Download as pdf or txt
Download as pdf or txt
You are on page 1of 24

BioNanoScience

https://doi.org/10.1007/s12668-024-01376-2

REVIEW

Harnessing Nanotechnology for Idarubicin Delivery in Cancer Therapy:


Current Approaches and Future Perspectives
Fatemeh Safari1 · Yeganeh Jalalian1 · Hamidreza Abdouss1 · Mehrab Pourmadadi2 · Payam Zahedi1 ·
Majid Abdouss3 · Abbas Rahdar4 · Sonia Fathi‑karkan5,6 · Sadanand Pandey7,8

Accepted: 25 March 2024


© The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2024

Abstract
The global burden of cancer, which constitutes approximately one-sixth of all worldwide mortalities, poses a significant
challenge for scientific researchers in their pursuit of effective therapeutic interventions. This discourse delves into the
potential of idarubicin (IDA), an anthracycline antibiotic with pronounced lipophilicity, known for its accelerated nuclear
sequestration, enhanced cellular permeability, and potent cytotoxicity. Despite IDA’s demonstrated effectiveness in com-
bating a spectrum of cancers, its therapeutic utility is significantly hampered by the onset of deleterious side effects such
as cardiotoxicity and myelosuppression. The manuscript provides an exhaustive analysis of novel methodologies that have
been developed to circumvent the impediments associated with IDA delivery, encompassing lipid-based nanoparticles (NPs),
polymeric nanocarriers, carbonaceous nanostructures, and inorganic NPs. It examines key parameters such as drug release,
encapsulation efficacy, loading proficiency, zeta potential, along with in vivo and in vitro characterizations, offering a com-
prehensive assessment of the advancements in disparate delivery systems for IDA. Furthermore, the manuscript probes into
co-delivery formulations of IDA in conjunction with other anti-cancer agents, underscoring their promising implications for
future oncological therapy. The discourse also highlights the paucity of empirical research in this domain. The revelations
gleaned from this review are anticipated to make a substantial contribution to the genesis of innovative strategies for future
oncological treatment.

Keywords Idarubicin · Anthracycline antibiotics · Nanoparticles · Drug delivery systems · Co-delivery ·


Nanoformulations · Cancer therapeutics

Abbreviations AF9 5-[2,4-Bis(azanyl)pyrimidin-5-yl]oxy-


4-DMDR 4-Demethoxydaunorubicin 2-methoxy-4-propan-2-yl-benzenesulfon-
A549 Lung carcinoma epithelial cells amide
AML Acute myeloid leukemia

4
* Mehrab Pourmadadi Department of Physics, Faculty of Sciences, University
mehrabpourmadadi@gmail.com of Zabol, Zabol 538‑98615, Iran
5
* Abbas Rahdar Natural Products and Medicinal Plants Research Center,
a.rahdar@uoz.ac.ir North Khorasan University of Medical Sciences,
Bojnurd 94531‑55166, Iran
* Sonia Fathi‑karkan
6
Soniafathi92@gmail.com Department of Advanced Sciences and Technologies
in Medicine, School of Medicine, North Khorasan University
* Sadanand Pandey
of Medical Sciences, Bojnurd 9414974877, Iran
sadanand.au@gmail.com; spandey@shooliniuniversity.com
7
School of Bioengineering and Food Technology, Faculty
1
Department of Polymer, School of Chemical Engineering, of Applied Sciences and Biotechnology, Shoolini University,
College of Engineering, University of Tehran, Tehran, Iran Solan 173229, Himachal Pradesh, India
2 8
Protein Research Center, Shahid Beheshti University, Department of Chemistry, College of Natural Science,
Tehran 1983963113, GC, Iran Yeungnam University, 280 Daehak‑Ro, Gyeongsan,
3 Gyeongbuk 38541, Republic of Korea
Department of Chemistry, Amirkabir University
of Technology, Tehran, Iran

Vol.:(0123456789)

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

AuMPn Gold shell-coated magnetic polyester MPE Maleate-polyester


nanoparticles mTHPP 5,10,15,20-Tetrakis(meso-hydroxyphe-
BILM Biodegradable idarubicin-loaded nyl)porphyrin
microsphere MTT 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphe-
BuCy Busulphan and cyclophosphamide nyltetrazolium bromide
combination NCI-H460 Human non-small cell lung carcinoma
C57BL/6 C57 black 6 NP Nanoparticle
Caco-2 Cancer coli-2 NLCs Nanostructured lipid carriers
CD β-Cyclodextrin NHL Non-Hodgkin’s lymphoma
CD8 Cluster of differentiation 8 NT Normothermia
CR Complete remission PBS Phosphate buffered saline
CNT Carbon nanotube PD1 Programmed cell death protein 1
DDC Dual drug conjugate PEG Polyethylene glycol
DLS Dynamic light scattering PGA Poly(glycolic acid)
DNA Deoxyribonucleic acid PSS Poly(styrene sulfonate)
DNR Daunorubicin POPC Glycerolphospholipid 1-palmitoyl-2-ole-
DSPC Distearoylphosphatidylcholine oyl-sn-glycero-3-phosphocholine
DPPC 1,2-Dipalmitoyl-sn-glycero-3-phospho- ptPVA Pteroic acid enhanced polyvinyl alcohol
choline PDT Photodynamic therapy
DPV Differential pulse voltammetry PTT Photothermal therapy
DXR Doxorubicin PGA Poly (α) L-glutamic acid
D-pen D-penicillamine SCARA5 Scavenger receptor class A member 5
DFT Density functional theory SDDS Smart drug delivery system
DL Drug loading SLNs Solid-lipid NPs
EDTA Ethylenediaminetetraacetic acid TACE Transarterial chemoembolization
EE Encapsulation efficiency TAM Tamoxifen
eNOS Endothelial nitric oxide synthase TfR1 Transferrin receptor protein 1
FA Folic acid TGI Tumor growth inhibition
GDY Graphydine tBA Tert-butyl acrylate
GDYO Graphydine oxide TEM Transmission electron microscopy
GFP Green fluorescent protein UV Ultraviolet
h Hour vdW Van der Waals
HBR Hyperbranched resin
HCC Hepatocellular carcinoma
HER Human epidermal growth factor receptor 1 Introduction
HPLC High-performance liquid
chromatography Cancer, stemming from genetic mutations within cells that
HT Hyperthermia control proliferation, differentiation, and apoptosis [1], has,
HsAFr Horse spleen apoferritin by statistical evidence, emerged as a significant cause of
IC50 Half maximal inhibitory concentration mortality in the USA, ranking second only to cardiovascular
IDA Idarubicin disease [2]. Despite substantial strides in cancer therapies
IgG4 Immunoglobulin G4 over the past five decades, it remains a persistent global
M Molar health conundrum, thereby necessitating the ongoing quest
MCF-7 Michigan Cancer Foundation-7 for innovative treatment methods [3]. Epidemiological
MD Molecular dynamics research underscores the efficacy of cancer cell reduction
MDR Multi-drug resistance by avoiding exposure to smoke, increasing consumption of
MLL Mixed lineage leukemia fruits and vegetables, minimizing sun exposure, enhancing
MMA Methyl methacrylate physical activity, and reducing alcohol and red meat intake
MNP Magnetic nanoparticle [4].
m-PEG-PLGA Methoxy poly(ethylene Current therapeutic interventions for cancer encompass
glycol)-b-poly(L-lactide-co-glycolide) radiotherapy, surgery, immunotherapy, targeted therapy, and
MPBH 4-(4-N-maleimidophenyl)butyric acid chemotherapy [5], all aimed at inhibiting cancer cell growth.
hydrazide However, these modalities often fall short in facilitating

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

complete cellular recovery [6]. However, it is important to to numerous anticancer drugs based on NPs progressing to
note that while targeting cancerous cells, these treatments the experimental phase since 2001 [25].
may inadvertently cause damage to healthy tissues [7]. Sur- At the moment, anthracycline antibiotics, such doxoru-
gical interventions come with an assortment of risks includ- bicin (DXR), epirubicin, daunorubicin (DNR), and IDA, are
ing pain, strength reduction, cognitive issues, and psychoso- some of the most often used anticancer medications. They
cial implications such as depression [8]. Chemotherapy, on are used to treat a variety of solid and medical tumors [26].
the other hand, is associated with a litany of side effects like However, their usage is significantly limited due to severe
hair loss, fatigue, and occasionally, cognitive impairment side effects such as cardiotoxicity and myelosuppression.
[9]. The emergence of resistance to current chemotherapeu- Notably, cumulative dose is the cause of cardiotoxicity, a
tic agents underscores the urgent need for novel, improved common adverse effect of these medications. Anthracyclines
treatment approaches [10]. Current modalities plagued by exert their cytotoxic effects primarily by traversing the cellu-
numerous of drawbacks including side effects, drug resist- lar membrane and forming covalent bonds with DNA pairs,
ance, and inadequate drug delivery to tumor sites, which thereby enhancing their cytotoxic potency [27]. This work,
underscores the pressing need for innovative functional as delineated in Fig. 1, primarily underscores the exploration
methods to circumvent these challenges [11]. of IDA and its varied delivery routes as feasible alternatives
The utility of nanoparticles (NPs) has been explored to conventional anthracycline treatment. The core objective
extensively in recent times, specifically for drug delivery of this study is to unravel methods that mitigate the detri-
purposes, owing to their idealistic size that permits seam- mental side effects associated with IDA use while potentiat-
less interaction with cellular and subcellular biomolecules ing its cytotoxic effect on cancerous cells. Given the limited
[12]. Since most of the drug molecules are at or near the cell empirical research in this area, our aim with this study is to
membrane surface, the uniform characteristics of these par- offer a substantial impetus for the development of innovative
ticles guarantee a large surface area, which is advantageous strategies in future oncological therapy.
for drug delivery. Hence, NPs with larger surface area facili-
tate rapid drug release, while larger particles encapsulate
additional drugs for slower distribution [13]. Nanotechnol- 2 Idarubicin as a Chemotherapeutic Agent
ogy provides plausible solutions for drug delivery, especially
harnessing NPs [14]. Nanotechnology-assisted devices typi- IDA is an antitumor drug primarily used to treat leukemia
cally meet the criteria of having a coverage ranging from 1 and other hematological cancers [28]. As an anthracycline
to 100 nm at the minimum scale [15]. This technology has combined with cytarabine, IDA was discovered by Arca-
found applications across diverse fields, including cancer mone et al. in 1976 [29] and was first used to treat cancer
diagnosis and treatment [16]. in the late 1980s. Since then, IDA has played a significant
In the near future, “targeted drug delivery” systems using role in the treatment of acute myeloid leukemia (AML) and
NPs to carry drugs to cancer cells are expected to gain prom- is presently the subject of extensive clinical study for the
inence. These modern cancer cures and innovations bear the treatment of various types of solid cancer lesions, includ-
potential to be less invasive and more patient-friendly com- ing carcinoma of the breast, plasmacytomas, and lympho-
pared to conventional chemotherapy. Their controlled drug mas [30]. Moreover, IDA has exhibited a remarkably higher
release process can enhance specificity while diminishing antileukemic efficacy compared to other drug combinations,
side effects in patients [17, 18]. Furthermore, enhanced drug such as busulphan and cyclophosphamide (BuCy) [31].
solubility and stability can be achieved via nanotechnology IDA, also referred to as 4-demethoxydaunorubicin
with benefits including optimal drug concentration at spe- (4-DMDR), is derived synthetically from DNR in which a
cific sites, mitigation of off-target effects, and enhancement hydrogen atom is substituted for the C-4 methoxyl group
of drug plasma half-life [19]. NPs provide protective capa- [32]. Having greater lipophilicity that results in more rapid
bilities against degradation caused by physical and chemi- accumulation in the nuclei [33], higher cell-permeability,
cal elements, enabling extended delivery and reduction of and cytotoxicity compared to similar anthracyclines, includ-
toxicity [20]. Additionally, they offer multiple administra- ing DNR and DXR [34], are among the most potent charac-
tion approaches and facilitate drug delivery across critical teristics of this chemotherapeutic agent. Furthermore, IDA
and specific barriers [21]. However, NPs do present with has demonstrated greater effectiveness in treating cancer cell
some disadvantages, primarily linked to the poor biocom- lines that exhibit resistance to DXR, DNR, and aclarubicin
patibility of the nanomaterials employed in their formation [35]. Additionally, IDA possesses lower relative cardiotoxic-
[22]. Various nanocarriers, such as polymers (nanoemul- ity than those of epirubicin and DXR [36].
sions, micelles), lipids (liposomes), and gold and metallic IDA has also undergone considerable evaluation for
NPs, are used for drug administration to cancerous sites [23, AML induction treatment and has been discovered to be
24]. Significant advancements in nanotechnology have led more beneficial than other anthracyclines in improving

