Enhancing The Therapeutic Efficacy of Nanoparticles For Cancer Treatment Using Versatile Targeted Strategies
Enhancing The Therapeutic Efficacy of Nanoparticles For Cancer Treatment Using Versatile Targeted Strategies
Enhancing The Therapeutic Efficacy of Nanoparticles For Cancer Treatment Using Versatile Targeted Strategies
Abstract
Poor targeting of therapeutics leading to severe adverse effects on normal tissues is considered one of the obstacles
in cancer therapy. To help overcome this, nanoscale drug delivery systems have provided an alternative avenue for
improving the therapeutic potential of various agents and bioactive molecules through the enhanced permeability
and retention (EPR) effect. Nanosystems with cancer-targeted ligands can achieve effective delivery to the tumor cells
utilizing cell surface-specific receptors, the tumor vasculature and antigens with high accuracy and affinity. Addition-
ally, stimuli-responsive nanoplatforms have also been considered as a promising and effective targeting strategy
against tumors, as these nanoplatforms maintain their stealth feature under normal conditions, but upon homing in
on cancerous lesions or their microenvironment, are responsive and release their cargoes. In this review, we compre-
hensively summarize the field of active targeting drug delivery systems and a number of stimuli-responsive release
studies in the context of emerging nanoplatform development, and also discuss how this knowledge can contribute
to further improvements in clinical practice.
Keywords: Drug delivery, Targeted strategies, Active targeting, Stimuli-responsive materials, Cancer treatment
© The Author(s) 2022. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which
permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the
original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or
other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line
to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this
licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativeco
mmons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 2 of 40
Fig. 2 Schematic representation of receptor-mediated active targeting and passive targeting through the EPR effect
accumulation have attracted extensive attention in recent agents in clinical use are monoclonal antibodies (mAbs)
years. Table 1 summarizes some of the specific receptors and small molecule tyrosine kinase inhibitors (TKIs).
overexpressed on various tumor cells along with their The mAbs can be directly applied to deliver therapeu-
related ligands. Utilizing cell surface active targeting tic agents to tumor cells through drug-Ab complexes or
strategies has greatly advanced tumor treatment. Some modified on the surface of the nanoplatform-loaded ther-
of these approaches are summarized in the following apeutic agents [29, 30].
sections. Recently, EGFR-based nanoplatforms have been widely
explored against cancers [31–35]. These nanoparticles
Epidermal growth factor receptor (EGFRs)‑based active are generally internalized into the cells through an EGFR-
targeting mediated endocytosis process, resulting in the formation
The EGFR, a transmembrane protein, is involved in the of lysosomes and release of encapsulated drugs for cancer
occurrence of several types of cancers, including lung, treatment. As an exemplar, Nan and co-workers prepared
pancreatic, colorectal, and breast cancers [24]. Activation versatile nanoplatforms capable of specific codeliv-
of the EGFR is triggered by the binding of ligands, includ- ery of DOX and cisplatin to tumor sites by utilizing an
ing EGF, transforming growth factor-α (TGF-α), epireg- EGFR-targeted approach [33]. These targeted nanopar-
ulin, heparin-binding EGF, betacellulin, amphiregulin, ticles showed high stability with sustained cargo release,
and neuregulin G2β. This enables protein kinase (PK) to showing satisfactory killing effects in lung cancer mod-
transfer a phosphate molecule from adenosine triphos- els. In a similar approach, Liang et al. prepared versatile
phate (ATP) to the tyrosine residues, resulting in phos- nanoplatforms functionalized with anti-EGFR Ab for
phorylation of the intracellular domain, which mediates lesion-specific delivery of carmustine to malignant glio-
a signaling cascade pathway [25, 26]. Eventually, this pro- blastomas for growth suppression [34]. Confirming the
cess can result in tumorigenesis and cancer progression, role of the EGFR, Shuai and co-workers reported higher
thereby making EGFR one of the main anticancer tar- internalization of an anti-EGFR monoclonal antibody-
gets [27, 28]. The most commonly used EGFR targeting conjugated nanoplatform in EGFR-positive human skin
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 4 of 40
Table 1 The overexpressed receptors on various tumor cells and their ligands
Receptor Ligands Tumor Refs.
Folate Folic acid Breast, lung, cervical cancer, hepatocellular carcinoma [414–419]
CD14 anti-CD14 mAb Prostatic cancer [420]
CD22 anti-CD22 mAb Lymphoma cancer [421, 422]
CD44 HA, chondroitin sulfate Breast, Melanoma [423–426]
αvβ3 integrin RGD peptide Endothelial, glioma, lung, melanoma, breast cancer [427–434]
Transferrin TfR ligand, transferrin Breast cancer, Glioblastoma [435–438]
HER2 Trastuzumab Breast anti-HER2 scFv neu peptide (FCDG- Breast cancer [439–441]
FYACYADV) KCCYSL (P6.1 peptide)
Estrogen Estrone, 17 β-Estradiol, tamoxifen Breast cancer [442–446]
Chemokine (CXCR4) LFC131 peptide, anti-CXCR4 mAb, Peptide R, Peptide Breast, lung cancer, hepatocellular carcinoma, Lym- [447–454]
T22 phoma
LHRH Peptide Breast cancer [106, 107]
Biotin Biotin Breast, lung, cervical cancer, hepatocellular carcinoma [455–460]
PSMA A10 PSMA Apt, anti-PSMA Prostatic cancer [461–463]
VEGF anti-VEGF mAb Pancreatic cancer [464, 465]
IL4 AP1 peptide Colon, glioblastoma [466–468]
IL4 Pep-1 Lung cancer [469–471]
IL13 IL13 peptide Glioblastoma [472–474]
Asialoglycoprotein Lactobionic acid, galactose Hepatocellular carcinoma [475, 476]
receptor (ASGPR)
squamous cell carcinoma compared to EGFR-negative gemcitabine (Gem) encapsulated nanoplatforms could
breast cancer [35]. Furthermore, Choi and co-work- effectively inhibit tumor growth [36]. Gupta and col-
ers demonstrated that binding EGFR-targeting Abs to leagues constructed a Gem encapsulated nanoplatform
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 5 of 40
against pancreatic cancer through covalent binding to targeted delivery of epirubicin to overexpressed αvβ3
EGFR antibodies [37], presenting higher cytotoxicity of integrin in esophageal cancer, resulting in the designed
the designed nanoplatform for EGFR-overexpressing nanoplatforms not only reducing epirubicin-induced
pancreatic cell lines. Moreover, anti-EGFR functionalized cardiotoxicity but also improving the therapeutic effect
Fe2O3 nanoparticles can be used as magnetic resonance in comparison to free agents [52]. Recently, Roy et al.
imaging contrast agents for tumor diagnosis [38, 39]. In prepared pH-responsive nanoparticles for the effec-
an interesting review article, Yi and colleagues discussed tive delivery of raloxifene to breast cancer cells through
the role of EGFR tyrosine kinase inhibitors in targeted RGD-modified nanocarriers. The designed nanoparticles
nanoplatforms for tumor treatment [40]. showed good cytotoxicity and antitumor efficacy toward
Supported by the rapid advancement of nanomedi- αvβ3 positive breast cancer cells and a 4T1-bearing
cine, these inhibitor-loaded nanoparticles are showing mouse model [53]. In another recent study, Wang’s group
improved bioavailability, prolonged blood circulation, reported a bispecific assembling peptide antiCD3-G7-
enhanced tumor accumulation and reduced off-tar- RGD for tumor immunotherapy [54]. The RGD was used
get side effects, leading to significant augmentation of to improve tumor accumulation and cell internalization
therapeutic efficacy [41, 42] supporting their continued via the integrin receptor-mediated endocytosis process.
development. The anti-CD3 was designed to target the CD3 receptor
on T lymphocytes and induce a T cell-mediated immune
αvβ3 integrin receptor‑mediated active targeting response against tumor cells overexpressing integrin
Integrin receptors, consisting primarily of transmem- αvβ3, resulting in satisfactory antitumor effects. In sum-
brane glycoproteins, can mediate cell–cell and cell- mary, nanoparticles can preferentially and effectively tar-
extracellular matrix adhesion [43]. More than 23 integrin get integrin binding sites in tumors (e.g., the RGD motif ),
heterodimers have been identified in humans to date thereby providing a solid basis for developing precision
[44]. These receptors control the connection between the tumor treatment strategies [55].
extracellular matrix (ECM) and the cell cytoskeleton as
well as maintaining communications between cells [43, Folate receptor (FR)‑mediated active targeting
45]. The extracellular domains of integrins have strong FRs, a class of glycoproteins, have been classified into
affinity for the proteins (collagen, fibronectin, laminin three subtypes namely FRα, FRβ and FRγ. It should be
and vitronectin) in the ECM. Furthermore, integrins noted that FRα and FRβ can closely bind to the tumor
can play a significant part in several signaling pathways cell membrane via a glycosylphosphatidylinositol anchor,
involved in cell proliferation after combining with the while FRγ has only been reported in hematopoietic cells
ECM [46]. It is possible to target integrin receptor-pos- [4, 56–58]. Among them, FRα is the most widely gener-
itive tumor cells through functionalized nanoparticles ated FR subtype and is overgenerated in various tumor
containing an integrin targeting motif (such as RGD- cells, especially in breast, lung, kidney, cervical, and ovar-
containing peptides and polymers). This approach has ian cancer [59–61]. Moreover, FR can transport folate
been extensively explored [47–53]. For example, Lu and into tumor cells via the receptor-mediated endocytosis
co-workers prepared cyclic RGD peptide-functionalized process [62]. For this reason, a number of FA-based nan-
nanoplatforms for paclitaxel (PTX) delivery to glio- oplatforms have been prepared for increased internaliza-
blastoma cells overexpressing αvβ3, resulting in antitu- tion of therapeutic agents by tumor cells [63–65]. In one
mor effects in in vivo models [49]. In another example, example, Murgia et al. prepared an organic/inorganic
Li et al. prepared RGD-conjugated resveratrol loaded hybrid nanoplatform modified by FA-chitosan conju-
human serum albumin nanoparticles, which showed gates to load upconverting NaYF4 nanoparticles and dau-
higher internalization efficiency (approximately 3.6-fold norubicin for tumor therapy [62]. The FA modification
higher) as well as improved tumor suppression features significantly improved the cellular uptake of the nano-
compared to the non-functionalized formulation [50]. particles, and an in vivo xenograft model also showed a
Amreddy and co-workers developed RGD-functional- positive antitumor effect. In another example, Wang et al.
ized nanoparticles for the delivery of therapeutic agents designed an FA-conjugated chitosan loaded rutin pre-
(PTX and cisplatin) to αvβ3 integrin receptor-over- pared palladium nanoplatform for FA-mediated target-
expressing lung cancer cells and found that the RGD- ing treatment. The introduction of FA into the designed
targeted nanoformulations showed higher endocytosis nanoplatform significantly improved the endocytosis
efficiency (approximately 1.4-fold higher) compared with efficiency of the nanoparticles in breast cancer cells. The
non-RGD-functionalized formulations [51]. Pan and designed nanoplatform was shown to considerably sup-
co-workers developed RGD-modified fluorescent nano- press cell proliferation as evidenced by a cell viability
platforms for simultaneous fluorescence-guided and assay [66]. Mechanistically, FRs can identify and bind to
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 6 of 40
extracellular FA-modified nanoparticles and then trans- integrated as targeting units on nanoparticles against
port them into the tumor cells through a FR-mediated HER2 overexpressing cancer. The introduction of trastu-
endocytosis process [67]. In these nanoparticles, the FA zumab (TZ), a humanized anti-HER2 Ab, endows nano-
portion is used as a tumor-targeting ligand. On bind- particles with excellent therapeutic efficacy for breast
ing to the FR on tumor cells, the cell membrane can cancer treatment [81]. It can block cell cycle arrest and
invaginate and pinch off to form endosomes which sub- reduce angiogenesis by disturbing downstream HER2
sequently reach lysosomes or other organelles. The drug- signaling activity. The interaction between TZ and HER
encapsulated nanoparticles can dissociate from the FR blocks receptor cleavage and activates the response of
and effectively release the encapsulated drug at the TME Ab-dependent cellular cytotoxicity and receptor degrada-
for tumor treatment. tion following internalization of the TZ-HER2 complex.
Pertuzumab (PZ), another humanized mAb, has been
Transferrin (Tf) receptor‑mediated active targeting used to suppress heregulin-mediated activation of HER2
e3+ pool in the body, Tf plays an impor-
As the critical F phosphorylation and tumor proliferation [82]. Nanopar-
tant role in Fe metabolism and delivery. To meet the ticles functionalized with anti-HER2 Abs or its fragments
growing requirements of Fe for maintaining cell growth can be effectively used for specific delivery of therapeu-
and division, transferrin receptors (TfR) are frequently tic agents to HER2-overexpressed tumor cells by the
overexpressed on the surface of a number of tumors HER2 receptor-mediated endocytosis process [83] which
including pancreas, breast, prostate, colon, and lung can- enhances therapeutic efficacy with fewer side effects.
cer, with high affinity to Tf [68–71]. This has prompted
scientists to use the TfR as an active targeting site in Estrogen receptor‑mediated active targeting
the design of novel anti-cancer delivery platforms. TfR Estrogen is a steroid hormone that plays a critical part in
can be employed either for Tf-mediated targeting and maintaining reproductive system function, bone homeo-
internalization of therapeutic agents or to block normal stasis, brain development, and cardiovascular remodeling
receptor function, resulting in cell death [72–74]. In an [84]. Among the three forms (estrone (E1), estradiol (E2),
interesting recent article, Zhang et al. developed a novel and estriol (E3)), E2 is the crucial for the progression of
transferrin protein corona (Tpc)-modified CuGd nano- breast, endometrial, and ovarian cancers [85, 86]. Estro-
platform (Tpc-CuGd) for tumor-targeting photothermal gen function relies primarily on its binding and subse-
and chemodynamic synergistic therapy [75]. quent activation of two structurally different estrogen
In summary, various Tf-modified nanoparticles have receptors (ERα and ERβ) [87]. Therefore, these related
been developed for the targeted delivery of therapeutic receptors are considered members of the nuclear recep-
agents to tumor sites, which can preferentially deliver tor superfamily.
therapeutics into TfR-overexpressing tumor cells by It has been reported that following intracellular uptake
receptor-mediated internalization [76], showing excellent of estrogen-modified nanoparticles by receptor-mediated
antitumor effects with few side effects. endocytosis, intracellular ERs can carry these nanopar-
ticles toward the nucleus for nuclear targeting [88]. Fur-
Human epidermal growth factor receptor 2 thermore, these receptors have been found overexpressed
(HER2)‑mediated active targeting on several tumor cell surfaces. In a recent application,
The HER family, comprising HER1, HER2, HER3, and Kapara and co-workers [89] reported a straightforward
HER4, plays a crucial part in the pathogenesis of vari- and non-destructive 3D surface-enhanced Raman spec-
ous tumors including gastric and breast cancer [77, 78]. troscopy (SERS) imaging strategy to track the cellular
HER-targeting-based strategies may address tumor internalization of AuNPs modified with an anti-ERα Ab
chemoresistance as their associated receptors usually in MCF-7 cells. It was found that these modified nano-
possess tyrosine kinase catalytic activity [79]. Among particles were effectively internalized by tumor cells
these, the HER2 receptor is commonly studied in breast using the ERα receptor-mediated endocytosis process for
cancer as it is overexpressed > 20% of patients [80]. While enhanced tumor treatment.
the HER2 receptor does not have a natural ligand, it can
dimerize with other ErbB family receptors, which results Cluster of differentiation (CD) receptor‑mediated active
in activation of the HER signaling pathways [79]. A sig- targeting
nificant challenge in developing targeted drugs has been The CD receptor family comprises surface receptors
the identification and preparation of HER2-specific mainly present on cancer stem cells (CSCs), including
artificial ligands with specificity and colloidal stability. CD14, CD22, CD36, CD44, and CD133, which can be
Recently, a variety of monoclonal antibodies (Abs) and used as promising delivery targets against tumor metas-
their fragments, as well as some peptide drugs, have been tasis. Among them, CD44, a transmembrane adhesion
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 7 of 40
glycoprotein, has been commonly used to target recep- chemokine signaling can modulate the normal immune
tors for targeted tumor treatment [90–92]. Hyaluronic function in epithelial cells because CXCR4 plays a signifi-
acid (HA), a ligand with good biocompatibility, has been cant role in normal cell growth and angiogenesis [105].
widely used in CD44 receptor-mediated active target- There is there an urgent need to design novel chemokine
ing delivery systems. It can be readily obtained due to inhibitors that do not disturb the function of healthy cells.
its abundance as a natural polymer compared with poly- A number of LHRH receptors have been found in breast,
mers that require multiple step chemical synthesis [93, ovarian and prostate cancer, but their expression is low
94]. HA-functionalized nanoplatforms can effectively or absent in the corresponding healthy tissues [106, 107].
deliver therapeutic agents to tumor cells through CD44 Therefore, several nanoplatforms modified with LHRH
receptor-mediated active targeting, with an excellent peptides have been explored for the targeted delivery of
cytotoxic profile and tumor kill. For example, Kim et al. therapeutic agents [108–111]. For instance, LHRH pep-
[94] reported a HA modified, trio-stimuli receptive and tide conjugated nanoparticles prepared by Tang and co-
on-demand triggerable nanoplatform for multimodal workers enhanced cellular uptake and tumor suppression
cancer treatment. These HA-enveloped nanoparticles in comparison to the non-LHRH targeted formulations
effectively suppressed tumor growth in comparison to [112]. Moreover, Taheri and co-workers designed LHRH
groups without HA modification. In general, HA is modi- peptide-functionalized methotrexate-encapsulated nano-
fied on the surface of nanoparticles to specifically bind particles with higher therapeutic efficacy against cancer
to CD44 receptors that are overexpressed in tumor cells, [113]. In addition, Zhang’s group reported the anti-cancer
thus mediating tumor endocytosis. In addition, HA has ability of LHRH receptor-targeted mitoxantrone-encapsu-
the tendency to be degraded to smaller fragments in lated versatile nanoplatforms in vivo, demonstrating aug-
the presence of hyaluronidase which is also abundantly mented tumor suppression with the targeted liposomes in
present in the TME [95]. The versatile characteristics of comparison to non-targeted formulations [111]. Although
HA as a targeted and enzyme-responsive ligand make these receptor-mediated strategies have shown potential
it a promising candidate for application in specific drug advantages for drug delivery, several factors, such as ligand
delivery systems. stability, orientation, and density, must be taken into con-
sideration to preserve the function of the targeting ligand.
Other receptor‑mediated active targeting systems
In addition to the receptors mentioned above, other recep- Stimuli‑responsive targeting strategies
tors have also been used to design targeted anti-cancer Unique features of the TME include an acidic envi-
nanoplatforms, including chemokine, biotin, and lutein- ronment, a high concentration of GSH and ROS, and
izing hormone-releasing hormone (LHRH) receptors [96– increased expression of specific enzymes (MMP-2/cath-
100]. For example, chemokine receptor type 4 (CXCR4) epsin B). Therefore, nanoparticles incorporating TME-
is a class of G-protein-coupled receptor that plays an responsive components can pave the way for targeted
important part in tumor metastasis by gathering tumor drug delivery and tumor treatment. In response to these
cells along chemokine gradients. Several peptide-func- endogenous stimuli, alterations in molecular function
tionalized nanoplatforms have been prepared for targeting and dispersion behavior, morphology, and degradation
CXCR4 receptor-positive cancers. For example, Alberi- kinetics can be induced. This facilitates either intracel-
cio et al. developed circular peptide T22-functionalized lular internalization or escape from endosome/lysoso-
mesoporous silica for the effective delivery of chemothera- mal degradation and release of pharmaceutical active
peutic agents to tumor cells [101]. Wang and co-workers ingredients [114]. In addition to endogenous respon-
prepared epirubicin-encapsulated polymeric nanoparticles sive nanosystems, some exogenous stimuli-responsive
that clearly improved therapeutic efficacy in hepatocellular nanoparticles also show beneficial targeting behavior by
carcinoma by conjugating the LFC131 peptide to increase utilizing controllable external factors, such as lasers, tem-
the affinity [102]. Similarly, Murakami’s group also devel- perature, ultrasound, and magnetism. Several examples
oped cellulose nanoparticles with the LFC131 peptide of endogenous and exogenous responsive nanoplatforms
for targeted tumor treatment [103]. Xiao et al. designed a are presented below (Fig. 4).
novel nanoplatform to target CXCR-4 to effectively induce
p53 expression in hepatocellular carcinoma models. Com- Endogenous Stimuli‑responsive targeting strategies
bining the CXCR4-targeted p53 mRNA nanoplatform with Redox‑responsive targeting strategies
anti-PD-1 treatment effectively induced cellular repro- Redox species in tumor cells form a complex antioxidant
gramming and immune components of the tumor micro- defense system to modulate redox homeostasis, play-
environment in established hepatocellular carcinoma ing an important role in the cell life cycle [115]. Com-
models [104]. It should be noted that the suppression of mon reactive oxygen species (ROS) include hydroxyl
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 8 of 40
radicals (·OH), singlet oxygen (1O2), and hydrogen perox- ROS‑responsive targeting strategies Hypoxia, which can
ide (H2O2). It should be noted that H2O2 is a stable and cause tumorigenesis and cancer progression, has been
nontoxic ROS, while others have a short half-life and can considered as a significant biomarker in cancer theranos-
be effectively transformed into powerful toxic reagents tics and targeted treatment. Moreover, it is controlled by
[116, 117]. On the one hand, H2O2 can be used as a sub- the overgeneration of VEGF and hypoxia induced factor
strate for O2 production with the aid of a specific enzyme (HIF-1α) in tumor cells [130], resulting in decreased sen-
to alleviate tumor microenvironment hypoxia in some sitivity of cancers to radiotherapy (RT), causing chemore-
O2-demanding therapeutic strategies [118]. On the other sistance and also greatly affecting the efficacy of O2-related
hand, the H 2O2 can also be converted into other highly treatments, such as photodynamic therapy (PDT) and
active ROS, including 1O2, O2·− and ·OH [116]. This sonodynamic therapy (SDT) [131]. Recently, researchers
increased ROS can result in oxidative stress, such as lipid have developed versatile ROS-responsive nanoparticles
peroxidation (LPO), and protein and DNA impairment through catalase (CAT)-mediated tumor site-specific
[119]. In addition, glutathione (GSH), as an antioxidant, O2 generation to alleviate hypoxia for enhancing tumor
is commonly distributed in tumor cells at concentrations treatment [132]. For instance, Zhang et al. developed
up to 2–10 mM, playing a significant role in consuming liposomes loaded with a cisplatin-prodrug functional-
ROS and modulating redox homeostasis [120, 121]. Fur- ized phospholipid and CAT [133], alleviating the chem-
thermore, a high GSH concentration can make tumor oresistance caused by hypoxia. Further, the liposome
cells resistant to various treatments [122]. Therefore, it encapsulation also endowed the prepared nanoplatforms
is advantageous to develop redox-sensitive nanoparti- with satisfactory biocompatibility and a high tumor accu-
cles for the delivery of therapeutic agents to trigger treat- mulation profile. Treatment with the designed liposomes
ments such as chemodynamic therapy (CDT) (Fig. 5). In induced the highest level of DNA impairment in tumor
addition, to further improve the therapeutic profile, ROS cells exposed to X-rays in comparison to the control
generation combined with GSH depletion can effectively group. In addition, a range of nanocarriers with CAT
disturb redox homeostasis to augment oxidative stress, mimicking activity, including MOF, M nO2, CeO2, Pt, and
thus resulting in tumor cell apoptosis [123]. ROS are gen- Pd [131, 134–138], have also shown greatly potential in
erated by the partial reduction of O2 which is necessary nanomedical applications. It should be noted that MnO2
for maintaining the normal function of aerobic organisms is well known to convert H 2O2 into O2 under the action of
using energy provided from four electron reduction reac- the acidic TME with the disruption of M nO2-based nano-
tion [124–129]. As shown in Table 2, most efforts have particles [138, 139]. These ROS-responsive nanoparticles
been to develop ROS-responsive building blocks, which capable of stimulating tumor site-specific O 2 production
can be combined with chemotherapeutics to achieve provide a practical strategy for improving the sensitivity
excellent antitumor activity with few side effects. of RT and chemotherapy.
