IMR Press / FBL / Volume 27 / Issue 5 / DOI: 10.31083/j.fbl2705145
Open Access Review
Recent Advancements of Nanotechnology-Based Strategies for Overcoming Tumor Microenvironment Hypoxia
Jiangchao Wu1,2,3,†Jinyuan Song1,2,3,†Xianghong Yin4Jianghui Tang1,2,3Junlei Zhang1,2,3Xun Wang1,2,3Yongtao Ji1,2,3Yaxing Zhao1,2,3Dong Chen5,6Jianpeng Sheng1,2,3,*Xueli Bai1,2,3,*Tingbo Liang1,2,3,*
Show Less
1 Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 310000 Hangzhou, Zhejiang, China
2 Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, 310000 Hangzhou, Zhejiang, China
3 Zhejiang University Cancer Center, Zhejiang University, 310000 Hangzhou, Zhejiang, China
4 Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310000 Hangzhou, Zhejiang, China
5 Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310000 Hangzhou, Zhejiang, China
6 College of Energy Engineering and State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University, 310027 Hangzhou, Zhejiang, China
*Correspondence: shengjp@zju.edu.cn (Jianpeng Sheng); shirleybai@zju.edu.cn (Xueli Bai); liangtingbo@zju.edu.cn (Tingbo Liang)
These authors contributed equally.
Academic Editors: Yoshikatsu Koga, Kenji Takashima and Takahiro Anzai
Front. Biosci. (Landmark Ed) 2022, 27(5), 145; https://doi.org/10.31083/j.fbl2705145
Submitted: 8 February 2022 | Revised: 27 February 2022 | Accepted: 4 March 2022 | Published: 6 May 2022
Copyright: © 2022 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Hypoxia is a typical characteristic of most solid malignancies, which has multiple effects on malignant phenotypes and biological behaviors of tumors including epithelial-mesenchymal-transition (EMT), invasion, migration, metastasis, autophagy, stem cell maintenance, pathological angiogenesis, drug resistance, and immunosuppression. Rcentlyumoand reversing the tumor hypoxic environment via nanotechnology has emerged as a novel therapeutic approach for the treatment of malignancies. The main strategies related to nanotechnology to alleviate or ameliorate hypoxic environment are as follows: (1) Bringing external oxygen to tumor hypoxic microenvironment; (2) Generating oxygen based on nanotechnology in situ; (3) Regulating the structure of the tumor microenvironment; (4) Decreasing oxygen consumption in the tumor microenvironment. In this review, we will discuss these nanotechnologies in detail.

Keywords
nanotechnology
tumor microenvironment
hypoxia
metal-organic frameworks
photodynamic therapy
1. Introduction

Most solid malignancies are characterized by hypoxic tumor microenvironment (TME) due to the malformed blood vessel that cannot provide adequate oxygen or the imbalance between oxygen support and consumption in tumor cells [1, 2, 3, 4, 5, 6]. Intratumoral hypoxia leads to increased activity of hypoxia-inducible factors (HIFs), which plays an important role in tumor progression and affect malignant tumor hallmarks including but not limited to cell proliferation, differentiation, apoptosis, genetic instability, tumor metabolism, vascularization/angiogenesis, immunosuppression, and metastasis [7]. In addition, tumor hypoxic microenvironment is a significant obstacle to oxygen-dependent cancer therapy such as radiotherapy, chemotherapy, photodynamic therapy (PDT), immunotherapy, and so on [8].

Overall, hypoxia is regarded as a critical role in tumor progression and resistance to tumor therapy, which makes it a novel target for cancer therapy [9]. Recently, many researchers have tried to target hypoxia TME via nanotechnology. In this review, we focused on the recent advancements of nanotechnology-based strategies for overcoming hypoxia in TME. The main strategies related to nanotechnology to alleviate or ameliorate hypoxic environment are as follows: (1) Bringing external oxygen to tumor hypoxia microenvironment; (2) Generating oxygen based on nanotechnology in situ; (3) Regulating the structure of the TME; (4) Decreasing consumption of oxygen in the TME (Fig. 1). These strategies are shown in Table 1 (Ref. [10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35] and will be discussed in detail in the following sections.