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

Fig. 1  A schematic overview of IDA and its main methods of delivery to cancer cell lines

overall survival [37]. For instance, nivolumab, a human chromatography (HPLC) in plasma, urine, saliva, and aque-
IgG4 anti-PD-1 antibody, was added to IDA and cytarabine- ous solutions with mass spectrometry [42] detectors, amper-
based induction therapy for AML in a study conducted by ometric [43], fluorescence [44], and UV [45]. Kurbanoglu et
Ravandi et al. [38]. The results indicated that the IDA-based al. [46] utilized a voltammetric method for examining the
formulation could extend the length of remission, eliminate electrochemical properties of IDA by employing pyrolytic
remaining malignancy, and increase the activity of cyto- graphite electrodes and a multi-walled glassy carbon elec-
toxic T cells. Furthermore, additional reductions of severe trode modified by carbon nanotube (CNT). The objective
graft-versus-host-disease could be more feasible with an was to develop the modified electrodes for the adsorptive
induction therapy based on IDA. Moreover, using IDA has stripping DPV test to determine IDA in the pharmaceutical
shown higher rates of complete remission (CR) in compari- dosage form.
son with DNR [39]. The current paper provides a comprehensive review of the
However, as an essential treatment for cancer therapy, characteristics, pharmacology, and recent developments in
the clinical utilization of IDA has detrimental effects, with the use of IDA in the delivery of drugs for cancer therapy.
cardiotoxicity being the most prominent among them. Moreover, medical uses, side effects, synthesis, and mech-
Therefore, its dosage is constrained and can put cancer anisms to overcome IDA drug resistance are reviewed. A
survivors at higher risk of cardiovascular morbidity and classification of this therapeutic agent is developed based on
mortality [33]. Additionally, topoisomerase II may be lipid-based and polymeric structures, as well as their stimuli-
inhibited by IDA’s non-covalent interactions with DNA, responsive and non-responsive formulations. Furthermore,
which prevent DNA unwinding and nucleic acid synthesis inorganic and carbon-based platforms, alongside co-delivery
[40]. Planar organic compounds, such as IDA, often bind formulations for encapsulating and delivering IDA to malig-
to DNA by intercalation due to their condensed aromatic nant cells, are discussed to comparatively show their advan-
rings [41]. tages and disadvantages. This study provides an overview of
In order to determine IDA and its metabolites, a few the most recent improvements in IDA, confirming it as one
methods have been documented in the literature by using of the most promising anthracyclines for treating different
capillary electrophoresis alongside high-performance liquid cancers, including AML.

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

3 IDA Delivery Systems transfersomes, solid-lipid NPs (SLNs), and nanostructured


lipid carriers (NLCs). Each of these groups possesses its
Nanotechnology has the potential to enable the delivery own set of benefits and drawbacks when used for drug
of IDA in significantly higher doses to cancer cells, pav- encapsulation. For instance, liposomes have the ability to
ing the way for a more powerful cancer treatment. This reduce the toxicity of chemotherapeutic agents and improve
strategy aims to enhance the therapeutic efficacy of the bioavailability but are unstable and difficult to synthesize
medication while minimizing any associated unwanted [53]. SLNs play a crucial role in regulating drug release by
effects by optimizing its delivery [47]. One strategy is to enhancing drug payload and stability, mitigating toxicity
use lipid-based NPs as IDA carriers, composed of bio- [54], and enabling the incorporation of significant quantities
compatible lipids that regulate drug-release properties and of both lipophilic and hydrophilic drugs. The advantages
enhance the medication’s pharmacokinetic characteristics of NLCs are comparable to those of SLNs, but they can
[48]. An alternate methodology involves the use of poly- also overcome some shortcomings including high moisture
meric nanocarriers, which are NPs composed of various concentration, drug leakage, and poor loading capacity [55].
biodegradable and biocompatible polymers. The use of Conventional drug carriers cannot deliver hydrophobic
these nanocarriers allows for the encapsulation of IDA, drugs. In that case, lipid-based NPs can be used instead,
enabling precise and sustained release, hence ensuring due to their lipophilic nature [56]. IDA is a hydrophobic
specific drug delivery while minimizing adverse effects drug [49], and both stimuli-responsive and non-responsive
on the whole system [49]. lipid-based NPs are deployed for the encapsulation and
The other approach entails employing carbon-based application of IDA in cancer therapy. Figure 2 depicts a clas-
nanostructures as carriers for the pharmaceutical agent. sification of lipid-based NPs, frequently employed in drug
The aforementioned structures, such as carbon nanotubes delivery approaches.
and fullerenes, exhibit notable characteristics in the realm
of drug delivery. These features include a substantial drug-
loading capacity, the ability to overcome cellular barriers, 3.1.1 Non‑responsive Nanoparticles Derived from Lipids
and the possibility for functionalization to regulate drug
release [50]. Inorganic NPs, such those composed of iron Few investigations on non-sensitive lipid-based NPs for IDA
oxide or gold, may be used as vehicles for specific intracel- delivery have been conducted. Cancerous cells possess the
lular drug delivery. NPs’ surfaces may be altered to allow capability to develop resistance against chemotherapy agents
for targeted delivery, and their properties can be used for and hinder drug absorption, thereby leading to treatment
concurrent imaging or diagnostic applications in connec- failure of the tumor. This phenomenon is called multi-drug
tion with cancer treatments [51]. Co-delivery platforms resistance (MDR) [57], and ways to overcome this problem
are an alternative approach that involves the simultaneous for cancer treatment must be sought. Ping Ma and cowork-
administration of IDA and other anti-cancer medications ers [58] developed an SLN formulation for IDA and DXR
via a singular nanocarrier. The principal purpose of these encapsulation to overcome MDR in leukemia. The NPs
systems is to optimize the therapeutic efficacy by promot- exhibited a diameter below 100 nm, accompanied by an
ing medication synergy, mitigating multidrug resistance, entrapment effectiveness over 80%. Furthermore, the meas-
and offering complete therapy for intricate and diverse ured zeta potential ranged from around − 5 to − 15 mV. In
cancer types [52]. most cell lines tested, the I­ C50 rates of IDA nanocarriers
The primary objective of these nanosystems is to were comparable to those of free IDA, as demonstrated by
enhance the cytotoxicity of IDA towards cancer cells in vitro studies. In the P388 animal model studied in vivo,
while mitigating its adverse effects, therefore augmenting IDA NPs was comparable to free IDA, but different in the
the overall efficacy of cancer therapy. A more thorough HCT-15 animal model.
examination of each drug delivery technique, including Gian Paolo Zara [59] invented an SLN-based IDA formu-
its benefits, underlying mechanisms, and potential applica- lation to treat cancerous cells in rats. The objective of this
tions in the treatment of cancer, is provided in the sections study was to enhance the bioavailability of the pharmaceuti-
that follow. cal compound. The size of the drug particles ranged from
around 70 to 90 nm, while the zeta potential was approxi-
mately 39.5 mV. Moreover, the release rate of the medica-
3.1 Lipid‑Based Nanoparticles tion was found to be less than 1% after a duration of 2 h. The
experiments indicated that administering IDA-SLN duode-
Lipid-based NPs fall into many groups according on nally resulted in improved tissue distribution and a decrease
their properties; they include liposomes, niosomes, in the release rate of IDA.

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

Fig. 2  Different subtypes of lipid-based NPs commonly used for drug encapsulation

In another work, Pakawadee Sermsappasuk [60] con- found to be − 19.31 mV and − 50.33 mV, respectively. Con-
ducted a synthesis of a liposomal formulation for the pur- sequently, there was a substantial increase in cellular absorp-
pose of encapsulating IDA. This was achieved by using tion and medication efficacy. The observed phenomenon
cholesterol, 1,2-distearoylsn-glicero-phosphocholine, and is likely attributed to trastuzumab-conjugated liposomes,
1,2-distearoyl-sn-glicerophosphoethanolamine-N- [poly which demonstrate a greater ability to target malignant cells
(ethylene glycol)2000. The objective of this study was compared to free IDA, as seen in Fig. 3. Table 1 provides a
to assess the use and limitations of liposomal IDA. The comprehensive overview of formulations based on lipids that
liposomes containing drugs exhibited a mean diameter of exhibit limited reactivity and are employed for the delivery
around 100 nm, accompanied by an entrapped efficiency of of IDA in the domain of cancer treatments. The application
approximately 90%. Additionally, the absorption of the free of these formulations holds significant promise in enhanc-
IDA decreased to 20%, and the new formulation exhibited a ing the efficacy of IDA by improving its targeted delivery,
decrease in coronary vasoconstriction. thereby enhancing therapeutic outcomes while mitigating
The human epidermal growth factor receptor (HER) is a non-specific effects.
significant approach used in the field of cancer therapy and
detection. This receptor plays a crucial role in regulating 3.1.2 Stimuli‑Responsive Nanoparticles Derived
cellular development and viability. The family unit com- from Lipids
prises four main individuals identified as HER-1, HER-2,
HER-3, and HER-4 [61]. Mansour Amin and coworkers [62] The performance of stimuli-responsive lipid-based NPs
designed two formulations, namely immunoliposomes and may be adjusted by altering certain parameters, such as
liposomes, for the targeted delivery of IDA to breast can- temperature and pH. This section primarily focuses on cur-
cer cells expressing the HER2 receptor. The dimensions of rent research concerning stimuli-responsive lipid-based
the immunoliposomes and liposomes were measured to be nanoparticles utilized for the transportation of IDA [64]. Tao
156.5 ± 2.7 nm and 169.5 ± 3.3 nm, respectively. The average Lu and colleagues [65] developed a novel IDA formulation
zeta potential of the immunoliposomes and liposomes was that was a combination of thermosensitive liposomes and