Fig. 4 Schematic representation of exogenous and endogenous stimuli-responsive nanoplatforms for tumor therapy
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 9 of 40
Fig. 5 (A) Schematic illustration of amplified oxidative stress based on intracellular ROS for incurring tumor cell apoptosis. B Schematic illustration
of the anti-metastasis performance of the GSH-responsive nanoplatforms
Hydroxyl radicals (·OH) are not only an important been devoted to replenishing H2O2 in tumor cells [143].
component of ROS, but also the main product of the Fen- Among these strategies, iron-based nanoparticles have
ton reaction for tumor-targeted therapy [140]. Moreover, been widely applied to generate highly toxic ·OH for
iron-free Fenton nanocatalysts including transition promotes H2O2-sensitive transformation from insoluble
metal-based, precious-metal-based, sulfide-based nano- to soluble forms. Poly (propylene sulfide) conjugated with
catalysts and their composites multifunctional radical PEG can rapidly self-assemble into nanoparticles and
therapeutics have been developed [145]. decompose upon confrontation with H 2O2, suggesting
Recently, Chen and colleagues prepared copper per- great promise as a delivery platform. Overall, nanopar-
oxide (Cu2O2) nanoparticles with the features of revers- ticles containing H2O2-sensitive groups are expected to
ible degradation to generate self-supplying H2O2 through become more widely used in stimuli-triggered disintegra-
changes in Ph [146]. The H2O2 could be effectively cata- tion and specific cancer treatment [150, 151].
lyzed by C u2+ to generate highly toxic ·OH. These nano-
particles showed improved tumor inhibition efficacy in Reactive nitrogen species (RNS)‑responsive targeting strat‑
comparison to the controls. Additionally, H2O2 has also egies NO, the first gas molecule for therapy, has attracted
been used for NO-based gas treatment. This has been attention because of its excellent diffusivity and cell mem-
called a “green” treatment approach for cancer therapy, as brane penetration, endowing it with broad biological
it shows minimal toxicity for normal tissues while offer- activities and therapeutic potential [152–158]. It has been
ing metabolic benefits that are not achievable through reported that matrix metalloproteinases (MMPs), which
chemotherapy or other traditional therapeutic modali- comprise a family of enzymes that can degrade matrix
ties [147]. For example, Chen and colleagues developed proteins, are capable of depletion of collagen through acti-
mesoporous silica nanoparticles as biocompatible nano- vation of NO, resulting in improved penetration ability of
vehicles for the delivery of arginine and glucose oxidase the prepared nanoparticles [159, 160]. In addition, NO
(Gox) [148]. These nanoparticles used encapsulated Gox can react with 1O2 to generate highly toxic peroxynitrite
to provide a degrading glucose reaction to increase intra- (ONOO−) which has a stronger tumor cell killing abil-
cellular H2O2 concentration, which can then oxidize argi- ity [161]. ONOO- can convert pro-MMPs into MMPs to
nine into NO under the action of specific NO synthase. degrade the extracellular matrix to enhance the penetra-
As the levels of glucose increase, the tumor microenvi- tion ability of nanoparticles and induce DNA impairment.
ronment became more acidic, allowing H 2O2 to facilitate NO and ONOO- can cause mitochondrial dysfunction by
the NO production. After treatment with the designed reducing mitochondrial membrane potential and inhib-
nanoparticles, tumor volumes were considerably reduced iting the generation of ATP, which effectively suppresses
and the mice had longer survival times. In another mode, ATP-related tumor-derived vesicles and tumor metas-
H2O2 can be used as a stimulus for the disruption of nan- tasis [162]. Moreover, the derived RNS and superoxide
oparticles, leading to controllable release. For instance, can effectively kill cancer cells by inducing nitrosative or
a novel oxidation-sensitive polymeric carrier has been oxidative stress, DNA or mitochondrial impairment and
used to prepare antitumors nanoplatforms [149]. Among improving inflammatory reactions, resulting in acceler-
them, poly (propylene sulfide) as a hydrophobic block ated cell apoptosis [147, 163–165]. However, there are still
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 11 of 40
some concerns regarding the delivery of NO by different novel nanoprodrug had a high CPT loading efficiency
nanomaterials due to difficulties in obtaining efficient and exhibited rapid drug release when exposed to GSH.
encapsulation and precise release [166]. To overcome the Additionally, the photothermal effect of the cocaine dyes
drawbacks of the current NO delivery carriers and NO further facilitated disulfide linker cleavage. The encap-
donors, an interesting approach was reported to transport sulated croconaine dyes endowed this nanoparticle with
NO for tumor treatment using prodrug self-assembling NIR fluorescence and photoacoustic imaging properties
nanoplatforms of NO donors. Briefly, phenylsulfonyl- for tumor treatment.
furoxan was used as the NO donor in the synthesis of a Platinum drugs (e.g., cisplatin, carboplatin and oxali-
prodrug using ester and disulfide bonds. The insertion of platin) currently remain the most commonly used
disulfide bonds facilitates the self-assembly of polymers chemotherapeutic agents against a number of tumors
in solution. Subsequently, the multiresponsive tumor- [190, 191]. However, there are numerous problems with
targeting NO nanoparticles can be obtained by adding FA these drugs in clinical use, such as lack of specificity and
onto the surface, which can achieve the effective delivery severe side effects on normal organs. Therefore is a grow-
of NO to tumor regions, leading to accurate NO release ing tendency to develop prodrug-based nontoxic Pt(IV)
and inducing tumor cell apoptosis [167]. Researchers have s that can be converted into highly toxic Pt(II) through
also developed other NO donors, such as Roussin’s black the reduction of GSH [192]. Farokhzad’s group developed
salt, metal NO complexes, and S-nitrosothiols [168–171]. self-assembled nanoparticles comprised of PEGlipid and
NO may also relieve hypoxia in the tumor area through Pt (IV) prodrug for tumor treatment [193]. On one hand,
vasodilation, which promotes PDT efficacy [172], further this nanoscale strategy facilitated the delivery of cargoes
improving the combined effects of PDT and NO in can- across cell membranes into cells by endocytosis. On
cer therapy. To improve penetration into tumor tissue in another other, these prodrugs had a GSH-depleting fea-
PDT-mediated tumor treatment, researchers typically ture, resulting in the release of Pt(II) to act on DNA and
combine rare-earth up-conversion nanomaterials with trigger tumor cell apoptosis.
different photosensitive therapeutic agents [173, 174]. In addition to the above GSH-responsive nanoparti-
However, this poses new risks in the preparation and cles, multivalent metal ions such as Fe2+ and Fe3+ Cu+
biosecurity of such nanoparticles. The combination of and Cu2+ and Mn2+ and Mn4+also show GSH-responsive
ROS and RNS responsive strategies into the same nano- behavior due to a shift in valency, [194]. These reduced
particles with good biological safety can be expected to metal ions can be further applied for diagnosis or
provide an efficient and all-in-one anticancer treatment. improved treatment. In one example, ultrasmall (4 nm)
cerium oxide nanoparticles (CeO2 NPs) were rapidly
GSH‑responsive targeting strategies A number of nano- etched, leading to the opening of nanochannels in the
carriers comprised of disulfide bonds, carbon-diselenide mesoporous silicon nanoparticles when exposed to vita-
bonds, diselenide bonds, or a sulfonyl group [175–182] min C or GSH, resulting in controlled antitumor drug
have been prepared by cross-linking reactions. Overgen- release [195]. Recently, our group prepared versatile Cu-
erated GSH can effectively break various disulfide bonds, MOF nanoparticles loaded with VK3 for enhanced CDT
thus causing disintegration of nanoparticles and accurate by regulating GSH and H 2O2 in the tumor microenviron-
cargo release in cancer cells. It should be noted that nano- ment [196, 197]. Cu+ and Cu2+ showed better catalytic
platforms with disulfide bonds embedded in mesoporous capability than classical Fe-dependent Fenton agents.
silica nanoparticles show fast biodegradation and are The satisfactory antitumor effects presented by these Cu-
emerging as promising nanovehicles [183]. based nanoparticles, and the cascade-enhanced chemo-
For the development of GSH-sensitive nanoplatforms, chemodynamic therapy approach provide an opportunity
the co-assembly of amphipathic block copolymers and for the application of such novel nanoplatforms for HCC
therapeutic agents with GSH-responsive groups into sev- treatment. Furthermore, future advancements, such
eral nanosystems (such as liposomes, nanoparticles, and as improved targeting, can effectively improve the effi-
micelles) has been considered as a potential application cacy and use of such approaches, which should be ben-
approach [184]. Nanoplatforms bearing GSH-cleavable eficial to cancer treatment [196, 197]. Additionally, the
prodrugs have also been developed, which can be effec- consumption of GSH plays an important role in metal-
tively modulated to toxic therapeutic agents by exces- based chemodynamic therapy. For instance, Liu et al.
sive intracellular GSH [185–188]. For example, Sun et al. developed advanced metal-based nanoparticles through
loaded a trimeric prodrug into FA functionalized polylac- chemodynamics for multimodal tumor treatment [198].
tic-coglycolic acid hybrid nanoparticles, where the chem- GSH acted on the designed nanoparticles and effectively
otherapeutic camptothecin (CPT) was conjugated to reduced Mn4+, Mn3+, and Cu2+ into Mn2+ and Cu+,
NIR croconaine dyes through disulfide bonds [189]. This accompanied by GSH consumption. Inductively coupled
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 12 of 40
plasma optical emission spectrometry (ICP-OES) was tumor accumulation of the loaded therapeutic agents and
used to support the rapid release of Cu and Mn from the facilitating the release of cargoes in the acidic tumoral
nanoparticles in an acidic environment containing GSH. microenvironment [205]. Currently, researchers typically
The nanoparticles displayed specific recognition and use changes in chemical structure (such as changes in
homotypic targeting profiles to MCF-7 cells. Combin- hydrophilicity through deprotonation and protonation)
ing metal ions with GSH-consumption in the TME could and acid-sensitive chemical bonds to design pH-respon-
become a more promising targeted strategy for CDT sive nanoplatforms (Fig. 6). Additionally, the designed
pH-sensitive formulations usually have the ability to pro-
ROS and GSH dual‑responsive tumor‑targeting strat‑ tect several therapeutic agents and vehicles for tumor
egy As mentioned above, intracellular redox regulation therapy from being trapped in endosomes [206, 207].
has been considered an effective strategy against cancer. Generally, the encapsulation of chemotherapeutics inside
However, the ROS produced from the catalytic oxida- pH-responsive nanoparticles is an efficient approach for
tion of H
2O2 can be removed by the overgenerated GSH, prolonging the blood circulation time of the encapsu-
compromising therapeutic interventions. To overcome lated agents and their retention inside the nanoparticles
this, selective enhancement of oxidative stress through in a physiological environment. Moreover, pH-responsive
depleting GSH levels and simultaneously elevating ROS nanoparticles are also able to improve the pharmacoki-
concentrations can be a specific and promising strategy in netics and biodistribution of the encapsulated payload.
cancer treatment [199]. For instance, Liang and colleagues This is essential for delaying metabolism and the subse-
designed an oxidative stress-amplified nanoplatform for quent release of drugs.
disturbing mitochondrial redox balance, which com-
prised atomically dispersed Au anchored onto a carbon- Protonation and deprotonation‑based nanoplat‑
dot surface modified with cinnamaldehyde and triph- forms Protonation and deprotonation are widely used
enylphosphine [200]. The acidity of endosomes facilitates mechanisms for pH-sensitive nanocarriers in tumor
the dissociation of cinnamaldehyde. Subsequently, the treatment. pH-responsive nanoplatforms including
nanoparticles rapidly react with GSH, accompanied by polyelectrolytes, such as poly(aspartic acid-graft-imi-
ROS generation, resulting in the elevation of ROS and the dazole), cationic poly(β-amino ester) (PBAE), anionic
simultaneous reduction of GSH. As a result, levels of mito- poly(Asp), PDMAEMA, polysulfonamide, poly(histidine)
chondrial GSH in tumor cells were obviously decreased (poly(His)), and poly(acrylic acid) (PAA), are shown in
after incubation with the prepared nanoparticles. In addi- Table 3. In an advanced strategy to design biocompatible
tion, the prepared nanoparticles with enhanced oxida- nanoparticles, it has also been proposed that biodegrad-
tive stress possessed excellent anticancer effects against able materials such as enzyme-responsive chitosan and
HepG-2 tumors. These groups of designed nanoparticles certain polypeptides can be used for protonation and
also showed prolonged survival times and few side effects deprotonation-based nanomaterials through function-
against various tumor models, which can be attributed to alization with an acid-responsive group to the backbone
the fact that normal tissues, unlike the TME, do not have of biodegradable materials [208–210]. These materials
high redox levels. generally contain –COOH as anionic groups and –NH2
as cationic groups combined with other hydrophobic
pH‑responsive targeting strategies or hydrophilic molecules, which can be further used in
pH-sensitive nanocarriers have been extensively explored pH-responsive nanoplatforms through protonation and
to design versatile nanoplatforms for targeted drug deliv- deprotonation.
ery. The TME usually has a lower extracellular pH (pHex) Cationic materials with -NH2 groups can effectively
with a mean value of ~ 6.5 in comparison to healthy tis- protonate in an acidic environment and show excellent
sue [201]. Generally, compared with healthy cells, tumor hydrophilicity, while they can deprotonate in a neu-
cells rapidly consume glucose for glycolysis with rapid tral environment to show hydrophobicity. In contrast,
lactate production to obtain the energy required for anionic materials with –COOH groups [211–214] can
maintaining their proliferation regardless of oxygen also deprotonate and protonate in the opposite way.
content; consequently, the higher metabolism rate of For instance, the groups of imidazole can be easily pro-
tumor cells has been recognized as a major cause of the tonated under acidic conditions, as they have a pair of
acidic TME [202]. Additionally, tumor cell, lysosomes electrons on the unsaturated N atoms, leading to the con-
and endosomes also have a lower pH (endosomal pH version of hydrophobic to hydrophilic states, which can
(pHen)) in comparison to pHex [203, 204]. Therefore, incur disintegration of the nanocarriers and consequently
pH-responsive nanoplatforms have been developed as release the loaded therapeutic agents. Besides, poly(His)-
an effective tumor treatment tool, greatly enhancing the PEG shows obvious nanoscale core–shell micelles in a
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 13 of 40
Fig. 6 Schematic illustration of pH-responsive nanoplatforms for the delivery of therapeutic agents. The nanoplatforms can effectively accumulate
in the tumor sites via the EPR effect. In the tumor microenvironment, acidic conditions can effectively trigger drug release for tumor treatment
neutral environment consisting of the hydrophilic PEG released in an acidic environment because of the destruc-
shell and hydrophobic cores of poly(His) by deprotona- tion of the hydrophobic cores. In another example, Oh
tion. However, the protonation of poly(His) responds et al. also prepared pH-responsive micelles using amphi-
to His groups and destabilizes micelles because of the philic polyelectrolytes for docetaxel delivery. Similarly,
reduced hydrophobicity of poly(His) at pHex. Further, the prepared micelles exhibited good colloidal stabil-
the poorly soluble therapeutic agents encapsulated in ity under physiological conditions, while they became
the core of the pH-responsive micelles can be effectively unstable due to protonation of the imidazole group under
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 14 of 40
acidic conditions. Therefore, the docetaxel-encapsulated ticular, acid-sensitive chemical bonds have been inten-
micelles have pH-responsive release behavior due to sively investigated for pH-responsive nanocarriers, such as
structural changes induced by protonation of the imi- esters, imines, and hydrazine. These acid-sensitive bond-
dazole groups in the amphiphilic polyelectrolytes. pH- based nanoplatforms have been proven to be relatively
responsive release can also lead to high stability in blood stable in physiological environments but are easily bro-
circulation, a decrease in the toxicity of healthy tissues, ken via nucleophilic substitution reactions under acidic
and increased drug availability. conditions [216]. Further, the acid-sensitive bonds can
Additionally, some materials with anionic polyelectro- be directly conjugated to the therapeutic agents as labile
lytes have also been used to prepare pH-sensitive nano- groups in nanoparticles. For example, with the protona-
carriers for drug delivery [215]. However, the strategy tion of labile compounds containing C=N bonds (such as
of taking advantage of anionic amphiphilic molecules hydrazone bonds, imine groups, and oxime bonds) under
to prepare pH-responsive and tumor-targeted nanocar- acidic conditions, they can be readily susceptible to nucle-
riers can be different from that utilizing cationic mate- ophilic substitution by H 2O because of the increased elec-
rials. Under acidic conditions, such as pHen and pHex, trophilicity of carbon atoms [217–220]. Hydrazone link-
anionic polymers containing -COOH groups can exist as ages in particular, with satisfactory acid responsiveness
protonated (hydrophobic) units and are not applicable and a fast degradation rate, have been widely used in dif-
to tumor-targeted micelles from amphiphilic polymer ferent pH-responsive nanoplatforms such as liposomes,
blocks rich in anionic groups. As a result, anionic poly- nanoparticles, and micelles [210, 221–225]. Additionally,
mers can be encapsulated with some chemotherapeutics acid-responsive groups can also be applied to improve
such as DOX using hydrophobic interactions in physi- the limited cargo release from the nanoparticle core and
ological environments, and therapeutic agents can be target-cell interactions because of PEGylation. For exam-
effectively released for specific tumor treatment under ple, Wu et al. synthesized hydrazone linker-functionalized
acidic conditions via weakened interactions owing to liposomes to address the problems with PEGylation. As
protonation. expected, the hydrazone bond-functionalized liposomes
exhibited satisfactory lysosomal escape properties and
Acid‑sensitive bond cleavage‑based nanoparticles As enhanced tumor accumulation in comparison to normal
previously described, the acidity differences between the liposomes [226]. However, imine bonds have poor stabil-
various compartments of tumor cells and between tumors ity under physiological environments due to the loss of
or healthy tissues have received widespread consideration mesomeric effects in comparison to hydrazone bonds
for designing pH-responsive chemical structures. In par- [227]. Therefore, researchers have made significant efforts
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 15 of 40
to improve the stability of imine bonds by introducing tion showed clearly enhanced DOX release under simu-
strong π–π conjugated systems such as benzene rings lated tumor microenvironment conditions and in tumor
[228, 229]. For example, Wang et al. prepared versatile cell models [235]. In addition, Zhang’s group synthesized
biomimetic nanoparticles based on formyl benzoic acid- camptothecin-encapsulated mesoporous silica nanopar-
PEG-maleimide functionalized mesoporous silica against ticles surface functionalized with targeting cRGD and
metastatic triple-negative breast cancer. After loading of MMP-2 responsive fluorescence imaging groups, acting as
immune adjuvant and photothermal therapeutic agents, a diagnostic platform as well as for tumor location. These
the immune peptide was then linked to the surface of advanced nanoplatforms were able to efficiently undergo
nanoparticles through acid-responsive benzoic-imine enzymatic hydrolysis in overexpressed MMP-2 environ-
bonds. It was applied to the therapy of metastatic triple- ments to improve tumor treatment by the release of their
negative breast cancer through immune remodeling and cargoes [236]. Further, some amphiphilic block copoly-
photothermal ablation [230]. Taken together, these low- mers (such as PCL-PEG) are suitable for the preparation
pH-responsive nanoparticles can effectively release the of versatile delivery platforms against cancer. For instance,
encapsulated chemotherapeutic agents upon encounter- PTX-encapsulated PEG-PCL nanoparticles function-
ing the acidic TME in targeted cancer therapy. alized with activated low molecular weight protamine
showed satisfactory targeted glioma effects. Further, these
Enzyme‑responsive targeting strategies advanced nanoparticles also exhibited enhanced MMP-
Enzymes, being a significant component of the nano- dependent cellular internalization, increased cytotoxic-
biotechnology toolbox, have exceptional biorecognition ity, and augmented tumor suppression in glioma models
abilities as well as excellent catalytic properties. Gener- [237]. In another study, Yang et al. prepared a versatile
ally, abnormal enzyme expression observed in cancer nanoparticle based on MMP-sensitive Au nanoparticles
provides many opportunities for designing targeted nan- for tumor-specific photoacoustic imaging-guided tumor
oparticles modified with enzyme-responsive linkages. treatment and drug delivery. The Au nanoparticles could
Recently, many smart nanoparticles have been prepared be further grafted with complementary DNA strands,
for intracellular as well as extracellular tumor-specific functionalized with PEG and conjugated with therapeutic
drug delivery based upon enzyme expression at the target agents through MMP-responsive peptides and thermal-
site. sensitive linkers, respectively. As a result, the developed
nanoparticles showed augmented efficiency in tumor
MMP‑responsive nanoplatforms Matrix metallopro- treatment and photoacoustic imaging in comparison to
teinases (MMPs), which are overexpressed in various MMP-inert nanoparticles [238]. Similarly, hydrophilic
types of tumors, are closely related to cancer patho- siRNA and poorly soluble drugs could be effectively code-
physiology. MMP-2 and MMP-9 in particular have been livered using versatile micelles prepared by MMP-2-re-
explored for preparing enzyme-responsive nanoplatforms sponsive copolymers. The prepared nanoplatform showed
[231, 232]. For example, Yamada et al. prepared two PTX satisfactory colloidal stability and enhanced endocytosis
prodrugs by conjugating an octapeptide (AcGPLGIAGQ) efficiency in different tumor cell lines and significant pas-
with PTX at different sites that could be effectively broken sive targeting behavior in tumor-bearing models. Mallik
down by MMP2 in the tumor microenvironment. These et al. prepared an MMP-9 responsive nanoplatform using
nanoparticles can effectively release PTX to inhibit can- collagen-simulated lipoprotein conjugated to PEG cleav-
cer cell proliferation [233]. Among various natural materi- able polymers to encapsulate Gem. The designed enzyme
als, gelatin is an example of a biocompatible polymer that stimuli-responsive nanoparticles demonstrated a faster
can be degraded by MMPs and promote cargo release in Gem release rate treated with MMP-9 and a higher tumor
tumor sites. For instance, Wang et al. developed MMP- inhibition ratio in comparison to MMP-inert nanoparti-
responsive PVA-peptide conjugates for achieving self- cles [239]. Yang’s group [240] designed an advanced MMP
assembly with enhanced tumor accumulation, capable stimuli-responsive nanoplatform encapsulated with the
of improving PD-L1 blocking efficiency for augmented chemotherapeutic agent curcumin using a block copoly-
immunotherapy. Once the self-assembled nanoplatforms mer with surface-adsorbed peptides that could improve
entered the TME, the enzyme-cleavable peptide could be endocytosis. The prepared nanoplatform showed a sus-
immediately degraded under the action of overexpressed tained curcumin release behavior under physiological
MMPs to effectively release cargoes for cancer treatment conditions, while release could be accelerated under con-
[234]. Furthermore, gelatin-functionalized DOX-loaded ditions that mimic the tumor microenvironment. There is
mesoporous silica nanoparticles have been applied for the no doubt that these designed MMP-responsive nanopar-
delivery of therapeutic agents against MMP-9 overgener- ticles present excellent tumor specificity and therapeutic
ated in cancer. As expected, the prepared nanoformula- efficacy in cancer models with few side effects
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 16 of 40
Heparanase‑responsive nanoplatforms Glycosamino- only in tumor cells but also in tumor-related endothelial,
glycans and structural proteins together constitute the fibroblast, myoepithelial, and osteoclast cells as well as
matrix of the tumor tissues, of which the main compo- leukocyte cells [248]. A great number of studies have been
nent of glycosaminoglycans is heparan sulfate proteogly- undertaken on the design and development of cathepsin-
can [241]. Furthermore, heparan sulfate proteoglycan is responsive nanoplatforms, particularly on Gly-Phe-Leu-
actively involved with various biological factors (including Gly, which has been commonly applied as a spacer that
VEGF, TGF-β, and b-FGF) that play an important role in can be effectively degraded in overexpressed cathepsin B
the interaction between normal cells and tumors. In addi- environments [249]. For example, Xia et al. prepared a pH
tion, some reports have pointed out that highly metastatic and cathepsin B dual-responsive nanovaccine that specif-
and malignant cancers frequently over generate hepara- ically targeted endosomal Toll-like receptors (TLRs) for
nase-1, which can degrade HSPGs in the tumor micro- enhanced tumor vaccination. In vivo results showed excel-
environment, causing enhanced secretion of the above lent prophylactic and antitumor effects of the nanovac-
bioactive factors and the consecutive triggering of related cine against tumor-bearing mice. This endosome-targeted
pathways resulting in cancer metastasis, epithelial-mes- responsive nanovaccine approach provides a promising
enchymal transition, and neovascularization. Addition- delivery platform for adjuvants to promote the design
ally, the new spaces formed in the matrix can also result and preparation of cancer nanovaccines [250]. Accurate
in cancer invasion and metastasis [242]. It has also been assessment of cathepsin B expression in vivo may pro-
reported that heparanase-1 can effectively degrade hepa- vide a potential approach for early tumor diagnosis [251].
rin, suggesting a potential novel nanovehicle with hepara- Taking advantage of precise photoacoustic imaging, an
nase sensitivity for drug delivery. intelligent photoacoustic probe Cypate-CBT, which could
An example of utilizing heparin-prepared nanoplat- effectively assemble into cypate-containing nanoprobes in
forms is to bind heparin molecules via GSH-responsive response to overgenerated GSH and cathepsin B in tumor
disulfide bonds to construct heparin-based nanogels cells, was prepared by Liang’s group [251] for the accurate
[243]. Another approach involved the construction of a and specific monitoring of cathepsin B. In comparison
nanocomplex through electrostatic interactions between to unmodified Cypate, this nanoprobe showed a higher
protamine and heparin for effective loading of positively photoacoustic signal in cathepsin B-positive breast can-
charged therapeutic agents. Researchers have devel- cer models, supporting the intracellular accumulation of
oped similar versatile nanoparticles with polyelectrolyte the nanoprobes after cathepsin B-triggered self-assembly.
complexes encapsulating small therapeutic agents [244]. The cathepsin B-responsive nanoprobe can be employed
Isothermal titration calorimetry and real-time dynamic as an efficient photoacoustic imaging agent for the early
swelling spectroscopy have been used to explore the diagnosis and targeted therapy of cancer.
underlying mechanisms and principles for the fabrication
of advanced nanoplatforms via intermolecular electro- Hypoxia‑responsive targeting strategies
static interactions [245]. During optimization of the man- Hypoxia, considered a significant hallmark of solid
ufacturing process, the polyelectrolyte nanocomplex can tumors, has been observed in more than 60% of cancers
be developed with appropriate negative surface charges [252, 253]. The partial pressure of O 2 (pO2) is generally
and particle size [246]. 1,2-Dioleoyl-3-trimethylammo- approximately 40–60 mm Hg in normal tissues while it
nium-propane (DOTAP), a positively charged phospho- is less than 10 mm Hg in tumor tissues, and even as low
lipid compound widely applied to construct cationic as 0–2.5 mm Hg in some cases [254–256]. The O2 con-
liposomes, has been applied to encapsulate hydrophobic sumed by tumor cells exceeds supply leading to this path-
chemotherapeutic agents through liposomes formation ological phenomenon. Abnormally vigorous metabolism
[247]. When codelivered therapeutic agents nanoplat- and cell growth in tumor cells can deplete intracellular
forms enter the tumor microenvironment, overexpressed O2. Secondly, the vascular system in the tumor tissue
heparanase-1 can rapidly recognize the outer heparin is disordered, resulting in an insufficient supply of O 2.
shell and cleave it, resulting in the release of cargoes Finally, the short O2 diffusion distance (less than 200 μm)
for tumor cell kill [242]. This also causes the positively cannot meet the demand of tumor cells further away
charged nanoparticle core to be exposed to the cancer from the blood vessels [257–259]. To better adapt to this
cells, and efficient endocytosis of nanoparticles by tumor harsh living environment, hypoxic cancer cells must alter
cells can be achieved with this approach. some of their biological characteristics, such as upregu-
lating the levels of HIF-1α, carbonic anhydrase IX (CA
Cathepsin‑sensitive nanocarriers Recently, researchers IX), and other enzymes [260].
