR positive
HeLa,
TfR positive
MCF-7 cells,
TfR partially
positive HEK-
293 and
TfR negative
Mouse embryo
broblast cell
lines
–Tf-CuNC-DOX-NPs
showed excellent
targeting efciency
on TfR overexpressed
cells (HeLa and MCF-
7) as compared with
the less TfR expressed
cells (HEK-293 and
3T3-L1)
–Confocal microscopy
images of HeLa cells
revealed successful
internalization of Tf-Cu
nanoclusters in the
cells
146
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 33 Manila Typesetting Company 06/21/2022 09:12AM
34 Ahmad et al.
TABLE 2: (continued)
Receptor
targeted
Targeting
ligand
Type of
nanomedicines
Type of
imaging
Imaging agent Therapeutic
agent
Breast cancer
model
Outcome Ref.
Tf
TPGS micelles
Fluorescence
Ultra-
bright gold
nanoclusters
Docetaxel
In vitro/MDA-
MB-231-luc
and NIH-3T3
broblast cells
In vivo/female
CB-17 mice
bearing MDA-
MB-231-luc
cells induced
tumor
Signicantly higher
cellular uptake and
theranostic effect
4.6-fold reduction in
IC50 value compared
with non-targeted
micelles
147
Tf PEGylated gold
NPs
PAI Fluorescein
isothiocyanate
Gold NPs In vitro/Hs578T
cancer cells
–6-fold higher cellular
uptake in breast cancer
cells
–Reduced in laser
power effective for
therapy from 1600 W/
cm2 to 7 W/cm2, which
is more than two orders
of magnitude lower
153
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 34 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 35
with targeting propensity toward breast cancer cells may encounter many challenges,
yet it will be benecial to develop these nanomedicines as the conventional techniques
used for diagnosis and management of breast cancer are not efcient enough to image
the breast tumors completely with minute detailing. Further, targeted theranostic medi-
cines, which can be sorted out using novel techniques and strategies, give a unique op-
portunity to maximize the accumulation of anticancer therapeutics to cancer cells which
could result in an improved therapeutic index with a signicant reduction in toxicity
which is one of the major hurdles in the treatment of cancer along with the development
of resistance to chemotherapeutic agents.
Cancer nanotechnology has advanced over the last two decades to reach a stage
where the novel approach of “cancer theranostics” has been conceptualized and devel-
oped for the diagnosis and therapy of breast cancer. It provides a unique combination
of drug and a diagnostic probe having controlled release and receptor-mediated target-
ing potential. Theranostics offers an effective and patient-friendly alternative. Various
polymeric, lipid-based and inorganic NPs have been considered for this approach. They
have shown therapeutically signicant and noteworthy results in various investigations.
However, the majority of theranostic applications in breast cancer have only been dem-
onstrated at the pre-clinical level. In addition, due to nanoscale sizing, complex syn-
thetic pathways can alter the physicochemical properties and consequently become a
source of toxicity at the cellular or subcellular level. Various investigations related to
the same have established the potential of nanomaterials for biochemical and physi-
ological alterations. Therefore, a correlation needs to be established between their ef-
cacy and safety. The study of in vitro/in vivo correlation has also become a challenge
due to the non-availability of suitable tools. Thus, there is an unmet need to match the
changing paradigm of breast cancer therapy using receptor-mediated nanomedicine with
advanced bio-pharmaceutical and toxicity assessment tools.
VI. CONCLUSION
Breast cancer is emerging as one of the most frequently diagnosed cancer in women in
developed and developing countries. Management of breast cancer is a difcult task if
not diagnosed at an early stage. Further, the most aggressive form of breast cancer, i.e.,
TNBS is very difcult to manage due to metastasis which is further worsened by poor
prognosis. Receptor-targeted nanomedicines are being explored to augment the selec-
tivity of drug delivery to cancer cells. With the help of suitable ligand and engineering
technology, promising nanomedicines may be designed to target the therapeutic, imag-
ing agents, or both simultaneously to cancer cells. The result of ongoing studies and
studies done in the last few decades are strongly supporting the candidature of receptor-
targeted nanomedicines for targeted delivery of therapeutic and diagnostic moieties to
breast cancer with an improved therapeutic index.
Further nanomedicine-based approaches are also being explored for theranostic ap-
plications in breast cancer with promising results which have been obtained in research
studies conducted in the last decade. These nanomedicines based theranostic approaches
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 35 Manila Typesetting Company 06/21/2022 09:12AM
36 Ahmad et al.
have revealed an ability to deliver the therapeutic agents to breast cancer, selectively, to
enhance the efcacy of cancer therapy with simultaneous imaging of cancer microen-
vironment for better prognosis and management of breast cancer. So, from the result of
these investigations, it could be expected that continuous and coherent investigation in
this direction may help in developing promising therapeutic approaches based on nano-
medicines for targeted delivery of diagnostic agents, therapeutic moieties, and theranos-
tic modalities to breast cancer, which may get approval for clinical use.
ACKNOWLEDGMENTS
T.S. and K.J. are thankful to National Institute of Pharmaceutical Education and Research
(NIPER), Raebareli, for providing the facilities to write this manuscript. Author, Dr.
Keerti Jain, acknowledges Indian Council of Medical Research (ICMR), New Delhi
for the nancial support in the form of ICMR Extramural Research Project (Project
ID: 2020-4686; Ref. No. 5/13/34/2020/NCD-III). The NIPER Raebareli communication
number for this manuscript is NIPER-R/Communication/177.
REFERENCES
1. World Health Organization. Cancer. Available from: https://www.who.int/news-room/fact-sheets/
detail/cancer.
2. Momenimovahed Z, Salehiniya H. Epidemiological characteristics of and risk factors for breast can-
cer in the world. Breast Cancer (Dove Med Press). 2019;11:151–64.
3. Cancer.Net. Breast cancer - metastatic: Statistics. Available from: https://www.cancer.net/cancer-types/
breast-cancer-metastatic/statistics.
4. Francis I, Altemaimi R, Al-Ayadhy B, Alath P, Jaragh M, Mothafar F, Kapila K. Hormone receptors
and human epidermal growth factor (HER2) expression in ne-needle aspirates from metastatic breast
carcinoma – Role in patient management. J Cytol. 2019;36(2):94–100.
5. Yao H, He G, Yan S, Chen C, Song L, Rosol TJ, Deng X. Triple-negative breast cancer: Is there a
treatment on the horizon? Oncotarget. 2017;8(1):1913–24.
6. Khan MA, Jain VK, Rizwanullah M, Ahmad J, Jain K. PI3K/AKT/mTOR pathway inhibitors in tri-
ple-negative breast cancer: A review on drug discovery and future challenges. Drug Discov Today.
2019;24(11):2181–91.
7. Ji X, Lu Y, Tian H, Meng X, Wei M, Cho WC. Chemoresistance mechanisms of breast cancer and their
countermeasures. Biomed Pharmacother. 2019;114:108800.
8. Ahmad J, Akhter S, Greig NH, Amjad Kamal M, Midoux P, Pichon C. Engineered nanoparticles
against mdr in cancer: The state of the art and its prospective. Curr Pharm Des. 2016;22(28):4360–73.
9. Ahmad J, Kohli K, Mir SR, Amin S. Lipid based nanocarriers for oral delivery of cancer chemothera-
peutics: An insight in the intestinal lymphatic transport. Drug Deliv Lett. 2013;3(1):38–46.
10. Ahmad J, Amin S, Rahman M, Rub R, Singhal M, Ahmad M, Rahman Z, Addo R, Ahmad F, Mushtaq
G, Amjad Kamal M, Akhter S. Solid matrix based lipidic nanoparticles in oral cancer chemotherapy:
Applications and pharmacokinetics. Curr Drug Metab. 2015;16(8):633–44.
11. Ahmad J, Akhter S, Rizwanullah M, Amin S, Rahman M, Ahmad MZ, Rizvi MA, Amjad Kamal M,
Ahmad FJ. Nanotechnology-based inhalation treatments for lung cancer: State of the art. Nanotechnol
Sci Appl. 2015;8:55–66.
12. Afsana, Jain V, Haider N, Jain K. 3D printing in personalized drug delivery. Curr Pharm Des.
2019;24(42):5062–71.