Fig. 1.

Schematic illustration of the strategies based on nanotechnology for overcoming tumor hypoxia microenvironment.

Table 1.Nanotechnology-based strategies for overcoming tumor microenvironment hypoxia and typical examples in vivo.
Strategy Nanoparticles Tumor model (cell line) References
Bring external oxygen to tumor hypoxia microenvironment Oxy-PDT Colon carcinoma (CT26) [10]
HMSNs-GOx-Ce6@CPPO-PFC/O2 Melanoma lung metastasis (B16 F10) [11]
PFC@PLGA-RBCM Breast carcinoma (4T1) [12]
IPH@RBC Colon carcinoma (CT26) [13]
PCN-224-Pt Liver carcinoma (H22) [14]
O2@UiO-66@ICG@RBC Breast carcinoma (MCF-7) [15]
IR780@O2-SFNs/iRGD Breast carcinoma (4T1) [16]
ZDZP@PDA–PEG Breast carcinoma (4T1) [17]
Polydopamine-nanoparticle-stabilized oxygen microcapsules Pancreatic carcinoma (KPC) [18]
Generating O2 based on nanotechnology in situ LipoMB/CaO2 Breast carcinoma (4T1) [19]
A-MnO2 Breast carcinoma (EMT6) [20]
BSA-MnO2/IR820@OCNC Esophageal carcinoma (KYSE 30) [21]
HSA-MnO2-Ce6&Pt Breast carcinoma (4T1) [22]
Fuco-MnO₂ Pancreatic carcinoma (BxPC-3) [23]
CAT@Pt (IV)-liposome Breast carcinoma (4T1) [24]
HA-CAT@aCe6 Breast carcinoma (MDA-MB-231) [25]
mCMSNs Breast carcinoma (MCF-7) [26]
PDA-Pt-CD@RuFc Breast carcinoma (4T1) [27]
PCCN Breast carcinoma (4T1) [28]
FA-WN-Ce6 Colon carcinoma (CT26) [29]
Regulating the structure of TME SPION@hMSN Breast carcinoma (4T1) [30]
α-CD-Ce6-NO Breast carcinoma (MCF-7) [31]
DiR-hCe6-liposome Breast carcinoma (4T1) [32]
Decreasing oxygen consumption in the TME PV-TS Breast carcinoma (4T1) [33]
Mito-OxE Breast carcinoma (4T1) [34]
SORgenTAM Breast carcinoma (4T1) [35]
2. Discussion
2.1 The Mechanisms of Hypoxia Promoting Tumor Progression

The hypoxic condition has pleiotropic effects on malignancy phenotypes and biological behaviors of tumors including such as EMT, invasion, migration, metastasis, cancer stem cell maintenance, autophagy, pathological angiogenesis, drug resistance, and immunosuppression [36, 37, 38, 39, 40]. For example, Hypoxia plays an important role in causing the EMT, invasion, migration, and metastasis of pancreatic ductal adenocarcinoma (PDAC) [37]. Tumor cells acquire and maintain cancer stem cell (CSC) phenotype under hypoxic conditions, which has increased self-renewal and invasion potential [41, 42]. Autophagy is associated with cellular response to stresses such as hypoxia in the TME [43, 44, 45, 46]. Evidence suggested that enhanced autophagy promoted tumor survival [45]. Angiogenesis is another important component in the TME under hypoxic conditions [47], and HIF-1α is essential to hypoxia-induced angiogenesis via the expression of VEGF in the tumor [48]. Under hypoxic conditions in PDAC, cancer-associated fibroblasts (CAFs) and dense extracellular matrix (ECM) components were deposited to induce extrinsic resistance for drugs [49]. The strategy of blocking immune checkpoints by immune checkpoint inhibitors (ICIs) has achieved great success in melanoma and lung cancer [50, 51, 52]. However, this strategy is limited due to its low response rate in some types of cancers in clinics such as PDAC, which is characterized by hypoxic conditions in TME [53]. Thus, tumor hypoxic microenvironment is a significant impediment to the efficacy of tumor therapy [8]. Relieving the hypoxia status of TME is a promising strategy for tumor treatment.