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

Fig. 3  This schematic representation depicts the methodical deliv- tion of each treatment was 4 h, and it was followed by a 72-h period
ery of IDA through liposomes coupled with trastuzumab to breast of incubation during which the assessment of cell viability was con-
tumor cells that excessively express the HER2 receptor. To ascertain ducted. Significant alterations in cellular viability were seen in all
the impact of various IDA formulations, the viability of the MCF-7 instances, as compared to both the control groups that were not sub-
and SK-BR-3 cell lines was assessed. In a series of three consecutive jected to any treatment and the MCF-7 cell line. Reprinted from ref.
tests, the cells were subjected to treatment with conventional IDA, [62] with permission from Elsevier, copyright 2018
liposomal IDA, and immunoliposomal IDA, respectively. The dura-

hyperthermia (HT) with the goal of enhancing IDA retention These formulations had a molar ratio of about 5 to 1 (lipid
and drug release in different temperatures. The size of the to drug) at pH 7, and the size of these particles was around
synthesized liposomes was around 85 to 130 nm. Moreover, 10 nm, with the drug entrapment efficiency of 87%. The
the drug entrapment efficiency was around 100%, and the observed nanoformulation demonstrated pH-responsive
zeta potential was between − 5 and − 9 mV. In vitro studies characteristics, leading to an accelerated drug release rate
showed low leakage at 37 °C for 1 h, whereas at 42 °C, the under acidic conditions (pH > 7). Moreover, experiments
experiments indicated an ultra-fast and total induced release showed that these formulations caused better uptake of drugs
of the drug. In addition, IDA-TSL was as cytotoxic as the and indicated more cytotoxicity in leukemia cells compared
free drug to tumor cells at 42 °C. However, the cytotoxic- to free drugs.
ity was found to be less at 37 °C. In vivo studies indicated Nancy Dos Santos and coworkers [67] synthesized pH-
high drug absorption by tumor cells, and studies using mice responsive liposome formulations to improve antitumor
models revealed that the combination of mild HT and IDA- activities. The objective of this research was to elevate the
TSL inhibited tumor growth and caused more cell survival plasma levels of IDA and encapsulate the drug in liposomes
compared to free HT and IDA. to prolong its circulation time. The novel compositions were
To increase the effectiveness of delivering IDA and DXR prepared using a molar ratio of about 5 to 1 (lipid to drug)
into tumors, Yongzhong Wang [66] developed a method to in an extracellular environment at pH 7.4 and an intracellu-
deliver the drug by using PazPC, an oxidized phospholipid. lar environment at pH 4. Furthermore, the drug entrapment

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

Table 1  Non-responsive lipid-based formulations for IDA-loaded cancer therapeutics


Formulation Particle size Zeta potential EE (%) DL (%) Release Characterization Case study Reference
(nm) (mV)

SLN-IDA 100 − 5 to − 15 > 80 10 100% in 6 h In vivo and Leukemia P388 [63]


in vitro and HCT-15
mouse model
SLN-IDA 80 ± 10 39.5 5 98 < 1% at pH 7.4 In vitro Rats [59]
in 2 h
Liposome-IDA 100 N/A 90 N/A N/A In vivo Rat heart [60]
Liposome-IDA 169.5 ± 3.3 − 50.33 N/A N/A N/A In vivo and HER2-positive [62]
in vitro breast cancer-
ous cell line
(SK-BR-3 and
MCF-7)
Immunolipo- 156.5 ± 2.7 − 19.31 N/A N/A N/A In vivo and HER2-positive [62]
some-IDA in vitro breast cancer-
ous cell line
(SK-BR-3 and
MCF-7)

efficiency of these formulations exceeded 95%. Experi- existence of PEG was an important factor in cholesterol-free
ments indicated the best formulation was achieved when liposomes, and it caused improvement in circulation lon-
the levels of polyethylene glycol (PEG)-lipid incorporation gevity. Additionally, this formulation exhibited an improved
increased under iso-osmotic conditions which caused more medication retention rate in comparison to liposomes con-
IDA release. In vivo, research conducted on some mice indi- taining cholesterol. The release of entrapped IDA (present
cated that compared to free IDA, the liposome formulation in both unbound and precipitated forms) from the aqueous
increased the lifespan. It was observed that the new formula- core of a liposome into the surrounding medium is depend-
tion improved the circulation longevity and antitumor activi- ent on the drug’s partitioning properties, as seen in Fig. 4.
ties in the drug but was ineffective in multidrug resistance. Table 2 presents a complete summary of several lipid-based
Jerzy Gubernator [68] developed a pH-sensitive liposome NPs that exhibit responsiveness to stimuli and are used for
formulation by utilizing a novel approach that dissolves the the purpose of delivering IDA to cancer cells. The NPs in
medicine in EDTA salt solutions. The present formulation question have been purposefully created and programmed to
demonstrated an extended retention period and was specifi- respond to certain triggers or events, hence increasing their
cally designed to inhibit the release of the medication from capacity to efficiently transport IDA to cancer cells that have
the PEGylated liposomes, which also contained cholesterol. been carefully targeted.
The primary objective of the study was to develop a novel
and stable liposomal formulation with the aim of decreasing 3.2 Idarubicin Polymeric Delivery Systems
the dissolution rate of IDA in an in vivo setting. The size of
these liposomes was between 95 and 120 nm, and they had a Due to anthracyclines’ ability to permeate across cell mem-
zeta potential ranging from 2 to 4.5 mV. Moreover, the drug branes, target topoisomerase II, and intercalate between
entrapment efficiency of these formulations was around 98% DNA base pairs, cytotoxicity is an inevitable barrier inhib-
with a 0.2 molar ratio of drug to lipid. This method indicated iting wider clinical use of idarubicin as an anticancer drug
higher stability and plasma level compared to other encap- [76]. Because of its much greater lipophilicity—which
sulation methods for IDA. results from the aglycone’s D ring missing a methoxy group
In another study, Nancy Dos Santos [69] developed a at position 4—IDA shows higher cytotoxicity in comparison
new cholesterol-free liposomal formulation to encapsulate with DXR and DNR [63]. IDA’s proper encapsulation is a
IDA. The formulation consisted of distearoylphosphati- crucial aspect of an effective application of this therapeutic
dylcholine (DSPC): distearoylphosphatidylethanolamine agent in cancer treatment. The most relevant polymeric car-
(DSPE)-PEG2000 liposomes. The liposomes that were pre- riers for achieving this purpose are included in this section.
pared exhibited an average size of around 110 nm, and a Utilizing polymeric NPs as drug carriers is a suitable
significant proportion of the encapsulated medication was approach since they possess certain beneficial features
released during a 15-min period at a temperature of 37 °C, such as biodegradability, stability, and the state of not
exceeding 95% in quantity. Experiments revealed that the being able to inflame. Another prominent characteristic of

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

Fig. 4  It is possible to control the drug partitioning process within bility is dependent on how the drug partitions; this behavior may vary
liposome membranes by using pH gradients and crystalline precipita- depending on a number of parameters, including pH, membrane sur-
tion development. The protonated (BH +) and unprotonated (B) forms face charge, and the particular characteristics of the lipid acyl chains.
of IDA enter a steady state of equilibrium as they move from the Reprinted from ref. [62] with permission from Elsevier, copyright
encapsulating precipitate inside the liposome’s aqueous core, over the 2002
bilayer interface, and finally through the bilayer itself. IDA’s availa-

these NPs is the crucial quality of nontoxicity [70], which 3.2.1 Non‑responsive Nanoparticles Derived
makes the use of these agents less hazardous for the body from Polymers
than that of other particles. These particles range in size
from 20 to 250 nm, with their surface consisting of non- Insensitive polymer-derived NPs are a promising vehicle
fouling water-soluble polymers, namely dextran and PEG for the delivery of anticancer drugs. In light of this, Liang
[71]. Polymeric NPs can be deployed in two different ways et al.’s study [49] examined the antileukemic effects and cell
to deliver drugs to the tumor site or inside the infected absorption of polymeric NPs loaded with IDA. In this work,
cells. Either the anticancer drug is joined to the NP’s sur- an amphiphilic diblock copolymer (m-PEG-PLGA) was
face or is enclosed and preserved in the polymeric bed. developed from methoxy poly(ethylene glycol)-b-poly(L-
Various physiochemical structures of these particles con- lactide-co-glycolide). The MLL-AF9-induced antileukemic
tribute to a diversity of morphologies and compositions in activity was put to the test using IDA-loaded mPEG-PLGA
the polymeric core and periphery, as illustrated in Fig. 5. A NPs in mice. The study discovered that IDA/mPEG-PLGA
variety of biological barriers can be surmounted by poly- NPs displayed exceptional encapsulation efficiency (EE)
meric NPs, which release the ideal dosage of the intended and drug loading (DL) values. It was suggested that PLGA’s
therapeutic payload. The functionalization of these medi- hydrophobic segments, which provide sufficient hydropho-
cations helps to target the surface antigens of tissues and bic force to encase a substantial IDA payload in the mPEG-
cells with a ligand (such as an aptamer, peptide, antibody- PLGA NPs’ cores, were responsible for the high EE and DL
antibody fragment, or small molecule) to reduce the agent’s values. Results also revealed that IDA/mPEG-PLGA dis-
nonspecific dispersion and increase the duration of blood played lower I­ C50 in leukemia blasts than pure IDA. Addi-
residence time [72, 73]. tionally, the cell viability of IDA/mPEG-PLGA was found

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

Table 2  Stimuli-responsive lipid-based formulations for IDA-loaded cancer therapeutics


Formulation Particle size Zeta potential EE (%) DL (%) Release Characteriza- Case study Reference
(nm) (mV) tion

Thermosensi- 85–130 − 5 to − 9 100 30 ~ 90% at pH In vivo and Hyperthermia [70]


tive liposome 5.5 and 42 °C in vitro (HT)
loaded with in 1 h
IDA (IDA- ~ 20% at pH
TSL) 5.5 and 37 °C
in 1 h
PazPC-IDA 10 − 24.3 ± 3.7 87 14.8 ~ 62.5% at pH In vitro Leukemia P388 [71]
6.5 in 10 h and P388/ADR
Cholesterol-free N/A N/A > 95 N/A N/A In vivo and Mice P388/ [72]
liposomes in vitro ADR and
loaded with MDA435LCC6/
IDA MDR1
PEGylated 95–120 2–4.5 98 N/A ~ 98% at pH 4 In vivo and Mice BALB/c [73]
cholesterol- and 37 °C in in vitro
containing 24 h
liposomes
loaded with
IDA
DSPC 110 ± 30 N/A N/A > 95 > 95 at 37 °C in In vivo Balb/c mice [74]
PEGylated 15 min
cholesterol-
free liposomes
loaded with
IDA
IDA-SDDS N/A N/A 13.0 ± 3.6 N/A 100% at 42 °C In vitro eNOS-Tag-GFP [75]
in 1 h mice

Fig. 5  A classification of polymer-based nanoformulations for drug delivery according to their physiochemical structures