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 36 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 37
13. Pardhi VP, Verma T, Flora SJS, Chandasana H, Shukla R. Nanocrystals: An overview of fabrica-
tion, characterization and therapeutic applications in drug delivery. Curr Pharm Des. 2019;24(43):
5129–46.
14. Joshi K, Chandra A, Jain K, Talegaonkar S. Nanocrystalization: An emerging technology to enhance
the bioavailability of poorly soluble drugs. Pharm Nanotechnol. 2019;7(4):259–78.
15. Jain K, Shukla R, Yadav A, Ujjwal RR, Flora SJS. 3D printing in development of nanomedicines.
Nanomaterials. 2021;11(2):1–24.
16. Jain K, Jain NK. Dendrimers as nanobiopolymers in cancer chemotherapy. In: Nanobiomedicine.
Houston, TX: Studium Press; 2015. p. 289–306.
17. Rizwanullah Md, Ahmad J, Amin S. Nanostructured lipid carriers: A novel platform for chemothera-
peutics. Curr Drug Deliv. 201;13(1):4–26.
18. Akhter MdH, Rizwanullah Md, Ahmad J, Ahsan MJ, Mujtaba MdA, Amin S. Nanocarriers in ad-
vanced drug targeting: Setting novel paradigm in cancer therapeutics. Artif Cells Nanomed Biotechnol.
2018;46(5):873–84.
19. Jain V, Jain K. Molecular targets and pathways for the treatment of visceral leishmaniasis. Drug
Discov Today. 2018;23(1):161–70.
20. Jain K, Mehra NK, Jain NK. Potentials and emerging trends in nanopharmacology. Curr Opin
Pharmacol. 2014;15(1):97–106.
21. Soni N, Jain K, Gupta U, Jain NK. Controlled delivery of gemcitabine hydrochloride using mannosyl-
ated poly(propyleneimine) dendrimers. J Nanoparticle Res. 2015;17(11):1–17.
22. Pardhi V, Chavan RB, Thipparaboina R, Thatikonda S, Naidu VGM, Shastri NR. Preparation, char-
acterization, and cytotoxicity studies of niclosamide loaded mesoporous drug delivery systems. Int J
Pharm. 2017;528(1-2):202–14.
23. Rizwanullah M, Amin S, Mir SR, Fakhri KU, Rizvi MMA. Phytochemical based nanomedicines
against cancer: Current status and future prospects. J Drug Target. 2018;26(9):731–52.
24. Jain K. Nanohybrids of dendrimers and carbon nanotubes: A benefaction or forfeit in drug delivery?
Nanosci Nanotechnol. 2018;9(1):21–9.
25. Dahiya S, Dahiya R, Hernández E. Nanocarriers for anticancer drug targeting: Recent trends and chal-
lenges. Crit Rev Ther Drug Carrier Syst. 2021;38(6):49–103.
26. Navya PN, Kaphle A, Srinivas SP, Bhargava SK, Rotello VM, Daima HK. Current trends and chal-
lenges in cancer management and therapy using designer nanomaterials. Nano Converg. 2019;6(2):23.
27. Jain K, Jain NK. Surface engineered dendrimers as antiangiogenic agent and carrier for anticancer
drug: Dual attack on cancer. J Nanosci Nanotechnol. 2014;14(7):5075–87.
28. Rahman M, Akhter S, Ahmad MZ, Ahmad J, Addo RT, Ahmad FJ, Pichon C. Emerging advances in
cancer nanotheranostics with graphene nanocomposites: Opportunities and challenges. Nanomedicine.
2015;10(15):2405–22.
29. Bulbake U, Doppalapudi S, Kommineni N, Khan W. Liposomal formulations in clinical use: An up-
dated review. Pharmaceutics. 2017;9(2):12.
30. Din FU, Aman W, Ullah I, Qureshi OS, Mustapha O, Shaque S, Zeb A. Effective use of nanocarriers
as drug delivery systems for the treatment of selected tumors. Int J Nanomed. 2017;12:7291–309.
31. Kirtane AR, Kalscheuer SM, Panyam J. Exploiting nanotechnology to overcome tumor drug resis-
tance: Challenges and opportunities. Advanced Drug Delivery Reviews. 2013;65(13-14):1731–47.
32. Maeda H, Nakamura H, Fang J. The EPR effect for macromolecular drug delivery to solid tumors:
Improvement of tumor uptake, lowering of systemic toxicity, and distinct tumor imaging in vivo. Adv
Drug Deliv Rev. 2013;65(1):71–9.
33. Schaaf MB, Garg AD, Agostinis P. Dening the role of the tumor vasculature in antitumor immunity
and immunotherapy article. Cell Death Dis. 2018;9(2):1–14.
34. Fang J, Nakamura H, Maeda H. The EPR effect: Unique features of tumor blood vessels for drug
delivery, factors involved, and limitations and augmentation of the effect. Adv Drug Deliv Rev.
2011;63(3):136–51.
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 37 Manila Typesetting Company 06/21/2022 09:12AM
38 Ahmad et al.
35. Khawar IA, Kim JH, Kuh HJ. Improving drug delivery to solid tumors: Priming the tumor microenvi-
ronment. J Control Release. 2015;201:78–89.
36. Shi Y, van der Meel R, Chen X, Lammers T. The EPR effect and beyond: Strategies to improve tumor
targeting and cancer nanomedicine treatment efcacy. Theranostics. 2020;10(17):7921–4.
37. Danhier F, Feron O, Préat V. To exploit the tumor microenvironment: Passive and active tumor target-
ing of nanocarriers for anti-cancer drug delivery. J Control Release. 2010;148(2):135–46.
38. Bertrand N, Wu J, Xu X, Kamaly N, Farokhzad OC. Cancer nanotechnology: The impact of passive
and active targeting in the era of modern cancer biology. Adv Drug Deliv Rev. 2014;66:2–25.
39. Shi J, Kantoff PW, Wooster R, Farokhzad OC. Cancer nanomedicine: Progress, challenges and op-
portunities. Nat Rev Cancer. 2017;17(1):20–37.
40. Fang J, Islam W, Maeda H. Exploiting the dynamics of the EPR effect and strategies to improve
the therapeutic effects of nanomedicines by using EPR effect enhancers. Adv Drug Deliv Rev.
2020;157:142–60.
41. Subhan MA, Yalamarty SSK, Filipczak N, Parveen F, Torchilin VP. Recent advances in tumor target-
ing via epr effect for cancer treatment. J Pers Med. 2021;11(6).
42. Islam R, Maeda H, Fang J. Factors affecting the dynamics and heterogeneity of the EPR effect: Patho-
physiological and pathoanatomic features, drug formulations and physicochemical factors. Expert
Opinion on Drug Delivery. 2021:1–14.
43. Halin C, Neri D. Antibody-based targeting of angiogenesis. Crit Rev Ther Drug Carrier Syst.
2001;18(3):299–339.
44. Golombek SK, May JN, Theek B, Appold L, Drude N, Kiessling F, Lammers T. Tumor targeting via
EPR: Strategies to enhance patient responses. Adv Drug Deliv Rev. 2018;130:17–38.
45. Patra JK, Das G, Fraceto LF, Campos EVR, Rodriguez-Torres MDP, Acosta-Torres LS, Diaz-Torres
LS, Grillo R, Swamy MK, Sharma S, Habtemariam S, Shin H. Nano based drug delivery systems:
Recent developments and future prospects. J Nanobiotechnol. 2018;16(1):71.
46. Fam SY, Chee CF, Yong CY, Ho KL, Mariatulqabtiah AR, Tan WS. Stealth coating of Nanoparticles
in drug-delivery systems. Nanomaterials. 2020;10(4):1–18.
47. Hillaireau H, Couvreur P. Nanocarriers’ entry into the cell: Relevance to drug delivery. Cell Mol Life
Sci. 2009;66(17):2873–96.
48. Pérez-Herrero E, Fernández-Medarde A. Advanced targeted therapies in cancer: Drug nanocarriers,
the future of chemotherapy. Eur J Pharm Biopharm. 2015;93:52–79.
49. Xie J, Yang Z, Zhou C, Zhu J, Lee RJ, Teng L. Nanotechnology for the delivery of phytochemicals in
cancer therapy. Biotechnology Advances. 2016;34(4):343–53.