have demonstrated that a variety of cathepsins are overex- As a result, enhanced cancer metastasis and poor ther-
pressed in different types of tumors. These are found not apeutic effects are usually evident in hypoxic-stimulated
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 17 of 40
tumors [261, 262]. For instance, the hypoxic microenvi- cells, such as azoreductase, nitroreductase, methio-
ronment can result in overexpression of HIF-1α, which nine synthase reductase, inducible nitric synthase and
is capable of modulating gene expression relevant to DT-diaphorase (DTD) [274, 275]. Hence, considerable
tumor proliferation, invasion and metastasis to facilitate efforts have been made to develop hypoxia-responsive
the resistance to O 2-dependent antitumor strategies, nanoplatforms that can be activated by these enzymes
such as chemotherapy, PDT and RT [263–266]. In addi- for enhanced tumor treatment based on the above find-
tion, hypoxia-adapted tumor cells generally do not have ings [274, 276, 277]. Hypoxia-responsive chemical bonds
the rapid division characteristics of normal tumor cells, (Table 4), including nitro groups, azo groups, quinone
so they are not sensitive to conventional chemotherapeu- and N-oxide compounds, are also applied in the con-
tic agents that interfere with DNA replication [267, 268]. struction of hypoxia-responsive nanoparticles. They can
Further, O2 plays an important part in repairing DNA change their conformation and physicochemical char-
dysfunction after radiation treatment (such as X-ray and acteristics such as hydrophobic features and electron
γ-ray) during RT, and it is the source of PDT or SDT- affinity by gaining or losing their electrons [278, 279]. As
mediated ROS applied to fight tumors [267, 269–273] anticipated, such hypoxia-responsive nanoparticles have
Hypoxia of tumor tissue is generally considered to been found to exhibit satisfactory performance for drug
indicate poor prognosis for tumor treatment, but par- delivery. They have great potential for tumor treatment
ticular biological features can make it a specific target for including hypoxia-responsive cargo release, prolonged
cancer therapy. In fact, tumor cells prefer aerobic glycol- blood circulation time, and enhanced tumor penetra-
ysis to obtain energy rather than the conventional oxida- tion and accumulation. Below we discuss the chemical
tive phosphorylation pathway due to the Warburg effect. structures that can be used to design effective hypoxia-
Therefore, many enzymes related to electron donation or responsive nanoparticles and the strategies for taking
reduction response are overgenerated in hypoxic tumor
codelivery of therapeutic agents to tumors, but also pro- et al. designed a novel nanoplatform capable of regulat-
vided novel ideas for designing and preparing advanced ing the tumor microenvironment via Fenton reaction-
materials for multimodality tumor treatment. based chemodynamic therapy. This nanoplatform utilizes
Azoreductase-sensitive organic ligands can also be glucose-mediated continual O2 consumption to create a
used to construct nanoscale coordination complexes and localized hypoxic microenvironment for enhanced tira-
bring hypoxia-sensitive characteristics to nanothera- pazamine-mediated chemotherapy. The production of
nostics for tumor diagnosis and treatment. For example, exogenous H2O2 by GOx facilitates the release of Fe3+
Huang et al. [306] prepared an azoreductase-triggered from the nanoparticles to convert H2O2 into the highly
nanocomplex where the encapsulated chemotherapeu- cytotoxic ∙OH. This versatile nanoplatform showed
tic DOX and siRNA were capable of downregulating the enhanced tumor accumulation and excellent antitumor
expression of HIF-1α, thus reducing multidrug resist- efficacy in tumor-bearing models [318]. Yang et al. pre-
ance. 4,4′-Azobisbenzoinc acid, as the main ligand of pared nanoparticles capable of enhancing tumor hypoxic
the nanocomposites, was effectively reduced by azore- levels by loading vascular disruption agents that cut off
ductase to release the encapsulated DOX and siRNA the O2 supply. As a result, the designed nanoparticles
in the hypoxic tumor cells. In another example, Zhou not only suppressed tumor proliferation but also effec-
et al. [292] reported on an aptamer/antibody nanofor- tively inhibited tumor metastasis [319]. Moreover, PDT
mulation functionalized with hypoxia-sensitive AZO can be used as an excellent strategy to enhance tumor
compounds capable of decreasing off-target effects. In hypoxia and improve tirapazamine-mediated chemo-
this nanoformulation, a conditional aptamer was conju- therapeutic effects through the transformation of 3O2 to
1
gated with hydrophilic polymers containing AZO groups O2. For example, Yan et al. encapsulated tirapazamine
which played an important role in preventing binding into the pores of porphyrinic-based MOFs on the sur-
to normal cells. The hydrophilic block polymers could face of lanthanide-doped upconversion nanoparticles to
be detached from the nanoparticles through the reduc- prepare a versatile nanotheranostic agent. The cell and
tion of AZO, allowing aptamer/antibody recognition of animal experiment data showed that the combination
the cancer cell surface in a hypoxic microenvironment. of tirapazamine and PDT yielded enhanced therapeu-
Mesoporous silica nanoparticles are another important tic efficacy. Further, the integration of nanotheranostic
platform as inorganic drug delivery carriers for tumor agents with anti-programmed death-ligand 1 (anti-PD-
treatment. They have the advantages of low side effects, L1) clearly decreased the tumor volume at distant sites by
good biocompatibility and stability, relatively uniform improving immune infiltration [320].
size and a large specific surface area [307, 308]. For exam- Banoxantrone dihydrochloride (AQ4N) can not only be
ple, Jang and colleagues developed hybrid mesoporous selectively activated in hypoxic tumor cells but can also
silica nanoparticles functionalized with β-cyclodextrin be reduced under the action of reductases [321, 322]. The
and 4-(phenylazo) benzoic acid for improved on-demand protonated form (1,4-bis([2-(dimethylamino-N-oxide)
drug release. As expected, the nanoparticles displayed ethyl]amino)5,8-dihydroxy-anthracene-9,10-dine (AQ4)
improved selective drug release and significant cytotoxic- containing two tertiary amine groups) can utilize DNA
ity in comparison to nonresponsive nanoparticles [309]. intercalation to strongly suppress topoisomerase II [323,
324]. Thus, Feng et al. prepared a multipurpose liposome
Oxide groups to encapsulate soluble banoxantrone dihydrochloride and
The N-oxide group can also be used in the design and poorly soluble 64Cu-hCe6 into the cavity and lipid layer of
preparation of hypoxia-responsive nanoplatforms for the liposomes, respectively. Severe local hypoxia induced
effective tumor treatment [310]. Tirapazamine and ban- by Ce6 under laser irradiation could activate the antican-
oxantrone dihydrochloride are the most studied agents. cer activity of banoxantrone dihydrochloride, resulting in
Tirapazamine is an aromatic N-oxide compound while improved therapeutic efficacy in tumor-bearing models
banoxantrone dihydrochloride is an aliphatic N-oxide [325].
derivative, exhibiting higher cytotoxicity in hypoxic can- In another study, Yang et al. prepared novel organo-
cer cells than in normal cells [311–315]. silica nanoparticles containing tetrasulfide bonds to
In hypoxic tumor cells, tirapazamine can produce radi- encapsulate banoxantrone dihydrochloride and GOx for
cal species that break DNA through a single-electron tumor treatment [326]. Overexpressed GSH in tumor
reduction reaction catalyzed by various intracellular cells can effectively cleave the tetrasulfide bonds to dis-
reductases, leading to irreversible damage and apoptosis rupt the nanoparticles leading to the release of banox-
[316]. Because of the specific responsive strategy, nano- antrone dihydrochloride and GOx. Subsequently, GOx
particles that elevate tumor hypoxia can significantly can consume O 2 and glucose to produce H 2O2, thereby
enhance the antitumor effects of tirapazamine [317]. Guo exacerbating the hypoxia and further promoting the
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 20 of 40
and H2O or hydroxyl radicals. Various theranostic nano- nanoparticles could also decompose water to generate
agents have been developed based on manganese (Mn) to O2 following laser irradiation, and subsequently trans-
induce Fenton-like reactions. form O2 into toxic 1O2 for tumor kill. The excellent pho-
Recently, Chen et al. designed an excellent TiO-porphy- tothermal conversion efficiency significantly enhanced
rin-based nanoplatform (FA-TiOPs) by loading TiO-por- blood circulation to the hypoxic tumor microenviron-
phyrin in FA-modified liposomes. These liposomes could ment. Additionally, Tang et al. designed a nanosensor for
effectively catalyze H 2O and overexpress H2O2, in situ the tracking and assessment of non-small cell lung can-
producing active ROS. Furthermore, TiO-porphyrin cer using near-infrared excited hypoxia imaging in which
could photo-split H 2O to generate H 2O2, ∙OH radicals, the acceptor and donor pairs within a biological MOF
and O2. The increased O2 concentration not only allevi- matrix are precisely controlled to rationalize upconver-
ated the hypoxic tumor microenvironment but could also sion Förster resonance energy transfer. It was found to be
be further converted by TiO-porphyrin into 1O2 to kill beneficial both in vitro and in in vivo zebrafish models.
cancer cells. Furthermore, researchers have reported that To overcome the limitations of hypoxia treatment
the high energy of TiO-porphyrin in the excited state and using photodynamic therapy, several radical generators
the narrow gap energy between the triplet excited state [354] that do not consume O2 have been developed. For
and the excited state may facilitate effective photocata- example, Dong et al. [355–357] have developed several
lytic reactions. In addition, overgenerated H2O2 in tumor advanced organic superoxide radical photo-generators
cells could also be catalyzed to produce 1O2, particu- to effectively address problems existing in hypoxia treat-
larly in an acidic environment, exerting active antitumor ment with PDT. In one instance, they formed highly
effects and preventing damage to healthy tissues. Overall, efficient photosensitizers to carry out type I PDT elimina-
tumor-targeted liposomes provided adequate ROS to the tion of hypoxic tumor tissues by vascular disruption. The
tumor through several in situ photocatalytic reactions in vitro and in vivo results showed that these nanopar-
that are O2 dependent and achieve effective cancer inhi- ticles could not only overcome the hypoxia paradox but
bition. Other therapeutic agents such as immunostimu- also suppress cancer metastasis through treatment with
latory or chemotherapy drugs can also be encapsulated type I PDT in 4TI breast cancer cell mouse models [355].
into such hypoxia-responsive nanoparticles through con- pH-sensitive zinc (II) metalated porphyrin nanoparti-
jugation or loading strategies for effective tumor-targeted cles were also prepared by this group to track and treat
therapy. cervical cancer tumor-bearing mice. Interestingly, they
observed that the phototherapy effects of the prepared
Other hypoxia‑responsive nanoplatforms With increas- nanoparticles could be effectively activated by increased
ing attention being paid to the hypoxic TME, a growing acidity [357]. Taken together, these hypoxia-responsive
number of strategies have been developed using external nanoparticles are making substantial progress in target-
stimuli, such as lasers. Xu et al. [351] designed an NIR ing tumor sites and enhancing therapeutic efficacy.
laser-controlled O2/Pt2+ self-producing prodrug (UCPP)
to enhance PDT efficacy in the hypoxic TME allow- Interstitial fluid pressure (IFP)‑related targeting strategies
ing combined photo-chemotherapy. The nanosystem The IFP in healthy tissues is only about 0–3 mm Hg,
included Pt4+ and Ce6, in which upconversion nanoparti- while tumors show an IFP of around 5–130 mm Hg [358].
cles were encapsulated to transform 980 nm near-infrared It should be noted that interstitial fibrosis and abnor-
light into 365 nm and 660 nm emissions to decompose mal lymph vessels and blood are considered the primary
Pt4+ and initiate Ce6-mediated PDT. The decomposition reasons for increased IFP [358, 359]. Elevated IFP can
of Pt4+ produced O 2 for depletion in the PDT process serve as an obstacle to the delivery of therapeutic agents,
and released Pt2+ for chemotherapy. Therefore, this novel because of drops in convection between the extravas-
nanosystem achieved enhanced tumor accumulation and cular and intravascular spaces, resulting in restricted
satisfactory tumor suppression in mouse xenograft mod- drug delivery to the tumor tissues. Further, it also cor-
els with no recurrence. relates with high recurrence rates in some tumors (such
Another emerging method is the use of lasers to as gynecological cancers) [360, 361]. Recently, some pre-
decompose abundant water molecules in living organ- liminary studies have reported that appropriate hyper-
isms to relieve hypoxic TME. Thus, Zheng et al. [352] thermia can effectively decrease intratumoral IFP to
prepared C3N4-based versatile nanoparticles to trigger facilitate tumor treatment. The intravenous administra-
the decomposition of H2O and produce O2 after expo- tion of chemotherapeutic agent-encapsulated liposomes
sure to 630 nm laser irradiation to reverse hypoxia- has been applied in combination with two ablative heat-
mediated PDT tolerance. In another example, Jiang et al. ing approaches to appropriate hyperthermia and coagu-
[353] reported that prepared ultrathin graphdiyne oxide lative ablation. For example, Zhao et al. reported that a
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 22 of 40
two-step ablation (45 ℃ for 2 min and 70 ℃ for 3 min) in mouse models [370]. Tang and co-workers prepared
conjunction with liposomes could obtain a survival ben- switchable aptamer micelle flares conjugated to a diacyl
efit in comparison to administering nanoformulations lipid chimera, which can monitor intracellular ATP [371].
with a single heating approach in a Balb/c mice bearing These micelles showed benefits for cell permeability and
4T1 tumor model [359]. Designing a versatile hyper- molecular imaging, with potential for tumor diagnosis
thermia therapeutic nanoparticle appears to provide a and targeted delivery. In summary, ATP can be consid-
promising potential approach for the improvement of the ered an efficient stimulus to promote release of preloaded
targeted drug delivery. drugs from nanoparticles for specific cancer treatments
and for diagnostic purposes
ATP‑responsive targeting strategies
ATP, which has been called “the energy currency of the Exogenous stimuli‑responsive targeting strategies
cell,” is fundamental to various cellular signal cascades.
ATP concentrations can reach up to 10 mM in tumor Temperature stimuli‑responsive targeting strategies
cells, while it is only approximately 5 mM in the extra- Temperature stimuli-responsive nanoplatforms have
cellular fluid. Therefore, a concentration gradient of ATP been designed for tumor treatment. Ideal tempera-
levels between extracellular and intracellular levels has ture-responsive materials with a lower critical solution
been used to develop ATP-responsive nanoplatforms for temperature include poly(2-oxazo line)s (POxs), poly-
tumor treatment. For instance, Kataoka’s group reported N-isopropylacrylamide (PNIPAAm), poly(methyl vinyl
ATP-responsive micelles for the delivery of siRNA to ether) (PMVE), and poly(vinyl caprolactam) (PNVCL),
tumors. Because of competitive binding between micelles which can readily undergo solid-to-liquid phase transi-
and ATP, the designed micelles could be crosslinked with tions according to external conditions [372–376]. Among
extracellular ATP but collapsed because of intracellular these materials, PNIPAAm has been most commonly
ATP, resulting in the efficient release of loaded siRNA used for preparing nanoplatforms as it has a lower critical
[362]. Aida et al. developed protein-based nanoplatforms solution temperature of approximately 30 ℃. As an exam-
to release ATP-sensitive agents for tumor treatment. ple, Grüll and co-workers prepared temperature stimuli-
The nanocarrier was prepared using various barrel- responsive liposomes loaded with therapeutic agents for
shaped chaperonin groups assembled via coordination high intensity focused ultrasound-mediated targeted
with Mg2+ into tubular structures that protected loaded delivery. As expected, temperature stimuli-responsive
therapeutic agents from biological metabolism and deg- release of cargoes was observed along with enhanced
radation [363]. Upon internalization by tumor cells, endocytosis of therapeutic agents by the tumor cells
hydrolysis of ATP to form ADP can trigger protein con- [377]. In another example, Deng et al. prepared DOX-
formational changes and collapse of the nanoparticles, encapsulated temperature stimuli-responsive liposomes
resulting in the selective release of the contents [363]. surface functionalized with iRGD peptide (CCRGDKG-
ATP ligands have been developed for monitoring ATP PDC) for targeted tumor treatment. In combination with
using several sensors, including electrochemical, colori- high intensity focused ultrasound-mediated temperature
metric, and fluorescent platforms [364–368]. Wang et al. stimuli-responsive DOX release, the designed liposomes
prepared nanoparticles complexed with PEI hybridized were specifically internalized by αvβ3-positive tumor
with the ATP-responsive ligands, siRNA and DOX. The cells, with good treatment efficacy [378]. As explained
prepared nanoparticles using a gradient of ATP concen- above, temperature stimuli-responsive nanoparticles
trations showed rapid cargo release in an ATP-respon- can have a significant impact at the cellular level, which
sive manner. An enhanced anti-proliferative effect was potentiates the cytotoxicity of certain active pharma-
observed, possibly due to enhanced cell apoptosis in ceutical ingredients, mostly explained by changes in
mitochondria-mediated pathways and cell cycle arrest the pharmacokinetics of the agent under hyperthermic
at the G2 phase [369]. In another example, Gu et al. conditions and associated cellular changes, resulting in
designed ATP-binding aptamer DNA functionalized increased nanoparticle uptake.
with polymeric nanocarriers encapsulated with chemo-
therapeutics for targeted delivery to ATP overgenerated Magnetic stimuli‑responsive targeting strategies
environments. In comparison to non-ATP responsive Over the past few decades, magnetic stim [379–381].
nanogels, the ATP-responsive nanogels achieved sig- Moreover, considering that magnetic nanoparticles have
nificant therapeutic effects in various tumor cell lines. excellent physiochemical performances and biological
Furthermore, functionalization with hyaluronic acid for effects, they have been proposed as ideal platforms for a
tumor-specific targeting accompanied by ATP respon- number of tumor theranostics [382]. Encapsulated super-
siveness improved tumor inhibition in tumor-bearing paramagnetic Fe2O3 nanoparticles (SPIONs) present an
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 23 of 40
excellent magnetic moment and satisfactory biocompat- guided by ultrasound stimuli. Under ultrasound treat-
ibility in comparison to other magnetic stimuli-respon- ment, the designed nanoparticles exhibited satisfactory
sive nanoplatforms. Furthermore, magnetic molybdenum drug release, cellular uptake and other pharmacokinetic
disulfide (mMoS2) can be modified by liposomes with a characteristics, as well as superior antitumor efficacy in
phospholipid bilayer membrane structure to construct glioma models [392]. Zheng’s group prepared ultrasound
magnetically responsive nanoplatforms which do not stimuli-responsive DOX-loaded mesoporous silica nano-
easily aggregate in physiological solutions, and have particles featuring the ultrasound-responsive release of
good biocompatibility, thereby showing great promise for cargoes for glioma treatment. They showed an obvious
nanomedicine applications [383]. suppression in tumor invasiveness and growth, as well as
Successful application of magnetically responsive nan- increased survival in a mouse glioma model [393]. Mura-
oplatforms includes encapsulation/immobilization of gaki’s group developed epirubicin-encapsulated micelles
therapeutic agents into magnetic-responsive nanoplat- as tumor sonosensitizers. Using HIFU, the nanoparticles
forms, injection of the magnetic stimuli-responsive nano- could be disrupted to allow drug release in canine spon-
platforms into the body and taking advantage of external taneous chondrosarcoma, osteosarcoma, hepatocellular
magnetic fields to recruit and activate the magnetic stim- and prostate cancer [394]. Biocompatible piezoelectric
uli-responsive nanoplatforms at the lesions of interest nanoparticles have also been encapsulated with DSPE-
[384–386]. Recently, Shuai et al. [387] reported a GSH- PEG and modified with anti-HER2 Ab for targeted breast
responsive MOF to effectively load IDO inhibitor, and cancer treatment. As anticipated, these designed ultra-
NO donor s-nitrosothiol groups for improving antitumor sound stimuli-responsive nanoplatforms can effectively
immunotherapy. In this nanoplatform, the high T1 relax- release encapsulated active pharmaceutical ingredients
ivity endows magnetic resonance (MR) imaging capabili- in a controlled manner, interfering with cell division and
ties to detect the in vivo biodistribution of nanoagents. inhibiting tumor proliferation
Shi et al. reported a versatile nanodiagnostic based on
DOX-encapsulated tannic acid-Fe networks (TAFs) Laser stimuli‑responsive targeting strategies
functionalized with fibronectin for combination cancer Laser stimuli can break light-sensitive functional bonds
treatment under the guidance of MR imaging. In this sys- or groups, including coumarinyl ester, truxylic acid and
tem, the TAF network allows the nanodiagnostic to have pyrenyl methyl ester. Much work has focused on employ-
excellent r1 relaxivity for T1-weighted MR cancer imag- ing these laser-responsive nanoplatforms to deliver
ing [388]. The development of magnetic stimuli-respon- chemotherapeutic agents by destroying the nanovehi-
sive nanoparticles with imaging properties will help to cle at the lesions [395–399]. For example, Chen et al.
determine when there is good tumor accumulation. prepared a photolabile spherical nucleic acid for light-
responsive codelivery of antisense oligonucleotide and
Ultrasound stimuli‑responsive targeting strategies siRNA. Upon exposure to an NIR laser, the prepared
Ultrasound has become an excellent external stimu- nanoplatforms rapidly oxidized and dissociated with con-
lus capable of facilitating the disruption of nanoparti- tinuous responsive release, resulting in a positive effect
cles and releasing their cargoes at the lesions of interest on tumor treatment [400]. Xu et al. designed a light stim-
[389]. Ultrasound stimuli-responsive nanoplatforms can uli-responsive nanoparticle to achieve long blood circu-
therefore be a valuable tool for enhancing therapeutic lation, enhanced tumor accumulation and penetration,
agent accumulation in tumors with low EPR effects. For and rapid body elimination in an imaging-guided treat-
example, SDT-based nanoparticles capable of continu- ment. After the nanoplatform accumulated in the tumor
ous production of CO2 have been recently developed to regions, the cargoes could be effectively released by laser
accomplish ultrasound-mediated inertial cavitation irradiation. Importantly, the released therapeutic agents
(UIC) to augment ROS accumulation against cancer could effectively penetrate the whole tumor tissue with a
[390]. The in vitro and vivo results indicated that con- diameter of approximately nine millimeters giving tumor
tinuous UIC accelerated a massive generation of ROS, suppression [401]. Additionally, Kim’s group prepared a
resulting in the improvement of SDT using a single laser-responsive and biomimetic nanoplatform for deep
nanoplatform. Furthermore, the highly-accumulative tumor penetration [402]. In this study, the in vitro drug
ROS arising from continuous UIC have been shown to release profile and tumor cell inhibition rate were signifi-
induce robust immunogenic cell death (ICD), which is cantly improved after laser irradiation. The use of lasers
typically represented by increased antigen exposure and as an exogenous stimulus can effectively improve the
presentation, enhanced DC maturation and more acti- therapeutic effect and reduce the side effects by control-
vated CD8+T cell infiltration in tumors [391]. Price et al. ling the drug release behavior.
prepared cisplatin encapsulated solid lipid nanoparticles
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 24 of 40
Hybridization and combination of cancer these stimuli will be employed to load the drug into the
nanomedicine nanoparticles and trigger the drug release. Additionally,
Designing smart targeting nanoparticles with stimuli- the activation of drug release under external stimuli,
responsive profiles has proved promising for providing including a magnetic field, temperature, light or ultra-
site-specific, accurate, and systemic drug administra- sound, can also be achieved at the targeted site. Owing
tion. Moreover, stimuli-responsiveness can substantially to the complex TME, including the abnormal expression
increase the diverse utility of such systems by integrating of multiple receptors, high redox potential, and abnor-
drug administration with other features, such as sensing, mal metabolic conditions, smart nanoparticles have been
imaging, or monitoring. The designed smart nanoparti- specially developed for anticancer medication. Studies
cles can accumulate in the tumor region through either using stimuli-responsiveness and targeting strategies are
passive targeting behavior (EPR effects) or receptor- detailed in Table 5.
mediated active targeting strategies. Subsequently, such For example, a smart dual-responsive and target-
nanoparticles provide yet another possibility to fine tune ing nanoplatform was prepared for the codelivery of
their response toward each stimulus individually, ena- chemotherapeutics (DOX and PTX) for treatment of
bling drug release to be precisely controlled under the lung adenocarcinoma [403] (Fig. 7). In this nanoplat-
cumulative effect of multiple stimuli. In these nanopar- form, FA was used as a receptor-mediated targeting mol-
ticles, multiple impulses are integrated to activate nano- ecule to facilitate the entry of these nanoplatforms into
particles in the TME by introducing exogenous stimuli, tumor cells. Moreover, acid-liable block copolymers and
such as laser and ultrasound. In such systems, one of disulfide bonds endowed the nanoplatform with pH and
Fig. 7 Schematic illustration of stimuli-responsive and targeted nanoplatforms for the specific delivery of therapeutic agents. A Preparation
of the smart nanoplatform via electrostatic and hydrophobic interaction, and the pH-responsive surface charge switch, and GSH-responsive
chemical degradation of polymer backbone. B Schematic illustration of and FA-mediated target and pH/GSH-responsive delivery processes:
(a) the nanoplatforms show high stability in blood circulation; (b) therefore, they can effectively accumulate in tumor lesions via the EPR effect
and receptor-mediated targeting; (c) acidic conditions can cause charge conversion of the nanoplatform; (d) endosome escape of the smart
nanoplatform via proton-sponge effect; (e) intracellular GSH stimulation will trigger the release of therapeutic agents for tumor treatment
GSH-responsive drug release behavior in the TME. It nanoplatform showed good biocompatibility, excellent
should be noted that the prepared nanoplatforms exhib- cellular internalization, and improved tumor cell inhibi-
ited a surface charge switch from negative to positive tion. Furthermore, the nanoplatform appeared synergis-
during transmission from physiological environment to tic and improved solid tumor killing efficiency compared
the TME, which can enhance tumor cells internalization. with mono-chemotherapy in tumor-bearing mice mod-
Subsequently, endosome escape of the nanoplatforms els. This suggests that hybridization and combination of
was achieved in the acidic endo/lysosome environment cancer nanomedicines present great promise for tumor
via the "proton-sponge" effect. As expected, this smart treatment. In another important example, Jeong Hoon
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 26 of 40
Byeon’s group [404] developed a platform for digitiz- their physiological environment. For example, most car-
able and continuous-flow manufacture in a compact bon nanomaterials (such as carbon nanoparticles and
and reconfigurable manner using a serial combination of nanotubes) and metals (such as MnO) can act efficiently
plug-in reactionwares. This platform comprised three dif- in acidic environments but generate ROS near tumors
ferent composite nanocompounds with photothermally which can lead to cancer progression and metastasis
modulatable and structurally degradable characteris- [410, 411]. Notably, ROS-responsive nanoplatforms tend
tics for cancer treatment. As expected, these nanocom- to be rapidly phagocytized due to their special surface
pounds used for NIR-triggered chemothermal cancer properties [412, 413].
therapy showed excellent anticancer efficacy with low In the future, there is an urgent need to control the
side effects and effective renal excretion. Taken together, physicochemical features of nanoparticles to improve
the hybridization and combination of nanomedicine their targeting ability, especially their morphology, parti-
appears to hold great promise for cancer treatment. cle size distribution and surface chemistry. For example,
new surface modification strategies need to be explored
Conclusions and prospects to confer novel multifunctionalities to the nanoparticles.
In this review, we have focused on recent advances in Moreover, in order to improve the antitumor effects of
receptor-mediated and stimuli-responsive active tar- nanoparticles the development of alternative reactions,
geting strategies for cancer treatment. These versatile formulations, or constructs containing stimulus compo-
nanoparticles effectively overcome undirected drug bio- nents aimed at producing multiple strategies for highly
distribution, undesired toxicity and high doses of admin- effective combination cancer treatment should be a
istration, and play an important role in the development focus. Importantly, these new generation targeting strat-
of novel chemotherapeutic agents and the understand- egies should be explored for an in-depth understanding
ing of their antitumor efficacy. Significant progress has of key parameters, such as their pharmacokinetics, bio-
been made in developing target-specific therapies lead- distribution and nano-bio interfacial interactions, as such
ing to better cellular internalization and site-specific outcomes have a significant impact on cancer treatment.
agent release by exploiting specific cancer cell surface Furthermore, there are possibilities to develop novel
receptors. These active targeting strategies not only stimuli-responsive modalities for better encapsulation
enhance the efficacy of the drug but also reduce poten- of agents as well as their controlled release to further
tial side effects. Additionally, stimuli-responsive target- increase their therapeutic index with few side effects. It
ing strategies with their unique characteristics have also is forecast that nanoscale biomaterials comprising bio-
shown high stability, enhanced tumor accumulation, compatible lipids, polymers or inorganic materials in
and rapid release behaviors in response to exogenous conjugation with targeting groups will have tremendous
or endogenous environmental stimuli both in vitro and scope for transporting pharmaceutical active ingredi-
in vivo. Taken together, precise delivery and specific ents to their specific target sites for improved therapeutic
release can be readily achieved by using the synergistic purposes. Such versatile targeted nanoparticles will find
effects between versatile receptor-mediated and stimulus broader application possibilities and will aid in the role
response targeting strategies, resulting in killing cancer out of personalized/precision medicine.
cells within the tumor without damaging healthy tissues.