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

to be higher than that of pure IDA, showing higher efficacy for branching, and ricinoleic acid for termination. In contrast
both in vivo and in vitro. The research demonstrates excel- to IDA, which had a chemical bond with the resin, TAM had
lent release properties of mPEG-PLGA for encapsulation of a physical one, according to the study results. The release
IDA chemotherapeutic agent to treat AML. velocity in the phosphate buffer was found to be boosted by
In one study, Jain, Ratnesh, et al. [74] modified the outer- triggering factors like Pseudomonas sp. lipase and sodium
most layer of propyl starch NPs laden with IDA in an attempt dodecyl sulfate. Conversely, an increase in loading concen-
to increase the anti-cancer agent’s intracellular absorption. trations was shown to lead to a reduction in release veloc-
The pteroic acid-enhanced polyvinyl alcohol (ptPVA) was ity. Comparing the administration of pure medications with
synthesized and afterwards used in the NP formulation as drug-loaded NPs showed significantly higher cell death rates
a stabilizing agent. The NPs treated with ptPVA exhibited in the cytotoxicity assays performed on MCF-7 breast cancer
enhanced protein adsorption, resulting in heightened cel- cells. Particles devoid of medicinal materials, on the other
lular internalization. The results of protein binding assays hand, showed no discernible toxicity. Table 3 provides an
demonstrated a greater degree of adhesion of the model pro- overview of non-responsive polymeric systems used in can-
teins onto NPs formed with ptPVA in comparison with those cer therapy.
prepared with PVA. The cellular uptake tests provided con-
firmation of the enhanced cellular uptake of NPs stabilized 3.2.2 Stimuli‑Responsive Nanoparticles Derived
by ptPVA (Fig. 6). from Polymers
The study conducted by Güç, Gündüz, and Gündüz [75]
focused on the use of nano-scale dendrimer/hyperbranched The term “stimuli-responsive” generally refers to environ-
polymer for drug delivery purposes. A novel hyperbranched ment-sensitive nanocarriers that undergo swelling, disinte-
resin (HBR) based on fatty acids was developed with the gration, or collapse in response to either an external pro-
particular purpose of facilitating the transfer of Tamoxifen vocative agent (such as temperature, light, magnetic field,
(TAM) and IDA. The HBR is constructed using dipentae- or ultrasound) or an internal stimulant (such as lysosomal
rythritol as the core component, dimethylolpropionic acid digestive enzymes, glucose, and redox potential). Recently,

Fig. 6  A Propyl starch NPs were investigated for their ability to have been treated with ptPVA in HT-29 cells. The red fluorescence
bind proteins, with bovine serum albumin serving as a representa- highlights the cell membranes, which have been stained with RRCA,
tive protein in the studies. PVA (identified as PVA 1–3) and ptPVA while the blue fluorescence depicts the cell nuclei, which have been
(also referred to as ptPVA 1–3) were used to create three distinct stained with DAPI. After a period of 2 h (C), the NPs start the pro-
batch of propyl starch NPs. B The given data illustrates the release cess of binding to the cell membrane, and this binding activity inten-
pattern of indocyanine green (IDA)-loaded propyl starch NPs modi- sifies after 4 h (D) with an increase in uptake. Conversely, there was
fied with ptPVA in an in vitro setting. The graph shown illustrates the no absorption of the propyl starch NPs made using unmodified PVA
pattern of release during a 24-h period. The present study illustrates (E). Reprinted from ref. [74] with permission from Elsevier, copy-
the investigation of the cellular absorption of propyl starch NPs that right 2011

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

Table 3  Non-responsive polymer nanoformulations for IDA encapsulation


Formulation Particle size Zeta potential EE (%) DL (%) Release Characteriza- Case Study Reference
(nm) (mV) tion

IDA/mPEG- 81.2 ± 3.4 − 34 ± 3.6 93.95 ± 3.2 8.59 ± 0.96 80.2 wt% at In vivo and MLL-AF9- [49]
PLGA 37 °C in in vitro induced
40 days, PBS murine
as the release leukemia
medium
IDA-loaded 218.5 ± 6.12 − 5.54 ± 0.88 84.2 ± 3.23 82 ± 4 46.7 ± 3.2%. In vitro HT-29 [74]
propyl starch at 37 °C in
NPs modified 7 days
with ptPVA
HBR-IDA 217.1 ± 5.6 − 41.4 ± 1.6 74 59.6 ± 4.3 < 0.7% at pH In vitro MCF-7 [75]
7.4, 37 °C in
67 days

these groups of NPs have garnered considerable interest compared to unconjugated IDA, it was found that the dual
because of their enhanced capacity to promote drug release drug conjugate (DDC) considerably increased drug exposure
at the malignant spot (spatial control) or at the appropri- by a factor of seven and extended the half-time of plasma
ate moment (temporal control) [77]. Recent research has circulation. Additionally, significant growth in tumor accu-
revealed a groundbreaking group of polymeric nanomedi- mulation was seen, which resulted in 89% tumor growth
cines that are bio-sensitive and adept at delivering drugs to inhibition (TGI) as opposed to 60% with unconjugated IDA.
specific sites with remarkable efficiency. These nanocarriers This resulted in a 17% enhancement in NCI-H460 tumor
maintain stability in extracellular environments and circula- viability in mice models (Fig. 7) [79].
tion, effectively addressing the challenge of balancing sta- Blaudszun et al. investigated cell-mediated drug deliv-
bility and drug release within polymer-based carriers [78]. ery that utilizes living cells to transport and dispense
According to research conducted by Wadhwa et al., the the medication, with the aim of minimizing premature
group used pH-responsive hydrazone bonds and disulfide cell death. To this end, the biocompatible poly(lactic-
to covalently bind IDA and D-penicillamine (D-pen) to the co-glycolic-acid) (PLGA) and maleate-polyester (MPE)
peripheral chain of poly (α) L-glutamic acid (PGA). When NPs were encapsulated with IDA and the effects of the

Fig. 7  A An illustration demonstrating the steps involved in forming rescence images, correspondingly, after 4 h (upper) and 12 h (lower)
an IDA-MPBH. B Examination of the PGA and DDC circular dichro- of treatment. D Evaluation of potential anticancer effectiveness using
ism spectra. Prior to coupling, the spectral observations are compared the NCI-H460 tumor model. A tumor development trajectory in
with the DDCs at room temperature (25 °C) and around 50 M PGA athymic nu/nu mice fed a bi-daily, three-dose regimen of NCI-H460
level. The figure shown is the average of two separate investigations. xenografts (50–100 mm.3). Reprinted from ref. [79] with permission
C The observation of HL-60 cells using confocal microscopy during from Elsevier, copyright 2012
DDC administration, displaying clear interference contrast and fluo-

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

stimuli-responsive and active medication were examined loading efficiency of 7.15% and an entrapment efficiency of
on human T cells. To further assess the uptake, T cells 84.7%. The MTT viability test was used to assess the anti-
were treated with the poly(styrene sulfonate) (PSS)-com- cancer effects of free and HsAFr-IDA on MCF-7 cancer cells
plex of 5,10,15,20-tetrakis(meso-hydroxyphenyl)porphyrin in humans [83]. An overview of the many stimuli-responsive
(mTHPP). The results revealed that MPE-IDA NPs were polymeric formulations used to administer IDA therapies is
more effective at preventing IDA toxicity than PLGA-IDA given in Table 4.
NPs in T lymphocytes, and PSS/mTHPP loaded lympho-
cytes could survive for 4 days minimum unless they are cul-
tivated in the light [80]. 3.3 Carbon‑Based and Inorganic Delivery Systems
Another study by Nafee et al. [81] proposed that employ-
ing pH-triggered polymeric NPs, star polymer deployment, The potential of carbon-oriented delivery systems to effi-
might help with chemotherapy. β-cyclodextrin (CD) formed ciently carry medications for the purpose of cancer treatment
the hydrophilic center of the amphiphilic star polymer is attracting wide attention. These structures, renowned for
known as MMA-tBA CD. The hydrophobic arms, moreo- their unique attributes, such as a vast surface area, thermal
ver, were made using tert-butyl acrylate (tBA) and methyl conductivity, and potent penetrative abilities, are an excel-
methacrylate (MMA). The resulting polymer exhibited a lent candidate for focused drug delivery, especially the con-
slightly negative zeta potential and varied in size from 130 veyance of IDA [84]. Researchers have been successfully
to 200 nm. When loaded with CD-NPs, the polymers were encapsulating IDA with the carbon-derived nanocarriers
observed to release their payload over a period of 48 h, with with an aim to lower the drug’s toxicity and enhance its anti-
an initial burst of 40%. Additionally, polymeric magnetic cancer properties [85]. The encapsulation process typically
NPs (MNPs) have recently drawn attention in cancer therapy involves a strategic disassembly and reassembly approach
due to their favorable properties, including reduced agglom- that maintains the tertiary structure of the carrier protein,
eration, improved biocompatibility, and cellular uptake. The thereby preserving its ideal function [86]. Groundbreaking
exterior section of these NPs consists of polymers, while research in this realm has yielded commendable results.
a magnetic core serves as the inner section. As a poten- For instance, investigations indicate that deploying these
tially effective delivery approach for the treatment of breast carbon-focused nanocarriers for IDA delivery attains high
cancer, Gunduz et al. [82] reported encapsulating IDA in loading and entrapment efficiencies [87]. Furthermore, vari-
PEG-coated, folic acid (FA) linked MNPs in MCF-7 cells. ous studies deployed molecular dynamics (MD) modeling
Researchers found that compared to pure IDA, there would to ascertain the graphydine oxide nanosheet’s ability to load
be an increase in cytotoxicity when using IDA-loaded FA- the IDA medication. The lowering van der Waals (vdW)
PEG-MNPs. Exposure of IDA-loaded magnetically sensitive interaction energy of IDA and graphydine oxide nanosheet
NPs to magnetic fields resulted in a successful uptake by explains their strong attraction, suggesting that the latter is a
MCF-7 cells. suitable carrier for IDA delivery. The simulation also looks
To conquer the rapid elimination of the therapeutic agent at how the loaded IDA-graphydine oxide nanosheet combi-
into the cardiovascular system using transarterial chem- nation penetrates the biomembrane, and it shows that IDA
oembolization (TACE) locoregional therapy, Zheng et al. adsorbed molecules continue to be conjugated with graphy-
[28] employed gelatin and carrageenan to prepare biode- dine oxide [88].
gradable microspheres capable of being loaded with IDA In order to encapsulate IDA, Lu et al. [89] created a smart
(BILMs), resulting in IDA-MS to treat hepatocellular car- drug delivery system (SDDS) that used 1,2-distearoyl-sn-
cinoma (HCC). TACE with IDA-MS significantly reduced glycero-3-phosphocholine (DSPC) and phospholipid-based
tumor growth while posing no additional significant adverse heat-triggered 1,2-dipalmitoyl-sn-glycero-3-phosphocho-
effects beyond those seen in the IDA group, according to line (DPPC). In both in vivo and in vitro settings, the drug
research conducted on an HCC model in mice and rabbits. release, biodistribution, and cell uptake of IDA-loaded
Furthermore, by promoting CD8 + T cell expression and delivery systems were evaluated and compared with those
boosting the effectiveness of anti-PD1 immunotherapy, of DXR. The findings demonstrated that as opposed to
TACE with IDA-MS improved the HCC tumor immuno- DXR-SDDS, IDA-SDDS exhibited quicker release and 10
logical environment (Fig. 8). Additionally, Ronak Rafipour times higher cellular uptake evaluated in vitro. Furthermore,
and her colleagues have utilized protein-based nanocarriers IDA appeared to have reduced drug redistribution and four
for carrying IDA with the goal of mitigating toxicity and times more absorption of entire tumor drugs than DXR
enhancing the drug’s anticancer properties. This procedure in vivo, collectively leading to improved therapeutic effi-
entailed demounting and remounting horse spleen apofer- cacy. According to the research, DXR-SDDS exhibits greater
ritin (HsAFr) in the presence of IDA, without modifying the tumor absorption and content release efficiency in vivo when
tertiary structure of the protein. The group achieved a drug heated at high temperatures (Fig. 9).