50. Attia MF, Anton N, Wallyn J, Omran Z, Vandamme TF. An overview of active and passive tar-
geting strategies to improve the nanocarriers efciency to tumour sites. J Pharm Pharmacol.
2019;71(8):1185–98.
51. Zhong Y, Meng F, Deng C, Zhong Z. Ligand-directed active tumor-targeting polymeric nanoparticles
for cancer chemotherapy. Biomacromolecules. 2014;15(6):1955–69.
52. Yoo J, Park C, Yi G, Lee D, Koo H. Active targeting strategies using biological ligands for nanopar-
ticle drug delivery systems. Cancers. 2019;11(5).
53. Bazak R, Houri M, el Achy S, Kamel S, Refaat T. Cancer active targeting by nanoparticles: A compre-
hensive review of literature. J Cancer Res Clin Oncol. 2015;141(5):769–84.
54. He K, Zeng S, Qian L. Recent progress in the molecular imaging of therapeutic monoclonal antibod-
ies. J Pharm Anal. 2020;10(5):397–413.
55. Vyas SP, Singh A, Sihorkar V. Ligand-receptor-mediated drug delivery: An emerging paradigm in cel-
lular drug targeting. Crit Rev Ther Drug Carrier Syst. 2001;18(1):1–76.
56. Danhier F. To exploit the tumor microenvironment: Since the EPR effect fails in the clinic, what is the
future of nanomedicine? J Control Release. 2016;244(Pt A):108–21.
57. Roma-Rodrigues C, Mendes R, Baptista PV, Fernandes AR. Targeting tumor microenvironment for
cancer therapy. Int J Mol Sci. 2019;20(4).
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 38 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 39
58. Ashfaq UA, Riaz M, Yasmeen E, Yousaf M. Recent advances in nanoparticle-based targeted drug-
delivery systems against cancer and role of tumor microenvironment. Crit Rev Ther Drug Carrier Syst.
2017;34(4):317–53.
59. Mallidi S, Luke GP, Emelianov S. Photoacoustic imaging in cancer detection, diagnosis, and treatment
guidance. Trends Biotechnol. 2011;29(5):213–21.
60. Lima ZS, Ebadi MR, Amjad G, Younesi L. Application of imaging technologies in breast cancer detec-
tion: A review article. Open Access Maced J Med Sci. 2019;7(5):838–48.
61. Yezhelyev MV, Gao X, Xing Y, Al-Hajj A, Nie S, O’Regan RM. Emerging use of nanoparticles in
diagnosis and treatment of breast cancer. Lancet Oncol. 2006;7(8):657–67.
62. Wang J, Xu B. Targeted therapeutic options and future perspectives for HER2-positive breast cancer.
Signal Transduct Target Ther. 2019;4(1):1–22.
63. Li Y, Yang X-Q, Chen C, Peng C-W, Hou J-X, Liu S-P, Qi C-B, Gong Y-P, Zhu X-B, Pang D-W.
Quantum dot-based quantitative immunouorescence detection and spectrum analysis of epidermal
growth factor receptor in breast cancer tissue arrays. Int J Nanomed. 2011;6:2265.
64. Wu L, Qu X. Cancer biomarker detection: Recent achievements and challenges. Chem Soc Rev.
2015;44(10):2963–97.
65. Chen C, Peng J, Xia HS, Yang GF, Wu QS, Chen LD, Zeng LB, Zhang ZL, Pang DW, Li Y. Quantum
dots-based immunouorescence technology for the quantitative determination of HER2 expression in
breast cancer. Biomaterials. 2009;30(15):2912–8.
66. Pelaz B, Alexiou C, Alvarez-Puebla RA, Alves F, Andrews AM, Ashraf S, Balogh LP, Ballerini
L, Bestetti A, Brendel C, Bosi S, Carril M, Chan WCW, Chen C, Chen X, Chen X, Cheng Z, Cui
D, Du J, Dullin C, Escudero A, Feliu N, Gao M, George M, Gogotsi Y, Grünweller A, Gu Z, Halas
NJ, Hampp N, Hartmann RK, Hersam MC, Hunziker P, Jian J, Jiang X, Jungebluth P, Kadhiresan
P, Kataoka K, Khademhosseini A, Kopeček J, Kotov NA, Krug HF, Lee DS, Lehr CM, Leong
KW, Liang X, Lim ML, Liz-Marzán LM, Ma X, Macchiarini P, Meng H, Möhwald H, Mulvaney
P, Nel AE, Nie S, Nordlander P, Okano T, Oliveira J, Park TH, Penner RM, Prato M, Puntes V, Rotello
VM, Samarakoon A, Schaak RE, Shen Y, Sjöqvist S, Skirtach AG, Soliman MG, Stevens MM, Sung
HW, Tang BZ, Tietze R, Udugama BN, VanEpps JS, Weil T, Weiss PS, Willner I, Wu Y, Yang L, Yue
Z, Zhang Q, Zhang Q, Zhang XE, Zhao Y, Zhou X, Parak WJ. Diverse applications of nanomedicine.
ACS Nano. 2017;11(3):2313–81.
67. Meier R, Henning TD, Boddington S, Tavri S, Arora S, Piontek G, Rudelius M, Corot C, Daldrup-Link
HE. Breast cancers: MR imaging of folate-receptor expression with the folate-specic nanoparticle
P1133. Radiology. 2010;255(2):527–35.
68. Lim EK, Kim HO, Jang E, Park J, Lee K, Suh JS, Huh YM, Haam S. Hyaluronan-modied mag-
netic nanoclusters for detection of CD44-overexpressing breast cancer by MR imaging. Biomaterials.
2011;32(31):7941–50.
69. Keshtkar M, Shahbazi-Gahrouei D, Khoshfetrat S, Mehrgardi M, Aghaei M. Aptamer-conjugated
magnetic nanoparticles as targeted magnetic resonance imaging contrast agent for breast cancer. J
Med Signals Sens. 2016;6(4):243–7.
70. Nian D, Shi P, Sun J, Ren L, Hao X, Han J. Application of luteinizing hormone-releasing hormone–
ferrosoferric oxide nanoparticles in targeted imaging of breast tumors. J Int Med Res. 2019;47(4):
1749–57.
71. Yang J, Lee CH, Park J, Seo S, Lim EK, Song YJ, Suh JS, Yoon HG, Huh YM, Haam S. Antibody
conjugated magnetic PLGA nanoparticles for diagnosis and treatment of breast cancer. J Mater Chem.
2007;17(26):2695–9.
72. Wu P, He X, Wang K, Tan W, Ma D, Yang W, He C. Imaging breast cancer cells and tissues using
peptide-labeled uorescent silica nanoparticles. J Nanosci Nanotechnol. 2008;
73. Sha Q, Guan R, Su H, Zhang L, Liu BF, Hu Z, Liu X. Carbohydrate-protein template synthesized
high mannose loading gold nanoclusters: A powerful uorescence probe for sensitive Concanavalin A
detection and specic breast cancer cell imaging. Talanta. 2020;218:121130.
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 39 Manila Typesetting Company 06/21/2022 09:12AM
40 Ahmad et al.
74. Tanaka S, Wadati H, Sato K, Yasuda H, Niioka H. Red-uorescent Pt nanoclusters for detecting and
imaging HER2 in breast cancer cells. ACS Omega. 2020;5(37):23718–23.
75. Sakamoto JH, Smith BR, Xie B, Rokhlin SI, Lee SC, Ferrari M. The molecular analysis of breast
cancer utilizing targeted nanoparticle based ultrasound contrast agents. Technol Cancer Res Treat.
2005;4(6):627–36.
76. Liu J, Li J, Rosol TJ, Pan X, Voorhees JL. Biodegradable nanoparticles for targeted ultrasound imag-
ing of breast cancer cells in vitro. Phys Med Biol. 2007;52(16):4739–47.
77. Xu L, Du J, Wan CF, Zhang Y, Xie SW, Li HL, Yang H, Li F. Ultrasound molecular imaging of breast
cancer in MCF-7 orthotopic mice using gold nanoshelled poly(lactic-co-glycolic acid) nanocapsules:
A novel dual-targeted ultrasound contrast agent. Int J Nanomed. 2018;13:1791–807.