While the abovementioned approaches have many ben-
Abbreviations
efits, there are also some caveats. The size and surface EPR: Enhanced permeability and retention; HA: Hyaluronic acid; TME: Tumor
characteristics of nanoplatforms can disrupt membranes microenvironment; FRs: Folate receptors; Tf: Transferrin; FA: Folate acid; EGFRs:
and interfere with protein folding and membrane activ- Epidermal growth factor receptors; HER2: Human epidermal growth factor
receptor 2; DOX: Doxorubicin; PTX: Paclitaxel; TGF-α: Transforming growth
ity. These intracellular dysfunctions can further trigger factor-α; IFP: Interstitial fluid pressure; MOF: Metal organic framework; UIC:
feedback mechanisms such as “frustrated phagocytosis” Ultrasound-mediated inertial cavitation; POxs: Poly(2-oxazo line)s; PMVE: Poly
[405]. Once administrated, the prepared nanoplatforms (methyl vinyl ether); NMR: Nuclear magnetic resonance; GOx: Glucose oxidase;
AZO: Azobenzene; Ce6: Chlorin e6; NQO1: NAD(P)H quinone dehydrogenase
circulate in the bloodstream to access various tissues 1; GSH: Glutathione; PEG: Polyethylene glycol; CA IX: Carbonic anhydrase IX;
or organs. During this circulation, these nanoplatforms Ab: Antibody; ATP: Adenosine triphosphate; TZ: Trastuzumab; PTT: Photo-
can interact with biomacromolecules (including carbo- thermal therapy; PDT: Photodynamic therapy; CDT: Chemodynamic therapy;
RT: Radiotherapy; CAT: Catalase; ROS: Reactive oxygen species; RNS: Reactive
hydrates, proteins, nucleic acids, and lipids) which can nitrogen species; MR: Magnetic resonance; MMP: Matrix metalloproteinase;
coat the nanoplatforms, leading to a surface or biomol- TaOx: Tantalum oxide; RBCs: Red blood cells; ICD: Immunogenic cell death;
ecule corona, which alters the surface properties of the PNIPAAm: Poly-N-isopropylacrylamide; PNVCL: Poly(vinyl caprolactam); PK: Pro-
tein kinase; ICP-OES: Inductively coupled plasma optical emission spectrome-
nanoplatforms, affects their therapeutic effects, and can try; CD: Cluster of differentiation; ECM: Extracellular matrix; CPT: Camptothecin;
induce protein unfolding [406–409]. Stimuli-responsive DOTAP: 1,2-Dioleoyl-3-trimethylammonium-propane; AQ4N: Banoxantrone
nanoplatforms are effective but can still be affected by dihydrochloride; DTD: DT-diaphorase; TKIs: Tyrosine kinase inhibitors.
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 27 of 40
28. Wang D, Zhou J, Fang W, Huang C, Chen Z, Fan M, Zhang MR, et al. A 49. Zhan C, Gu B, Xie C, Li J, Liu Y, Lu W. Cyclic RGD conjugated
multifunctional nanotheranostic agent potentiates erlotinib to EGFR poly(ethylene glycol)-co-poly(lactic acid) micelle enhances paclitaxel
wild-type non-small cell lung cancer. Bioact Mater. 2022;13:312–23. anti-glioblastoma effect. J Control Release. 2010;143(1):136–42.
29. Maruani A. Bispecifics and antibody-drug conjugates: a positive syn- 50. Long Q, Zhu W, Guo L, Pu L. RGD-conjugated resveratrol HSA nanopar-
ergy. Drug Discov Today Technol. 2018;30:55–61. ticles as a novel delivery system in ovarian cancer therapy. Drug Des
30. Matusewicz L, Filip-Psurska B, Psurski M, Tabaczar S, Podkalicka J, Devel Ther. 2020;14:5747–56.
Wietrzyk J, Ziółkowski P, et al. EGFR-targeted immunoliposomes as a 51. Babu A, Amreddy N, Muralidharan R, Pathuri G, Gali H, Chen A, Zhao
selective delivery system of simvastatin, with potential use in treatment YD, et al. Chemodrug delivery using integrin-targeted PLGA-chitosan
of triple-negative breast cancers. Int J Pharm. 2019;569: 118605. nanoparticle for lung cancer therapy. Sci Rep. 2017;7(1):14674.
31. Wang J, Zhang Y, Zhang G, Xiang L, Pang H, Xiong K, Lu Y, et al. 52. Fan Z, Chang Y, Cui C, Sun L, Wang DH, Pan Z, Zhang MJ. Near infrared
Radiotherapy-induced enrichment of EGF-modified doxorubicin nano- fluorescent peptide nanoparticles for enhancing esophageal cancer
particles enhances the therapeutic outcome of lung cancer. Drug Deliv. therapeutic efficacy. Nat Commun. 2018;9(1):2605.
2022;29(1):588–99. 53. Yadav AS, Radharani NNV, Gorain M, Bulbule A, Shetti D, Roy G, Baby
32. Rusdin A, Wathoni N, Motoyama K, Joni IM, Lesmana R, Muchtaridi M. T, et al. RGD functionalized chitosan nanoparticle mediated targeted
Nanoparticles targeted drug delivery aystem via epidermal growth delivery of raloxifene selectively suppresses angiogenesis and tumor
factor receptor: a review. J Pharm. 2019;1(3):77–91. growth in breast cancer. Nanoscale. 2020;12(19):10664–84.
33. Nan Y. Lung carcinoma therapy using epidermal growth factor recep- 54. Wang MD, Lv GT, An HW, Zhang NY, Wang H. In situ self-assembly of
tor-targeted lipid polymeric nanoparticles co-loaded with cisplatin and bispecific peptide for cancer immunotherapy. Angew Chem Int Ed
doxorubicin. Oncol Rep. 2019;42(5):2087–96. Engl. 2022;61(10):e202113649.
34. Kuo YC, Liang CT. Inhibition of human brain malignant glioblastoma 55. Yang RQ, Wang PY, Lou KL, Dang YY, Tian HN, Li Y, Gao YY, Huang WH,
cells using carmustine-loaded catanionic solid lipid nanoparticles Zhang YQ, Liu XL, Zhang GJ. Biodegradable nanoprobe for NIR-II
with surface anti-epithelial growth factor receptor. Biomaterials. fluorescence image-guided surgery and enhanced breast cancer
2011;32(12):3340–50. radiotherapy efficacy. Adv Sci. 2022;9(12):e2104728.
35. Liao C, Sun Q, Liang B, Shen J, Shuai X. Targeting EGFR-overexpressing 56. Holm J, Hansen SI. Characterization of soluble folate receptors (folate
tumor cells using cetuximab-immunomicelles loaded with doxorubicin binding proteins) in humans. Biological roles and clinical potentials
and superparamagnetic iron oxide. Eur J Radiol. 2011;80(3):699–705. in infection and malignancy. Biochim Biophys Acta Proteins Proteom.
36. Kim IY, Kang YS, Lee DS, Park HJ, Choi EK, Oh YK, Son HJ, et al. Antitumor 2020;1868(10):140466.
activity of EGFR targeted pH-sensitive immunoliposomes encapsu- 57. Lynn RC, Poussin M, Kalota A, Feng Y, Low PS, Dimitrov DS, Powell
lating gemcitabine in A549 xenograft nude mice. J Control Release. DJ Jr. Targeting of folate receptor β on acute myeloid leukemia
2009;140(1):55–60. blasts with chimeric antigen receptor-expressing T cells. Blood.
37. Aggarwal S, Yadav S, Gupta S. EGFR targeted PLGA nanoparticles using 2015;125(22):3466–76.
gemcitabine for treatment of pancreatic cancer. J Biomed Nanotechnol. 58. Shen F, Wu M, Ross JF, Miller D, Ratnam M. Folate receptor type gamma
2011;7(1):137–8. is primarily a secretory protein due to lack of an efficient signal for
38. Salehnia Z, Shahbazi-Gahrouei D, Akbarzadeh A, Baradaran B, Farajnia S, glycosylphosphatidylinositol modification: protein characterization and
Naghibi M. Synthesis and characterisation of iron oxide nanoparticles cell type specificity. Biochemistry. 1995;34(16):5660–5.
conjugated with epidermal growth factor receptor (EGFR) monoclonal 59. Scaranti M, Cojocaru E, Banerjee S, Banerji U. Exploiting the folate recep-
antibody as MRI contrast agent for cancer detection. IET Nanobiotech- tor α in oncology. Nat Rev Clin Oncol. 2020;17(6):349–59.
nol. 2019;13(4):400–6. 60. Assaraf YG, Leamon CP, Reddy JA. The folate receptor as a rational thera-
39. Shahbazi-Gahrouei D, Abdi N, Shahbazi-Gahrouei S, Hejazi SH, Salehnia peutic target for personalized cancer treatment. Drug Resist Updat.
Z. In vivo study of anti-epidermal growth factor receptor antibody- 2014;17(4–6):89–95.
based iron oxide nanoparticles (anti-EGFR-SPIONs) as a novel MR 61. Chen C, Ke J, Zhou X, Yi W, Brunzelle JS, Li J, Yong EL, et al. Structural
imaging contrast agent for lung cancer (LLC1) cells detection. IET basis for molecular recognition of folic acid by folate receptors. Nature.
Nanobiotechnol. 2020;14(5):369–74. 2013;500(7463):486–9.
40. Zhou X, Shi K, Hao Y, Yang C, Zha R, Yi C, Qian Z. Advances in nanotech- 62. Fernández M, Javaid F, Chudasama V. Advances in targeting the
nology-based delivery systems for EGFR tyrosine kinases inhibitors in folate receptor in the treatment/imaging of cancers. Chem Sci.
cancer therapy. Asian J Pharm Sci. 2020;15(1):26–41. 2017;9(4):790–810.
41. Xue EY, Yang C, Fong WP, Ng DKP. Site-specific displacement-driven 63. Hao Y, Li H, Zhao H, Liu Y, Ge X, Li X, Chen H, et al. An intelligent nano-
activation of supramolecular photosensitizing nanoassemblies for vehicle armed with multifunctional navigation for precise delivery of
antitumoral photodynamic therapy. ACS Appl Mater Interfaces. Toll-like receptor 7/8 agonist and immunogenic cell death amplifiers
2022;14(13):14903–15. to eliminate solid tumors and trigger durable antitumor immunity. Adv
42. Ganthala PD, Alavala S, Chella N, Andugulapati SB, Bathini NB, Sistla R. Healthc Mater. 2022;11(12):e2102739.
Co-encapsulated nanoparticles of Erlotinib and Quercetin for targeting 64. Janardhanam LSL, Bandi SP, Venuganti VVK. Functionalized LbL film for
lung cancer through nuclear EGFR and PI3K/AKT inhibition. Colloids localized delivery of STAT3 siRNA and oxaliplatin combination to treat
Surf B Biointerfaces. 2022;211:112305. colon cancer. ACS Appl Mater Interfaces. 2022;14(8):10030–46.
43. Wen L, Moser M, Ley K. Molecular mechanisms of leukocyte β2 integrin 65. Kefayat A, Hosseini M, Ghahremani F, Jolfaie NA, Rafienia M. Biodegrad-
activation. Blood. 2022;139(24):3480–92. able and biocompatible subcutaneous implants consisted of pH-sensi-
44. Hynes RO. Integrins: bidirectional, allosteric signaling machines. Cell. tive mebendazole-loaded/folic acid-targeted chitosan nanoparticles for
2002;110(6):673–87. murine triple-negative breast cancer treatment. J Nanobiotechnology.
45. Backer MV, Backer JM. Imaging key biomarkers of tumor angiogenesis. 2022;20(1):169.
Theranostics. 2012;2(5):502–15. 66. Sathiyaseelan A, Saravanakumar K, Manivasagan P, Jeong MS, Jang E-S,
46. Cabodi S, Di Stefano P, Leal Mdel P, Tinnirello A, Bisaro B, Morello V, Wang M-H. Folic acid conjugated chitosan encapsulated palladium
Damiano L, et al. Integrins and signal transduction. Adv Exp Med Biol. nanoclusters for NIR triggered photothermal breast cancer treatment.
2010;674:43–54. Carbohydr Polym. 2022;280:119021.
47. Sofias AM, Toner YC, Meerwaldt AE, van Leent MM, Soultanidis G, 67. Barz M, Canal F, Koynov K, Zentel R, Vicent MJ. Synthesis and in vitro
Elschot M, Gonai H, et al. Tumor targeting by αvβ3-integrin-specific evaluation of defined HPMA folate conjugates: influence of aggrega-
lipid nanoparticles occurs via phagocyte hitchhiking. ACS Nano. tion on folate receptor (FR) mediated cellular uptake. Biomacromol.
2020;14(7):7832–46. 2010;11(9):2274–82.
48. Rios De La Rosa JM, Spadea A, Donno R, Lallana E, Lu Y, Puri S, Caswell 68. Byrne JD, Betancourt T, Brannon-Peppas L. Active targeting schemes
P, et al. Microfluidic-assisted preparation of RGD-decorated nanopar- for nanoparticle systems in cancer therapeutics. Adv Drug Deliv Rev.
ticles: exploring integrin-facilitated uptake in cancer cell lines. Sci Rep. 2008;60(15):1615–26.
2020;10(1):14505.
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 29 of 40
69. Khan I, Gril B, Steeg PS. Metastasis suppressors NME1 and NME2 pro- 90. Hou Y, Jin J, Duan H, Liu C, Chen L, Huang W, Gao Z, et al. Targeted
mote dynamin 2 oligomerization and regulate tumor cell endocytosis, therapeutic effects of oral inulin-modified double-layered nanopar-
motility, and metastasis. Cancer Res. 2019;79(18):4689–702. ticles containing chemotherapeutics on orthotopic colon cancer.
70. Wu Y, Xu J, Chen J, Zou M, Rusidanmu A, Yang R. Blocking transferrin Biomaterials. 2022;283:121440.
receptor inhibits the growth of lung adenocarcinoma cells in vitro. 91. Zhu J, Guo M, Cui Y, Meng Y, Ding J, Zeng W, Zhou W. Surface
Thorac Cancer. 2018;9(2):253–61. coating of pulmonary siRNA delivery vectors enabling mucus
71. Luck AN, Mason AB. Structure and dynamics of drug carriers and their penetration, cell targeting, and intracellular radical scavenging for
interaction with cellular receptors: focus on serum transferrin. Adv Drug enhanced acute lung injury therapy. ACS Appl Mater Interfaces.
Deliv Rev. 2013;65(8):1012–9. 2022;14(4):5090–100.
72. Ge P, Liu Y, Chen Q, Su Z, Du Y, Luo S, Zhao X, et al. Transferrin recep- 92. Zhang Z, Qin S, Chen Y, Zhou L, Yang M, Tang Y, Zuo J, Zhang J, et al.
tors/magnetic resonance dual-targeted nanoplatform for precise Inhibition of NPC1L1 disrupts adaptive responses of drug-tolerant
chemo-photodynamic synergistic cancer therapy. Nanomedicine. persister cells to chemotherapy. EMBO Mol Med. 2022;14(2):e14903.
2022;39:102467. 93. Montanari E, Di Meo C, Oates A, Coviello T, Matricardi P. Pursu-
73. Zhang K, Li J, Xin X, Du X, Zhao D, Qin C, Han X, et al. Dual targeting ing intracellular pathogens with hyaluronan from a “Pro-Infection”
of cancer cells and MMPs with self-assembly hybrid nanoparticles polymer to a biomaterial for “Trojan Horse” systems. Molecules.
for combination therapy in combating cancer. Pharmaceutics. 2018;23(4):939.
2021;13(12):1990. 94. Poudel K, Banstola A, Tran TH, Thapa RK, Gautam M, Ou W, Pham
74. Ramalho MJ, Loureiro JA, Coelho MAN, Pereira MC. Transferrin receptor- LM, et al. Hyaluronic acid wreathed, trio-stimuli receptive and
targeted nanocarriers: overcoming barriers to treat glioblastoma. on-demand triggerable nanoconstruct for anchored combinatorial
Pharmaceutics. 2022;14(2):279. cancer therapy. Carbohydr Polym. 2020;249:116815.
75. Qi X, Wang G, Wang P, Pei Y, Zhang C, Yan M, Wei P, et al. Transferrin 95. Lei C, Liu XR, Chen QB, Li Y, Zhou JL, Zhou LY, Zou T. Hyaluronic acid
protein corona-modified CuGd core-shell nanoplatform for tumor- and albumin based nanoparticles for drug delivery. J Control Release.
targeting photothermal and chemodynamic synergistic therapies. ACS 2021;331:416–33.
Appl Mater Interfaces. 2022;14(6):7659–70. 96. Zheng N, Liu W, Li B, Nie H, Liu J, Cheng Y, Wang J, et al. Co-delivery of
76. Arumov A, Liyanage PY, Trabolsi A, Roberts ER, Li L, Ferreira BCLB, Gao sorafenib and metapristone encapsulated by CXCR4-targeted PLGA-
Z, Ban Y, Newsam AD, Taggart MW, Vega F, Bilbao D, Leblanc RM, Schatz PEG nanoparticles overcomes hepatocellular carcinoma resistance to
JH. Optimized doxorubicin chemotherapy for diffuse large B-cell sorafenib. J Exp Clin Cancer Res. 2019;38(1):232.
lymphoma exploits nanocarrier delivery to transferrin receptors. Cancer 97. Xiao K, Liu Q, Suby N, Xiao W, Agrawal R, Vu M, Zhang H, et al. LHRH-
Res. 2021;81(3):763–75. targeted redox-responsive crosslinked micelles impart selective drug
77. Shi F, Liu Y, Zhou X, Shen P, Xue R, Zhang M. Disitamab vedotin: a delivery and effective chemotherapy in triple-negative breast cancer.
novel antibody-drug conjugates for cancer therapy. Drug Deliv. Adv Healthc Mater. 2021;10(3):e2001196.
2022;29(1):1335–44. 98. Tian H, Zhang J, Zhang H, Jiang Y, Song A, Luan Y. Low side-effect and
78. Liu Y, Ye Z, Yang W, Hu Y, Zhao Y, Li Z, Xu B, Chen D, Tu J, Shen Y. A triple heat-shock protein-inhibited chemo-phototherapy nanoplatform via
enhanced permeable gold nanoraspberry designed for positive feed- co-assembling strategy of biotin-tailored IR780 and quercetin. Chem
back interventional therapy. J Control Release. 2022;345:120–37. Eng J. 2020;382:123043.
79. Ringhieri P, Mannucci S, Conti G, Nicolato E, Fracasso G, Marzola P, 99. Setrerrahmane S, Xu H. Tumor-related interleukins: old validated targets
Morelli G, et al. Liposomes derivatized with multimeric copies of for new anti-cancer drug development. Mol Cancer. 2017;16(1):153.
KCCYSL peptide as targeting agents for HER-2-overexpressing tumor 100. Zilio S, Bicciato S, Weed D, Serafini P. CCR1 and CCR5 mediate cancer-
cells. Int J Nanomed. 2017;12:501–14. induced myelopoiesis and differentiation of myeloid cells in the tumor.
80. Mitri Z, Constantine T, O’Regan R. The HER2 receptor in breast cancer: J Immunother Cancer. 2022;10(1):e003131.
pathophysiology, clinical use, and new advances in therapy. Chemother 101. De La Torre C, Casanova I, Acosta G, Coll C, Moreno MJ, Albericio F,
Res Pract. 2012;2012:743193. Aznar E, et al. Gated mesoporous silica nanoparticles using a double-
81. Mazzucchelli S, Truffi M, Fiandra L, Sorrentino L, Corsi FJWJoP. Targeted role circular peptide for the controlled and target-preferential release
approaches for HER2 breast cancer therapy: news from nanomedicine? of doxorubicin in CXCR4-expresing lymphoma cells. Adv Funct Mater.
World J Pharmacol. 2014;3(4):72–85. 2015;25(5):687–95.
82. Clark AS, Yau C, Wolf DM, Petricoin EF, van’t Veer LJ, Yee D, Moulder SL, 102. Di-Wen S, Pan G, Hao L, Zhang J, Xue Q, Wang P, Yuan Q. Improved
Wallace AM, et al. Neoadjuvant T-DM1/pertuzumab and paclitaxel/ antitumor activity of epirubicin-loaded CXCR4-targeted polymeric
trastuzumab/pertuzumab for HER2+ breast cancer in the adaptively nanoparticles in liver cancers. Int J Pharm. 2016;500(1–2):54–61.
randomized I-SPY2 trial. Nat Commun. 2021;12(1):6428. 103. Chittasupho C, Kewsuwan P, Murakami T. CXCR4-targeted nanoparticles
83. Jäger S, Wagner TR, Rasche N, Kolmar H, Hecht S, Schröter C. Generation reduce cell viability, induce apoptosis and inhibit SDF-1α induced BT-
and biological evaluation of Fc antigen binding fragment-drug con- 549-Luc cell migration in vitro. Curr Drug Deliv. 2017;14(8):1060–70.
jugates as a novel antibody-based format for targeted drug delivery. 104. Xiao Y, Chen J, Zhou H, Zeng X, Ruan Z, Pu Z, Jiang X, et al. Combining
Bioconjug Chem. 2021;32(8):1699–710. p53 mRNA nanotherapy with immune checkpoint blockade repro-
84. Scabia V, Ayyanan A, De Martino F, Agnoletto A, Battista L, Laszlo C, grams the immune microenvironment for effective cancer therapy. Nat
Treboux A, Zaman K, et al. Estrogen receptor positive breast cancers Commun. 2022;13(1):758.
have patient specific hormone sensitivities and rely on progesterone 105. Xue L, Mao X, Ren L, Chu X. Inhibition of CXCL12/CXCR4 axis as a
receptor. Nat Commun. 2022;13(1):3127. potential targeted therapy of advanced gastric carcinoma. Cancer Med.
85. Hamilton KJ, Hewitt SC, Arao Y, Korach KS. Estrogen hormone biology. 2017;6(6):1424–36.
Curr Top Dev Biol. 2017;125:109–46. 106. Nian D, Shi P, Sun J, Ren L, Hao X, Han J. Application of luteinizing
86. Folkerd EJ, Dowsett M. Influence of sex hormones on cancer progres- hormone-releasing hormone-ferrosoferric oxide nanoparticles in
sion. J Clin Oncol. 2010;28(26):4038–44. targeted imaging of breast tumors. J Int Med Res. 2019;47(4):1749–57.
87. Thomas C, Gustafsson JÅ. The different roles of ER subtypes in cancer 107. Obayemi J, Salifu A, Eluu S, Uzonwanne V, Jusu S, Nwazojie C, Onye-
biology and therapy. Nat Rev Cancer. 2011;11(8):597–608. kanne C, et al. LHRH-conjugated drugs as targeted therapeutic agents
88. Kurmi BD, Paliwal R, Paliwal SR. Dual cancer targeting using estrogen for the specific targeting and localized treatment of triple negative
functionalized chitosan nanoparticles loaded with doxorubicin- breast cancer. Sci Rep. 2020;10(1):8212.
estrone conjugate: A quality by design approach. Int J Biol Macromol. 108. Li X, Taratula O, Taratula O, Schumann C, Minko T. LHRH-targeted
2020;164:2881–94. Drug delivery systems for cancer therapy. Mini Rev Med Chem.
89. Kapara A, Brunton V, Graham D, Faulds K. Investigation of cellular 2017;17(3):258–67.
uptake mechanism of functionalised gold nanoparticles into breast 109. Garbuzenko OB, Kuzmov A, Taratula O, Pine SR, Minko T. Strategy to
cancer using SERS. Chem Sci. 2020;11(22):5819–29. enhance lung cancer treatment by five essential elements: inhalation
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 30 of 40
delivery, nanotechnology, tumor-receptor targeting, chemo- and gene 132. Yang Z, Wang J, Ai S, Sun J, Mai X, Guan W. Self-generating oxygen
therapy. Theranostics. 2019;9(26):8362–76. enhanced mitochondrion-targeted photodynamic therapy for tumor
110. Pan Q, Tian J, Zhu H, Hong L, Mao Z, Oliveira JM, Reis RL, et al. Tumor- treatment with hypoxia scavenging. Theranostics. 2019;9(23):6809–23.
targeting polycaprolactone nanoparticles with codelivery of paclitaxel 133. Zhang R, Song X, Liang C, Yi X, Song G, Chao Y, Yang Y, Yang K, Feng
and IR780 for combinational therapy of drug-resistant ovarian cancer. L, Liu Z. Catalase-loaded cisplatin-prodrug-constructed liposomes to
ACS Biomater Sci Eng. 2020;6(4):2175–85. overcome tumor hypoxia for enhanced chemo-radiotherapy of cancer.
111. Zhang L, Ren Y, Wang Y, He Y, Feng W, Song C. Pharmacokinetics, Biomaterials. 2017;138:13–21.
distribution and anti-tumor efficacy of liposomal mitoxantrone modi- 134. Wen M, Ouyang J, Wei C, Li H, Chen W, Liu YN. Artificial enzyme cata-
fied with a luteinizing hormone-releasing hormone receptor-specific lyzed cascade reactions: antitumor immunotherapy reinforced by NIR-II
peptide. Int J Nanomed. 2018;13:1097–105. light. Angew Chem Int Ed Engl. 2019;58(48):17425–32.
112. Li M, Tang Z, Zhang Y, Lv S, Yu H, Zhang D, Hong H, et al. LHRH-peptide 135. Chen S, Jia Q, Zheng X, Wen Y, Liu W, Zhang H, Ge J, Wang P. PEGylated
conjugated dextran nanoparticles for targeted delivery of cisplatin to carbon dot/MnO2 nanohybrid: a new pH/H2O2-driven, turn-on cancer
breast cancer. J Mater Chem B. 2014;2(22):3490–9. nanotheranostics. Sci China Mater. 2018;61(10):1325–38.
113. Taheri A, Dinarvand R, Atyabi F, Ahadi F, Nouri FS, Ghahremani MH, 136. Jia T, Xu J, Dong S, He F, Zhong C, Yang G, Bi H, Xu M, Hu Y, Yang D.
Ostad SN, et al. Enhanced anti-tumoral activity of methotrexate- Mesoporous cerium oxide-coated upconversion nanoparticles for
human serum albumin conjugated nanoparticles by targeting with tumor-responsive chemo-photodynamic therapy and bioimaging.
luteinizing hormone-releasing hormone (LHRH) peptide. Int J Mol Sci. Chem Sci. 2019;10(37):8618–33.
2011;12(7):4591–608. 137. Ma Z, Wu L, Han K, Han H. Pt nanozyme for O 2 self-sufficient, tumor-
114. Banstola A, Poudel K, Kim JO, Jeong JH, Yook S. Recent progress in specific oxidative damage and drug resistance reversal. Nanoscale
stimuli-responsive nanosystems for inducing immunogenic cell death. Horiz. 2019;4(5):1124–31.
J Control Release. 2021;337:505–20. 138. Liu Y, Wu H, Li M, Yin J, Nie Z. pH dependent catalytic activities of
115. Diaz-Vivancos P, de Simone A, Kiddle G, Foyer CH. Glutathione- platinum nanoparticles with respect to the decomposition of hydrogen
linking cell proliferation to oxidative stress. Free Radic Biol Med. peroxide and scavenging of superoxide and singlet oxygen. Nanoscale.
2015;89:1154–64. 2014;6(20):11904–10.