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

Fig. 8  IDA-MS reduced tumor


development and improved anti-
PD1 efficacy in the H22 carci-
noma model. A The plan of the
carried-out research. B Tumor
images taken from the IDA,
IDA-MS, IDA + PD1, and IDA-
MS + PD1 cohorts. Size (C)
and assessment of each group’s
subcutaneous lesions’ mass (D).
E PET scans demonstrating the
tumor-ridden animals in each
group. F Differences in the
maximum ID%/g. From Ref.
[28], used under Creative Com-
mons CC-BY license

Via deploying MD simulation, graphydine oxide research on assessing the transfer efficacy of graphydine
(GDYO) nanosheets were found to be useful for loading nanosheets loaded with IDA, using MD simulation and den-
IDA [88]. As a membrane model, Shahabi and Raissi exam- sity functional theory (DFT) [90]. The IDA molecules and
ined the use of graphydine (GDY) to transport IDA in an the nanosheet interacted weakly, with an adsorbed strength
environment of glycerolphospholipid 1-palmitoyl-2-ole- value of 81.305 kJ/mol, according to the Non-covalent
oyl-sn-glycero-3-phosphocholine (POPC). The primary Interaction technique of DFT calculation. The results imply
reason for decent IDA adsorption on GDYO nanosheets that applying electric fields in the xy and z directions may
was associated with van der Walls interaction energy, as it enhance the contact between the adsorbed medication and
was negatively changed during the first 12 ns of simulation the porous framework of the nanosheet by raising the vdW
run time. Results also showed that the stable IDA-GDYO interaction energy. The findings support those of the earlier
complex proceeded towards the POPC membrane sponta- research and demonstrate that the hydrophobic characteris-
neously at the early stages of simulation, demonstrating tics of the graphene nanosheet lead to a higher vdW interac-
GDYO nanosheets as highly potential carriers for IDA at the tion’s energy value. Table 5 summarizes the aforementioned
near cell membrane. Shahabi and Raissi carried out similar carbon-based formulations and their major characteristics.

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

Table 4  Stimuli-responsive polymeric formulations for IDA-loaded cancer therapeutics


Formulation Particle size Zeta potential EE (%) DL (%) Release Characterization Case study Reference
(nm) (mV)

DDCs from IDA N/A N/A N/A N/A 90% at pH 5.2, In vivo and NCI-H460 [79]
and D-pen 10 mM glu- in vitro
conjugated to tathione, and
PGA in 14–16 h
< 10% at pH 7.4
in 16 h
PLGA-IDA 215.8–9.6 − 20.57 ± 0.76 92.6–0.2 2.26 50% at pH 7.4 in Ex vivo Human T lym- [80]
48 h phocytes
MPE-IDA 226.8–7.1 − 1.88 ± 0.28 81.4–1.34 1.97 15% at pH 7.4 in Ex vivo Human T lym- [80]
24 h phocytes
MMA-tBA CD 130–200 0 to − 2 ~ 40 N/A 80% at pH 7.4 in In vitro Caco-2 and [81]
star copoly- 48 h A549
mers
FA–PEG–MNP 10–40 N/A 36.1 ± 3.4 N/A ~ 80% at pH 5.4 In vitro MCF-7 [82]
in 12 h
~ 60% at pH 7.0
in 12 h
IDA-MS from 1.5–2.9 e + 5 N/A 99.12 N/A 38.4% in 0.9% In vivo and VX2 rabbit and [28]
gelatin and NaCl solution in vitro C57BL/6 mice
carrageenan- in 12 h HCC
synthesized
BILMs
HsAFr-IDA 6–8 N/A 84.75 7.15 Through interac- In vitro MCF-7 [83]
tion with the
SCARA5 and
TfR1 receptors

3.4 Inorganic Formulations interactions with cell membranes, and drug delivery.


Rafipour et al. [83] used HsAFr as nanocages for IDA and
Researchers have formulated IDA into inorganic medica- examined their efficacy on human breast cancer tumors
tions or formulations to enhance its therapeutic potential (MCF-7). Encapsulation efficiency (EE), as a parameter to
and delivery. These formulations, such as IDA-loaded NPs, evaluate drug-loading capacity, was found to be 84.75%,
utilize the advantages of inorganic materials, including showing high compatibility between IDA and the protein.
enhanced stability, controlled release, and targeted deliv- Additionally, it was determined that IDA and HsAFr-IDA
ery, in addition to possessing a high surface-to-volume ratio. had ­IC50 values of 5.57 and 1.6 ng/mL, correspondingly,
By improving the drug’s solubility and cellular absorption, which led to increased toxicity in MCF-7 cells. To opti-
this approach may boost therapeutic effectiveness. Further- mize antileukemic synergy and minimize cardiotoxicity in
more, by carefully controlling the NPs’ size and form, one patients with acute promyelocytic leukemia (APML4), a
may significantly influence their distribution and clearance combination of IDA, arsenic trioxide (ATO), and all-trans-
levels, which will increase the medication’s effectiveness retinoic acid (ATRA) was created [93]. ATO was put on an
even more [91]. Due to their biocompatibility and ease of IDA and ATRA backbone for two cycles of consolidation
surface functionalization for targeted delivery, IDA has been after introduction. Consequently, APML4 patients exhibited
incorporated into silica or mesoporous silica NPs in certain a remarkable rise in freedom from relapse and failure-free
instances [92]. The design of multifunctional inorganic for- survival compared with previously studied ATRA/IDA-
mulations containing IDA, in which NPs are not only drug based protocol (APML3) [94].
carriers but also possess therapeutic properties, is gaining In another study, the drug-eluting embolic (DEE) agent
popularity. For example, IDA-carrying MNPs may be tar- ONCOZENE (CeloNova BioSciences) appeared to be a
geted to the tumor location using an external magnetic field, viable platform for loading IDA which could be used to
and their magnetic features can also be used to enhance the treat patients with HCC [95]. To form the exterior shell,
overall therapeutic impact. DEE agents were coated with poly[bis(trifluroexthoxy)
Among inorganic structures, apoferritin was found to phosphazene], an inorganic polymer consisting of negatively
have suitable properties, such as self-assembly, decent charged sodium polymethacrylate as the hydrogel core.

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

Fig. 9  When combined with


hyperthermia, the IDA-SDDS
exhibits a greater effectiveness
of release and greater assimila-
tion by tumors in living bodies
compared to the DXR-SDDS. A
A depiction of the hyperthermia
(HT) therapy method and the
blood sample collection process
for rodents is provided. B, C
Three tumor-bearing BFS-1
mice are administered with a
low or high dosage of SDDS
(A for low dose and B for high
dose) in order to assess the lev-
els of medicines in the plasma
before and after 1-h HT (42 °C)
or normothermia (NT) (37 °C)
treatment. There is a complete
release when IDA-SDDS and
HT are used in tandem. D Com-
pared to DXR, tumors in an HT
environment absorb the released
IDA more effectively, sustaining
greater drug levels 48 h after
treatment. A nonparametric
Mann–Whitney test on a mini-
mum of three rats per cohort
yielded a *p < 0.03 result. E It
has been shown that during the
first 48 h of therapy, the ratio of
IDA to DXR concentration in
the tumor, which is generated
from D, steadily increases. This
suggests that IDA is kept in
cells after absorption for a con-
siderable amount of time. From
Ref. [89], used under Creative
Commons CC-BY license

Table 5  Carbon-based formulations for IDA loading


Formulation Particle size Zeta potential EE (%) DL (%) Release Characterization Case study/plat- Reference
(nm) (mV) form

IDA-SDDS N/A N/A 13.0 ± 3.6 N/A 100% at 42 °C In vivo and eNOS-Tag-GFP [89]
composed of in vitro mice
DPPC and
DSPC
IDA-GDYO 6 (via simula- N/A N/A N/A 150 ns (produc- MD simulation POPC bilayer [88]
tion) tion run-time)
IDA-GDY 7 (via simula- N/A N/A N/A ~ 10 ns (produc- DFT and MD Liquid water [90]
complex tion) tion run-time) simulation
IDA-loaded 218.5 ± 6.12 − 5.54 ± 0.88 84.2 ± 3.23 82 ± 4 46.7 ± 3.2%. at In vitro HT-29 [74]
propyl starch 37 °C in 7 days
NPs stabilized
with ptPVA

Results indicated that 10 mg of IDA could be loaded onto since IDA exhibits strong cytotoxicity against HCC cancer
the ONCOZENE DEE agents within 10 min, proving a rapid cells even at low doses. Rui Meng’s [96] research led to in
loading process. Conversely, the slow release of IDA from the development of AuMPn, an innovative, multipurpose NP
the ONCOZENE DEE agents was seen as advantageous encased in a gold nanoshell. The production of AuMPn was

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

achieved by combining the processes of emulsion solvent of the spheroid-shaped AuMPn particles that were produced
evaporation and hydroxylamine reduction. The researchers was 235.71 ± 34 nm, with an encapsulation efficiency of
employed IDA to assess the drug loading and release capa- 72.08% and drug loading efficiency of 1.73%. The gold
bilities of the NPs through dialysis. The average dimension exterior of the AuMPn emitted a spectrum resembling a

Fig. 10  Discoveries concerning


apoptosis in controlling the cells
and IDA. The combination of
MNP-GA-IDA indicates that
the majority of control cells are
still viable. Upon application of
MNP-GA-IDA, cells begin to
accumulate in areas related to
apoptosis, yet they do not pro-
gress to necrosis. These results
show that the MNP-GA-IDA
combination does not induce
necrosis-mediated cell demise.
Instead, it induces cell death by
activating the apoptotic path-
way. From Ref. [97], used under
Creative Commons CC-BY
license

Table 6  Inorganic structures developed for IDA delivery


Formulation Particle size (nm) Zeta EE (%) DL (%) Release Characterization Case study Reference
potential
(mV)

HsAFr-IDA 6–8 N/A 84.75 7.15 Through interaction In vitro MCF-7 [83]
with the SCARA5
and TfR1 receptors
IDA-loaded ATRA N/A N/A N/A N/A N/A AMPL4 Protocol APML4 [93]
and ATO complex
IDA-loaded ATRA​ N/A N/A N/A N/A N/A AMPL3 Protocol APML4 [94]
IDA with ONCOZ- 106 ± 11 N/A ≥ 99.5 N/A 75.9 ± 2.3% In vivo and in vitro HCC carcinoma [95]
ENE DEE agents
AuMPn 235.71 ± 34 N/A 72.08% 1.73% N/A In vitro N/A [96]
MNP-GA-IDA 21 ± 2.5 N/A N/A N/A N/A In vitro HL-60 [97]