2.2 Recent Advancements about the Strategies for Overcoming Tumor Hypoxic Microenvironment Based on Nanotechnology
2.2.1 Bring External Oxygen to Tumor Hypoxia Microenvironment

Delivering oxygen to a hypoxic microenvironment by nanomaterials is the most common method to alleviate tumor hypoxia. Recently, various nanotechnology-based methods have been developed to bring external oxygen to the tumor hypoxic microenvironment.

2.2.1.1 Artificial Red Blood Cells Substitutes (RBCSs)

The most important function of natural Red Blood Cells (RBCs) is to transfer oxygen (O2) from the oxygen-enriched tissues to oxygen-deficient tissues. Each RBC contains thousands of hemoglobin (Hb) molecules and each Hb molecule combines with four oxygen molecules. In a hypoxic microenvironment, Hb releases oxygen very quickly to improve the condition of hypoxia [54].

Many researchers are devoted to assembling semi-synthetic RBCs with Hb as the oxygen carrier, which are called Hb-based oxygen carriers (HBOCs) (Fig. 2i). There are several different HBOC systems such as cell-free Hb [55, 56] and particle-encapsulated Hb [57, 58, 59]. Cell-free Hb has some disadvantages including short circulation time, poor stability, and potential side effects, due to the dissociation of Hb tetramer, binding to plasma haptoglobin, and clearance by liver, kidney, and spleen [60, 61]. By chemical modification or encapsulation with biodegradable materials, Hb-based O2 carriers could reduce the disadvantages of cell-free Hb system and the capability to carry oxygen was similar to natural RBCs [62, 63, 64, 65, 66].

Fig. 2.

Schematic illustration of the strategies based on nanotechnology for overcoming tumor hypoxia microenvironment. (i) The morphology and structure of artificial RBCs. (ii) The morphology and structure of PFC-based oxygen carriers. PFC was wrapped in PLGA shell and then coated with RBCM. (iii) The illustration of construction of a MOF and its building components. (iv) The morphology and structure of polydopamine-nanoparticle-stabilized oxygen microcapsules. (v) The illustration of Chemical decomposition like metallic oxides producing oxygen. (vi) The schematic diagram of water-splitting strategy breaks down water to produce oxygen. (vii) The schematic diagram of catalase breaks down H2O2 to produce oxygen. (viii) The schematic diagram of regulating the structure by increasing blood flow. (ix) The schematic diagram of decreasing oxygen consumption.

Compared with natural RBCs, Hb-based O2 carriers can infiltrate into tumor tissues through the narrow vascular structure and provide enough O2 in hypoxic tumors [67]. Nonetheless, the switch of Hb into non-functional methemoglobin (metHb) occurs in the HBOC system, which is prevented in natural RBCs by a multi-enzyme system. Therefore, next-generation HBOCs are desired to create a nanomaterial system with excellent oxygen-carrying capacity and reduced conversion of Hb into metHb [68].

2.2.1.2 PFC-Based Oxygen Carriers (PFOCs) and Fe2+ Porphyrin Systems

To alleviate the disadvantages of HBOCs, many investigators have focused on synthetic molecules to replace the function of natural Hb. Perfluorocarbons (PFCs) have a great potential to carry gases and are used as oxygen carriers due to their stability and biosafety [10, 11].

PFCs are inert liquids, and the shape can be cyclic or linear so that PFCs can easily dissolve tremendous amounts of O2 and exchange gases with high efficiency. Nevertheless, PFCs are not water-soluble and will formulate into emulsions in vivo. Another advantage of PFCs is that oxygen load can be precisely controlled [69].