116. Wu M, Ding Y, Li L. Recent progress in the augmentation of reac- 139. Ding B, Zheng P, Ma Pa, Lin J. Manganese oxide nanomaterials:
tive species with nanoplatforms for cancer therapy. Nanoscale. synthesis, properties, and theranostic applications. Adv Mater.
2019;11(42):19658–83. 2020;32(10):e1905823.
117. Hu X, Tian H, Jiang W, Song A, Li Z, Luan Y. Rational design of IR820- and 140. Gligorovski S, Strekowski R, Barbati S, Vione D. Environmental implica-
Ce6-based versatile micelle for single NIR laser-induced imaging and tions of hydroxyl radicals (·OH). Chem Rev. 2015;115(24):13051–92.
dual-modal phototherapy. Small. 2018;14(52):e1802994. 141. Tang Z, Liu Y, He M, Bu W. Chemodynamic therapy: tumour microenvi-
118. Yang N, Xiao W, Song X, Wang W, Dong X. Recent advances in tumor ronment-mediated fenton and fenton-like reactions. Angew Chem Int
microenvironment hydrogen peroxide-responsive materials for cancer Ed Engl. 2019;58(4):946–56.
photodynamic therapy. Nanomicro Lett. 2020;12(1):15. 142. Yang B, Chen Y, Shi J. Nanocatalytic medicine. Adv Mater.
119. Dong Z, Feng L, Chao Y, Hao Y, Chen M, Gong F, Han X, et al. Amplifica- 2019;31(39):e1901778.
tion of tumor oxidative stresses with liposomal fenton catalyst and 143. Wang W, Jin Y, Xu Z, Liu X, Bajwa SZ, Khan WS, Yu H. Stimuli-activatable
glutathione inhibitor for enhanced cancer chemotherapy and radio- nanomedicines for chemodynamic therapy of cancer. Wiley Interdiscip
therapy. Nano Lett. 2019;19(2):805–15. Rev Nanomed Nanobiotechnol. 2020;12(4):e1614.
120. Fu L, Wan Y, Qi C, He J, Li C, Yang C, Xu H, et al. Nanocatalytic theranos- 144. Gao S, Lin H, Zhang H, Yao H, Chen Y, Shi J. Nanocatalytic tumor therapy
tics with glutathione depletion and enhanced reactive oxygen species by biomimetic dual inorganic nanozyme-catalyzed cascade reaction.
generation for efficient cancer therapy. Adv Mater. 2021;33(7):e2006892. Adv Sci. 2019;6(3):1801733.
121. Zhong X, Wang X, Cheng L, Tang Ya, Zhan G, Gong F, Zhang R, et al. 145. Chen Q, Yang D, Yu L, Jing X, Chen Y. Catalytic chemistry of iron-free
GSH-depleted PtCu3 nanocages for chemodynamic-enhanced sonody- Fenton nanocatalysts for versatile radical nanotherapeutics. Mater
namic cancer therapy. Adv Funct Mater. 2020;30(4):1907954. Horiz. 2020;7(2):317–37.
122. Corso CR, Acco A. Glutathione system in animal model of solid 146. Lin LS, Huang T, Song J, Ou XY, Wang Z, Deng H, Tian R, et al. Synthesis
tumors: from regulation to therapeutic target. Crit Rev Oncol Hematol. of copper peroxide nanodots for H 2O2 self-supplying chemodynamic
2018;128:43–57. therapy. J Am Chem Soc. 2019;141(25):9937–45.
123. Hu J, Liu S. Modulating intracellular oxidative stress via engineered 147. Fan W, Yung BC, Chen X. Stimuli-responsive NO release for on-demand
nanotherapeutics. J Control Release. 2020;319:333–43. gas-sensitized synergistic cancer therapy. Angew Chem Int Ed Engl.
124. Yang S, Wong KH, Hua P, He C, Yu H, Shao D, Shi Z, et al. ROS-responsive 2018;57(28):8383–94.
fluorinated polyethyleneimine vector to co-deliver shMTHFD2 and 148. Fan W, Lu N, Huang P, Liu Y, Yang Z, Wang S, Yu G, et al. Glucose-respon-
shGPX4 plasmids induces ferroptosis and apoptosis for cancer therapy. sive sequential generation of hydrogen peroxide and nitric oxide for
Acta Biomater. 2022;140:492–505. synergistic cancer starving-like/gas therapy. Angew Chem Int Ed Engl.
125. D’Autréaux B, Toledano MB. ROS as signalling molecules: mechanisms 2017;56(5):1229–33.
that generate specificity in ROS homeostasis. Nat Rev Mol Cell Biol. 149. Zeng Q, Zhang R, Zhang T, Xing D. HO-responsive biodegradable
2007;8(10):813–24. nanomedicine for cancer-selective dual-modal imaging guided precise
126. Winston GW, Di Giulio RT. Prooxidant and antioxidant mechanisms in photodynamic therapy. Biomaterials. 2019;207:39–48.
aquatic organisms. Aquat Toxicol. 1991;19(2):137–61. 150. Lu K, Lin P, Chuang E, Shih C, Cheng T, Lin T, Sung H, et al.
127. Yang B, Chen Y, Shi J. Reactive oxygen species (ROS)-based nanomedi- H2O2-depleting and O 2-generating selenium nanoparticles for fluo-
cine. Chem Rev. 2019;119(8):4881–985. rescence imaging and photodynamic treatment of proinflammatory-
128. Panieri E, Santoro M. ROS homeostasis and metabolism: a dangerous activated macrophages. ACS Appl Mater Interfaces. 2017;9(6):5158–72.
liason in cancer cells. Cell Death Dis. 2016;7:e2253. 151. Ma N, Li Y, Xu H, Wang Z, Zhang X. Dual redox responsive assem-
129. Raza MH, Siraj S, Arshad A, Waheed U, Aldakheel F, Alduraywish S, blies formed from diselenide block copolymers. J Am Chem Soc.
Arshad M. ROS-modulated therapeutic approaches in cancer treat- 2010;132(2):442–3.
ment. J Cancer Res Clin Oncol. 2017;143(9):1789–809. 152. Reis AKCA, Stern A, Monteiro HP. S-nitrosothiols and HS donors: Poten-
130. Walsh JC, Lebedev A, Aten E, Madsen K, Marciano L, Kolb HC. The tial chemo-therapeutic agents in cancer. Redox Biol. 2019;27:101190.
clinical importance of assessing tumor hypoxia: relationship of tumor 153. Duan Y, Wang Y, Li X, Zhang G, Zhang G, Hu J. Light-triggered nitric
hypoxia to prognosis and therapeutic opportunities. Antioxid Redox oxide (NO) release from photoresponsive polymersomes for corneal
Signal. 2014;21(10):1516–54. wound healing. Chem Sci. 2020;11(1):186–94.
131. Sahu A, Kwon I, Tae G. Improving cancer therapy through the nanoma-
terials-assisted alleviation of hypoxia. Biomaterials. 2020;228:119578.
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 31 of 40
154. Yang S, Gao H. Nanoparticles for modulating tumor microenviron- 176. Zheng X, Pan D, Zhu G, Zhang L, Bhamra A, Chen R, Zhang H, et al. A
ment to improve drug delivery and tumor therapy. Pharmacol Res. dendritic polymer-based nanosystem mediates drug penetration and
2017;126:97–108. irreversible endoplasmic reticulum stresses in tumor via neighboring
155. Hu C, Cun X, Ruan S, Liu R, Xiao W, Yang X, Yang Y, et al. Enzyme- effect. Adv Mater. 2022;34:e2201200.
triggered size shrink and laser-enhanced NO release nanoparticles 177. Zhang L, Wang K, Huang Y, Zhang H, Zhou L, Li A, Sun Y. Photosensi-
for deep tumor penetration and combination therapy. Biomaterials. tizer-induced HPV16 E7 nanovaccines for cervical cancer immuno-
2018;168:64–75. therapy. Biomaterials. 2022;282:121411.
156. Qin L, Gao H. The application of nitric oxide delivery in nanoparticle- 178. Yang J, Pan S, Gao S, Dai Y, Xu H. Anti-recurrence/metastasis and che-
based tumor targeting drug delivery and treatment. Asian J Pharm Sci. mosensitization therapy with thioredoxin reductase-interfering drug
2019;14(4):380–90. delivery system. Biomaterials. 2020;249:120054.
157. Liu R, Xiao W, Hu C, Xie R, Gao H. Theranostic size-reducible and no 179. Yan H, Dong J, Huang X, Du X. Protein-gated upconversion nanoparti-
donor conjugated gold nanocluster fabricated hyaluronic acid nano- cle-embedded mesoporous silica nanovehicles via diselenide linkages
particle with optimal size for combinational treatment of breast cancer for drug release tracking in real time and tumor chemotherapy. ACS
and lung metastasis. J Control Release. 2018;278:127–39. Appl Mater Interfaces. 2021;13(24):29070–82.
158. Zhang X, Du J, Guo Z, Yu J, Gao Q, Yin W, Zhu S, et al. Efficient near infra- 180. Shao D, Zhang F, Chen F, Zheng X, Hu H, Yang C, Tu Z, et al. Biomimetic
red light triggered nitric oxide release nanocomposites for sensitizing diselenide-bridged mesoporous organosilica nanoparticles as an
mild photothermal therapy. Adv Sci. 2019;6(3):1801122. X-ray-responsive biodegradable carrier for chemo-immunotherapy. Adv
159. Page-McCaw A, Ewald AJ, Werb Z. Matrix metalloproteinases Mater. 2020;32(50):e2004385.
and the regulation of tissue remodelling. Nat Rev Mol Cell Biol. 181. Han L, Zhang X, Wang Y, Li X, Yang X, Huang M, Hu K, et al. Redox-
2007;8(3):221–33. responsive theranostic nanoplatforms based on inorganic nanomateri-
160. Dong X, Liu H, Feng H, Yang S, Liu X, Lai X, Lu Q, et al. Enhanced drug als. J Control Release. 2017;259:40–52.
delivery by nanoscale integration of a nitric oxide donor to induce 182. Suto N, Kamoshita S, Hosoya S, Sakurai K. Exploration of the reactiv-
tumor collagen depletion. Nano Lett. 2019;19(2):997–1008. ity of multivalent electrophiles for affinity labeling: sulfonyl fluoride
161. Wan S, Zeng J, Cheng H, Zhang X. ROS-induced NO generation for gas as a highly efficient and selective label. Angew Chem Int Ed Engl.
therapy and sensitizing photodynamic therapy of tumor. Nano Lett. 2021;60(31):17080–7.
2018;185:51–62. 183. Zhang L, Wang L, Yao H, Xu F, Chen Y. Biodegradable and biocompatible
162. Shi M, Zhang J, Wang Y, Peng C, Hu H, Qiao M, Zhao X, et al. Tumor- monodispersed hollow mesoporous organosilica with large pores for
specific nitric oxide generator to amplify peroxynitrite based on highly delivering biomacromolecules. J Mater Chem B. 2017;5(39):8013–25.
penetrable nanoparticles for metastasis inhibition and enhanced 184. Chen H, Zhang W, Zhu G, Xie J, Chen X. Rethinking cancer nanothera-
cancer therapy. Biomaterials. 2022;283: 121448. nostics. Nat Rev Mater. 2017;2(7):1–18.
163. Liu Y, Shi J. Antioxidative nanomaterials and biomedical applications. 185. Peng J, Xiao Y, Yang Q, Liu Q, Chen Y, Shi K, Hao Y, et al. Intracellular
Nano Today. 2019;27:146–77. aggregation of peptide-reprogrammed small molecule nanoassem-
164. Jin Q, Deng Y, Jia F, Tang Z, Ji J. Gas therapy: an emerging “green” strat- blies enhances cancer chemotherapy and combinatorial immuno-
egy for anticancer therapeutics. Adv Ther. 2018;1(6):1800084. therapy. Acta Pharm Sin B. 2021;11(4):1069–82.
165. Huang Z, Fu J, Zhang Y. Nitric oxide donor-based cancer therapy: 186. Wang N, Wang Z, Xu Z, Chen X, Zhu G. A cisplatin-loaded immuno-
advances and prospects. J Med Chem. 2017;60(18):7617–35. chemotherapeutic nanohybrid bearing immune checkpoint inhibitors
166. Levy ES, Morales DP, Garcia JV, Reich NO, Ford PC. Near-IR mediated for enhanced cervical cancer therapy. Angew Chem Int Ed Engl.
intracellular uncaging of NO from cell targeted hollow gold nanoparti- 2018;57(13):3426–30.
cles. Chem Commun. 2015;51(100):17692–5. 187. Cheng R, Feng F, Meng F, Deng C, Feijen J, Zhong Z. Glutathione-
167. Gu G, Chen C, Zhang S, Yin B, Wang J. Self-assembly dual-responsive responsive nano-vehicles as a promising platform for targeted intracel-
NO donor nanoparticles for effective cancer therapy. ACS Appl Mater lular drug and gene delivery. J Control Release. 2011;152(1):2–12.
Interfaces. 2021;13(43):50682–94. 188. Zhu Z, Wang Z, Hao Y, Zhu C, Jiao Y, Chen H, Wang Y, et al. Glutathione
168. Luo Z, Zhou Y, Yang T, Gao Y, Kumar P, Chandrawati R. Ceria nanoparti- boosting the cytotoxicity of a magnetic platinum(iv) nano-prodrug in
cles as an unexpected catalyst to generate nitric oxide from S-nitroso- tumor cells. Chem Sci. 2016;7(4):2864–9.
glutathione. Small. 2022;18(11):e2105762. 189. Yu F, Zhang F, Tang L, Ma J, Ling D, Chen X, Sun X. Redox-responsive
169. Li W, Wang D, Lao KU, Wang X. Buffer concentration dramatically dual chemophotothermal therapeutic nanomedicine for imaging-
affects the stability of S-nitrosothiols in aqueous solutions. Nitric Oxide. guided combinational therapy. J Mater Chem B. 2018;6(33):5362–7.
2022;118:59–65. 190. Yao H, Shen N, Ji G, Huang J, Sun J, Wang G, Tang Z, et al. Cisplatin nano-
170. Zhao C, Zhang H, Zhou J, Lu Q, Zhang Y, Yu X, Wang S, et al. Metabo- particles promote intratumoral C D8+ T cell priming via antigen presen-
lomics-based molecular signatures reveal the toxic effect of co- tation and T cell receptor crosstalk. Nano Lett. 2022;22(8):3328–39.
exposure to nitrosamines in drinking water. Environ Res. 2022;204(Pt B): 191. Ding K, Wang L, Zhu J, He D, Huang Y, Zhang W, Wang Z, et al. Photo-
111997. enhanced chemotherapy performance in bladder cancer treatment
171. Xiang H, Deng Q, An L, Guo M, Yang S, Liu J. Tumor cell specific and via albumin coated AIE aggregates. ACS Nano. 2022. https://doi.org/10.
lysosome-targeted delivery of nitric oxide for enhanced photodynamic 1021/acsnano.1c10770.
therapy triggered by 808 nm near-infrared light. Chem Commun. 192. Wang Z, Wang N, Cheng S, Xu K, Deng Z, Chen S, Xu Z, et al. Phorbipl-
2016;52(1):148–51. atin, a highly potent Pt (IV) antitumor prodrug that can be controllably
172. Zhang S, Li Q, Yang N, Shi Y, Ge W, Wang W, Huang W, et al. Phase- activated by red light. Chem. 2019;5(12):3151–65.
change materials based nanoparticles for controlled hypoxia 193. Ling X, Tu J, Wang J, Shajii A, Kong N, Feng C, Zhang Y, et al. Glu-
modulation and enhanced phototherapy. Adv Funct Mater. tathione-responsive prodrug nanoparticles for effective drug delivery
2019;29(49):1906805. and cancer therapy. ACS Nano. 2018;13(1):357–70.
173. Sun X, Sun J, Dong B, Huang G, Zhang L, Zhou W, Lv J, et al. Nonin- 194. Wang X, Wang X, Jin S, Muhammad N, Guo Z. Stimuli-responsive thera-
vasive temperature monitoring for dual-modal tumor therapy based peutic metallodrugs. Chem Rev. 2018;119(2):1138–92.
on lanthanide-doped up-conversion nanocomposites. Biomaterials. 195. Muhammad F, Wang A, Qi W, Zhang S, Zhu G. Intracellular antioxidants
2019;201:42–52. dissolve man-made antioxidant nanoparticles: using redox vulnerability
174. Shi G, Monro S, Hennigar R, Colpitts J, Fong J, Kasimova K, Yin H, et al. of nanoceria to develop a responsive drug delivery system. ACS Appl
Ru (II) dyads derived from α-oligothiophenes: a new class of potent and Mater Interfaces. 2014;6(21):19424–33.
versatile photosensitizers for PDT. Chem Rev. 2015;282:127–38. 196. Tian H, Zhang M, Jin G, Jiang Y, Luan Y. Cu-MOF chemodynamic
175. Prasetyanto EA, Bertucci A, Septiadi D, Corradini R, Castro-Hartmann nanoplatform via modulating glutathione and H2O2 in tumor micro-
P, De Cola L. Breakable hybrid organosilica nanocapsules for protein environment for amplified cancer therapy. J Colloid Interface Sci.
delivery. Angew Chem Int Ed Engl. 2016;55(10):3323–7. 2021;587:358–66.
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 32 of 40
197. Tian H, Zhao S, Nice EC, Huang C, He W, Zou B, Lin J, et al. A cascaded 216. Shin Y, Husni P, Kang K, Lee D, Lee S, Lee E, Youn Y, et al. Recent
copper-based nanocatalyst by modulating glutathione and cyclooxy- advances in pH- or/and photo-responsive nanovehicles. Pharmaceutics.
genase-2 for hepatocellular carcinoma therapy. J Colloid Interface 2021;13(5):725.
Sci. 2022;607:1516–26. 217. Xue Y, Bai H, Peng B, Tieu T, Jiang J, Hao S, Li P, et al. Porous silicon nano-
198. Liu C, Wang D, Zhang S, Cheng Y, Yang F, Xing Y, Xu T, et al. Biodegrad- carriers with stimulus-cleavable linkers for effective cancer therapy. Adv
able biomimic copper/manganese silicate nanospheres for chemo- Healthc Mater. 2022;11:e2200076.
dynamic/photodynamic synergistic therapy with simultaneous glu- 218. Qiu L, Xu J, Ahmed KS, Zhu M, Zhang Y, Long M, Chen W, et al.
tathione depletion and hypoxia relief. ACS Nano. 2019;13(4):4267–77. Stimuli-responsive, dual-function prodrug encapsulated in hyaluronic
199. Wu W, Chen M, Luo T, Fan Y, Zhang J, Zhang Y, Zhang Q, et al. ROS acid micelles to overcome doxorubicin resistance. Acta Biomater.
and GSH-responsive S-nitrosoglutathione functionalized polymeric 2022;140:686–99.
nanoparticles to overcome multidrug resistance in cancer. Acta 219. Xu M, Zhao D, Chen Y, Chen C, Zhang L, Sun L, Chen J, et al. Charge
Biomater. 2020;103:259–71. reversal polypyrrole nanocomplex-mediated gene delivery and
200. Gong N, Ma X, Ye X, Zhou Q, Chen X, Tan X, Yao S, et al. Carbon- photothermal therapy for effectively treating papillary thyroid cancer
dot-supported atomically dispersed gold as a mitochondrial and inhibiting lymphatic metastasis. ACS Appl Mater Interfaces.
oxidative stress amplifier for cancer treatment. Nat Nanotechnol. 2022;14(12):14072–86.
2019;14(4):379–87. 220. Jin Y, Song L, Su Y, Zhu L, Pang Y, Qiu F, Tong G, et al. Oxime linkage:
201. Stubbs M, McSheehy PM, Griffiths JR, Bashford CL. Causes and conse- a robust tool for the design of pH-sensitive polymeric drug carriers.
quences of tumour acidity and implications for treatment. Mol Med Biomacromol. 2011;12(10):3460–8.
Today. 2000;6(1):15–9. 221. Xu X, Li L, Zhou Z, Sun W, Huang Y. Dual-pH responsive micelle
202. Warburg O, Wind F, Negelein E. The metabolism of tumors in the platform for co-delivery of axitinib and doxorubicin. Int J Pharm.
body. J Gen Physiol. 1927;8(6):519–30. 2016;507(1–2):50–60.
203. Lee ES, Oh KT, Kim D, Youn YS, Bae YH. Tumor pH-responsive flower- 222. Gurav DD, Kulkarni AS, Khan A, Shinde VS. pH-responsive targeted and
like micelles of poly(l-lactic acid)-b-poly(ethylene glycol)-b-poly(l- controlled doxorubicin delivery using hyaluronic acid nanocarriers.
histidine). J Control Release. 2007;123(1):19–26. Colloids Surf B Biointerfaces. 2016;143:352–8.
204. Yang Q, He C, Zhang Z, Tan L, Liu B, Zhu Z, Shao Z, et al. Redox- 223. She W, Pan D, Luo K, He B, Cheng G, Zhang C, Gu Z. PEGylated
responsive flower-like micelles of poly (l-lactic acid)-b-poly (ethylene dendrimer-doxorubicin cojugates as pH-sensitive drug delivery
glycol)-b-poly (l-lactic acid) for intracellular drug delivery. Polymer. systems: synthesis and in vitro characterization. J Biomed Nanotechnol.
2016;90:351–62. 2015;11(6):964–78.
205. Xu Z, Yang D, Long T, Yuan L, Qiu S, Li D, Mu C, et al. pH-sensitive 224. Long Y, Gu W, Pang C, Ma J, Gao H. Construction of coumarin-based
nanoparticles based on amphiphilic imidazole/cholesterol modified cross-linked micelles with pH responsive hydrazone bond and tumor
hydroxyethyl starch for tumor chemotherapy. Carbohydr Polym. targeting moiety. J Mater Chem B. 2016;4(8):1480–8.
2022;277:118827. 225. Ding Y, Sun D, Wang G, Yang H, Xu H, Chen J, Xie Y, et al. An efficient
206. Bae YM, Park YI, Nam SH, Kim JH, Lee K, Kim HM, Yoo B, et al. Endo- PEGylated liposomal nanocarrier containing cell-penetrating peptide
cytosis, intracellular transport, and exocytosis of lanthanide-doped and pH-sensitive hydrazone bond for enhancing tumor-targeted drug
upconverting nanoparticles in single living cells. Biomaterials. delivery. Int J Nanomed. 2015;10:6199.
2012;33(35):9080–6. 226. Kanamala M, Palmer BD, Jamieson SM, Wilson WR, Wu Z. Dual pH-sen-
207. Iversen TG, Skotland T, Sandvig K. Endocytosis and intracellular sitive liposomes with low pH-triggered sheddable PEG for enhanced
transport of nanoparticles: present knowledge and need for future tumor-targeted drug delivery. Nanomedicine. 2019;14(15):1971–89.
studies. Nano Today. 2011;6(2):176–85. 227. Zhang Z, He C, Chen X. Hydrogels based on pH-responsive reversible
208. Qu J, Peng S, Wang R, Yang S, Zhou Q, Lin J. Stepwise pH-sensitive carbon-nitrogen double-bond linkages for biomedical applications.
and biodegradable polypeptide hybrid micelles for enhanced cel- Mater Chem Front. 2018;2(10):1765–78.
lular internalization and efficient nuclear drug delivery. Biointerfaces. 228. Gu J, Cheng W, Liu J, Lo SY, Smith D, Qu X, Yang Z. pH-triggered revers-
2019;181:315–24. ible “stealth” polycationic micelles. Biomacromol. 2008;9(1):255–62.
209. Lin W, Ma G, Yuan Z, Qian H, Xu L, Sidransky E, Chen S. Develop- 229. Ding C, Gu J, Qu X, Yang Z. Preparation of multifunctional drug car-
ment of zwitterionic polypeptide nanoformulation with high rier for tumor-specific uptake and enhanced intracellular delivery
doxorubicin loading content for targeted drug delivery. Langmuir. through the conjugation of weak acid labile linker. Bioconjug Chem.
2018;35(5):1273–83. 2009;20(6):1163–70.
210. Piątkowski M, Janus Ł, Radwan-Pragłowska J, Bogdał D, Matysek D. 230. Cheng Y, Chen Q, Guo Z, Li M, Yang X, Wan G, Chen H, et al. An intel-
Biodegradable, pH-sensitive chitosan beads obtained under micro- ligent biomimetic nanoplatform for holistic treatment of metastatic
wave radiation for advanced cell culture. Colloids Surf B Biointerfaces. triple-negative breast cancer photothermal ablation and immune
2018;164:324–31. remodeling. ACS Nano. 2020;14(11):15161–81.
211. Lo Y, Huang X, Chen H, Huang Y, Liao Z, Wang L. ROP and ATRP 231. Jhaveri A, Deshpande P, Torchilin V. Stimuli-sensitive nanopreparations
fabricated redox sensitive micelles based on PCL-SS-PMAA diblock for combination cancer therapy. J Control Release. 2014;190:352–70.
copolymers to co-deliver PTX and CDDP for lung cancer therapy. 232. Eatemadi A, Aiyelabegan HT, Negahdari B, Mazlomi MA, Daraee H,
Biointerfaces. 2021;198:111443. Daraee N, Eatemadi R, et al. Role of protease and protease inhibi-
212. Rasib S, Ahmad Z, Khan A, Akil H, Othman M, Hamid Z, Ullah F. Syn- tors in cancer pathogenesis and treatment. Biomed Pharmacother.
thesis and evaluation on pH-and temperature-responsive chitosan-p 2017;86:221–31.
(MAA-co-NIPAM) hydrogels. Int J Biol Macromol. 2018;108:367–75. 233. Yamada R, Kostova MB, Anchoori RK, Xu S, Neamati N, Khan S. Biological
213. Zeinali E, Haddadi-Asl V, Roghani-Mamaqani H. Synthesis of dual evaluation of paclitaxel-peptide conjugates as a model for MMP2-
thermo- and pH-sensitive poly(N-isopropylacrylamide-co-acrylic targeted drug delivery. Cancer Biol Ther. 2010;9(3):192–203.
acid)-grafted cellulose nanocrystals by reversible addition-frag- 234. Wang T, He Z, Yuan C, Deng Z, Li F, Chen X, Liu Y. MMP-responsive
mentation chain transfer polymerization. J Biomed Mater Res A. transformation nanomaterials with IAP antagonist to boost immune
2018;106(1):231–43. checkpoint therapy. J Control Release. 2022;343:765–76.
214. Kuo C, Don T, Lin Y, Hsu S, Chiu W. Synthesis of pH-sensitive sulfona- 235. Xu J, Gao F, Li L, Ma H, Fan Y, Liu W, Guo S, et al. Gelatin-mesoporous
mide-based hydrogels with controllable crosslinking density by post silica nanoparticles as matrix metalloproteinases-degradable drug
thermo-curing. J Polym Res. 2019;26(1):1–9. delivery systems in vivo. Micropor Mesopor Mat. 2013;182:165–72.
215. Song Y, Sun Y, Tang M, Yue Z, Ni J, Zhao J, Wang W, et al. Polyoxometa- 236. Zeng W, Wu L, Sun Y, Wang Y, Wang J, Ye D. Ratiometric imaging of
late modified by zeolite imidazole framework for the pH-responsive MMP-2 activity facilitates tumor detection using activatable near-
electrodynamic/chemodynamic therapy. ACS Appl Mater Interfaces. infrared fluorescent semiconducting polymer nanoparticles. Small.