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

fingerprint when exposed to near-infrared light. Additional double strands (utilized to load the drugs). The results indi-
research indicated that the release of IDA from AuMPn cated that when AS1411NTrs was employed for IDA deliv-
could be controlled using near-infrared irradiation. ery, a greater capacity of IDA was loaded compared to free
Hasan Ulusal and colleagues examined the efficacy and drug. Furthermore, the link between IDA and AO caused
toxicity of IDA by stabilizing this therapeutic agent on an improved stability in AS1411NTrs. Additionally, in vitro
MNPs coated with glutaraldehyde (GA) based on their cost- experiments using the MCF-7/ADR and MCF-7 lineages
effectiveness and toxicity-free nature. The study found that showed that 37 °C enhanced the cytotoxicity and synergistic
the ­IC50 value decreased by 5 to 7 due to MNP usage. Flow effects of IDA/(MTR)2Zn2 + /AO. Furthermore, loading the
cytometry and PCR analysis revealed that MNPs increased AS1411NTrs resulted in improved drug-induced apoptosis.
apoptosis, while suppressing the production of MDR1 and MCF-7/ADR demonstrated superior absorption of the com-
genes preventing apoptosis (Fig. 10) [97]. This suggests that bination of drugs in the cell uptake assays.
MNP mechanisms could help curb drug resistance by con- The current body of research on novel interventions
trolling drug release and preventing drug exit from cells. for non-Hodgkin’s lymphoma (NHL) has predominantly
Table 6 demonstrates inorganic formulations developed for focused on the utilization of IR780. This dye operates in
IDA encapsulation. the near-infrared spectrum and acts as a photosensitizer in
both photothermal therapy (PTT) and photodynamic therapy
(PDT). Researchers created a NP system that could transport
4 Idarubicin Co‑delivery Platforms IR780 and IDA, a chemotherapeutic drug, to bridge PTT/
PDT and chemotherapy for a more comprehensive treat-
Co-delivery nanosystems for cancer treatment represent a ment. The NPs, made of PEG-b-PTMC, were individually
rapidly evolving area of medical research. They are intended loaded with IR780 and IDA, exhibiting potent PTT/PDT
to enhance the therapeutic efficacy of medications by facili- efficiency when exposed to a near-infrared laser. Addition-
tating the simultaneous delivery of various therapeutic ally, they exhibited the process of endocytosis and displayed
agents. These systems can deliver conventional chemother- heightened formation of reactive oxygen species. The cell
apy drugs, gene-based molecules, and plant-derived mate- lines Raji and U937 exhibited decreased cell proliferation
rials, achieving synergistic effects and leading to enhanced upon exposure to the NPs. Furthermore, in vivo experiments
desired responses compared to single drug strategies [98]. demonstrated a significant decrease in growth and enhanced
Co-delivery can enhance therapeutic effects, decrease side biological safety, as seen in Fig. 11 [102].
effects, and to some extent, prevent multidrug resistance In a previous work, the synthesis of PEG-b-PTMC was
(MDR) [99]. However, few studies have been presented on conducted, whereby the amphiphilic mPEG-b-PTMC169
the co-delivery of IDA with other drugs. This section dis- was employed for the purpose of encapsulating ABT-
cusses the studies and experiments conducted on the co- 199, resulting in enhanced effectiveness and drug loading
loading of IDA with other medications. [103]. The results obtained from dynamic light scattering
Drug conjugation is a mechanism that enables several (DLS) and transmission electron microscopy (TEM) images
drugs onto the same carrier by covalently preconjugating revealed that the size of the nanocarriers containing both
through hydrolyzable linkers [100]. Saurabh Wadhwa and agents was approximately 138 nm and 210 nm, respectively.
coworkers [79] synthesized a DDC of PGA to co-deliver This disparity is attributed to the contraction of PEG seg-
IDA and D-penicillamine (D-pen). PGA was covalently ments in dry conditions. Furthermore, the zeta potential of
bound to the medications. According to the findings, the NPs was improved showing a better encapsulation rate of
DDC increased the exposure of IDA by sevenfold and positively charged IR780. The loading efficiency of anthra-
caused an extension in the circulation half-time of plasma. cycline in nanocarriers was about 7.38 ± 0.46% and its EE
Moreover, enhanced tumor accumulation was observed. The percentage was almost 78.73 ± 6.8%. Experiments showed
increase in antitumor activity was attributed to 89% in tumor the release rate of IDA was about 39.3 ± 8.0% after 4 h in
growth inhibition (TGI), which was 60% in free drugs and PBS, as the release medium. Raji and U937 lymphoma cells
contributed to a notable improvement of 17% regarding the showed an excellent uptake of NPs which changed over time
survival of NCI-H460 tumors in mouse models. in vitro. These NPs have also obtained better cytocompat-
In another study, Danfeng Wanga and colleagues [101] ibility and biological safety in vivo. The findings indicate
used DNA nanostructures to co-load mithramycin-acridine that the simultaneous administration of all three therapeutic
orange and IDA. This research aimed to explore the impact approaches led to a notable eradication of lymphoma cells.
of various types of binding in drugs on drug release and Despite the ongoing research, co-delivery mechanisms of
cytotoxicity. The nanostructured DNA that was used in this IDA with other therapeutic agents are still relatively obscure
experiment was AS1411NTrs, which was a combination of and demand more comprehensive investigations. Current
aptamer AS1411 (used to attack cancerous cells) and DNA studies have indicated the potential for improved therapeutic

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

Fig. 11  The study investigated


the in vivo tumor prevention
effects on U937 tumor-bearing
mice. A The development of
tumor volume after therapy
was seen in a sample size of
5. B The impact of various
therapies on the body weight of
mice afflicted with U937 tumor.
C Photographs depicting the
distinct tumor tissues in isola-
tion after to a 13-day period of
exposure to various treatment
modalities. D The histologi-
cal and immunohistochemical
examination of the tumor cells
after various treatment regimens
was conducted using hema-
toxylin and eosin staining. From
Ref. [102], used under Creative
Commons CC-BY license

efficiency and durability due to mechanisms like drug conju- anthracycline counterparts. Despite these favorable qualities,
gation and DNA nanostructure utilization. These integrated the full potential of IDA is currently hampered by significant
systems demonstrate increased drug exposure, enhanced side effects, namely cardiotoxicity, myelosuppression, poor
tumor accumulation, and improved in vitro cytotoxicity. absorption, rapid metabolism, and inadequate distribution.
Similarly, the co-delivery of the photosensitizer IR780 with Hence, with advancements in nanotechnology, novel meth-
IDA also exhibited promising outcomes in terms of inhibit- odologies have been developed to overcome such delivery
ing cell proliferation and enhancing biosecurity in the con- impediments. Lipid-based NPs, polymeric nanocarriers,
text of non-Hodgkin’s lymphoma. Nevertheless, in order carbonaceous nanostructures, and inorganic NPs have been
for these co-delivery systems to be widely used in the field proven to revitalize IDA’s therapeutic potential. Among the
of cancer therapy, more research is necessary to enhance mentioned structures, lipid-based NPs, stimuli-responsive
the synthesis techniques and to verify their effectiveness, or not, and polymeric nanocarriers appear to be particu-
safety, and stability in both laboratory settings (in vitro) and larly promising in encapsulating IDA, exhibiting significant
living organisms (in vivo). Undoubtedly, the integration of encapsulation efficiency, drug loading values, and release
nanotechnology and drug delivery has significant potential characteristics.
in enhancing the efficacy of cancer therapies, hence demand- Although strides have been made, the non-responsive and
ing sustained research efforts to achieve further refinement. stimuli-responsive lipid-based and polymeric nanocarriers
systems still face multiple challenges. The issues included
in this context are to the assurance of therapeutic levels at
5 Conclusion the designated treatment site, regulation of drug release, and
effective encapsulation of the medication. Additionally, it is
The escalating need for effective intervention for malig- crucial to improve the manufacturing process of nanocarriers
nant cancers has motivated profound exploration of differ- to enhance their stability and extend their circulation time.
ent strategies to transform oncological treatment. Recently, For example, in carriers that respond to stimuli, controlling
IDA, an anthracycline antibiotic, has shown much promise the triggers for drug release can be challenging in a clinical
with its potential in cancer therapy. This is due to its anti- setup, which requires more research and investigation.
leukemic activity, increased lipophilicity, higher cell per- Carbon-oriented delivery platforms have also demon-
meability, and improved cytotoxic effect compared to its strated potential as effective IDA delivery systems due

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

to their vast surface area and penetrative properties. The References


unique advantages of carbon-based NPs include low-
ered toxicity and enhanced anticancer characteristics of 1. La Vecchia, S. and C. Sebastián. Metabolic pathways regulat-
the encapsulated IDA. Notwithstanding these promising ing colorectal cancer initiation and progression. in Seminars
in cell & developmental biology. 2020. Elsevier.
results, more empirical investigation is required to com- 2. Sathishkumar, K., et al. (2022). Cancer incidence estimates
prehend and enhance the potential of these NPs as a deliv- for 2022 & projection for 2025: Result from National Cancer
ery mechanism for IDA. Registry Programme, India. The Indian Journal of Medical
In contrast, inorganic NPs have shown the capacity Research, 156(4&5), 598–607.
3. Wang, W., et al. (2023). Curcumin in cancer therapy: Explor-
for precise medication administration, improved durabil- ing molecular mechanisms and overcoming clinical challenges.
ity, and regulated release. The platform offered by these Cancer Letters, 570, 216332.
entities is very commendable owing to their versatility in 4. Lazarus, E., & Bays, H. E. (2022). Cancer and obesity: An
terms of dimensions, configurations, and physiochemical obesity medicine association (OMA) clinical practice state-
ment (CPS) 2022. Obesity Pillars, 3, 100026.
characteristics. Nevertheless, the assembly and modifica- 5. Ali, H., et al. (2023). Emergence of nanohybrids in hormonal
tion of the surfaces of these NPs continue to provide a cancer-targeted therapy. Hormone related cancer mechanistic
significant difficulty, necessitating more comprehensive and nanomedicines: Challenges and prospects (pp. 71–88).
study to address these concerns. Springer.
6. Bhattacharya, S., et al. (2023). Advances and challenges in thy-
Another promising field is the use of combination ther- roid cancer: The interplay of genetic modulators, targeted ther-
apy techniques with IDA along with additional chemo- apies, and AI-driven approaches. Life Sciences, 332, 122110.
therapy drugs. The enhancement of medication therapeu- 7. Qi, K., Sun, B., Liu, S.-Y., & Zhang, M. (2023). Research
tic effectiveness using co-delivery nanosystems and IDA progress on carbon materials in tumor photothermal therapy.
Biomedicine & Pharmacotherapy, 165, 115070.
has shown encouraging outcomes. Some of the possibili- 8. Lu, Y., Bai, X., & Pan, C. (2024). Impact of exercise interven-
ties that exist within the co-delivery nanosystems are the tions on quality of life and depression in lung cancer patients:
SDDS that uses phospholipids and the co-delivery of pho- A systematic review and meta-analysis. International Journal
tosensitizer IR780 with IDA. To verify these techniques’ of Psychiatry in Medicine, 59(2), 199–217.
9. Cheville, A., Smith, S., Barksdale, T., & Asher, A. (2020).
effectiveness, security, and general stability, further inves- Cancer Rehabilitation. In Braddom's Physical Medicine and
tigation is necessary. Rehabilitation (pp. 568–593.e7). Elsevier.
This multidisciplinary study may give rise to new para- 10. Mengarda, A. C., Iles, B., Longo, J. P. F., & de Moraes, J.
digms in cancer therapy in the years to come. The foundation (2023). Recent approaches in nanocarrier-based therapies for
neglected tropical diseases. Wiley Interdisciplinary Reviews:
has been laid for lipid-based NPs, polymeric nanocarriers, Nanomedicine and Nanobiotechnology, 15(2), e1852.
carbon-based NPs, and even inorganic NPs to potentially 11. Craig, M., et al. (2020). Engineering in medicine to address the
revolutionize cancer therapeutics. However, further preclini- challenge of cancer drug resistance: From micro-and nanotech-
cal studies and clinical trials are required to evaluate the effi- nologies to computational and mathematical modeling. Chemi-
cal Reviews, 121(6), 3352–3389.
cacy and safety of these intriguing nano-delivery technolo- 12. Fathi Karkan, S., Davaran, S., & Akbarzadeh, A. (2019). Cis-
gies. It is essential that legislation and procedures pertaining platin-loaded superparamagnetic nanoparticles modified with
to medication development adapt to the unique problems PCL-PEG copolymers as a treatment of A549 lung cancer
presented by these innovative treatments. cells. Nanomedicine Research Journal, 4(4), 209–219.
13. Kumar, M., et al. (2023). Micro and nano-carriers-based pul-
monary drug delivery system: Their current updates, chal-
lenges, and limitations – A review. JCIS Open. 12. 100095.
Author Contribution F.S., Y. J, H. A., M.P.: investigation, data cura- 14. Fatima Qizilbash, F., et al. (2023). Nanotechnology revolution-
tion. P.Z., M. A.: writing, review, and editing. A. R., S. F-k., S. P: ises breast cancer treatment: Harnessing lipid-based nanocar-
validation, writing, review, and editing. All authors reviewed the riers to combat cancer cells. Journal of Drug Targeting, 31(8),
manuscript. 794–816.
15. Rabiee, N., et al. (2020). Nanotechnology-assisted microflu-
Funding Abbas Rahdar thanks from University of Zabol for funding idic systems: From bench to bedside. Nanomedicine, 16(3),
(UOZ-GR-8906). The authors (Sadanand Pandey) express profound 237–258.
gratitude to Shoolini University for their invaluable support, which 16. Kong, X., et al. (2023). Advances of medical nanorobots for
played a pivotal role in facilitating the completion of work. future cancer treatments. Journal of Hematology & Oncology,
16(1), 74.
Data Availability No datasets were generated or analyzed during the 17. Abdelkader, H., et al. (2021). Polymeric long-acting drug
current study. delivery systems (LADDS) for treatment of chronic diseases:
Inserts, patches, wafers, and implants. Advanced Drug Deliv-
Declarations ery Reviews, 177, 113957.
18. Azizi, M., et al. (2023). Multifunctional plant virus nanoparti-
Ethical Approval Not applicable. cles: An emerging strategy for therapy of cancer. Wiley inter-
disciplinary reviews. Nanomedicine and Nanobiotechnology,
Competing Interests The authors declare no competing interests. 15(6), e1872.