Because of the rapid growth of the tumor, the center of the tumor is often hypoxic, which prohibits the production of reactive oxygen species (ROS) and leads to the tolerance of tumor cells to radiotherapy [70, 71]. PFOCs can promote oxygen concentration at the hypoxic site of the tumor to increase the sensitivity of tumor cells to radiotherapy [8, 72, 73]. For example, Gao et al. [12] reported that biocompatible PFC@poly (d,l-lactide-co-glycolide) (PLGA) nanoparticles, which were further coated with a red-blood-cell membrane (RBCM) to create PFC@PLGA-RBCM nanoparticles (Fig. 2ii).

PFC@PLGA-RBCM nanoparticles can availably deliver oxygen to the oxygen-starved interior of the tumor, which greatly relieves tumor hypoxia and thus enhance the treatment efficacy of radiotherapy [12, 74]. Furthermore, PFOCs can be used in enhancing the efficacy of PDT and chemotherapy. Although PFOCs have the outstanding oxygen-carrying ability, the hydrophobic features of PFOCs block their application in tumors [11]. In addition, extensive and enduring exposure to PFOCs may lead to some adverse reactions, including elevated central venous pressure, cutaneous flushing, pulmonary hypertension, chest tightness, fever, and hypotension [75].

Fe2+ protoporphyrin IX (heme), which comes from Hb, can stably bind oxygen. Therefore, another design of oxygen carriers was to explore the Fe2+ porphyrins. Many studies synthesized an amphiphilic Fe2+ porphyrin and encapsulated Fe2+ porphyrin into liposomes. These vesicles showed reversibly binding and releasing oxygen, similar to Hb [76].

2.2.1.3 Metal–Organic Frameworks (MOFs)

In recent years, metal-organic frameworks (MOFs) (Fig. 2iii) are a useful material in many domains for their catalytically active sites, large porosity, and flexible structure [77, 78]. In addition, MOFs are appropriate candidates for gas storage and separation due to the large surface area and uniform pore size.

DeCoste et al. [79] systematically explored MOFs for the adsorption of oxygen. The working capacity of MOFs was determined by high-pressure oxygen isotherms. Simulations were performed on the oxygen capacity of 10,000 hypothetical MOF materials in a database created using established high-speed MOF generation techniques, NU-125 was selected as prime candidate based on its superior ability to absorb oxygen and currently known synthesis techniques, and NU-125 was proved to have more capacity for oxygen than Norit activated carbon and zeolite NaX. These MOFs were proved to be useful for oxygen storage in different fields such as aerospace, military, and medicine [79]. Recently, Moghadam et al. [80] introduced a visualization concept to clarify the relationships between structural properties and oxygen adsorption ability. They employed high-throughput screening (HTS) techniques to explore oxygen storage in a database of 2,932 existing MOFs and discovered a top MOF material (UMCM-152). They further proved that UMCM-152 delivered 22.5% more oxygen than the material known.

Furthermore, MOFs have been explored to improve the therapeutic efficacy of PDT in cancers in combination with photosensitizers (PS). Zhang et al. [14] reported a nanoplatform that could promote the production of O2 in the tumor hypoxic microenvironment via H2O2-activated release of O2, which achieved better therapeutic effects.

Gao et al. [15] reported a PDT nanoplatform that took full advantage of MOFs. As an oxygen carrier, Zirconium (IV)-based MOF (UiO-66) was combined with indocyanine green (ICG) and further loaded with the cytomembrane of RBCs. After laser irradiation, ICG would degrade the cytomembrane of RBC. Meanwhile, the photothermal characteristic of ICG could accelerate the release of oxygen from UiO-66 in the hypoxic TME. Subsequently, the production of O2 could improve the effects of PDT in antitumor therapy, which provides a new therapy to kill hypoxic tumors. Ren et al. [17] reported a nanoplatform (ZDZP@PP) based on the zeolitic imidazolate framework-67 (ZIF-67) core with a pH-responsive catalase, and a polydopamine-poly (ethylene glycol) (PDA-PEG) layer for improved biocompatibility, which was proved to boost the efficiency of chemo-photodynamic therapy.