2022;14(4):4914–20. 2021;17(36):e2101924.
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 33 of 40
237. Gu G, Xia H, Hu Q, Liu Z, Jiang M, Kang T, Miao D, et al. PEG-co-PCL nan- activatable phototrigger conjugated chitosan nanoparticles. Adv Mater.
oparticles modified with MMP-2/9 activatable low molecular weight 2013;25(14):1981–6.
protamine for enhanced targeted glioblastoma therapy. Biomaterials. 257. Sahu A, Choi WI, Tae G. Recent progress in the design of hypoxia-
2013;34(1):196–208. specific nano drug delivery systems for cancer therapy. Adv Ther.
238. Yang K, Liu Y, Wang Y, Ren Q, Guo H, Matson JB, Chen X, et al. Enzyme- 2018;1(4):1800026.
induced in vivo assembly of gold nanoparticles for imaging-guided 258. Bertout JA, Patel SA, Simon MC. The impact of O2 availability on human
synergistic chemo-photothermal therapy of tumor. Biomaterials. cancer. Nat Rev Cancer. 2008;8(12):967–75.
2019;223:119460. 259. Thambi T, Park JH, Lee D. Hypoxia-responsive nanocarriers for cancer
239. Kulkarni PS, Haldar MK, Nahire RR, Katti P, Ambre AH, Muhonen WW, imaging and therapy: recent approaches and future perspectives.
Shabb JB, et al. MMP-9 responsive PEG cleavable nanovesicles for effi- Chem Commun. 2016;52(55):8492–500.
cient delivery of chemotherapeutics to pancreatic cancer. Mol Pharm. 260. Trastour C, Benizri E, Ettore F, Ramaioli A, Chamorey E, Pouysségur J,
2014;11(7):2390–9. Berra E. HIF-1alpha and CA IX staining in invasive breast carcinomas:
240. Guo F, Fu Q, Zhou K, Jin C, Wu W, Ji X, Yan Q, Yang Q, et al. Matrix met- prognosis and treatment outcome. Int J Cancer. 2007;120(7):1451–8.
alloprotein-triggered, cell penetrating peptide-modified star-shaped 261. Zhang B, Huang X, Wang H, Gou S. Promoting antitumor efficacy by
nanoparticles for tumor targeting and cancer therapy. J Nanobiotech- suppressing hypoxia via nano self-assembly of two irinotecan-based
nol. 2020;18(1):48. dual drug conjugates having a HIF-1α inhibitor. J Mater Chem B.
241. Bertolesi GE, Su HY, Michaiel G, Dueck SM, Hehr CL, McFarlane S. Two 2019;7(35):5352–62.
promoters with distinct activities in different tissues drive the expres- 262. Prasad P, Gordijo CR, Abbasi AZ, Maeda A, Ip A, Rauth AM, DaCosta
sion of heparanase in Xenopus. Dev Dyn. 2011;240(12):2657–72. RS, et al. Multifunctional albumin-MnO2 nanoparticles modulate solid
242. Huang Y, Shi Y, Wang Q, Qi T, Fu X, Gu Z, Zhang Y, et al. Enzyme tumor microenvironment by attenuating hypoxia, acidosis, vascular
responsiveness enhances the specificity and effectiveness of nano- endothelial growth factor and enhance radiation response. ACS Nano.
particles for the treatment of B16F10 melanoma. J Control Release. 2014;8(4):3202–12.
2019;316:208–22. 263. Kumari R, Sunil D, Ningthoujam R. Hypoxia-responsive nanoparticle
243. Bae KH, Mok H, Park TG. Synthesis, characterization, and intracellular based drug delivery systems in cancer therapy: an up-to-date review. J
delivery of reducible heparin nanogels for apoptotic cell death. Bioma- Control Release. 2020;319:135–56.
terials. 2008;29(23):3376–83. 264. Rankin EB, Giaccia AJ. Hypoxic control of metastasis. Science.
244. Al-Jamal KT, Al-Jamal WT, Wang J, Rubio N, Buddle J, Gathercole D, Zloh 2016;352(6282):175–80.
M, et al. Cationic poly-l-lysine dendrimer complexes doxorubicin and 265. Liu P, Xie X, Shi X, Peng Y, Ding J, Zhou W. Oxygen-self-supplying and
delays tumor growth in vitro and in vivo. ACS Nano. 2013;7(3):1905–17. HIF-1α-inhibiting core-shell nanosystem for hypoxia-resistant photody-
245. Aumiller WM, Keating CD. Phosphorylation-mediated RNA/peptide namic therapy. ACS Appl Mater Interfaces. 2019;11(51):48261–70.
complex coacervation as a model for intracellular liquid organelles. Nat 266. Jászai J, Schmidt MH. Trends and challenges in tumor anti-angiogenic
Chem. 2016;8(2):129–37. therapies. Cells. 2019;8(9):1102.
246. Umerska A, Paluch KJ, Martinez M, Corrigan OI, Medina C, Tajber L. 267. Harris AL. Hypoxia-a key regulatory factor in tumour growth. Nat Rev
Self-assembled hyaluronate/protamine polyelectrolyte nanoplexes: Cancer. 2002;2(1):38–47.
synthesis, stability, biocompatibility and potential use as peptide carri- 268. Hubbi ME, Semenza GL. Regulation of cell proliferation by hypoxia-
ers. J Biomed Nanotechnol. 2014;10(12):3658–73. inducible factors. Am J Physiol Cell Physiol. 2015;309(12):C775–82.
247. Fan Y, Wang Q, Lin G, Shi Y, Gu Z, Ding T. Combination of using 269. Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nat Rev Can-
prodrug-modified cationic liposome nanocomplexes and a potentiat- cer. 2011;11(6):393–410.
ing strategy via targeted co-delivery of gemcitabine and docetaxel 270. Lomax ME, Folkes LK, O’Neill P. Biological consequences of radiation-
for CD44-overexpressed triple negative breast cancer therapy. Acta induced DNA damage: relevance to radiotherapy. Clin Oncol.
Biomater. 2017;62:257–72. 2013;25(10):578–85.
248. Kallunki T, Olsen OD, Jäättelä M. Cancer-associated lysosomal changes: 271. Xing L, Gong J-H, Wang Y, Zhu Y, Huang Z, Zhao J, Li F, et al. Hypoxia
friends or foes? Oncogene. 2013;32(16):1995–2004. alleviation-triggered enhanced photodynamic therapy in combina-
249. Dheer D, Nicolas J, Shankar R. Cathepsin-sensitive nanoscale drug tion with IDO inhibitor for preferable cancer therapy. Biomaterials.
delivery systems for cancer therapy and other diseases. Adv Drug Deliv 2019;206:170–82.
Rev. 2019;151–152:130–51. 272. Li C, Yang X, An J, Cheng K, Hou X, Zhang X, Hu Y, et al. Red blood cell
250. Xia H, Qin M, Wang Z, Wang Y, Chen B, Wan F, Tang M, et al. A pH-/ membrane-enveloped O2 self-supplementing biomimetic nanopar-
enzyme-responsive nanoparticle selectively targets endosomal ticles for tumor imaging-guided enhanced sonodynamic therapy.
Toll-like receptors to potentiate robust cancer vaccination. Nano Lett. Theranostics. 2020;10(2):867–79.
2022;22(7):2978–87. 273. Gao M, Liang C, Song X, Chen Q, Jin Q, Wang C, Liu Z. Erythrocyte-
251. Wang C, Du W, Wu C, Dan S, Sun M, Zhang T, Wang B, et al. Cathespin membrane-enveloped perfluorocarbon as nanoscale artificial red
B-initiated cypate nanoparticle formation for tumor photoacoustic blood Cells to relieve tumor hypoxia and enhance cancer radiotherapy.
imaging. Angew Chem Int Ed Engl. 2022;61(5):e202114766. Adv Mater. 2017;29(35):1701429.
252. Huang C, Chia W, Chung M, Lin K, Hsiao C, Jin C, Lim W, et al. An 274. Wang Y, Shang W, Niu M, Tian J, Xu K. Hypoxia-active nanoparticles used
implantable depot that can generate oxygen in situ for overcoming in tumor theranostic. Int J Nanomed. 2019;14:3705.
hypoxia-induced resistance to anticancer drugs in chemotherapy. J Am 275. Xue T, Shen J, Shao K, Wang W, Wu B, He Y. Strategies for tumor hypoxia
Chem Soc. 2016;138(16):5222–5. imaging based on aggregation-induced emission fluorogens. Chemis-
253. Li Y, Lu A, Long M, Cui L, Chen Z, Zhu L. Nitroimidazole derivative try. 2020;26(12):2521–8.
incorporated liposomes for hypoxia-triggered drug delivery and 276. Huang W, Chen S, Chiang W, Huang C, Lo C, Chern C, Chiu H. Tumor
enhanced therapeutic efficacy in patient-derived tumor xenografts. microenvironment-responsive nanoparticle delivery of chemotherapy
Acta Biomater. 2019;83:334–48. for enhanced selective cellular uptake and transportation within tumor.
254. Shen X, Gates K. Enzyme-activated generation of reactive oxygen Biomacromol. 2016;17(12):3883–92.
species from heterocyclic N-oxides under aerobic and anaerobic condi- 277. Okuda K, Okabe Y, Kadonosono T, Ueno T, Youssif BG, Kizaka-Kondoh
tions and its relevance to hypoxia-selective prodrugs. Chem Res Toxicol. S, Nagasawa H. 2-Nitroimidazole-tricarbocyanine conjugate as a
2019;32(3):348–61. near-infrared fluorescent probe for in vivo imaging of tumor hypoxia.
255. Vaupel P, Schlenger K, Knoop C, Höckel M. Oxygenation of human Bioconjug Chem. 2012;23(3):324–9.
tumors: evaluation of tissue oxygen distribution in breast can- 278. Sun Y, Zhao D, Wang G, Wang Y, Cao L, Sun J, Jiang Q, et al. Recent
cers by computerized O 2 tension measurements. Cancer Res. progress of hypoxia-modulated multifunctional nanomedicines to
1991;51(12):3316–22. enhance photodynamic therapy: opportunities, challenges, and future
256. Lin Q, Bao C, Yang Y, Liang Q, Zhang D, Cheng S, Zhu L. Highly development. Acta Pharm Sin B. 2020;10(8):1382–96.
discriminating photorelease of anticancer drugs based on hypoxia
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 34 of 40
279. Liang J, Wei X, Yin T, Huo M. Advances in the hypoxia-responsive antitu- 301. Zhu J, Guo T, Wang Z, Zhao Y. Triggered azobenzene-based prodrugs
mor drug nanocarriers and tumor hypoxia relieve. J China Pharm Univ. and drug delivery systems. J Control Release. 2022;345:475–93.
2018;49(3):255–62. 302. Liu Y, Xie L, Gao M, Zhang R, Gao J, Sun J, Chai Q, et al. Super-assembled
280. Thambi T, Deepagan V, Yoon HY, Han HS, Kim S, Son S, Jo D, et al. periodic mesoporous organosilica frameworks for real-time hypoxia-
Hypoxia-responsive polymeric nanoparticles for tumor-targeted drug triggered drug release and monitoring. ACS Appl Mater Interfaces.
delivery. Biomaterials. 2014;35(5):1735–43. 2021;13(42):50246–57.
281. Cui D, Huang J, Zhen X, Li J, Jiang Y, Pu K. A semiconducting polymer 303. Tang J, Huang C, Shu J, Zheng J, Ma D, Li J, Yang R. Azoreductase and
nano-prodrug for hypoxia-activated photodynamic cancer therapy. target simultaneously activated fluorescent monitoring for cytochrome
Angew Chem Int Ed In Engl. 2019;58(18):5920–4. c release under hypoxia. Anal Chem. 2018;90(9):5865–72.
282. Luo R, Zhang Z, Han L, Xue Z, Zhang K, Liu F, Feng F, et al. An albumin- 304. Zha Y, Xin R, Zhang M, Cui X, Li N. Stimuli-responsive azobenzene-quan-
binding dimeric prodrug nanoparticle with long blood circulation and tum dots for multi-sensing of dithionite, hypochlorite, and azoreduc-
light-triggered drug release for chemo-photodynamic combination tase. Mikrochim Acta. 2020;187(8):481.
therapy against hypoxia-induced metastasis of lung cancer. Biomater 305. Huang X, Chen T, Mu N, Lam HW, Sun C, Yue L, Cheng Q, et al. Supra-
Sci. 2021;9(10):3718–36. molecular micelles as multifunctional theranostic agents for synergistic
283. Bera H, Abosheasha MA, Ito Y, Ueda M. Hypoxia-responsive pullu- photodynamic therapy and hypoxia-activated chemotherapy. Acta
lan-based nanoparticles as erlotinib carriers. Int J Biol Macromol. Biomater. 2021;131:483–92.
2021;191:764–74. 306. Huang C, Tan W, Zheng J, Zhu C, Huo J, Yang R. Azoreductase-respon-
284. Tseng SJ, Kempson IM, Huang K, Li H, Fa Y, Ho Y, Liao Z, et al. Targeting sive metal-organic framework-based nanodrug for enhanced cancer
tumor microenvironment by bioreduction-activated nanoparticles for therapy via breaking hypoxia-induced chemoresistance. ACS Appl
light-triggered virotherapy. ACS Nano. 2018;12(10):9894–902. Mater Interfaces. 2019;11(29):25740–9.
285. Li Y, Ding J, Xu X, Shi R, Saw PE, Wang J, Chung S, et al. Dual hypoxia- 307. He X, Chen F, Chang Z, Waqar K, Hu H, Zheng X, Wang Y, et al. Silver
targeting RNAi nanomedicine for precision cancer therapy. Nano Lett. mesoporous silica nanoparticles: fabrication to combination therapies
2020;20(7):4857–63. for cancer and infection. Chem Rec. 2022;22(4):e202100287.
286. Ma D, Huang C, Zheng J, Zhou W, Tang J, Chen W, Li J, et al. Azoreduc- 308. Wu F, Du Y, Yang J, Shao B, Mi Z, Yao Y, Cui Y, et al. Peroxidase-like active
tase-responsive nanoprobe for hypoxia-induced mitophagy imaging. nanomedicine with dual glutathione depletion property to restore
Anal Chem. 2019;91(2):1360–7. oxaliplatin chemosensitivity and promote programmed cell death. ACS
287. Zhang H, Liang F, Yang Y. Dual-stimuli responsive 2D supramolecular Nano. 2022;16(3):3647–63.
organic framework for the detection of azoreductase activity. Chemis- 309. Jang EH, Kim GL, Park MG, Shim MK, Kim JH. Hypoxia-responsive,
try. 2020;26(1):198–205. organic-inorganic hybrid mesoporous silica nanoparticles for triggered
288. Liu X, Wu Z, Guo C, Guo H, Su Y, Chen Q, Sun C, et al. Hypoxia respon- drug release. J Drug Deliv Sci Tec. 2020;56:101543.
sive nano-drug delivery system based on angelica polysaccharide for 310. Kang D, Cheung ST, Wong-Rolle A, Kim J. Enamine-oxides: synthesis
liver cancer therapy. Drug Deliv. 2022;29(1):138–48. and application to hypoxia-responsive prodrugs and imaging agents.
289. Cabré G, Garrido-Charles A, Moreno M, Bosch M, Porta-de-la-Riva M, ACS Cent Sci. 2021;7(4):631–40.
Krieg M, Gascón-Moya M, et al. Rationally designed azobenzene pho- 311. Sharma A, Arambula JF, Koo S, Kumar R, Singh H, Sessler JL, Kim JS.
toswitches for efficient two-photon neuronal excitation. Nat Commun. Hypoxia-targeted drug delivery. Chem Soc Rev. 2019;48(3):771–813.
2019;10(1):907. 312. Hang L, Zhang T, Wen H, Li M, Liang L, Tang X, Zhou C, et al. Rational
290. Xu Z, Pan C, Yuan W. Light-enhanced hypoxia-responsive and azoben- design of non-toxic GOx-based biocatalytic nanoreactor for multimodal
zene cleavage-triggered size-shrinkable micelles for synergistic photo- synergistic therapy and tumor metastasis suppression. Theranostics.
dynamic therapy and chemotherapy. Biomater Sci. 2020;8(12):3348–58. 2021;11(20):10001–111.
291. Li X, Wei Y, Wu Y, Yin L. Hypoxia-induced pro-protein therapy assisted 313. He Y, Guo S, Zhang Y, Liu Y, Ju H. NIR-II reinforced intracellular cyclic
by a self-catalyzed nanozymogen. Angew Chem Int Ed Engl. reaction to enhance chemodynamic therapy with abundant H2O2 sup-
2020;59(50):22544–53. ply. Biomaterials. 2021;275:120962.
292. Zhou F, Fu T, Huang Q, Kuai H, Mo L, Liu H, Wang Q, et al. Hypoxia- 314. He Y, Guo S, Zhang Y, Liu Y, Ju H. Near-infrared photo-controlled
activated PEGylated conditional aptamer/antibody for cancer imaging permeability of a biomimetic polymersome with sustained drug
with improved specificity. J Am Chem Soc. 2019;141(46):18421–7. release and efficient tumor therapy. ACS Appl Mater Interfaces.
293. Zhang R, Li Y, Zhang M, Tang Q, Zhang X. Hypoxia-responsive drug-drug 2021;13(13):14951–63.
conjugated nanoparticles for breast cancer synergistic therapy. RSC 315. Ji Y, Lu F, Hu W, Zhao H, Tang Y, Li B, Hu X, et al. Tandem activated photo-
Adv. 2016;6(36):30268–76. dynamic and chemotherapy: using pH-Sensitive nanosystems to realize
294. Li J, Meng X, Deng J, Lu D, Zhang X, Chen Y, Zhu J, et al. Multifunctional different tumour distributions of photosensitizer/prodrug for amplified
micelles dually responsive to hypoxia and singlet oxygen: enhanced combination therapy. Biomaterials. 2019;219: 119393.
photodynamic therapy via interactively triggered photosensitizer 316. Ma Z, Zhang Y, Dai X, Zhang W, Foda MF, Zhang J, Zhao Y, et al. Selective
delivery. ACS Appl Mater Interfaces. 2018;10(20):17117–28. thrombosis of tumor for enhanced hypoxia-activated prodrug therapy.
295. Li Z, Wu M, Bai H, Liu X, Tang G. Light-enhanced hypoxia-responsive Adv Mater. 2021;33(41):e2104504.
nanoparticles for deep tumor penetration and combined chemo- 317. Pan W, Tan Y, Meng W, Huang N, Zhao Y, Yu Z, Huang Z, et al. Microen-
photodynamic therapy. Chem Commun. 2018;54(93):13127–30. vironment-driven sequential ferroptosis, photodynamic therapy, and
296. Liu H, Zhang R, Niu Y, Li Y, Qiao C, Weng J, Li J, et al. Development of chemotherapy for targeted breast cancer therapy by a cancer-cell-
hypoxia-triggered prodrug micelles as doxorubicin carriers for tumor membrane-coated nanoscale metal-organic framework. Biomaterials.
therapy. RSC Adv. 2015;5(27):20848–57. 2022;283:121449.
297. Kulkarni P, Haldar MK, You S, Choi Y, Mallik S. Hypoxia-responsive 318. Guo Y, Jia HR, Zhang X, Zhang X, Sun Q, Wang S, Zhao J, et al. A glucose/
polymersomes for drug delivery to hypoxic pancreatic cancer cells. oxygen-exhausting nanoreactor for starvation- and hypoxia-activated
Biomacromol. 2016;17(8):2507–13. sustainable and cascade chemo-chemodynamic therapy. Small.
298. Li X, Pan Y, Chen C, Gao Y, Liu X, Yang K, Luan X, et al. Hypoxia-respon- 2020;16(31):2000897.
sive gene editing to reduce tumor thermal tolerance for mild-photo- 319. Yang S, Tang Z, Hu C, Zhang D, Shen N, Yu H, Chen X. Selectively poten-
thermal therapy. Angew Chem Int Ed Engl. 2021;60(39):21200–4. tiating hypoxia levels by combretastatin A4 nanomedicine: toward
299. Cheng H, Zhang S, Qi J, Liang X, Yoon J. Advances in Application of highly enhanced hypoxia-activated prodrug tirapazamine therapy for
azobenzene as a trigger in biomedicine: molecular design and sponta- metastatic tumors. Adv Mater. 2019;31(11):e1805955.
neous assembly. Adv Mater. 2021;33(26):e2007290. 320. Shao Y, Liu B, Di Z, Zhang G, Sun L, Li L, Yan C. Engineering of upcon-
300. Zhang X, Wu M, Li J, Lan S, Zeng Y, Liu X, Liu J, et al. Light-enhanced verted metal-organic frameworks for near-infrared light-triggered com-
hypoxia-response of conjugated polymer nanocarrier for successive binational photodynamic/chemo-/immunotherapy against hypoxic
synergistic photodynamic and chemo-therapy. ACS Appl Mater Inter- tumors. J Am Chem Soc. 2020;142(8):3939–46.
faces. 2018;10(26):21909–19.
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 35 of 40
321. McKeown SR, Hejmadi MV, McIntyre IA, McAleer JJ, Patterson LH. 341. Chen Z, Liu L, Liang R, Luo Z, He H, Wu Z, Tian H, et al. Bioinspired
AQ4N: an alkylaminoanthraquinone N-oxide showing bioreductive Hybrid protein oxygen nanocarrier amplified photodynamic therapy
potential and positive interaction with radiation in vivo. Br J Cancer. for eliciting anti-tumor immunity and abscopal effect. ACS Nano.
1995;72(1):76–81. 2018;12(8):8633–45.
322. Albertella MR, Loadman PM, Jones PH, Phillips RM, Rampling R, Burnet 342. Liu W, Liu T, Zou M, Yu W, Li C, He Z, Zhang M, et al. Aggressive man-
N, Alcock C, et al. Hypoxia-selective targeting by the bioreductive made red blood cells for hypoxia-resistant photodynamic therapy. Adv
prodrug AQ4N in patients with solid tumors: results of a phase I study. Mater. 2018;30(35):1802006.
Clin Cancer Res. 2008;14(4):1096–104. 343. Yang J, Li W, Luo L, Jiang M, Zhu C, Qin B, Yin H, et al. Hypoxic tumor
323. Steward WP, Middleton M, Benghiat A, Loadman PM, Hayward C, Waller therapy by hemoglobin-mediated drug delivery and reversal of
S, Ford S, et al. The use of pharmacokinetic and pharmacodynamic end hypoxia-induced chemoresistance. Biomaterials. 2018;182:145–56.
points to determine the dose of AQ4N, a novel hypoxic cell cytotoxin, 344. Ano T, Maitani MM, Sato Y, Tsubaki S, Wada Y. Drastic microwave heating
given with fractionated radiotherapy in a phase I study. Ann Oncol. of percolated Pt metal nanoparticles supported on A l2O3 substrate.
2007;18(6):1098–103. Processes. 2020;8(1):72.
324. Nishida CR, Lee M, De Montellano PR. Efficient hypoxic activation of 345. Xu Y, Shi X, Hua R, Zhang R, Yao Y, Zhao B, Liu T, et al. Remarkably
the anticancer agent AQ4N by CYP2S1 and CYP2W1. Mol Pharmacol. catalytic activity in reduction of 4-nitrophenol and methylene blue by
2010;78(3):497–502. Fe3O4@COF supported noble metal nanoparticles. Appl Catal B Environ.
325. Feng L, Cheng L, Dong Z, Tao D, Barnhart TE, Cai W, Chen M, et al. 2020;260:118142.
Theranostic liposomes with hypoxia-activated prodrug to effectively 346. Jin R, Lin B, Li D, Ai H. Superparamagnetic iron oxide nanoparticles for
destruct hypoxic tumors post-photodynamic therapy. ACS Nano. MR imaging and therapy: design considerations and clinical applica-
2017;11(1):927–37. tions. Curr Opin Pharmacol. 2014;18:18–27.
326. Yang Y, Lu Y, Abbaraju PL, Azimi I, Lei C, Tang J, Jambhrunkar M, et al. 347. Neuwelt A, Sidhu N, Hu C, Mlady G, Eberhardt SC, Sillerud LO.
Stepwise degradable nanocarriers enabled cascade delivery for syner- Iron-based superparamagnetic nanoparticle contrast agents
gistic cancer therapy. Angew Chem Int Ed Engl. 2018;28(28):1800706. for MRI of infection and inflammation. AJR Am J Roentgenol.
327. Shen S, Wu Y, Li K, Wang Y, Wu J, Zeng Y, Wu D. Versatile hyaluronic acid 2015;204(3):W302-313.
modified AQ4N-Cu(II)-gossypol infinite coordination polymer nanopar- 348. Shao C, Shen A, Zhang M, Meng X, Song C, Liu Y, Gao X, et al. Oxygen
ticles: multiple tumor targeting, highly efficient synergistic chemo- vacancies enhanced CeO(2): Gd nanoparticles for sensing a tumor vas-
therapy, and real-time self-monitoring. Biomaterials. 2018;154:197–212. cular microenvironment by Magnetic resonance imaging. ACS Nano.
328. Dias GG, King A, de Moliner F, Vendrell M, da Silva Júnior EN. Quinone- 2018;12(12):12629–37.
based fluorophores for imaging biological processes. Chem Soc Rev. 349. Song M, Liu T, Shi C, Zhang X, Chen X. Bioconjugated manganese diox-
2018;47(1):12–27. ide nanoparticles enhance chemotherapy response by priming tumor-
329. Xin F, Wu M, Cai Z, Zhang X, Wei Z, Liu X, Liu J. Tumor microenviron- associated macrophages toward M1-like phenotype and attenuating
ment triggered cascade-activation nanoplatform for synergistic and tumor hypoxia. ACS Nano. 2016;10(1):633–47.
precise treatment of hepatocellular carcinoma. Adv Healthc Mater. 350. Liu J, Chen Q, Zhu W, Yi X, Yang Y, Dong Z, Liu Z. Nanoscale-coordina-
2021;10(10):e2002036. tion-polymer-shelled manganese dioxide composite nanoparticles: a
330. Xu S, Yao H, Pei L, Hu M, Li D, Qiu Y, Wang G, et al. Design, synthesis, and multistage redox/pH/H2O2-responsive cancer theranostic nanoplat-
biological evaluation of NAD(P)H: quinone oxidoreductase (NQO1)- form. Adv Funct Mater. 2017;27(10):1605926.
targeted oridonin prodrugs possessing indolequinone moiety for 351. Xu S, Zhu X, Zhang C, Huang W, Zhou Y, Yan D. Oxygen and Pt(II) self-
hypoxia-selective activation. Eur J Med Chem. 2017;132:310–21. generating conjugate for synergistic photo-chemo therapy of hypoxic
331. Tanabe K, Harada H, Narazaki M, Tanaka K, Inafuku K, Komatsu H, Ito T, tumor. Nat Commun. 2018;9(1):2053.
et al. Monitoring of biological one-electron reduction by (19)F NMR 352. Zheng D, Li B, Li C, Fan J, Lei Q, Li C, Xu Z, et al. Carbon-dot-deco-
using hypoxia selective activation of an (19)F-labeled indolequinone rated carbon nitride nanoparticles for enhanced photodynamic
derivative. J Am Chem Soc. 2009;131(44):15982–3. therapy against hypoxic tumor via water splitting. ACS Nano.