78. Dong Q, Yang H, Wan C, Zheng D, Zhou Z, Xie S, Xu L, Du J, Li F. HER2-functionalized gold-
nanoshelled magnetic hybrid nanoparticles: A theranostic agent for dual-modal imaging and photo-
thermal therapy of breast cancer. Nanoscale Res Lett. 2019;14(1):235.
79. Galanzha EI, Kim JW, Zharov VP. Nanotechnology-based molecular photoacoustic and photother-
mal ow cytometry platform for in-vivo detection and killing of circulating cancer stem cells. J
Biophotonics. 2009;2(12):725–35.
80. Xi L, Grobmyer SR, Zhou G, Qian W, Yang L, Jiang H. Molecular photoacoustic tomography of breast
cancer using receptor targeted magnetic iron oxide nanoparticles as contrast agents. J Biophotonics.
2014;7(6):401–9.
81. Zhang T, Cui H, Fang CY, Cheng K, Yang X, Chang HC, Forrest ML. Targeted nanodiamonds as
phenotype-specic photoacoustic contrast agents for breast cancer. Nanomedicine. 2015;10(4):
573–87.
82. Liu Y, Wu PC, Guo S, Chou PT, Deng C, Chou SW, Yuan Z, Liu TM. Low-toxicity FePt nanoparticles
for the targeted and enhanced diagnosis of breast tumors using few centimeters deep whole-body
photoacoustic imaging. Photoacoustics. 2020;19:100179.
83. Kievit FM, Stephen ZR, Veiseh O, Arami H, Wang T, Lai VP, Park JO, Ellenbogen RG, Disis ML,
Zhang M. Targeting of primary breast cancers and metastases in a transgenic mouse model using ra-
tionally designed multifunctional SPIONs. ACS Nano. 2012;6(3):2591–601.
84. Nima ZA, Watanabe F, Jamshidi-Parsian A, Sarimollaoglu M, Nedosekin DA, Han M, Watts JA, Biris
AS, Zharov VP, Galanzha EI. Bioinspired magnetic nanoparticles as multimodal photoacoustic, pho-
tothermal and photomechanical contrast agents. Sci Rep. 2019;9(1):1–12.
85. Du J, Zhang Y, Jin Z, Wu H, Cang J, Shen Y, Miao F, Zhang A, Zhang Y, Zhang J, Teng G. Targeted
NIRF/MR dual-mode imaging of breast cancer brain metastasis using BRBP1-functionalized ultra-
small iron oxide nanoparticles. Mater Sci Engin C Mater Biol Appl. 2020;116:111188.
86. Luk BT, Zhang L. Current advances in polymer-based nanotheranostics for cancer treatment and di-
agnosis. ACS Appl Mater Interfaces. 2014;6(24):21859–73.
87. Grover VPB, Tognarelli JM, Crossey MME, Cox IJ, Taylor-Robinson SD, McPhail MJW. Magnetic
resonance imaging: Principles and techniques: Lessons for clinicians. J Clin Exp Hepatol. 2015;5(3):
246–55.
88. Estelrich J, Sánchez-Martín MJ, Busquets MA. Nanoparticles in magnetic resonance imaging: From
simple to dual contrast agents. Int J Nanomed. 2015;10:1727–41.
89. Fern I, Muñoz-Hernando M, Ruiz-Cabello J. Iron oxide nanoparticles: An alternative for positive
contrast in magnetic resonance imaging. Inorganics. 2020;1–22.
90. Li DL, Tan JE, Tian Y, Huang S, Sun PH, Wang M, Han YJ, Li HS, Wu HB, Zhang XM, Xu YK,
Wang QS. Multifunctional superparamagnetic nanoparticles conjugated with uorescein-labeled
designed ankyrin repeat protein as an efcient HER2-targeted probe in breast cancer. Biomaterials.
2017;147:86–98.
91. Ding N, Sano K, Kanazaki K, Ohashi M, Deguchi J, Kanada Y, Ono M, Saji H. In vivo HER2-targeted
magnetic resonance tumor imaging using iron oxide nanoparticles conjugated with anti-HER2 frag-
ment antibody. Mol Imaging Biol. 2016;18(6):870–6.
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 40 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 41
92. Lee T, Lim EK, Lee J, Kang B, Choi J, Park HS, Suh JS, Huh YM, Haam S. Efcient CD44-targeted
magnetic resonance imaging (MRI) of breast cancer cells using hyaluronic acid (HA)-modied
MnFe2O4 nanocrystals. Nanoscale Res Lett. 2013;8(1):1–9.
93. Meng J, Fan J, Galiana G, Branca RT, Clasen PL, Ma S, Zhou J, Leuschner C, Kumar C, Hormes J,
Otiti T, Beye A, Harmer M, Kiely C, Warren W, Haataja M, Soboyejo W. LHRH-functionalized super-
paramagnetic iron oxide nanoparticles for breast cancer targeting and contrast enhancement in MRI.
Mater Sci Engin C. 2009;29(4):1467–79.
94. Cubeddu R, Comelli D, D’Andrea C, Taroni P, Valentini G. Time-resolved uorescence imaging in
biology and medicine. J Phys D Appl Phys. 2002;35(9):R61.
95. Sarkar I, Mishra AK. Fluorophore tagged bio-molecules and their applications: A brief review. Appl
Spectrosc Rev. 2018;53(7):552–601.
96. Sarder P, Yazdanfar S, Akers WJ, Tang R, Sudlow GP, Egbulefu C, Achilefu S. All-near-infrared mul-
tiphoton microscopy interrogates intact tissues at deeper imaging depths than conventional single- and
two-photon near-infrared excitation microscopes. J Biomed Opt. 2013;18(10):106012.
97. Sajedi S, Sabet H, Choi HS. Intraoperative biophotonic imaging systems for image-guided interven-
tions. Nanophotonics. 2018;8(1):99–116.
98. Basu S, Alavi A. SPECT-CT and PET-CT in oncology – An overview. Curr Med Imaging Rev.
2011;7(3):202–9.
99. Chopra A, Shan L, Eckelman WC, Leung K, Menkens AE. Important parameters to consider for the
characterization of PET and SPECT imaging probes. Nucl Med Biol. 2011;38(8):1079–84.
100. Miletich RS. Positron emission tomography and single-photon emission computed tomography in
neurology. Continuum (Minneap Minn). 2016;22:1636–54.
101. Frangioni JV. In vivo near-infrared uorescence imaging. Curr Opin Chem Biol. 2003;7(5):626–34.
102. Ntziachristos V, Bremer C, Weissleder R. Fluorescence imaging with near-infrared light: New techno-
logical advances that enable in vivo molecular imaging. Eur Radiol. 2003;13(1):195–208.
103. Janib SM, Moses AS, MacKay JA. Imaging and drug delivery using theranostic nanoparticles. Adv
Drug Deliv Rev. 2010;62(11):1052–63.
104. Yamaguchi H, Tsuchimochi M, Hayama K, Kawase T, Tsubokawa N. Dual-labeled near-infrared/99mTc
imaging probes using PAMAM-coated silica nanoparticles for the imaging of HER2-expressing can-
cer cells. Int J Mol Sci. 2016;17(7).
105. Li K, Jiang Y, Ding D, Zhang X, Liu Y, Hua J, Feng S, Liu B. Folic acid-functionalized two-photon
absorbing nanoparticles for targeted MCF-7 cancer cell imaging. Chem Commun. 2011;47(26):
7323–5.
106. Christensen-Jeffries K, Couture O, Dayton PA, Eldar YC, Hynynen K, Kiessling F, O’Reilly M, Pinton
GF, Schmitz G, Tang MX, Tanter M, Sloun R. Super-resolution ultrasound imaging. Ultrasound Med
Biol. 2020;46(4):865–91.
107. Guo R, Lu G, Qin B, Fei B. Ultrasound imaging technologies for breast cancer detection and manage-
ment: A review. Ultrasound Med Biol. 2018;44(1):37–70.
108. Hoelen CGA, de Mul FFM, Pongers R, Dekker A. Three-dimensional photoacoustic imaging of blood
vessels in tissue. Opt Lett. 1998; 3(8):648.
109. Copland JA, Eghtedari M, Popov VL, Kotov N, Mamedova N, Motamedi M, Oraevsky A.
Bioconjugated gold nanoparticles as a molecular based contrast agent: Implications for imaging of
deep tumors using optoacoustic tomography. Mol Imaging Biol. 2004;6(5):341–9.