2.2.1.4 Oxygen Microcapsules

Recently, with the help of interfacial polymerization, our group created an effective oxygen delivery vehicle of polydopamine-nanoparticle-stabilized oxygen microcapsules (Fig. 2iv).

To prepare nanoparticle-stabilized oxygen microcapsules, at first, under vigorous and rapid shearing, dopamine, a neurotransmitter that assists cells to transmit pulses, was oxidized to form polydopamine nanoparticles at the oxygen/water interface. With the help of amino-rich polylysine and chitosan, uniform polydopamine nanoparticles were achieved and the overoxidation of polydopamine nanoparticles was prevented. By optimizing the interfacial adsorption kinetics of polydopamine nanoparticles, chitosan increases the life of oxygen microbubbles. Due to the decline of the interfacial tension, polydopamine nanoparticles temporarily assembled at the surface of oxygen microbubbles. Then, to achieve stable polydopamine-nanoparticle-stabilized oxygen microcapsules, glutaraldehyde was added to permanently crosslink polydopamine nanoparticles. After removing excess solvents and residual polydopamine nanoparticles, nanoparticle-stabilized oxygen microcapsules were well dispersed in oxygen-enriched water. Due to the solid shells availably enclosing oxygen in the core and effectively keeping oxygen microbubbles from coalescing, the oxygen microcapsules keep stable without observable changes for at least one week (Fig. 3, Ref. [1]). We created an effective oxygen delivery vehicle of polydopamine-nanoparticle-stabilized oxygen microcapsules, which effectively improves the hypoxic microenvironment. Gemcitabine (GEM) is a first-line chemotherapy drug for pancreatic cancer. Oxygen microcapsules combined with GEM drugs achieve a synergetic therapeutic effect in pancreatic cancer treatment [18].

Fig. 3.

Preparation of polydopamine-nanoparticle-stabilized oxygen microcapsules. Reproduced with permission [1].

In addition, we reported that oxygen microcapsules can markedly improve radiotherapy efficacy in a mouse model of hepatocellular carcinoma (HCC). Combined treatments of oxygen microcapsules and radiotherapy can effectively decrease tumor-associated macrophages (TAMs) infiltration in tumors while promoting the transformation of pro-tumor type M2 TAMs to anti-tumor type M1 TAMs, which activates the immune responses of T cell against the tumor (Fig. 4, Ref. [2]) [81].

Fig. 4.

Improving the efficacy of radiotherapy and anti-tumor immunity via using oxygen microcapsules. Reproduced with permission [2].

2.2.2 Generating O2 Based on Nanotechnology In Situ

Another way to improve hypoxia in the TME is to produce oxygen in situ. Three major strategies are discussed here, including chemical decomposition system, catalase, and water splitting.

2.2.2.1 Chemical Decomposition System

The chemical decomposition system involves a highly reactive compound that can decompose H2O2 to generate oxygen, which increases the oxygen concentration in the tumor. These compounds are usually metallic oxides, such as CaO2 [19, 82], MnO [83, 84], CeO2 [85, 86, 87], MnO2 [20, 21, 22, 88], etc. (Fig. 2v). MnO2 is the most common metal oxide to produce oxygen for tumor treatment in situ. Furthermore, MnO2 can be degraded into harmless substances-hydrosoluble Mn2+ ions, which can improve T1-weighted magnetic resonance imaging (MRI) [89]. Different morphologies and structures of MnO2 are used to generate O2, such as nanodots, nanotubes, nanoparticles, nanosheets, and so on [23, 90, 91, 92, 93]. For example, Prasad et al. [20] created nanoparticles of polyelectrolyte-albumin complex with MnO2 (A-MnO2). These nanoparticles could produce O2 by decomposing H2O2 in a hypoxic TME of murine breast tumor model. In addition, these nanoparticles decreased the expression of HIF-1α, which restrains tumor progression and aggressiveness. Compare with radiation therapy alone, combination treatment of nanoparticles and radiation dramatically restrained breast tumor growth.