332. Jiho Y, Kurihara R, Kawai K, Yamada H, Uto Y, Tanabe K. Enzymatic activa- 2016;10(9):8715–22.
tion of indolequinone-substituted 5-fluorodeoxyuridine prodrugs in 353. Jiang W, Zhang Z, Wang Q, Dou J, Zhao Y, Ma Y, Liu H, et al. Tumor reoxy-
hypoxic cells. Bioorg Med Chem Lett. 2019;29(11):1304–7. genation and blood perfusion enhanced photodynamic therapy using
333. Cho H, Bae J, Garripelli VK, Anderson JM, Jun HW, Jo S. Redox- ultrathin graphdiyne oxide nanosheets. Nano Lett. 2019;19(6):4060–7.
sensitive polymeric nanoparticles for drug delivery. Chem Commun. 354. Wang X, Gao F, Zhang X. Initiator-loaded gold nanocages as a light-
2012;48(48):6043–5. induced free-radical generator for cancer therapy. Angew Chem Int Ed
334. Dische S, Anderson P, Sealy R, Watson E. Carcinoma of the cervix- Engl. 2017;56(31):9029–33.
anaemia, radiotherapy and hyperbaric oxygen. Br J Radiol. 355. Chen D, Yu Q, Huang X, Dai H, Luo T, Shao J, Chen P, et al. A highly-effi-
1983;56(664):251–5. cient type I photosensitizer with robust vascular-disruption activity for
335. Dische S. The hyperbaric oxygen chamber in the radiotherapy of carci- hypoxic-and-metastatic tumor specific photodynamic therapy. Small.
noma of the bladder. Br J Radiol. 1973;46(541):13–7. 2020;16(23):e2001059.
336. Weaver LK, Hopkins RO, Chan KJ, Churchill S, Elliott CG, Clemmer TP, 356. Chen D, Wang Z, Dai H, Lv X, Ma Q, Yang D, Shao J, et al. Boosting O2·−
Orme JF Jr, et al. Hyperbaric oxygen for acute carbon monoxide poison- photogeneration via promoting intersystem-crossing and electron-
ing. N Engl J Med. 2002;347(14):1057–67. donating efficiency of Aza-BODIPY-based nanoplatforms for hypoxic-
337. Chen Q, Huang Z, Chen H, Shapiro H, Beckers J, Hetzel FW. Improve- tumor photodynamic therapy. Small. 2020;4(7):2000013.
ment of tumor response by manipulation of tumor oxygenation during 357. Liang P, Tang H, Gu R, Xue L, Chen D, Wang W, Yang Z, et al. A
photodynamic therapy. Photochem Photobiol. 2002;76(2):197–203. pH-responsive zinc (II) metalated porphyrin for enhanced photo-
338. Gundersen SI, Palmer AF. Hemoglobin-based oxygen carrier enhanced dynamic/photothermal combined cancer therapy. Sci China Mater.
tumor oxygenation: a novel strategy for cancer therapy. Biotechnol 2019;62(8):1199–209.
Prog. 2008;24(6):1353–64. 358. Heldin CH, Rubin K, Pietras K, Ostman A. High interstitial fluid pressure-
339. Karagoz B, Suleymanoglu S, Uzun G, Bilgi O, Aydinoz S, Haholu A, an obstacle in cancer therapy. Nat Rev Cancer. 2004;4(10):806–13.
Turken O, et al. Hyperbaric oxygen therapy does not potentiate 359. Zhao KMD, Wu HMD, Yang WMD, Cheng YMD, Wang SMD, Jiang
doxorubicin-induced cardiotoxicity in rats. Basic Clin Pharmacol Toxicol. ANMD, Yan KMD, Goldberg SNMD. Can two-step ablation com-
2008;102(3):287–92. bined with chemotherapeutic liposomes achieve better outcome
340. Song G, Ji C, Liang C, Song X, Yi X, Dong Z, Yang K, et al. TaOx decorated than traditional RF ablation? A solid tumor animal study Nanoscale.
perfluorocarbon nanodroplets as oxygen reservoirs to overcome 2022;14(17):6312–22.
tumor hypoxia and enhance cancer radiotherapy. Biomaterials. 360. Milosevic M, Fyles A, Hedley D, Pintilie M, Levin W, Manchul L, Hill R.
2017;112:257–63. Interstitial fluid pressure predicts survival in patients with cervix cancer
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 36 of 40
independent of clinical prognostic factors and tumor oxygen measure- 382. Jiao W, Zhang T, Peng M, Yi J, He Y, Fan H. Design of magnetic nano-
ments. Cancer Res. 2001;61(17):6400–5. platforms for cancer theranostics. Biosensors. 2022;12(1):38.
361. Roh HD, Boucher Y, Kalnicki S, Buchsbaum R, Bloomer WD, Jain RK. Inter- 383. Li J, Yang N, Yang M, Lu C, Xie M. Development of a magnetic MoS
stitial hypertension in carcinoma of uterine cervix in patients: possible system camouflaged by lipid for chemo/phototherapy of cancer.
correlation with tumor oxygenation and radiation response. Cancer Colloids Surf B Biointerfaces. 2022;213:112389.
Res. 1991;51(24):6695–8. 384. Dobson J. Magnetic nanoparticles for drug delivery. Eur J Vasc Endo-
362. Naito M, Ishii T, Matsumoto A, Miyata K, Miyahara Y, Kataoka K. A phe- vasc Surg. 2006;67(1):55–60.
nylboronate-functionalized polyion complex micelle for ATP-triggered 385. Liu Z, Zhao M, Yu L, Peng W, Chen Y, Zhang S. Redox chemistry-ena-
release of siRNA. Angew Chem Int Ed Engl. 2012;51(43):10751–5. bled stepwise surface dual nanoparticle engineering of 2D MXenes
363. Biswas S, Kinbara K, Niwa T, Taguchi H, Ishii N, Watanabe S, Miyata K, for tumor-sensitive and MRI-guided photonic breast-cancer hyper-
et al. Biomolecular robotics for chemomechanically driven guest deliv- thermia in the NIR-II biowindow. Biomater Sci. 2022;10(6):1562–74.
ery fuelled by intracellular ATP. Nat Chem. 2013;5(7):613–20. 386. Fang Z, Yang E, Du Y, Gao D, Wu G, Zhang Y, Shen Y. Biomimetic smart
364. He X, Zhao Y, He D, Wang K, Xu F, Tang J. ATP-responsive controlled nanoplatform for dual imaging-guided synergistic cancer therapy. J
release system using aptamer-functionalized mesoporous silica nano- Mater Chem B. 2022;10(6):966–76.
particles. Langmuir. 2012;28(35):12909–15. 387. Du L, He H, Xiao Z, Xiao H, An Y, Zhong H, Lin M, et al. GSH-
365. Wu C, Chen T, Han D, You M, Peng L, Cansiz S, Zhu G, et al. Engineering responsive metal-organic framework for intratumoral release of NO
of switchable aptamer micelle flares for molecular imaging in living and IDO inhibitor to enhance antitumor immunotherapy. Small.
cells. ACS Nano. 2013;7(7):5724–31. 2022;18(15):e2107732.
366. Zuo X, Song S, Zhang J, Pan D, Wang L, Fan C. A target-responsive 388. Xu Y, Guo Y, Zhang C, Zhan M, Jia L, Song S, Jiang C, et al. Fibronectin-
electrochemical aptamer switch (TREAS) for reagentless detection of coated metal-phenolic networks for cooperative tumor chemo-/
nanomolar ATP. J Am Chem Soc. 2007;129(5):1042–3. chemodynamic/immune therapy via enhanced ferroptosis-mediated
367. Cheng A, Sen D, Yu H. Design and testing of aptamer-based electro- immunogenic cell death. ACS Nano. 2022. https://doi.org/10.1021/
chemical biosensors for proteins and small molecules. Bioelectrochem- acsnano.1c08585.
istry. 2009;77(1):1–12. 389. Tharkar P, Varanasi R, Wong WSF, Jin CT, Chrzanowski W. Nano-
368. Zayats M, Huang Y, Gill R, Ma C, Willner I. Label-free and reagent- enhanced drug delivery and therapeutic ultrasound for cancer treat-
less aptamer-based sensors for small molecules. J Am Chem Soc. ment and beyond. Front Bioeng Biotechnol. 2019;7:324.
2006;128(42):13666–7. 390. Gao H, Wang Z, Tan M, Liu W, Zhang L, Huang J, Cao Y, et al. pH-
369. Zhang J, Wang Y, Chen J, Liang X, Han H, Yang Y, Li Q, et al. Inhibition responsive nanoparticles for enhanced antitumor activity by high-
of cell proliferation through an ATP-responsive co-delivery system of intensity focused ultrasound therapy combined with sonodynamic
doxorubicin and Bcl-2 siRNA. Int J Nanomed. 2017;12:4721–32. therapy. Int J Nanomedicine. 2022;17:333–50.
370. Mo R, Jiang T, DiSanto R, Tai W, Gu Z. ATP-triggered anticancer drug 391. Yin Y, Jiang X, Sun L, Li H, Su C, Zhang Y, Xu G, et al. Continuous
delivery. Nat Commun. 2014;5:3364. inertial cavitation evokes massive ROS for reinforcing sonodynamic
371. Zhao M, Liu Y, Hsieh RS, Wang N, Tai W, Joo KI, Wang P, et al. Clickable therapy and immunogenic cell death against breast carcinoma. Nano
protein nanocapsules for targeted delivery of recombinant p53 protein. Today. 2021;36:101009.
J Am Chem Soc. 2014;136(43):15319–25. 392. Timbie KF, Afzal U, Date A, Zhang C, Song J, Miller GW, Suk JS, et al.
372. Zheng D, Wang K, Bai B, Hu N, Wang H. Swelling and glyphosate- MR image-guided delivery of cisplatin-loaded brain-penetrating
controlled release behavior of multi-responsive alginate-g-P(NIPAm-co- nanoparticles to invasive glioma with focused ultrasound. J Control
NDEAm)-based hydrogel. Carbohydr Polym. 2022;282:119113. Release. 2017;263:120–31.
373. Xu L, Zhong S, Gao Y, Cui X. Thermo-responsive poly(N- 393. Wu M, Chen W, Chen Y, Zhang H, Liu C, Deng Z, Sheng Z, et al.
isopropylacrylamide)-hyaluronic acid nano-hydrogel and its multiple Focused ultrasound-augmented delivery of biodegradable multi-
applications. Int J Biol Macromol. 2022;194:811–8. functional nanoplatforms for imaging-guided brain tumor treatment.
374. Tang L, Wang L, Yang X, Feng Y, Li Y, Feng W. Poly Adv Sci. 2018;5(4):1700474.
(N-isopropylacrylamide)-based smart hydrogels: design, properties and 394. Horise Y, Maeda M, Konishi Y, Okamoto J, Ikuta S, Okamoto Y, Ishii H,
applications. Prog Mater Sci. 2021;115:100702. et al. Sonodynamic therapy with anticancer micelles and high-
375. Liu R, Ding T, Deng P, Yan X, Xiong F, Chen J, Wu Z. Preparation of LCST intensity focused ultrasound in treatment of canine cancer. Front
regulable DES-lignin-g-PNVCL thermo-responsive polymer by ARGET- Pharmacol. 2019;10:545.
ATRP. Int J Biol Macromol. 2022;194:358–65. 395. Wang Y, Deng Y, Luo H, Zhu A, Ke H, Yang H, Chen H. Light-responsive
376. Narang P, Venkatesu P. A molecular interplay for osmolytes-induced nanoparticles for highly efficient cytoplasmic delivery of anticancer
phase behaviour of poly (vinyl methyl ether). Polymer. 2017;131:224–33. agents. ACS Nano. 2017;11(12):12134–44.
377. de Smet M, Heijman E, Langereis S, Hijnen NM, Grüll H. Magnetic 396. Jin H, Zhu T, Huang X, Sun M, Li H, Zhu X, Liu M, et al. ROS-responsive
resonance imaging of high intensity focused ultrasound mediated drug nanoparticles based on amphiphilic hyperbranched polyphospho-
delivery from temperature-sensitive liposomes: an in vivo proof-of- ester for drug delivery: Light-triggered size-reducing and enhanced
concept study. J Control Release. 2011;150(1):102–10. tumor penetration. Biomaterials. 2019;211:68–80.
378. Deng Z, Xiao Y, Pan M, Li F, Duan W, Meng L, Liu X, et al. Hyperthermia- 397. Feng G, Ding D, Li K, Liu J, Liu B. Reversible photoswitching conju-
triggered drug delivery from iRGD-modified temperature-sensitive gated polymer nanoparticles for cell and ex vivo tumor imaging.
liposomes enhances the anti-tumor efficacy using high intensity Nanoscale. 2014;6(8):4141–7.
focused ultrasound. J Control Release. 2016;243:333–41. 398. Dunkel P, Ilaš J. Targeted cancer therapy using compounds activated
379. Yang Y, Yang T, Chen F, Zhang C, Yin B, Yin X, Han L, et al. Degradable by light. Cancers. 2021;13(13):3237.
magnetic nanoplatform with hydroxide ions triggered photoacoustic, 399. Zhang B, Shao C, Zhou K, Li Q, Duan Y, Yang Y, Zhu H. A NIR-trig-
MR imaging, and photothermal conversion for precise cancer theranos- gered multifunctional nanoplatform mediated by Hsp70 siRNA for
tic. Nano Lett. 2022;22(8):3228–35. chemo-hypothermal photothermal synergistic therapy. Biomater Sci.
380. Zhang S, Liu Y, Cao Y, Zhang S, Sun J, Wang Y, Song S, et al. Target- 2021;9(19):6501–9.
ing the Microenvironment of Vulnerable Atherosclerotic Plaques: an 400. Chen L, Li G, Wang X, Li J, Zhang Y. Spherical nucleic acids for near-
emerging diagnosis and therapy strategy for atherosclerosis. Adv Mater. infrared light-responsive self-delivery of small-interfering RNA and
2022;34:e2110660. antisense oligonucleotide. ACS Nano. 2021;15(7):1199–239.
381. Dhas N, Kudarha R, Pandey A, Nikam AN, Sharma S, Singh A, Garkal 401. Xu Z, Huang H, Xiong X, Wei X, Guo X, Zhao J, Zhou S. A near-
A, et al. Stimuli responsive and receptor targeted iron oxide based infrared light-responsive extracellular vesicle as a “Trojan horse” for
nanoplatforms for multimodal therapy and imaging of cancer: conjuga- tumor deep penetration and imaging-guided therapy. Biomaterials.
tion chemistry and alternative therapeutic strategies. J Control Release. 2021;269:120647.
2021;333:188–245.
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 37 of 40
402. Poudel K, Banstola A, Gautam M, Soe Z, Phung CD, Pham LM, Jeong 422. Liu X, Xia T, Fang Y, Zuo H, Dong X, Xu P, Ouyang J. Overcoming the
J-H, et al. Macrophage-membrane-camouflaged disintegrable and blood-brain barrier by using a multistage exosome delivery sys-
excretable nanoconstruct for deep tumor penetration. ACS Appl Mater tem to inhibit central nervous system lymphoma. Nanomedicine.
Interfaces. 2020;12(51):56767–81. 2022;41:102523.
403. Wang Q, Yali X, Hu Q, Huang S, Lin J, Zhou Q. Surface charge switchable 423. Dong S, Bi Y, Sun X, Zhao Y, Sun R, Hao F, Sun Y, et al. Dual-loaded
nano-micelle for pH/redox-triggered and endosomal escape mediated liposomes tagged with hyaluronic acid have synergistic effects in triple-
co-delivery of doxorubicin and paclitaxel in treatment of lung adeno- negative breast cancer. Small. 2022;18(16):e2107690.
carcinoma. Colloids Surf B Biointerfaces. 2022;216:112588. 424. Curcio M, Brindisi M, Cirillo G, Frattaruolo L, Leggio A, Rago V, Nicoletta
404. Poudel K, Park S, Hwang J, Ku SK, Yong CS, Kim JO, Byeon JH. Photother- FP, et al. Smart lipid-polysaccharide nanoparticles for targeted delivery
mally modulatable and structurally disintegratable Sub-8-nm Au1Ag9 of doxorubicin to breast Cancer Cells. Int J Mol Sci. 2022;23(4):2386.
embedded nanoblocks for combination cancer therapy produced by 425. Wang W, Chen C, Ying Y, Lv S, Wang Y, Zhang X, Cai Z, et al. Smart PdH@
plug-in assembly. ACS Nano. 2020;14(9):11040–54. MnO yolk-shell nanostructures for spatiotemporally synchronous
405. Ravi KA, Kusuma KG, Krishnamurthy PT. Carbon nanotubes in targeted hydrogen delivery and oxygen-elevated phototherapy of
drug delivery: focus on anticancer therapies. J Drug Deliv Sci Tec. melanoma. ACS Nano. 2022. https://doi.org/10.1021/acsnano.1c10450.
2020;59:101892. 426. Wu S, Zhang K, Liang Y, Wei Y, An J, Wang Y, Yang J, et al. Nano-enabled
406. Elsaesser A, Howard CV. Toxicology of nanoparticles. Adv Drug Deliv tumor systematic energy exhaustion via Zinc (II) interference medi-
Rev. 2012;64(2):129–37. ated glycolysis inhibition and specific GLUT1 depletion. Adv Sci.
407. Cedervall T, Lynch I, Lindman S, et al. Understanding the nanopar- 2022;9(7):e2103534.
ticle-protein corona using methods to quantify exchange rates 427. He S, Walimbe T, Chen H, Gao K, Kumar P, Wei Y, Hao D, et al. Bioac-
and affinities of proteins for nanoparticles. Proc Natl Acad Sci USA. tive extracellular matrix scaffolds engineered with proangiogenic
2007;104(7):2050–5. proteoglycan mimetics and loaded with endothelial progenitor cells
408. Neagu M, Piperigkou Z, Karamanou K, Engin AB, Docea AO, Constantin promote neovascularization and diabetic wound healing. Bioact Mater.
C, Negrei C, et al. Protein bio-corona: critical issue in immune nanotoxi- 2022;10:460–73.
cology. Arch Toxicol. 2017;91(3):1031–48. 428. Zhang W, Chen X, Ding D, Zhang G, Zhu Z, Yang X, Li M, et al. Real-time
409. Yang W, Wang L, Mettenbrink EM, DeAngelis PL, Wilhelm S. Nanoparti- imaging reveals specific nanoparticle target binding in a syngeneic
cle toxicology. Annu Rev Pharmacol Toxicol. 2021;61:269–89. glioma mouse model. Nanoscale. 2022;14(15):5678–88.
410. Zheng C, Wang Y, Phua SZF, Lim WQ, Zhao Y. ZnO-DOX@ZIF-8 core-shell 429. Liao L, Cen B, Li G, Wei Y, Wang Z, Huang W, He S, et al. A bivalent cyclic
nanoparticles for pH-responsive drug delivery. ACS Biomater Sci Eng. RGD-siRNA conjugate enhances the antitumor effect of apatinib via co-
2017;3(10):2223–9. inhibiting VEGFR2 in non-small cell lung cancer xenografts. Drug Deliv.
411. Cheng W, Pang H, Campen MJ, Zhang J, Li Y, Gao J, Ren D, Ji X, et al. Cir- 2021;28(1):1432–42.
culatory metabolites trigger ex vivo arterial endothelial cell dysfunction 430. Todaro B, Achilli S, Liet B, Laigre E, Tiertant C, Goyard D, Berthet N, et al.
in population chronically exposed to diesel exhaust. Part Fibre Toxicol. Structural influence of antibody recruiting glycodendrimers (ARGs) on
2022;19(1):20. antitumoral cytotoxicity. Biomater Sci. 2021;9(11):4076–85.
412. Yu Z, Li Q, Wang J, Yu Y, Wang Y, Zhou Q, Li P. Reactive oxygen species- 431. Gao J, Wang S, Dong X, Wang Z. RGD-expressed bacterial membrane-
related nanoparticle toxicity in the biomedical field. Nanoscale Res Lett. derived nanovesicles enhance cancer therapy multiple tumorous
2020;15(1):115. targeting. Theranostics. 2021;11(7):3301–16.
413. Aggarwal V, Tuli HS, Varol A, Thakral F, Yerer MB, Sak K, Varol M, et al. Role 432. Kim HR, Cho YS, Chung SW, Choi JU, Ko YG, Park SJ, Kim SY, et al.
of reactive oxygen species in cancer progression: molecular mecha- Caspase-3 mediated switch therapy of self-triggered and long-acting
nisms and recent advancements. Biomolecules. 2019;9(11):735. prodrugs for metastatic TNBC. J Control Release. 2022;346:136–47.
414. Li Y, Zhang P, Tang W, McHugh KJ, Kershaw SV, Jiao M, Huang X, et al. 433. Dou W-T, Xu F, Xu C-X, Gao J, Ru H-B, Luan X, Zhang J, et al. Graphene
Bright, magnetic NIR-II quantum dot probe for sensitive dual-modality nanoribbon-based supramolecular ensembles with dual-receptor
imaging and intensive combination therapy of cancer. ACS Nano. 2022. targeting function for targeted photothermal tumor therapy. Chem Sci.
https://doi.org/10.1021/acsnano.2c01153. 2021;12(33):11089–97.
415. Essa ML, Elashkar AA, Hanafy NAN, Saied EM, El-Kemary M. Dual target- 434. Ouyang Z, Li D, Xiong Z, Song C, Gao Y, Liu R, Shen M, et al. Antifouling
ing nanoparticles based on hyaluronic and folic acids as a promising dendrimer-entrapped copper sulfide nanoparticles enable photoa-
delivery system of the encapsulated 4-Methylumbelliferone (4-MU) coustic imaging-guided targeted combination therapy of tumors and
against invasiveness of lung cancer in vivo and in vitro. Int J Biol Macro- tumor metastasis. ACS Appl Mater Interfaces. 2021;13(5):6069–80.
mol. 2022;206:467–80. 435. Luo Z, Lu L, Xu W, Meng N, Wu S, Zhou J, Xu Q, et al. In vivo self-assem-
416. Alhalabi O, Chen J, Zhang Y, Lu Y, Wang Q, Ramachandran S, Tidwell bled drug nanocrystals for metastatic breast cancer all-stage targeted
RS, et al. MTAP deficiency creates an exploitable target for antifolate therapy. J Control Release. 2022;346:32–42.
therapy in 9p21-loss cancers. Nat Commun. 2022;13(1):1797. 436. Ning S, Zheng Y, Qiao K, Li G, Bai Q, Xu S. Laser-triggered combination
417. Ramezani Farani M, Azarian M, Heydari Sheikh Hossein H, Abdolvahabi therapy by iron sulfide-doxorubicin@functionalized nanozymes for
Z, Mohammadi Abgarmi Z, Moradi A, Mousavi SM, et al. Folic acid- breast cancer therapy. J Nanobiotechnology. 2021;19(1):344.
adorned curcumin-loaded iron oxide nanoparticles for cervical cancer. 437. Sandbhor P, Goda J, Mohanty B, Chaudhari P, Dutt S, Banerjee R. Non-
ACS Appl Bio Mater. 2022;5(3):1305–18. invasive transferrin targeted nanovesicles sensitize resistant glioblas-
418. Shen W, Ge S, Liu X, Yu Q, Jiang X, Wu Q, Tian Y, et al. Folate-function- toma multiforme tumors and improve survival in orthotopic mouse
alized SMMC-7721 liver cancer cell membrane-cloaked paclitaxel models. Nanoscale. 2021;14(1):108–26.
nanocrystals for targeted chemotherapy of hepatoma. Drug Deliv. 438. Janjua TI, Ahmed-Cox A, Meka AK, Mansfeld FM, Forgham H, Ignacio
2022;29(1):31–42. RMC, Cao Y, et al. Facile synthesis of lactoferrin conjugated ultra small
419. Jia L, Li J, Li P, Liu D, Li J, Shen J, Zhu B, et al. Site-specific glycoproteomic large pore silica nanoparticles for the treatment of glioblastoma.
analysis revealing increased core-fucosylation on FOLR1 enhances Nanoscale. 2021;13(40):16909–22.
folate uptake capacity of HCC cells to promote EMT. Theranostics. 439. Ha MJ, Singareeka Raghavendra A, Kettner NM, Qiao W, Damodaran S,
2021;11(14):6905–21. Layman RM, Hunt KK, et al. Palbociclib plus endocrine therapy signifi-
420. Hellsten R, Lilljebjörn L, Johansson M, Leandersson K, Bjartell A. The cantly enhances overall survival of H R+/HER2- metastatic breast cancer
STAT3 inhibitor galiellalactone inhibits the generation of MDSC-like patients compared to endocrine therapy alone in the second-line
monocytes by prostate cancer cells and decreases immunosuppressive setting: A large institutional study. Int J Cancer. 2022;150(12):2025–37.
and tumorigenic factors. Prostate. 2019;79(14):1611–21. 440. Wu H-X, Zhuo K-Q, Wang K. Efficacy of targeted therapy in patients
421. Nakase K, Tanaka I, Tawara I, Shiku H. CD22 expression in acute myeloid with HER2-positive non-small cell lung cancer: a systematic review and
leukemia: close correlation with interleukin-2 receptor α-chain (CD25) meta-analysis. Br J Clin Pharmacol. 2022;88(5):2019–34.
expression and poor prognosis. Leuk Lymphoma. 2022. https://doi.org/
10.1080/10428194.2022.2062346.
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 38 of 40
441. Pang Z, Dong X, Deng H, Wang C, Liao X, Liao C, Liao Y, et al. MUC1 trig- 460. Das M, Joshi A, Devkar R, Seshadri S, Thakore S. Vitamin-H channeled
gers lineage plasticity of Her2 positive mammary tumors. Oncogene. self-therapeutic P-gp inhibitor curcumin-derived nanomicelles for
2022;41(22):3064–78. targeting the tumor milieu by pH- and enzyme-triggered hierarchical
442. Raveendran R, Chen F, Kent B, Stenzel MH. Estrone-decorated polyion disassembly. Bioconjug Chem. 2022;33(2):369–85.
complex micelles for targeted melittin delivery to hormone-responsive 461. Hao Z, Fan W, Hao J, Wu X, Zeng G, Zhang L, Nie S, et al. Efficient
breast cancer cells. Biomacromol. 2020;21(3):1222–33. delivery of micro RNA to bone-metastatic prostate tumors by using
443. Madamsetty VS, Tavakol S, Moghassemi S, Dadashzadeh A, Schneible aptamer-conjugated atelocollagen in vitro and in vivo. Drug Deliv.