110. Yuan Z, Jiang H. Quantitative photoacoustic tomography. Philos Trans A Math Phys Eng Sci.
2009;367(1900):3043–54.
111. Manohar S, Dantuma M. Current and future trends in photoacoustic breast imaging. Photoacoustics.
2019;16:100134.
112. Steinberg I, Huland DM, Vermesh O, Frostig HE, Tummers WS, Gambhir SS. Photoacoustic clinical
imaging. Photoacoustics. 2019;14:77–98.
113. Balasundaram G, Ho CJH, Li K, Driessen W, Dinish US, Wong CL, Ntziachristos V, Liu B, Olivo M.
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 41 Manila Typesetting Company 06/21/2022 09:12AM
42 Ahmad et al.
Molecular photoacoustic imaging of breast cancer using an actively targeted conjugated polymer. Int
J Nanomed. 2015;10:387–97.
114. Kanazaki K, Sano K, Makino A, Shimizu Y, Yamauchi F, Ogawa S, Ding N, Yano T, Temma T, Ono
M, Saji H. Development of anti-HER2 fragment antibody conjugated to iron oxide nanoparticles for
in vivo HER2-targeted photoacoustic tumor imaging. Nanomedicine. 2015;11(8):2051–60.
115. Xi L, Grobmyer SR, Zhou G, Qian W, Yang L, Jiang H. Molecular photoacoustic tomography of breast
cancer using receptor targeted magnetic iron oxide nanoparticles as contrast agents. J Biophotonics.
2014;7(6):401–9.
116. Wang H, Liu C, Gong X, Hu D, Lin R, Sheng Z, Zheng C, Yan M, Chen J, Cai L, Song L. In vivo
photoacoustic molecular imaging of breast carcinoma with folate receptor-targeted indocyanine green
nanoprobes. Nanoscale. 2014;6(23):14270–9.
117. Lee DE, Koo H, Sun IC, Ryu JH, Kim K, Kwon IC. Multifunctional nanoparticles for multimodal
imaging and theragnosis. Chem Soc Rev. 2012;41(7):2656–72.
118. Forte S, Dellas S, Stieltjes B, Bongartz B. Multimodal ultrasound tomography for breast imaging: A
prospective study of clinical feasibility. Eur Radiol Exp. 2017;1(1):27.
119. Han C, Zhang A, Kong Y, Yu N, Xie T, Dou B, Li K, Wang Y, Li J, Xu K. Multifunctional iron oxide-
carbon hybrid nanoparticles for targeted uorescent/MR dual-modal imaging and detection of breast
cancer cells. Anal Chim Acta. 2019;1067:115–28.
120. Song W, Luo Y, Zhao Y, Liu X, Zhao J, Luo J, Zhang Q, Ran H, Wang Z, Guo D. Magnetic nano-
bubbles with potential for targeted drug delivery and trimodal imaging in breast cancer: An in vitro
study. Nanomedicine. 2017;12(9):991–1009.
121. Yang RM, Fu CP, Fang JZ, Xu XD, Wei XH, Tang WJ, Jiang XQ, Zhang LM. Hyaluronan-modied
superparamagnetic iron oxide nanoparticles for bimodal breast cancer imaging and photothermal ther-
apy. Int J Nanomed. 2017;12:197–206.
122. Lim EK, Kim HO, Jang E, Park J, Lee K, Suh JS, Huh YM, Haam S. Hyaluronan-modied mag-
netic nanoclusters for detection of CD44-overexpressing breast cancer by MR imaging. Biomaterials.
2011;32(31):7941–50.
123. Rainone P, Riva B, Belloli S, Sudati F, Ripamonti M, Verderio P, Colombo M, Colzani B, Gilardi MC,
Moresco RM, Prosperi D. Development of 99mtc-radiolabeled nanosilica for targeted detection of
HER2-positive breast cancer. Int J Nanomed. 2017;12:3447–61.
124. Zhang M, Kim HS, Jin T, Yi A, Moon WK. Ultrasound-guided photoacoustic imaging for the se-
lective detection of EGFR-expressing breast cancer and lymph node metastases. Biomed Opt Exp.
2016;7(5):1920.
125. Heidari Z, Sariri R, Salouti M. Gold nanorods-bombesin conjugate as a potential targeted imaging
agent for detection of breast cancer. J Photochem Photobiol B Biol. 2014;130:40–6.
126. Stephen BJ, Suchanti S, Mishra R, Singh A. Cancer nanotechnology in medicine: A promising ap-
proach for cancer detection and diagnosis. Crit Rev Ther Drug Carrier Syst. 2020;37(4):375–405.
127. Chen H, Zhang W, Zhu G, Xie J, Chen X. Rethinking cancer nanotheranostics. Nat Rev Mater.
2017;2:17024.
128. Muthu MS, Feng SS. Theranostic liposomes for cancer diagnosis and treatment: Current development
and pre-clinical success. Exp Opin Drug Deliv. 2013;10(2):151–5.
129. Jokerst JV, Gambhir SS. Molecular imaging with theranostic nanoparticles. Acc Chem Res.
201;44(10):1050–60.
130. Chen F, Ehlerding EB, Cai W. Theranostic nanoparticles. J Nucl Med. 2014;55(12):1919–22.
131. Chen F, Cai W. Tumor vasculature targeting: A generally applicable approach for functionalized nano-
materials. Small. 2014;10(10):1887–93.
132. Khaniabadi PM, Shahbazi-Gahrouei D, Aziz AA, Dheyab MA, Khaniabadi BM, Mehrdel B, Jameel
MS. Trastuzumab conjugated porphyrin-superparamagnetic iron oxide nanoparticle: A poten-
tial PTT-MRI bimodal agent for herceptin positive breast cancer. Photodiagnosis Photodyn Ther.
2020;31:101896.
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 42 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
Volume 39, Issue 6, 2022
Receptor-Targeted Surface-Engineered Nanomaterials 43
133. Choi W il, Lee JH, Kim JY, Heo SU, Jeong YY, Kim YH, Tae G. Targeted antitumor efcacy and
imaging via multifunctional nano-carrier conjugated with anti-HER2 trastuzumab. Nanomedicine.
2015;11(2):359–68.
134. Parhi P, Sahoo SK. Trastuzumab guided nanotheranostics: A lipid based multifunctional nanofor-
mulation for targeted drug delivery and imaging in breast cancer therapy. J Colloid Interface Sci.
2015;451:198–211.
135. Day ES, Bickford LR, Slater JH, Riggall NS, Drezek RA, West JL. Antibody-conjugated gold-
gold sulde nanoparticles as multifunctional agents for imaging and therapy of breast cancer. Int J
Nanomed. 2010;5(1):445–54.
136. Yang J, Lee CH, Park J, Seo S, Lim EK, Song YJ, Suh JS, Yoon HG, Huh YM, Haam S. Antibody
conjugated magnetic PLGA nanoparticles for diagnosis and treatment of breast cancer. J Mater Chem.
2007;17(26):2695–9.
137. Soleymani M, Khalighfard S, Khodayari S, Khodayari H, Kalhori MR, Hadjighassem MR, Shaterabadi
Z, Alizadeh M. Effects of multiple injections on the efcacy and cytotoxicity of folate-targeted mag-
netite nanoparticles as theranostic agents for MRI detection and magnetic hyperthermia therapy of
tumor cells. Sci Rep. 2020;10(1):1–14.
138. Alibolandi M, Abnous K, Sadeghi F, Hosseinkhani H, Ramezani M, Hadizadeh F. Folate receptor-
targeted multimodal polymersomes for delivery of quantum dots and doxorubicin to breast adenocar-
cinoma: In vitro and in vivo evaluation. Int J Pharm. 2016;500(1-2):162–78.
139. Zhou F, Feng B, Yu H, Wang D, Wang T, Liu J, Meng Q, Wang S, Zhang P, Zhang Z, Li Y. Cisplatin
prodrug-conjugated gold nanocluster for uorescence imaging and targeted therapy of the breast can-
cer. Theranostics. 2016;6(5):679–87.