Yang et al. [94] developed a multifunctional nanoplatform (MONs-GOx@MnO2-Ce6) consisting of glucose oxidase (GOx)-coated mesoporous organosilica nanoparticles (MONPs) and MnO2 nanosheets-chlorin e6 (Ce6). When MONPs-GOx@MnO2-Ce6 entered the TME, it could catalyze the glucose to H2O2, which was decomposed by MnO2 nanosheets to produce O2 in the TME. Tseng et al. [95] designed a hypoxia-responsive carrier based on the characteristics of lactate accumulation in tumors to achieve effective and specific delivery of antitumor viruses. The carrier was loaded with lactate oxidase (LOX), and lactate in tumors could penetrate into the carrier and be oxidized into pyruvic acid and H2O2. We think these produced H2O2 can be used as materials for the O2-producing compounds of chemical decomposition system. This provides a mechanistic basis for the combined application of LOX-loaded carriers with H2O2-related O2-producing chemical decomposition system in tumor therapy.

2.2.2.2 Catalase Breaks Down H2O2 to Produce Oxygen

In living cells, catalase is a critical enzyme that prevents cells from oxidative damage via catalyzing H2O2 to generate O2 and water (Fig. 2vii). It is well-known that tumor cells tend to contain higher levels of H2O2 than normal cells, which may result in DNA damage, metastasis, abnormal proliferation, and angiogenesis [96, 97, 98]. However, the instability of catalase limits its use in the production of oxygen due to its short half-life in vivo [89].

A lot of researchers are dedicated to improving the stability of catalase via encapsulating them to various nano-platforms [24, 99, 100]. For example, Phua et al. [25] created catalase-encapsulated hyaluronic-acid (HA)-based nanoparticles loaded with an adamantane-modified photosensitizer to improve the therapeutic effect of PDT. The adamantane-modified Ce6 (aCe6) was created by combining Ce6 with adamantane. The HA-CAT@aCe6 nanoparticles containing catalase can increase the therapeutic effect of PDT by decomposing endogenous H2O2 into water and oxygen. The cell viability experiment manifested that HA-CAT@aCe6 showed high cytotoxicity under 660 nm light exposure at normoxic conditions. HA-CAT@aCe6 also showed high cytotoxicity in a hypoxic microenvironment due to the oxygen production capability of catalase. Therefore, HA-CAT@aCe6 showed an outstanding potential to overcome hypoxic conditions for the treatment effect of PDT.

Liu et al. [101] wrapped the catalase in outer layers of MOFs. The integrated MOF system with characteristics of tandem catalysts can decompose endogenous H2O2 into water and oxygen. The treatment effect of PDT was improved by 8.7-fold than the treatment lack of catalase, which displayed an increased therapeutic effect against tumor cells in tumor hypoxic microenvironment. They reported tumor cell membrane coated mesoporous copper/manganese silicate nanospheres (mCMSNs). mCMSNs can increase ROS generation and reduce glutathione-activated Fenton reaction, which shows outstanding synergistic therapeutic effects. mCMSNs can reduce tumor hypoxic microenvironment via decomposing endogenous H2O2 into water and oxygen and further react with oxygen to release ROS under laser irradiation. Importantly, alleviating hypoxia conditions and reducing ROS scavenger achieves additional therapy effects in vitro and in vivo. In addition, the Mn2+ can play an important role in tumor MRI [26].

Li et al. [100] created a tumor-targeted cascade bioreactor (mCGP) which can increase targetability and retention capacities in the tumor. Once mCGP entered the tumor cell, it can catalyze the endogenous H2O2 to produce oxygen and water, which would next increase the disintegration of intracellular glucose and fortify the generation of cytotoxic singlet oxygen (1O2) under photon exposure. As a result, mCGP showed enhanced therapeutic efficacy due to the combined effect of PDT and cell starvation. Carbon dots (CDs) can effectively produce 1O2 for PDT in oncotherapy. Jia et al. [102] created magnetofluorescent Mn-CDs by utilizing manganese (II) phthalocyanine as a precursor. Mn-CDs can not only produce 1O2 but also decompose H2O2 to release oxygen.