JD, Fatemi I, Shirvani A, et al. Chitosan: a versatile bio-platform for breast 2016;23(3):874–81.
cancer theranostics. J Control Release. 2022;341:733–52. 462. Meher N, Seo K, Wang S, Bidkar AP, Fogarty M, Dhrona S, Huang X,
444. Moin A, Wani S, Osmani RA, Abu LA, Khafagy ES, Arab HH, Gangad- et al. Synthesis and preliminary biological assessment of carborane-
harappa HV, et al. Formulation, characterization, and cellular toxicity loaded theranostic nanoparticles to target prostate-specific mem-
assessment of tamoxifen-loaded silk fibroin nanoparticles in breast brane antigen. ACS Appl Mater Interfaces. 2021;13(46):54739–52.
cancer. Drug Deliv. 2021;28(1):1626–36. 463. Ma G, Severic M, Barker M, Pereira S, Ruiz A, Cheung CCL, Al-Jamal
445. Chauhan PS, Kumarasamy M, Sosnik A, Danino D. Enhanced thermo- WT. Dually targeted bioinspired nanovesicle delays advanced pros-
stability and anticancer activity in breast cancer cells of laccase immo- tate cancer tumour growth in vivo. Acta Biomater. 2021;134:559–75.
bilized on pluronic-stabilized nanoparticles. ACS Appl Mater Interfaces. 464. Yang M, Li J, Gu P, Fan X. The application of nanoparticles in cancer
2019;11(43):39436–48. immunotherapy: Targeting tumor microenvironment. Bioact Mater.
446. Chauhan K, Hernandez-Meza JM, Rodríguez-Hernández AG, Juarez- 2021;6(7):1973–87.
Moreno K, Sengar P, Vazquez-Duhalt R. Multifunctionalized biocatalytic 465. Savla SR, Laddha AP, Kulkarni YA. Pharmacology of apocynin: a natural
P22 nanoreactor for combinatory treatment of ER+ breast cancer. J acetophenone. Drug Metab Rev. 2021;53(4):542–62.
Nanobiotechnol. 2018;16(1):17. 466. Tan C, Fan H, Ding J, Han C, Guan Y, Zhu F, Wu H, et al. ROS-responsive
447. Dong J, Zhu C, Zhang F, Zhou Z, Sun M. “Attractive/adhesion force” dual- nanoparticles for oral delivery of luteolin and targeted therapy of
regulatory nanogels capable of CXCR4 antagonism and autophagy ulcerative colitis by regulating pathological microenvironment. Mater
inhibition for the treatment of metastatic breast cancer. J Control Today Bio. 2022;14:100246.
Release. 2022;341:892–903. 467. Yang C, Liu H, Tsai YC, Tseng JY, Liang S, Chen C, Lian W, et al. Inter-
448. Bhattarai S, Mackeyev Y, Venkatesulu BP, Krishnan S, Singh PK. CXC leukin-4 receptor-targeted liposomal doxorubicin as a model for
chemokine receptor 4 (CXCR4) targeted gold nanoparticles potently enhancing cellular uptake and antitumor efficacy in murine colorec-
enhance radiotherapy outcomes in breast cancer. Nanoscale. tal cancer. Cancer Biol Ther. 2015;16(11):1641–50.
2021;13(45):19056–65. 468. Yang F, Wong T, Teng MC, Liu R, Lu M, Liang H, Wei M. Focused ultra-
449. Li Z, Chen G, Ding L, Wang Y, Zhu C, Wang K, Li J, et al. Increased sound and interleukin-4 receptor-targeted liposomal doxorubicin for
survival by pulmonary treatment of established lung metastases enhanced targeted drug delivery and antitumor effect in glioblas-
with dual STAT3/CXCR4 inhibition by siRNA nanoemulsions. Mol Ther. toma multiforme. J Control Release. 2012;160(3):652–8.
2019;27(12):2100–10. 469. Orozco RC, Marquardt K, Mowen K, Sherman LA. Proautoimmune
450. Chen G, Wang Y, Wu P, Zhou Y, Yu F, Zhu C, Li Z, et al. Reversibly stabi- allele of tyrosine phosphatase, PTPN22, enhances tumor immunity. J
lized polycation nanoparticles for combination treatment of early- and Immunol. 2021;207(6):1662–71.
late-stage metastatic breast cancer. ACS Nano. 2018;12(7):6620–36. 470. Hao T, Fu Y, Yang Y, Yang S, Liu J, Tang J, Ridwan KA, et al. Tumor
451. Li Z, Wang F, Li Y, Wang X, Lu Q, Wang D, Qi C, et al. Combined anti- vasculature-targeting PEGylated peptide-drug conjugate prodrug
hepatocellular carcinoma therapy inhibit drug-resistance and metas- nanoparticles improve chemotherapy and prevent tumor metastasis.
tasis via targeting “substance P-hepatic stellate cells-hepatocellular Eur J Med Chem. 2021;219:113430.
carcinoma” axis. Biomaterials. 2021;276:121003. 471. D’Alterio C, Buoncervello M, Ieranò C, Napolitano M, Portella L, Rea G,
452. Wang H, Ellipilli S, Lee WJ, Li X, Vieweger M, Ho YS, Guo P. Multivalent Barbieri A, et al. Targeting CXCR4 potentiates anti-PD-1 efficacy modi-
rubber-like RNA nanoparticles for targeted co-delivery of paclitaxel fying the tumor microenvironment and inhibiting neoplastic PD-1. J
and MiRNA to silence the drug efflux transporter and liver cancer drug Exp Clin Cancer Res. 2019;38(1):432.
resistance. J Control Release. 2021;330:173–84. 472. Karakaş N, Stuckey D, Revai-Lechtich E, Shah K. IL13Rα2-and EGFR-
453. Chittasupho C, Anuchapreeda S, Sarisuta N. CXCR4 targeted dendrimer targeted pseudomonas exotoxin potentiates the TRAIL-mediated
for anti-cancer drug delivery and breast cancer cell migration inhibi- death of GBM cells. Int J Mol Med. 2021;48(1):145.
tion. Eur J Pharm Biopharm. 2017;119:310–21. 473. Pituch KC, Zannikou M, Ilut L, Xiao T, Chastkofsky M, Sukhanova M,
454. Falgàs A, Pallarès V, Unzueta U, Núñez Y, Sierra J, Gallardo A, Alba-Cas- Bertolino N, et al. Neural stem cells secreting bispecific T cell engager
tellón L, et al. Specific cytotoxic effect of an auristatin nanoconjugate to induce selective antiglioma activity. Proc Natl Acad Sci U S A.
towards CXCR4 diffuse large B-cell lymphoma cells. Int J Nanomedicine. 2021;118(9):e2015800118.
2021;16:1869–88. 474. Kemmerer CL, Schittenhelm J, Dubois E, Neumann L, Häsler LM,
455. Nerantzaki M, Michel A, Petit L, Garnier M, Ménager C, Griffete N. Lambert M, Renovanz M, et al. Cerebrospinal fluid cytokine levels are
Biotinylated magnetic molecularly imprinted polymer nanoparticles associated with macrophage infiltration into tumor tissues of glioma
for cancer cell targeting and controlled drug delivery. Chem Commun. patients. BMC Cancer. 2021;21(1):1108.
2022;58(37):5642–5. 475. Abdelmoneem MA, Elnaggar MA, Hammady RS, Kamel SM, Helmy
456. Pathak V, Nolte T, Rama E, Rix A, Dadfar SM, Paefgen V, Banala S, et al. MW, Abdulkader MA, Zaky A, et al. Dual-targeted lactoferrin shell-oily
Molecular magnetic resonance imaging of Alpha-v-Beta-3 integrin core nanocapsules for synergistic targeted/herbal therapy of hepato-
expression in tumors with ultrasound microbubbles. Biomaterials. cellular carcinoma. ACS Appl Mater Interfaces. 2019;11(30):26731–44.
2021;275:120896. 476. Zhang X, Chen X, Li B, Xu S, Wu Z, Hu M, Zhao Z, et al. Active targeted
457. Vinciguerra D, Degrassi A, Mancini L, Mura S, Mougin J, Couvreur P, Janus nanoparticles enable anti-angiogenic drug combining chemo-
Nicolas J. Drug-initiated synthesis of heterotelechelic polymer prodrug therapy agent to prevent postoperative hepatocellular carcinoma
nanoparticles for in vivo imaging and cancer cell targeting. Biomacro- recurrence. Biomaterials. 2022;281:121362.
mol. 2019;20(7):2464–76. 477. Xu X, Saw PE, Tao W, Li Y, Ji X, Yu M, Mahmoudi M, et al. Tumor micro-
458. Liu L, Liu F, Liu D, Yuan W, Zhang M, Wei P, Yi T. A smart theranostic environment-responsive multistaged nanoplatform for systemic RNAi
prodrug system activated by reactive oxygen species for regional and cancer therapy. Nano Lett. 2017;17(7):4427–35.
chemotherapy of metastatic cancer. Angew Chem Int Ed Engl. 478. Wang S, Yu G, Wang Z, Jacobson O, Tian R, Lin L, Zhang F, et al.
2022;61(12):e202116807. Hierarchical tumor microenvironment-responsive nanomedi-
459. Yan C, Liang N, Li Q, Yan P, Sun S. Biotin and arginine modified cine for programmed delivery of chemotherapeutics. Adv Mater.
hydroxypropyl-β-cyclodextrin nanoparticles as novel drug delivery 2018;30:e1803926.
systems for paclitaxel. Carbohydr Polym. 2019;216:129–39.
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 39 of 40
479. Yang Z, Dai Y, Shan L, Shen Z, Wang Z, Yung BC, Jacobson O, et al. 499. Zhang D, Cui P, Dai Z, Yang B, Yao X, Liu Q, Hu Z, et al. Tumor microen-
Tumour microenvironment-responsive semiconducting polymer-based vironment responsive FePt/MoS nanocomposites with chemotherapy
self-assembly nanotheranostics. Nanoscale Horiz. 2019;4(2):426–33. and photothermal therapy for enhancing cancer immunotherapy.
480. Wang F, Fan Z, Zhu Q, Tian H, Yao J, Jiang B, Zhu F, et al. Tumor Nanoscale. 2019;11(42):19912–22.
microenvironment-activated and viral-mimicking nanodrugs driven by 500. Yang K, Yue L, Yu G, Rao L, Tian R, Wei J, Yang Z, et al. A hypoxia respon-
molecular precise recognition for dNTP inhibition-induced synergistic sive nanoassembly for tumor specific oxygenation and enhanced
cancer therapy. ACS Biomater Sci Eng. 2019;5(9):4442–54. sonodynamic therapy. Biomaterials. 2021;275:120822.
481. Liu G, Tsai HI, Zeng X, Zuo Y, Tao W, Han J, Mei L. Phosphorylcholine- 501. Chen Q, Liang C, Sun X, Chen J, Yang Z, Zhao H, Feng L, et al. HO-
based stealthy nanocapsules enabling tumor microenvironment- responsive liposomal nanoprobe for photoacoustic inflammation imag-
responsive doxorubicin release for tumor suppression. Theranostics. ing and tumor theranostics via in vivo chromogenic assay. Proc Natl
2017;7(5):1192–203. Acad Sci USA. 2017;114(21):5343–8.
482. Li Y, Lin J, Wang P, Luo Q, Lin H, Zhang Y, Hou Z, et al. Tumor microen- 502. Wang X, Zhong X, Bai L, Xu J, Gong F, Dong Z, Yang Z, et al. Ultrafine
vironment responsive Shape-Reversal Self-Targeting Virus-Inspired titanium monoxide (TiO) nanorods for enhanced sonodynamic therapy.
nanodrug for imaging-guided near-infrared-II photothermal chemo- J Am Chem Soc. 2020;142(14):6527–37.
therapy. ACS Nano. 2019;13(11):12912–28. 503. Wen M, Yu N, Wu S, Huang M, Qiu P, Ren Q, Zhu M, et al. On-demand
483. Su Z, Xu Y, Wang Y, Shi W, Han S, Shuai X. A pH and reduction dual- assembly of polymeric nanoparticles for longer-blood-circulation and
sensitive polymeric nanomicelle for tumor microenvironment triggered disassembly in tumor for boosting sonodynamic therapy. Bioact Mater.
cellular uptake and controlled intracellular drug release. Biomater Sci. 2022;18:242–53.
2019;7(9):3821–31. 504. Wu M, Wu L, Li J, Zhang D, Lan S, Zhang X, Lin X, et al. Self-luminescing
484. Han X, Li Y, Xu Y, Zhao X, Zhang Y, Yang X, Wang Y, et al. Reversal of pan- theranostic nanoreactors with intraparticle relayed energy transfer
creatic desmoplasia by re-educating stellate cells with a tumour micro- for tumor microenvironment activated imaging and photodynamic
environment-activated nanosystem. Nat Commun. 2018;9(1):3390. therapy. Theranostics. 2019;9(1):20–33.
485. Yu Z, Wang M, Pan W, Wang H, Li N, Tang B. Tumor microenvironment- 505. Qian C, Yu J, Chen Y, Hu Q, Xiao X, Sun W, Wang C, et al. Light-activated
triggered fabrication of gold nanomachines for tumor-specific hypoxia-responsive nanocarriers for enhanced anticancer therapy. Adv
photoacoustic imaging and photothermal therapy. Chem Sci. Mater. 2016;28(17):3313–20.
2017;8(7):4896–903. 506. Yin W, Qiang M, Ke W, Han Y, Mukerabigwi JF, Ge Z. Hypoxia-responsive
486. Peng H, Zhang X, Yang P, Zhao J, Zhang W, Feng N, Yang W, et al. Defect block copolymer radiosensitizers as anticancer drug nanocarriers for
self-assembly of metal-organic framework triggers ferroptosis to over- enhanced chemoradiotherapy of bulky solid tumors. Biomaterials.
come resistance. Bioact Mater. 2021;19:1–11. 2018;181:360–71.
487. Zhang J, Huang H, Xue L, Zhong L, Ge W, Song X, Zhao Y, et al. 507. Liu C, Ewert KK, Yao W, Wang N, Li Y, Safinya CR, Qiao W. A multifunc-
On-demand drug release nanoplatform based on fluorinated aza- tional lipid incorporating active targeting and dual-control release
BODIPY for imaging-guided chemo-phototherapy. Biomaterials. capabilities for precision drug delivery. ACS Appl Mater Interfaces.
2020;256:120211. 2020;12(1):70–85.
488. Zhang P, Wang J, Chen H, Zhao L, Chen B, Chu C, Liu H, et al. Tumor 508. He H, Zhu R, Sun W, Cai K, Chen Y, Yin L. Selective cancer treatment
microenvironment-responsive ultrasmall nanodrug generators via photodynamic sensitization of hypoxia-responsive drug delivery.
with enhanced tumor delivery and penetration. J Am Chem Soc. Nanoscale. 2018;10(6):2856–65.
2018;140(44):14980–9. 509. Deng J, Liu F, Wang L, An Y, Gao M, Wang Z, Zhao Y. Hypoxia- and
489. Wu M, Xue Y, Li N, Zhao H, Lei B, Wang M, Wang J, et al. Tumor- singlet oxygen-responsive chemo-photodynamic micelles featured
microenvironment-induced degradation of ultrathin gadolinium oxide with glutathione depletion and aldehyde production. Biomater Sci.
nanoscrolls for magnetic-resonance-imaging-monitored, activatable 2018;7(1):429–41.
cancer chemotherapy. Angew Chem Int Ed Engl. 2019;58(21):6880–5. 510. Hua L, Wang Z, Zhao L, Mao H, Wang G, Zhang K, Liu X, et al.
490. Li F, Li T, Zhi D, Xu P, Wang W, Hu Y, Zhang Y, et al. Novel ultrasmall Hypoxia-responsive lipid-poly-(hypoxic radiosensitized polyprodrug)
multifunctional nanodots for dual-modal MR/NIR-II imaging-guided nanoparticles for glioma chemo- and radiotherapy. Theranostics.
photothermal therapy. Biomaterials. 2020;256:120219. 2018;8(18):5088–105.
491. Kim J, Jo C, Lim W, Jung S, Lee YM, Lim J, Lee H, et al. Programmed 511. Fan Y, Tu W, Shen M, Chen X, Ning Y, Li J, Chen T, et al. Targeted
nanoparticle-loaded nanoparticles for deep-penetrating 3D cancer tumor hypoxia dual-mode CT/MR imaging and enhanced radiation
therapy. Adv Mater. 2018;30:e1707557. therapy using dendrimer-based nanosensitizers. Adv Funct Mater.
492. Ye W, Chen X, Li X, Liu Y, Jia F, Jin Q, Ji J. Structure-switchable DNA 2020;30(13):1909285.
programmed disassembly of nanoparticles for smart size tun- 512. Wang W, Lin L, Ma X, Wang B, Liu S, Yan X, Li S, et al. Light-induced
ability and cancer-specific drug release. ACS Appl Mater Interfaces. hypoxia-triggered living nanocarriers for synergistic cancer therapy.
2020;12(20):22560–71. ACS Appl Mater Interfaces. 2018;10(23):19398–407.
493. Liu Y, Jiang M, Tu Y, Wang K, Zong Q, Yuan Y. Time-programmed activa- 513. Perche F, Biswas S, Wang T, Zhu L, Torchilin VP. Hypoxia-targeted siRNA
tion of dual polyprodrugs for synergistic cascade oxidation-chemother- delivery. Angew Chem Int Ed Engl. 2014;53(13):3362–6.
apy. Biomaterials. 2021;278:121136. 514. Long M, Liu X, Huang X, Lu M, Wu X, Weng L, Chen Q, et al. Alen-
494. Wang Z, Guo Y, Fan Y, Chen J, Wang H, Shen M, Shi X. Metal-phenolic- dronate-functionalized hypoxia-responsive polymeric micelles for
network-coated dendrimer-drug conjugates for tumor MR imaging targeted therapy of bone metastatic prostate cancer. J Control Release.
and chemo/chemodynamic therapy via amplification of endoplasmic 2021;334:303–17.
reticulum stress. Adv Mater. 2022;34(7):e2107009. 515. Guo Y, Jia H, Zhang X, Zhang X, Sun Q, Wang S, Zhao J, et al. A glucose/
495. Kim S, Lee J, Im S, Kim WJ. Injectable immunogel based on polymerized oxygen-exhausting nanoreactor for starvation- and hypoxia-activated
phenylboronic acid and mannan for cancer immunotherapy. J Control sustainable and cascade chemo-chemodynamic therapy. Small.
Release. 2022;345:138–46. 2020;16(31):e2000897.
496. Lee Y, Lee S, Jon S. Biotinylated bilirubin nanoparticles as a tumor 516. Chen D, Tang Y, Zhu J, Zhang J, Song X, Wang W, Shao J, et al.
microenvironment-responsive drug delivery system for targeted cancer Photothermal-pH-hypoxia responsive multifunctional nanoplatform
therapy. Adv Sci. 2018;5(6):1800017. for cancer photo-chemo therapy with negligible skin phototoxicity.
497. Chen S, Xue F, Kuang Y, Chen S, Sheng D, Chen H. A self-activating nan- Biomaterials. 2019;221:119422.
ovesicle with oxygen-depleting capability for efficient hypoxia-respon- 517. Zhang X, Chen X, Wang H, Jia H, Wu F. Supramolecular nanogel-based
sive chemo-thermo cancer therapy. Biomaterials. 2021;269:120533. universal drug carriers formed by “soft-hard” co-assembly: accurate
498. Wang W, Guo Z, Lu Y, Shen X, Chen T, Huang R-T, Zhou B, et al. Receptor- cancer diagnosis and hypoxia-activated cancer therapy. Adv Ther.
mediated and tumor-microenvironment combination-responsive Ru 2019;2(5):1800140.
nanoaggregates for enhanced cancer phototheranostics. ACS Appl
Mater Interfaces. 2019;11(19):17294–305.
Tian et al. Journal of Hematology & Oncology (2022) 15:132 Page 40 of 40
518. Zhang K, Zhang Y, Meng X, Lu H, Chang H, Dong H, Zhang X. Light- photothermal-differentiation-chemotherapy of breast cancer stem
triggered theranostic liposomes for tumor diagnosis and combined cells. Biomaterials. 2022;286:121598.
photodynamic and hypoxia-activated prodrug therapy. Biomaterials. 537. Xiong Z, Wang Y, Zhu W, Ouyang Z, Zhu Y, Shen M, Xia J, et al. A dual-
2018;185:301–9. responsive platform based on antifouling dendrimer-CuS nanohybrids
519. He Z, Dai Y, Li X, Guo D, Liu Y, Huang X, Jiang J, et al. Hybrid nanomedi- for enhanced tumor delivery and combination therapy. Small Methods.
cine fabricated from photosensitizer-terminated metal-organic frame- 2021;5(6):e2100204.
work nanoparticles for photodynamic therapy and hypoxia-activated 538. Borbély AA, Achermann P, Trachsel L, Tobler I. Sleep initiation and initial
cascade chemotherapy. Small. 2019;15(4):e1804131. sleep intensity: interactions of homeostatic and circadian mechanisms.
520. Luan X, Guan Y, Liu H, Lu Q, Zhao M, Sun D, Lovell JF, et al. A tumor J Biol Rhythms. 1989;4(2):149–60.
vascular-targeted interlocking trimodal nanosystem that induces and 539. Deng C, Liu Y, Zhou F, Wu M, Zhang Q, Yi D, Yuan W, et al. Engineering
exploits hypoxia. Adv Sci. 2018;5(8):1800034. of dendritic mesoporous silica nanoparticles for efficient delivery of
521. Zhang D, Cai Z, Liao N, Lan S, Wu M, Sun H, Wei Z, et al. pH/hypoxia water-insoluble paclitaxel in cancer therapy. J Colloid Interface Sci.
programmable triggered cancer photo-chemotherapy based on a 2021;593:424–33.
semiconducting polymer dot hybridized mesoporous silica framework. 540. Metawea ORM, Abdelmoneem MA, Haiba NS, Khalil HH, Teleb M,
Chem Sci. 2018;9(37):7390–9. Elzoghby AO, Khafaga AF, et al. A novel ’smart’ PNIPAM-based copoly-
522. Zhang H, Jiang W, Liu R, Zhang J, Zhang D, Li Z, Luan Y. Rational design mer for breast cancer targeted therapy: synthesis, and characterization
of metal organic framework nanocarrier-based codelivery system of of dual pH/temperature-responsive lactoferrin-targeted PNIPAM-co-AA.
doxorubicin hydrochloride/verapamil hydrochloride for overcoming Colloids Surf B Biointerfaces. 2021;202:111694.
multidrug resistance with efficient targeted cancer therapy. ACS Appl 541. Wang Z, He Q, Zhao W, Luo J, Gao W. Tumor-homing, pH- and
Mater Interfaces. 2017;9(23):19687–97. ultrasound-responsive polypeptide-doxorubicin nanoconjugates
523. Yao W, Yao J, Qian F, Que Z, Yu P, Luo T, Zheng D, et al. Paclitaxel- overcome doxorubicin resistance in cancer therapy. J Control Release.
loaded and folic acid-modified PLGA nanomedicine with glutathione 2017;264:66-75.
response for the treatment of lung cancer. Acta Biochim Biophys Sin. 542. Shu M, Tang J, Chen L, Zeng Q, Li C, Xiao S, Jiang Z, et al. Tumor micro-
2021;53(8):1027–36. environment triple-responsive nanoparticles enable enhanced tumor
524. Wang Y, Wang Q, Feng W, Yuan Q, Qi X, Chen S, Yao P, et al. Folic acid- penetration and synergetic chemo-photodynamic therapy. Biomateri-
modified ROS-responsive nanoparticles encapsulating luteolin for als. 2021;268:120574.
targeted breast cancer treatment. Drug Deliv. 2021;28(1):1695–708. 543. Wang F, Huang Q, Wang Y, Shi L, Shen Y, Guo S. NIR-light and GSH acti-
525. Gao W, Meng T, Shi N, Zhuang H, Yang Z, Qi X. Targeting and vated cytosolic p65-shRNA delivery for precise treatment of metastatic
microenvironment-responsive lipid nanocarrier for the enhancement cancer. J Control Release. 2018;288:126–35.
of tumor cell recognition and therapeutic efficiency. Adv Healthc Mater.
2015;4(5):748–59.
526. Tian H, Zhou L, Wang Y, Nice EC, Huang C, Zhang H. A targeted Publisher’s Note
nanomodulator capable of manipulating tumor microenvironment Springer Nature remains neutral with regard to jurisdictional claims in pub-
against metastasis. J Control Release. 2022;348:590–600. lished maps and institutional affiliations.
527. Xi L, Wang J, Wang Y, Ge Z. Dual-Ttargeting polymeric nanocarriers to
deliver ROS-responsive prodrugs and combat multidrug resistance of
cancer cells. Macromol Biosci. 2021;21(9):e2100091.
528. Huang Y, Xue Z, Zeng S. Hollow mesoporous Bi@PEG-FA nanoshell
as a novel dual-stimuli-responsive nanocarrier for synergistic
chemo-photothermal cancer therapy. ACS Appl Mater Interfaces.
2020;12(28):31172–81.
529. Zhang H, Zhang J, Li Q, Song A, Tian H, Wang J, Li Z, et al. Site-specific
MOF-based immunotherapeutic nanoplatforms via synergistic tumor
cells-targeted treatment and dendritic cells-targeted immunomodula-
tion. Biomaterials. 2020;245:119983.
530. Li X, Pan Y, Zhou J, Yi G, He C, Zhao Z, Zhang Y. Hyaluronic acid-modified
manganese dioxide-enveloped hollow copper sulfide nanoparticles
as a multifunctional system for the co-delivery of chemotherapeutic
drugs and photosensitizers for efficient synergistic antitumor treat-
ments. J Colloid Interface Sci. 2022;605:296–310.
531. Yuan Y, Liu J, Yu X, Liu X, Cheng Y, Zhou C, Li M, et al. Tumor-targeting
pH/redox dual-responsive nanosystem epigenetically reverses cancer
drug resistance by co-delivering doxorubicin and GCN5 siRNA. Acta
Biomater. 2021;135:556–66.
532. Mo C, Wang Z, Yang J, Ouyang Y, Mo Q, Li S, He P, et al. Rational assem-
bly of RGD/MoS/Doxorubicin nanodrug for targeted drug delivery,
GSH-stimulus release and chemo-photothermal synergistic antitumor
Ready to submit your research ? Choose BMC and benefit from:
activity. J Photochem Photobiol B. 2022;233:112487.
533. Meng X, Lu Z, Lv Q, Jiang Y, Zhang L, Wang Z. Tumor metabolism
• fast, convenient online submission
destruction via metformin-based glycolysis inhibition and glucose
oxidase-mediated glucose deprivation for enhanced cancer therapy. • thorough peer review by experienced researchers in your field
Acta Biomater. 2022;145:222–34. • rapid publication on acceptance
534. Shen Y, Wang M, Wang H, Zhou J, Chen J. Multifunctional human
• support for research data, including large and complex data types
serum albumin fusion protein as a docetaxel nanocarrier for chemo-
photothermal synergetic therapy of ovarian cancer. ACS Appl Mater • gold Open Access which fosters wider collaboration and increased citations
Interfaces. 2022;14(17):19907–17. • maximum visibility for your research: over 100M website views per year
535. Li X, Wang Z, Ma M, Chen Z, Tang XL, Wang Z. Self-Assembly iron oxide
nanoclusters for photothermal-mediated synergistic chemo/chemody- At BMC, research is always in progress.
namic therapy. J Immunol Res. 2021;2021:9958239.
536. Xie X, Jiang K, Li B, Hou S, Tang H, Shao B, Ping Y, et al. A small- Learn more biomedcentral.com/submissions
molecule self-assembled nanodrug for combination therapy of