140. Heidari Majd M, Asgari D, Barar J, Valizadeh H, Kal V, Abadpour A, Moumivand E, Mojarrad JS,
Rashidi MR, Coukos G, Omidi Y. Tamoxifen loaded folic acid armed PEGylated magnetic nanopar-
ticles for targeted imaging and therapy of cancer. Colloids Surf B Biointerfaces. 2013;106:117–25.
141. Muthu MS, Kulkarni SA, Raju A, Feng SS. Theranostic liposomes of TPGS coating for targeted co-
delivery of docetaxel and quantum dots. Biomaterials. 2012;33(12):3494–501.
142. Liu R, Xiao W, Hu C, Xie R, Gao H. Theranostic size-reducible and no donor conjugated gold nano-
cluster fabricated hyaluronic acid nanoparticle with optimal size for combinational treatment of breast
cancer and lung metastasis. J Control Release. 2018;278:127–39.
143. Wang L, Zhang H, Qin A, Jin Q, Tang BZ, Ji J. Theranostic hyaluronic acid prodrug micelles
with aggregation-induced emission characteristics for targeted drug delivery. Sci China Chem.
2016;59(12):1609–15.
144. Kim MW, Jeong HY, Kang SJ, Jeong IH, Choi MJ, You YM, Im CS, Song IH, Lee TS, Lee JS, Lee A,
Park YS. Anti-EGF receptor aptamer-guided co-delivery of anti-cancer siRNAs and quantum dots for
theranostics of triple-negative breast cancer. Theranostics. 2019;9(3):837–52.
145. Wang Y, Wang Y, Chen G, Li Y, Xu W, Gong S. Quantum-dot-based theranostic micelles conjugated
with an Anti-EGFR nanobody for triple-negative breast cancer therapy. ACS Appl Mater Interfaces.
2017;9(36):30297–305.
146. Goswami U, Dutta A, Raza A, Kandimalla R, Kalita S, Ghosh SS, Chattopadhyay A. Transferrin-
copper nanocluster-doxorubicin nanoparticles as targeted theranostic cancer nanodrug. ACS Appl
Mater Interfaces. 2018;10(4):3282–94.
147. Muthu MS, Kutty RV, Luo Z, Xie J, Feng SS. Theranostic vitamin E TPGS micelles of transferrin
conjugation for targeted co-delivery of docetaxel and ultra bright gold nanoclusters. Biomaterials.
2015; 39:234–48.
148. Dong Q, Wan C, Yang H, Zheng D, Xu L, Zhou Z, Xie S, Du J, Li F. Targeted gold nanoshelled hybrid
nanocapsules encapsulating doxorubicin for bimodal imaging and near-infrared triggered synergistic
therapy of HER2-positve breast cancer. J Biomater Appl. 2020;35(3):430–45.
149. Ko NR, Naujjaman M, Lee JS, Lim HN, Lee YK, Kwon IK. Graphene quantum dot-based theranos-
tic agents for active targeting of breast cancer. RSC Adv. 2017;7(19):11420–7.
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 43 Manila Typesetting Company 06/21/2022 09:12AM
44 Ahmad et al.
150. Milgroom A, Intrator M, Madhavan K, Mazzaro L, Shandas R, Liu B, Park D. Mesoporous silica
nanoparticles as a breast-cancer targeting ultrasound contrast agent. Colloids Surf B Biointerfaces.
2014;116:652–7.
151. Diaz-Diestra D, Thapa B, Badillo-Diaz D, Beltran-Huarac J, Morell G, Weiner BR. Graphene oxide/
ZnS:Mn nanocomposite functionalized with folic acid as a nontoxic and effective theranostic platform
for breast cancer treatment. Nanomaterials. 2018;8(7):1–18.
152. Pan C, Liu Y, Zhou M, Wang W, Shi M, Xing M, Liao W. Theranostic pH-sensitive nanoparticles
for highly efcient targeted delivery of doxorubicin for breast tumor treatment. Int J Nanomed.
2018;13:1119–37.
153. Li JL, Wang L, Liu XY, Zhang ZP, Guo HC, Liu WM, Tang SH. In vitro cancer cell imaging and
therapy using transferrin-conjugated gold nanoparticles. Cancer Lett. 2009;274(2):319–26.
154. Ma X, Zhao Y, Liang XJ. Theranostic nanoparticles engineered for clinic and pharmaceutics. Acc
Chem Res. 2011;44(10):1114–22.
155. Singh D, Dilnawaz F, Sahoo SK. Challenges of moving theranostic nanomedicine into the clinic.
Nanomedicine. 2020;15(2):111–4.
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
CRT-40686.indd 44 Manila Typesetting Company 06/21/2022 09:12AM
Critical Reviews™ in Therapeutic Drug Carrier Systems
... Further, nanostructure-based molecules can be utilized to effectively deliver the diagnostic, therapeutic, or theranostic agents to the brain. Theranostics are nano-sized agents that offer highly sensitive molecular detection and efficient drug targeting by serving as both diagnostic and therapeutic agents, simultaneously Ahmad et al. 2022;Chauhan et al. 2022;Bajwa et al. 2015). In this chapter, we have discussed various biosensors and biomarkers in AD, advancements in the detection of neurotransmitters, and applications of nanostructure-based molecules in diagnosis and theranostics of AD. ...
... Theranostics also provides sufficient pharmacokinetic information of the targeted drug throughout the treatment and thus offers a huge number of aided advantages over concrete diagnosis or treatment (Ahmad et al. 2022;Jain and Zhong 2022). Various nanostructure-based molecules can be used as carriers for delivering imaging modalities and therapeutic agents to the target sites. ...
Chapter
Alzheimer’s disease (AD), a neurodegenerative disorder characterized by progressive, continuous, and irreversible degeneration of structure and function of neurons, is a major cognitive dysfunction amongst elderly population. Drugs with different pharmacology have been developed and approved for the treatment of AD, but these treatments are not curative and merely provide symptomatic relief. Successful treatment of AD requires drug targeting to the central nervous system in effective therapeutic concentrations. Blood-brain barrier (BBB) is a major hurdle in transporting drugs to the brain. Nanostructure-based materials can cross the BBB by virtue of their very small size. Further, surface modification of these materials could aid in targeting the drugs to specific cells/organs, and to achieve higher drug concentrations at the required site. Nanostructure-based imaging modalities, therapeutic agents, and theranostics provide highly sensitive detection at molecular levels, optimal drug targeting, and combined effect. Recent advancements in the field of nanotechnology have enabled the combined delivery of diagnostic as well as therapeutic agents to remarkably improve the outcomes of AD treatment. In this chapter, we have discussed a brief overview of AD, recent advancements in the detection of neurotransmitters and applications of nanostructure-based molecules in the early detection of AD, targeted nanomedicines, and theranostic nanomaterials in the management of AD. We have also discussed the toxicity concerns with nanostructure-based materials and future perspectives of theranostics as personalized medicine.
... These biomarkers are involved in several molecular pathways associated with angiogenesis in cancer. Various agents, such as specific antibodies, aptamers, small interfering ribonucleic acids (siRNAs), and therapeutic agents, have been developed to target these biomarkers [5][6][7]. ...
Article
Full-text available
Angiogenesis plays a significant role in the human body, from wound healing to tumor progression. "Angiogenic switch" indicates a time-restricted event where the imbalance between pro-and antiangiogenic factors results in the transition from prevascular hyperplasia to outgrowing vascularized tumor, which eventually leads to the malignant cancer progression. In the last decade, molecular players, i.e., angiogenic biomarkers and underlying molecular pathways involved in tumorigenesis, have been intensely investigated. Disrupting the initiation and halting the progression of angiogenesis by targeting these biomarkers and molecular pathways has been considered as a potential treatment approach for tumor angiogenesis. This review discusses the currently known biomarkers and available antiangiogenic therapies in cancer, i.e., monoclonal antibodies, aptamers, small molecular inhibitors, miRNAs, siRNAs, angiostatin, endostatin, and melatonin analogues, either approved by the U.S. Food and Drug Administration or currently under clinical and preclinical investigations.
... Furthermore, the sub-equation Frontiers in Physics frontiersin.org 04 method [19], modified Kudryashov method [20], and F-expansion method [21][22][23] are just a few of the methods that have been used. ...