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

19. Handa, M., et al. (2021). Recent advances in lipid-engineered with acute myeloid leukemia in first complete remission. Bone
multifunctional nanophytomedicines for cancer targeting. Jour- Marrow Transplantation, 22(1), 13–19.
nal of Controlled Release, 340, 48–59. 38. Ravandi, F., et al. (2019). Idarubicin, cytarabine, and nivolumab
20. Wang, G., et al. (2020). Advances in nanotechnology-based strat- in patients with newly diagnosed acute myeloid leukaemia or
egies for the treatments of amyotrophic lateral sclerosis. Materi- high-risk myelodysplastic syndrome: A single-arm, phase 2
als Today Bio, 6, 100055. study. The Lancet Haematology, 6(9), e480–e488.
21. Delbreil, P., Rabanel, J.-M., Banquy, X., & Brambilla, D. (2022). 39. Sherif, H. A., et al. (2021). Treatment outcome of doxorubicin
Therapeutic nanotechnologies for Alzheimer’s disease: A critical versus idarubicin in adult acute myeloid leukemia. Leukemia
analysis of recent trends and findings. Advanced Drug Delivery Research Reports, 16, 100272.
Reviews, 187, 114397. 40. Foroughi, M. M., & Jahani, S. (2022). Investigation of a high-
22. Zhou, F., Peterson, T., Fan, Z., & Wang, S. (2023). The com- sensitive electrochemical DNA biosensor for determination of
monly used stabilizers for phytochemical-based nanoparticles: idarubicin and studies of DNA-binding properties. Microchemi-
Stabilization effects, mechanisms, and applications. Nutrients, cal Journal, 179, 107546.
15(18), 3881. 41. Rauf, S., et al. (2005). Electrochemical approach of anticancer
23. Fathi-Karkan, S., et al. (2023). Recent advancements in the tar- drugs–DNA interaction. Journal of Pharmaceutical and Bio-
geted delivery of etoposide nanomedicine for cancer therapy: A medical Analysis, 37(2), 205–217.
comprehensive review. European Journal of Medicinal Chemis- 42. Lachatre, F., et al. (2000). Simultaneous determination of four
try, 259, 115676. anthracyclines and three metabolites in human serum by liq-
24. Fathi-Karkan, S., Ghavidel-Kenarsari, F., & Maleki-Baladi, uid chromatography–electrospray mass spectrometry. Journal
R. (2022). Pullulan as promoting endothelialization capacity of Chromatography B: Biomedical Sciences and Applications,
of electrospun PCL-PU scaffold. The International Journal of 738(2), 281–291.
Artificial Organs, 45(12), 1013–1020. 43. Hu, Q., Hu, G., Zhou, T., & Fang, Y. (2003). Determination of
25. Sahu, T., et al. (2021). Nanotechnology based drug delivery sys- dissociation constants of anthrocycline by capillary zone electro-
tem: Current strategies and emerging therapeutic potential for phoresis with amperometric detection. Journal of Pharmaceuti-
medical science. Journal of Drug Delivery Science and Technol- cal and Biomedical Analysis, 31(4), 679–684.
ogy, 63, 102487. 44. Kuhlmann, O., Hofmann, S., & Weiss, M. (1999). Determination
26. Leso, V., et al. (2022). Exposure to antineoplastic drugs in occu- of idarubicin and idarubicinol in rat plasma using reversed-phase
pational settings: A systematic review of biological monitoring high-performance liquid chromatography and fluorescence detec-
data. International Journal of Environmental Research and Pub- tion. Journal of Chromatography B: Biomedical Sciences and
lic Health, 19(6), 3737. Applications, 728(2), 279–282.
27. Abbas, A. A., & AlAzmi, A. A. M. (2019). Anthracycline- 45. Badea, I., et al. (2005). A HPLC method for the simultaneous
induced cardiac toxicity: A clinical review. Indian Journal of determination of seven anthracyclines. Journal of Pharmaceuti-
Medical and Paediatric Oncology, 40(04), 465–475. cal and Biomedical Analysis, 39(1–2), 305–309.
28. Zheng, Z., et al. (2023). Idarubicin-loaded biodegradable micro- 46. Kurbanoglu, S., et al. (2013). Electrochemical investigations of
spheres enhance sensitivity to anti-PD1 immunotherapy in tran- the anticancer drug idarubicin using multiwalled carbon nano-
scatheter arterial chemoembolization of hepatocellular carci- tubes modified glassy carbon and pyrolytic graphite electrodes.
noma. Acta Biomaterialia, 157, 337–351. Electroanalysis, 25(6), 1473–1482.
29. Arcamone, F., et al. (1976). Synthesis and antitumor activity of 47. Li, W., Jiang, Y., & Lu, J. (2023). Nanotechnology-enabled
4-demethoxydaunorubicin, 4-demethoxy-7, 9-diepidaunorubicin, immunogenic cell death for improved cancer immunotherapy.
and their anomers. Cancer Treatment Reports, 60, 829–834. International Journal of Pharmaceutics, 634, 122655.
30. Borchmann, P., Hübel, K., Schnell, R., & Engert, A. (1997). 48. Behravan, N., et al. (2022). Lipid-based nanoparticulate delivery
Idarubicin: A brief overview on pharmacology and clinical use. systems for HER2-positive breast cancer immunotherapy. Life
International Journal of Clinical Pharmacology and Therapeu- Sciences, 291, 120294.
tics, 35(2), 80–83. 49. Liang, B., et al. (2019). Idarubicin-loaded methoxy poly(ethylene
31. Gotzhein, F. (2021). Tracking clonal composition of stem cell glycol)-b-poly(l-lactide-co-glycolide) nanoparticles for enhanc-
transplants by genetic barcoding and implementation of a tar- ing cellular uptake and promoting antileukemia activity. Inter-
geted barcoding procedure [Doctoral dissertation, University of national Journal of Nanomedicine, 14, 543–556.
Hamburg]. ediss.sub.hamburg. 50. Najafi, F., Pourali, S., & Kamran, A. (2021). Comparison of ther-
32. Li, X., Xu, S., Tan, Y., & Chen, J. (2015). The effects of idaru- modynamic functions of idarubicin cancer drug interaction with
bicin versus other anthracyclines for induction therapy of patients carbon nanotube: QM/MM studies. International Journal of New
with newly diagnosed leukaemia. The Cochrane Database of Sys- Chemistry, 8(1), 30–40.
tematic Reviews, (6), CD010432. 51. Alavi, M., & Varma, R. S. (2020). Overview of novel strategies
33. McGowan, J. V., et al. (2017). Anthracycline chemotherapy and for the delivery of anthracyclines to cancer cells by liposomal
cardiotoxicity. Cardiovascular Drugs and Therapy, 31, 63–75. and polymeric nanoformulations. International Journal of Bio-
34. Das, S., et al. (2022). Aptamers functionalized biomolecular logical Macromolecules, 164, 2197–2203.
nano-vehicles for applications in cancer diagnostics & thera- 52. Ashrafizadeh, M., et al. (2021). Hyaluronic acid-based nanoplat-
peutics. Applied NanoMedicine, 2(2), 360–360. forms for doxorubicin: A review of stimuli-responsive carriers,
35. Tsuruo, T., Oh-Hara, T., Sudo, Y., & Naito, M. (1993). Antitu- co-delivery and resistance suppression. Carbohydrate Polymers,
mor activity of idarubicin, a derivative of daunorubicin, against 272, 118491.
drug sensitive and resistant P388 leukemia. Anticancer Research, 53. Tapeinos, C., Battaglini, M., & Ciofani, G. (2017). Advances
13(2), 357–361. in the design of solid lipid nanoparticles and nanostructured
36. Kim, J. S., et al. (2022). Anthracycline derivatives inhibit cardiac lipid carriers for targeting brain diseases. Journal of Controlled
CYP2J2. Journal of Inorganic Biochemistry, 229, 111722. Release, 264, 306–332.
37. Jerjis, S., et al. (1998). Idarubicin to intensify the conditioning 54. Yingchoncharoen, P., Kalinowski, D. S., & Richardson, D. R.
regimens of autologous bone marrow transplantation for patients (2016). Lipid-based drug delivery systems in cancer therapy:

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

What is available and what is yet to come. Pharmacological 73. Abad, F., et al. (2023). Chitosan-Carbon nanotube Composite:
Reviews, 68(3), 701–787. An approach for controlled release of Quercetin, Modified with
55. Sheoran, S., Arora, S., Samsonraj, R., & Govindaiah, P. (2022). carboxymethyl Cellulose, for potential Anti-Cancer therapy.
Lipid-based nanoparticles for treatment of cancer. Heliyon, Inorganic Chemistry Communications, 158, 111621.
8(5), e09403. 74. Jain, R., et al. (2011). Enhanced cellular delivery of idarubicin
56. Mitchell, M. J., et al. (2021). Engineering precision nanoparti- by surface modification of propyl starch nanoparticles employing
cles for drug delivery. Nature Reviews Drug Discovery, 20(2), pteroic acid conjugated polyvinyl alcohol. International Journal
101–124. of Pharmaceutics, 420(1), 147–155.
57. Xu, Q., et al. (2023). Tumor microenvironment-regulating 75. Güç, E., Gündüz, G., & Gündüz, U. (2010). Fatty acid based
nanomedicine design to fight multi-drug resistant tumors. hyperbranched polymeric nanoparticles for hydrophobic drug
Wiley Interdisciplinary Reviews: Nanomedicine and Nano- delivery. Drug Development and Industrial Pharmacy, 36(10),
biotechnology, 15(1), e1842. 1139–1148.
58. Sarangi, B., et al. (2019). Solid lipid nanoparticles: A potential 76. Minotti, G., et al. (2004). Anthracyclines: Molecular advances
approach for drug delivery system. Nanoscience & Nanotech- and pharmacologic developments in antitumor activity and car-
nology-Asia, 9(2), 142–156. diotoxicity. Pharmacological reviews, 56(2), 185–229.
59. Zara, G. P., et al. (2002). Pharmacokinetics and tissue dis- 77. Zhang, Y., Li, J., & Pu, K. (2022). Recent advances in dual- and
tribution of idarubicin-loaded solid lipid nanoparticles after multi-responsive nanomedicines for precision cancer therapy.
duodenal administration to rats. Journal of Pharmaceutical Biomaterials, 291, 121906.
Sciences, 91(5), 1324–1333. 78. Das, B. C., et al. (2023). Stimuli-responsive boron-based materi-
60. Sermsappasuk, P., Hrynyk, R., Gubernator, J., & Weiss, M. als in drug delivery. International Journal of Molecular Sciences,
(2008). Reduced uptake of liposomal idarubicin in the perfused 24(3), 2757.
rat heart. Anti-cancer Drugs, 19(7), 729–732. 79. Wadhwa, S., & Mumper, R. J. (2012). Polypeptide conjugates of
61. Iqbal, N., & Iqbal, N. (2014). Human Epidermal Growth Factor d-penicillamine and idarubicin for anticancer therapy. Journal of
Receptor 2 (HER2) in Cancers: Overexpression and Thera- Controlled Release, 158(2), 215–223.
peutic Implications. Molecular Biology International, 2014, 80. Blaudszun, A.-R., et al. (2014). Polyester-idarubicin nanopar-
852748. ticles and a polymer-photosensitizer complex as potential drug
62. Amin, M., et al. (2018). Specific delivery of idarubicin to HER2- formulations for cell-mediated drug delivery. International Jour-
positive breast cancerous cell line by trastuzumab-conjugated nal of Pharmaceutics, 474(1–2), 70–79.
liposomes. Journal of Drug Delivery Science and Technology, 81. Nafee, N., et al. (2015). Cyclodextrin-based star polymers as a
47, 209–214. versatile platform for nanochemotherapeutics: Enhanced entrap-
63. Ma, P., et al. (2009). Development of idarubicin and doxorubicin ment and uptake of idarubicin. Colloids and Surfaces B: Bioint-
solid lipid nanoparticles to overcome Pgp-mediated multiple erfaces, 129, 30–38.
drug resistance in leukemia. Journal of Biomedical Nanotech- 82. Gunduz, U., et al. (2014). Idarubicin-loaded folic acid conjugated
nology, 5(2), 151–161. magnetic nanoparticles as a targetable drug delivery system for
64. Tang, Q., et al. (2018). Stimuli responsive nanoparticles for con- breast cancer. Biomedicine & Pharmacotherapy, 68(6), 729–736.
trolled anti-cancer drug release. Current Medicinal Chemistry, 83. Rafipour, R., Mousavi, A., & Mansouri, K. (2022). Apoferri-
25(16), 1837–1866. tin nanocages for targeted delivery of idarubicin against breast
65. Lu, T., et al. (2015). Formulation and optimization of idarubicin cancer cells. Biotechnology and Applied Biochemistry, 69(3),
thermosensitive liposomes provides ultrafast triggered release 1061–1067.
at mild hyperthermia and improves tumor response. Journal of 84. Tang, L., et al. (2021). Insights on functionalized carbon nano-
Controlled Release, 220, 425–437. tubes for cancer theranostics. Journal of Nanobiotechnology, 19,
66. Wang, Y., Chen, L., Ding, Y., & Yan, W. (2012). Oxidized phos- 1–28.
pholipid based pH sensitive micelles for delivery of anthracy- 85. Dehdashtian, S., Behbahanian, N., Mohammadi Taherzadeh, K.,
clines to resistant leukemia cells in vitro. International Journal & Hashemi, B. (2019). Development of electrochemical sensor
of Pharmaceutics, 422(1–2), 409–417. based on multiwall carbon nanotube for determination of anti-
67. Dos Santos, N., et al. (2005). Substantial increases in idaru- cancer drug idarubicin in biological samples. Advances in Nano-
bicin plasma concentration by liposome encapsulation mediates chemistry, 1(1), 22–28.
improved antitumor activity. Journal of Controlled Release, 86. Chandra, K., Dutta, S., Kolya, H., & Kang, C.-W. (2023). Struc-
105(1–2), 89–105. tural aspect of hydroxyethyl-starch–anticancer-drug-conjugates as
68. Gubernator, J., et al. (2010). The encapsulation of idarubicin state-of-the-art drug carriers. Scientia Pharmaceutica, 91(3), 32.
within liposomes using the novel EDTA ion gradient method 87. Matyszewska, D. (2020). The influence of charge and lipophilic-
ensures improved drug retention in vitro and in vivo. Journal of ity of daunorubicin and idarubicin on their penetration of model
Controlled Release, 146(1), 68–75. biological membranes–Langmuir monolayer and electrochemical
69. Dos Santos, N., et al. (2002). Improved retention of idaru- studies. Biochimica et Biophysica Acta (BBA)-Biomembranes,
bicin after intravenous injection obtained for cholesterol-free 1862(2), 183104.
liposomes. Biochimica et Biophysica Acta (BBA)-Biomembranes, 88. Shahabi, M., & Raissi, H. (2021). The transport of idarubicin
1561(2), 188–201. therapeutic agent using a novel graphene sheet as a drug delivery
70. Caro, C., et al. (2022). Nanomaterials loaded with Quercetin as platform through a biomembrane. Journal of Molecular Liquids,
an advanced tool for cancer treatment. Journal of Drug Delivery 323, 115050.
Science and Technology, 78, 103938. 89. Lu, T., et al. (2021). Externally triggered smart drug delivery
71. Cheng, R., et al. (2013). Dual and multi-stimuli responsive poly- system encapsulating idarubicin shows superior kinetics and
meric nanoparticles for programmed site-specific drug delivery. enhances tumoral drug uptake and response. Theranostics,
Biomaterials, 34(14), 3647–3657. 11(12), 5700.
72. Prabhu, R. H., Patravale, V. B., & Joshi, M. D. (2015). Polymeric 90. Shahabi, M., & Raissi, H. (2020). Two dimensional porous
nanoparticles for targeted treatment in oncology: current insights. frameworks of graphyne family as therapeutic delivery vehicles
International Journal of Nanomedicine, 10, 1001–1018. for Idarubicin biomolecule in silico: Density functional theory

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


BioNanoScience

and molecular dynamics simulation. Journal of Molecular Liq- 98. Eftekhari, R. B., et al. (2019). Co-delivery nanosystems for can-
uids, 319, 114334. cer treatment: A review. Pharmaceutical Nanotechnology, 7(2),
91. Li, C., et al. (2022). Molecular recognition-driven supramolecu- 90–112.
lar nanoassembly of a hydrophobic uracil prodrug and hydro- 99. Li, Y., Thambi, T., & Lee, D. S. (2018). Co-delivery of drugs and
philic cytarabine for precise combination treatment of solid and genes using polymeric nanoparticles for synergistic cancer thera-
non-solid tumors. Nanoscale Horizons, 7(2), 235–245. peutic effects. Advanced Healthcare Materials, 7(1), 1700886.
92. Pérez-Ruiz, T., Martínez-Lozano, C., Sanz, A., & Bravo, E. 100. Aryal, S., Hu, C. M. J., & Zhang, L. (2010). Combinatorial drug
(2001). Simultaneous determination of doxorubicin, daunoru- conjugation enables nanoparticle dual-drug delivery. Small,
bicin, and idarubicin by capillary electrophoresis with laser- 6(13), 1442–1448.
induced fluorescence detection. Electrophoresis, 22(1), 134–138. 101. Wang, D., et al. (2022). Idarubicin/mithramycin-acridine orange
93. Iland, H. J., et al. (2012). All-trans-retinoic acid, idarubicin, and combination drugs co-loaded by DNA nanostructures: Different
IV arsenic trioxide as initial therapy in acute promyelocytic leu- effects of intercalation and groove binding on drug release and
kemia (APML4). Blood, The Journal of the American Society of cytotoxicity. Journal of Molecular Liquids, 355, 118947.
Hematology, 120(8), 1570–1580. 102. Weng, S., et al. (2023). Idarubicin and IR780 co-loaded PEG-
94. Iland, H., et al. (2012). Results of the APML3 trial incorporating b-PTMC nanoparticle for non-Hodgkin’s lymphoma therapy by
all-trans-retinoic acid and idarubicin in both induction and con- photothermal/photodynamic strategy. Materials & Design, 230,
solidation as initial therapy for patients with acute promyelocytic 112008.
leukemia. Haematologica, 97(2), 227. 103. Nia, H. T., Munn, L. L., & Jain, R. K. (2020). Physical traits of
95. Guiu, B., et al. (2015). Idarubicin-loaded ONCOZENE drug-eluting cancer. Science, 370(6516), eaaz0868.
embolic agents for chemoembolization of hepatocellular carci-
noma: In vitro loading and release and in vivo pharmacokinetics. Publisher's Note Springer Nature remains neutral with regard to
Journal of Vascular and Interventional Radiology, 26(2), 262–270. jurisdictional claims in published maps and institutional affiliations.
96. Meng, R. (2016). Multifunctional gold nanoshell-coated mag-
netic nanoparticles: preparation, characterization, two-photon Springer Nature or its licensor (e.g. a society or other partner) holds
optical properties and drug release exploration. Chinese Phar- exclusive rights to this article under a publishing agreement with the
maceutical Journal, 24, 1394–1404. author(s) or other rightsholder(s); author self-archiving of the accepted
97. Ulusal, H., et al. (2022). Attachment of idarubicin to glutaralde- manuscript version of this article is solely governed by the terms of
hyde-coated magnetic nanoparticle and investigation of its effect such publishing agreement and applicable law.
in HL-60 cell line. International Journal of Chemistry and Tech-
nology, 6(2), 154–163.

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:

1. use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
2. use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
3. falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
4. use bots or other automated methods to access the content or redirect messages
5. override any security feature or exclusionary protocol; or
6. share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at

onlineservice@springernature.com

You might also like