Article
Full-text available
The fractional-order nonlinear Gardner and Cahn-Hilliard equations are often used to model ultra-short burst beams of light, complex fields of optics, photonic transmission systems, ions, and other fields of mathematical physics and engineering. This study has two main objectives. First, the main objective of this investigation is to solve the fractional-order nonlinear Gardner and Cahn-Hilliard equations by using the modified auxiliary equation method, which is not found in the literature. Second, the exact and approximate solutions of these equations are obtained by utilizing the fractional conformable residual power series algorithm and the modified auxiliary equation method. For the analytical and numerical solutions to two equations, we employ two separate techniques and establish consistency between the precise answers that are derived and the compatible numerical solution. To the best of our knowledge, this method of solving equations has never been investigated in this manner. The 2D and 3D contours have been defined using appropriate parametric values to support the physical compatibility of the results. The assessed findings suggested that the approach used in this study to recover inclusive and standard solutions is approachable, efficient, and faster in computing and can be considered a useful tool in resolving more complex phenomena that arise in the field of engineering, mathematical physics, and optical fiber. KEYWORDS fractional conformable residual power series algorithm, nonlinear partial differential equations, fractional-order nonlinear Cahn-Hilliard equation, modified auxiliary equation method, approximate solution CITATION Ali A, Nigar A, Nadeem M, Jat Baloch MY, Farooq A, Alrefaei AF and Hussain R (2023), Complex solutions for nonlinear fractional partial differential equations via the fractional conformable residual power series technique and modified auxiliary equation method.
... The contrast modalities of MRI are classified as T1 positive, for instance, gadolinium (Gd) chelates, which accelerate the longitudinal relaxation rate and produce brighter image, and T2 negative, which includes superparamagnetic iron that provides darker signal in T2 images by facilitating transverse relaxation rate. Paramagnetic agents such as Gd or manganese can be labeled onto the nanocarrier, while iron oxide nanosystem are innately superparamagnetic that can be tagged over nanocarriers' surface (Shu et al. 2021;Ahmad et al. 2022). The ligand employed in Gd complexes are macrocyclic polyamino carboxylates such as asdiethylenetriamine-penta-acetic acid (DTPA) and 1,4,7,10-tetraazacyclododecane-N, N′,N′′,N′′′-tetra-acetic acids (DOTA) (Yan and Zhuo 2001). ...
Chapter
“Nanotheranostic” is an integrative approach to achieve diagnosis and therapeutic effect simultaneously, using nanocarriers. Nanotheranostic platform can be engineered to overcome biological barriers, to target the therapeutic agent at the required locus, and to support the monitoring of drug delivery. Dendrimers are well-defined 3D globular nanoarchitectured monodisperse system, pliable to precise size control and surface modification, which could serve as a potential nanotheranostic platform to achieve image-guided therapy, distribution monitoring, and drug targeting. Dendrimers exhibit biodegradable, biocompatible, and stimuli-responsive features that could ensure the anticipated biodistribution and efficacy. In this chapter, the utility of dendrimer as a theranostic nanoplatform embodying diversified category of therapeutic, imaging, and targeting moieties has been explored. Dendrimer-based administration of imaging agents (magnetic resonance imaging, computed tomography, single-photon emission computed tomography) along with its application in chemotherapy, pharmacodynamics therapy, and gene therapy has also been discussed.KeywordsDendrimersTheranosticsChemotherapyComputed tomographyMagnetic resonance imaging
Article
Aim: The present research focused on development and optimization of ligand decorated theranostic nanocarrier encapsulating paclitaxel and carbon quantum dots (CQDs). Methods: CQDs were prepared by microwave-assisted pyrolysis and were characterized for particle size and fluorescence behavior. Ligand decorated zein nanoparticles, coloaded with paclitaxel and CQDs, were formulated using a one-step nanoprecipitation method and optimized for various process parameters. Results: Particle size for coated and uncoated nanoparticles was 90.16 ± 1.65 and 179.26 ± 3.61 nm, respectively, and entrapment efficiency was >80%. The circular dichroism spectroscopy showed zein retained its secondary structure and release study showed biphasic release behavior. Conclusion: The prepared theranostic nanocarrier showed optimal fluorescence and desired release behavior without altering the secondary structure of zein.
Article
For more than a decade, researchers have been working to achieve new strategies and smart targeting drug delivery techniques and technologies to treat breast cancer (BC). Nanotechnology presents a hopeful strategy for targeted drug delivery into the building of new therapeutics using the properties of nanomaterials. Nanoparticles are of high regard in the field of diagnosis and the treatment of cancer. The use of these nanoparticles as an encouraging approach in the treatment of various cancers has drawn the interest of researchers in recent years. In order to achieve the maximum therapeutic effectiveness in the treatment of BC, combination therapy has also been adopted, leading to minimal side effects and thus an enhancement in the quality of life for patients. This review article compares, discusses and criticizes the approaches to treat BC using novel design strategies and smart targeting of site-specific drug delivery systems.
Chapter
In the last decade, nanotechnology has shown incredible growth and established itself as valuable technology to prepare highly specific biomedical appliances. Quantum dots (QDs) are one such example of nanotechnological product, which is a crystalline semiconductor material able to produce bright fluorescent light upon excitation. The size of the QDs ranges from 2 to 10 nm, which are mostly made up of inorganic compounds like cadmium (Cd), selenium (Se), tellurium (Te), zinc (Zn), sulphur (S), etc. or organic materials like citric acid, various amino acid, carbohydrates, etc. Contrasting with commonly used organic dyes, it has unique optical and electrical properties that mainly assist in bioimaging, cellular imaging, biosensing, drug delivery, or gene delivery. Functionalization of QDs with various surface modification strategies and bioconjugation with the bioactive molecule enhance its biomedical applications by controlling the cellular toxicity exerted by the core of QDs. In this chapter, we have reviewed the QDs, starting from its method of preparation to derivatives, functionalization strategies, and detailed discussion on its theranostic applications.KeywordsQuantum dotsCarbon quantum dotsCancer imagingNanoparticlesChemotherapyTheranostic nanocarriers
Article
Breast cancer (BC) is one of the primary causes of death among females worldwide. It can affect a woman at any age after puberty, but the risk of developing the disease increases with age. An early diagnosis and the implementation of an appropriate therapeutic strategy are the two most essential aspects in assuring a favorable prognosis for patients diagnosed with any cancer. There has been significant development in cancer immunotherapy over the past few years. It is among the most effective approaches to fighting cancer and boosts the immune system. In the preclinical setting, immunotherapy using checkpoint blockade antibodies and antigen receptor T cells has shown promising results in BC. Despite this, developing safe and effective immunotherapy against breast cancer is challenging because several novel antigens are poorly immunogenic. Regrettably, conventional immunotherapy confronts further obstacles, such as its inability to trigger the anti-tumor response sufficiently. Most tumors have low immunogenicity due to their origin in healthy cells, making it difficult for the immune system to recognize them as foreign invaders. Additionally, the clinical use of immunotherapy for BC has experienced significant drawbacks, including poor immune responses due to insufficient antigen delivery to the immune cells and uncontrolled immune system regulation, which can promote autoimmunity and nonspecific inflammation. To address these challenges, nanomaterial-based immunotherapy has recently emerged as a potent tool against BC. Scientists have been enthralled by the potential of nanomaterial in BC immunotherapy for decades due to its significant benefits over traditional immunotherapy. Over the past few decades, there has been a considerable increase in the research and application of nanomaterial-based antigens/adjuvants in BC immunotherapy. This review focuses on current advances in BC immunotherapy strategies by focusing on recent breakthroughs in nano immunotherapy.
Article
Full-text available
Cancer causes the second-highest rate of death world-wide. A major shortcoming inherent in most of anticancer drugs is their lack of tumor selectivity. Nanodrugs for cancer therapy administered intravenously escape renal clearance, are unable to penetrate through tight endothelial junctions of normal blood vessels and remain at a high level in plasma. Over time, the concentration of nanodrugs builds up in tumors due to the EPR effect, reaching several times higher than that of plasma due to the lack of lymphatic drainage. This review will address in detail the progress and prospects of tumor-targeting via EPR effect for cancer therapy.
Article
Full-text available
Three-dimensional (3D) printing is gaining numerous advances in manufacturing approaches both at macro- and nanoscales. Three-dimensional printing is being explored for various biomedical applications and fabrication of nanomedicines using additive manufacturing techniques, and shows promising potential in fulfilling the need for patient-centric personalized treatment. Initial reports attributed this to availability of novel natural biomaterials and precisely engineered polymeric materials, which could be fabricated into exclusive 3D printed nanomaterials for various biomedical applications as nanomedicines. Nanomedicine is defined as the application of nanotechnology in designing nanomaterials for different medicinal applications, including diagnosis, treatment, monitoring, prevention, and control of diseases. Nanomedicine is also showing great impact in the design and development of precision medicine. In contrast to the “one-size-fits-all” criterion of the conventional medicine system, personalized or precision medicines consider the differences in various traits, including pharmacokinetics and genetics of different patients, which have shown improved results over conventional treatment. In the last few years, much literature has been published on the application of 3D printing for the fabrication of nanomedicine. This article deals with progress made in the development and design of tailor-made nanomedicine using 3D printing technology.
Article
Full-text available
Chemoresistance is one of the major challenges for the breast cancer treatment. Owing to its heterogeneous nature, the chemoresistance mechanisms of breast cancer are complicated, and not been fully elucidated. The existing treatments fall short of offering adequate solution to drug resistance, so more effective approaches are desperately needed to improve existing therapeutic regimens. To overcome this hurdle, a number of strategies are being investigated, such as novel agents or drug carriers and combination treatment. In addition, some new therapeutics including gene therapy and immunotherapy may be promising for dealing with the resistance. In this article, we review the mechanisms of chemoresistance in breast cancer. Furthermore, the potential therapeutic methods to overcome the resistance were discussed.
Article
Full-text available
Overexpression of human epidermal growth factor receptor 2 (HER2) is associated with more frequent cancer recurrence and metastasis. Sensitive sensing of HER2 in living breast cancer cells is crucial in the early stages of cancer and to further understand its role in cells. Biomedical imaging has become an indispensable tool in the fields of early cancer diagnosis and therapy. In this study, we designed and synthesized platinum (Pt) nanocluster bionanoprobes with red emission (Ex/Em = 535/630 nm) for fluorescence imaging of HER2. Our Pt nanoclusters, which were synthesized using polyamidoamine (PAMAM) dendrimer and preequilibration, exhibited approximately 1% quantum yield and possessed low cytotoxicity, ultrasmall size, and excellent photostability. Furthermore, combined with ProteinA as an adapter protein, we developed Pt bionanoprobes with minimal nonspecific binding and utilized them as fluorescent probes for highly sensitive optical imaging of HER2 at the cellular level. More importantly, molecular probes with long-wavelength emission have allowed visualization of deep anatomical features because of enhanced tissue penetration and a decrease in background noise from tissue scattering. Our Pt nanoclusters are promising fluorescent probes for biomedical applications.
Article
Full-text available
Therapeutic monoclonal antibodies have become one of the central components of the healthcare system and continuous efforts are made to bring innovative antibody therapeutics to patients in need. It is equally critical to acquire sufficient knowledge of their molecular structure and biological functions to ensure the efficacy and safety by incorporating new detection approaches since new challenges like individual differences and resistance are presented. Conventional techniques for determining antibody disposition including plasma drug concentration measurements using LC-MS or ELISA, and tissue distribution using immunohistochemistry and immunofluorescence are now complemented with molecular imaging modalities like positron emission tomography and near-infrared fluorescence imaging to obtain more dynamic information, while methods for characterization of antibody’s interaction with the target antigen as well as visualization of its cellular and intercellular behavior are still under development. Recent progress in detecting therapeutic antibodies, in particular, the development of methods suitable for illustrating the molecular dynamics, is described here.
Article
Full-text available
Following its discovery more than 30 years ago, the enhanced permeability and retention (EPR) effect has become the guiding principle for cancer nanomedicine development. Over the years, the tumor-targeted drug delivery field has made significant progress, as evidenced by the approval of several nanomedicinal anticancer drugs. Recently, however, the existence and the extent of the EPR effect-particularly in patients-have become the focus of intense debate. This is partially due to the disbalance between the huge number of preclinical cancer nanomedicine papers and relatively small number of cancer nanomedicine drug products reaching the market. To move the field forward, we have to improve our understanding of the EPR effect, of its cancer type-specific pathophysiology, of nanomedicine interactions with the heterogeneous tumor microenvironment, of nanomedicine behavior in the body, and of translational aspects that specifically complicate nanomedicinal drug development. In this virtual special issue, 24 research articles and reviews discussing different aspects of the EPR effect and cancer nanomedicine are collected, together providing a comprehensive and complete overview of the current state-of-the-art and future directions in tumor-targeted drug delivery.
Article
Nanocarriers are nanostructured vehicles employed to deliver anticancer drugs to the targeted tumor sites in the body. Nanocarriers have been successfully employed to circumvent certain limitations of conventional anticancer drug delivery while providing greater bioavailability, prolonged circulation time and higher tumor accumulation for enhanced therapeutic outcomes in cancer treatment. Nanocarriers are also responsive to functionalization to tailor their pharmaco-kinetics and achieve enhanced therapeutic outcomes in cancer therapy. Among organic, inorganic and hybrid type, several nanocarriers have gained approval for use in cancer patients, while many more are under clinical development. For the last two decades, cancer immunotherapy-based advanced targeting approaches such as monoclonal antibodies, antibody drug conjugates and immune checkpoint inhibitors that utilize human immune system functions have vastly developed which furnish better treatment options in several intractable cancers compared with traditional cancer therapies. This review discusses the imperative role of tumor vasculature in passive and active targeting of anticancer drugs using organic and inorganic nanocarriers and the current research efforts underway. The advanced targeting approaches for treatment of various cancers and their most recent clinical development scenario have been comprehensively explored. Further, potential challenges associated with each type of nanocarrier, and their translational obstacles are addressed.
Article
Introduction The enhanced permeability and retention (EPR) effect serves as the foundation of anticancer nanomedicine design. EPR effect-based drug delivery is an effective strategy for most solid tumors. However, the degree of efficacy depends on the pathophysiological conditions of tumors, drug formulations, and other factors. Areas covered Vascular mediators including nitric oxide (NO), bradykinin (BK), and prostaglandins (PGs) are vital for facilitating and maintaining EPR effect dynamics. Progression to large, advanced cancers may induce activated blood coagulation cascades, which lead to thrombus formation in tumor vasculature. Rapidly growing tumors cause obstructed or suppressed blood flow in tumor vasculature related to embolism or occluded blood vessels. The resulting limited tumor blood flow leads to less drug delivered to tumors, i.e. no or poor EPR effect. High stromal content also suppresses vascular permeability and drug diffusion. Restoring obstructed tumor blood flow and improving tumor vascular permeability via vascular mediators will improve drug delivery and the EPR effect. Physicochemical features of nanomedicines also influence therapeutic outcomes and are vital for the EPR effect. Expert opinion The tumor microenvironment, especially tumor blood flow, is critical for a potent EPR effect. A rational strategy for circumventing EPR effect barriers must include restoring tumor blood flow.
Article
Despite extraordinary advances that have been made in cancer therapy, the number of cancer cases continue to surge, making it the leading cause of death across the world. As a result, early detection is one of the key aspects in the battle against the disease. Screening and early diagnosis play a pivotal role for effective treatment and to lower the cancer mortality rate. Cancer nanotechnology is a new branch in biology that provides a link between nanotechnology and clinical cancer research. Moreover, it also aims to integrate the advancements made in the manufacture of nanoscale devices with cellular and molecular components associated with cancer diagnosis and therapy. Understanding these new technologies is crucial to integrating these practices into clinical settings. This novel approach has facilitated the conjugation of nanoscale devices with agents such as tumor-specific li-gands, antibodies, and imaging probes. This review summarizes the advancements made in nanotechnology based approaches in diagnosing cancer. Coupling of nanoparticles with targeting molecules enables an efficient interaction between biological systems with extraordinary accuracy. The progress associated with nanoscale devices such as metal based nanomaterials, exosomes, magnetic nanoparticles, in addition to quantum dots and lab on chip devices with regard to diagnostic applications has been discussed. We summarize how nanoparticles take advantage of the tumor microenvironment for targeting cancer cells. Further, the review outlines the drawbacks, challenges, and future prospects associated with these techniques as effective strategies to replace current clinical trends.