Recently, a multifunctional nano-platform PDA-Pt-CD@RuFc was developed, which consisted of platinum-decorated and cyclodextrin (CD)-modified polydopamine (PDA) nanoparticles combined with a ferrocene-appended ruthenium complex (RFC). PDA-Pt-CD@RuFc nanoparticles combined PDT with photothermal therapy (PTT), which can kill tumor cells in an anoxic microenvironment in several aspects. Firstly, the PDA-Pt-CD@RuFc nanoparticles can decompose H2O2 to release oxygen and water. Secondly, photothermal heating can stretch the blood vessels and then increase oxygen supplement. Thirdly, PDT based on RuFc can also occur through a non-oxygen-dependent Fenton reaction. In addition, combined therapy can reverse the hypoxic microenvironment of the tumor to acquire a better combination therapy effect [27].

2.2.2.3 Generating O2 by Water Splitting

Catalase and catalase-like material can effectively decompose H2O2 to release oxygen and water in the tumor hypoxic microenvironment. However, in bioactive cells, the levels of endogenous H2O2 are limited, which limits the use of H2O2 decomposition strategies in tumors [103, 104]. Therefore, recently, some studies attempted to produce oxygen via splitting water rather than H2O2 to enhance the level of oxygen in the tumor hypoxic microenvironment [105, 106, 107]. The water-splitting strategy can take full advantage of unlimited raw materials to generate oxygen in vivo (Fig. 2vi). Moreover, the usage of water-splitting materials shows a powerful ability to raise oxygen levels in vivo, which improves the PDT effect. For example, by combining carbon dots with C3N4, a carbon nitride (C3N4)-based multifunctional nanocomposite (PCCN) can perform water splitting. Carbon dots were first combined with C3N4 to increase its red-light absorption. Under light exposure, PCCN could increase the intracellular level of oxygen and ROS in the tumor hypoxic microenvironment [28]. Analogously, Ce6 combined with tungsten nitride nanoparticles (WN NPs) decorated with folic acid (FA) modification to produce FA-WN-Ce6 (FWC) nanoparticles. FWC nanoparticles can increase PDT against the hypoxic TME. The WN in FWC plays an important role in water splitting to produce oxygen, which markedly improves the ROS generation ability of Ce6 in hypoxic TME both in vitro and in vivo. Furthermore, FWC nanoparticles can decrease the invasion and metastasis of cancer cells due to increased oxygen levels in the hypoxic TME [29]. Nonetheless, the water-splitting strategy can’t be applied in tissues with limited light source penetration.

2.2.3 Regulating the Structure of TME

In addition to directly carrying oxygen to the tumor hypoxic microenvironment or producing oxygen in situ, regulating the structure of TME can elevate oxygen levels in the tumor hypoxic microenvironment [89].

Increasing Blood Flow

Due to the abnormal blood vessels in the area of the tumor, inadequate blood flow is a common characteristic of solid tumors. Therefore, improving blood flow would be an effective method to increase the oxygen level in the hypoxic TME [108]. For example, Nitric oxide (NO) can mediate the relaxation of the blood vessel. Blood flow can be regulated by controlling the release of NO (Fig. 2viii).

Jin et al. [30] synthesized a novel BNN-type NO-releasing molecule (NORM) activated by ultrasound and a nano-carrier of superparamagnetic iron oxide-encapsulated mesoporous silica nanoparticles (SPION@hMSN) to create a novel nanomaterial (BNN6-SPION@hMSN). BNN6-SPION@hMSN demonstrated excellent tumor-killing ability and ultrasound-triggered NO release to mediate relaxation of the blood vessel. BNN6-SPION@hMSN can efficiently control the release of NO to improve the hypoxia of the TME.

Analogously, Deng et al. [31] invented a nanocarrier with GSH-sensitive NO prodrug. The nanocarrier was fabricated by LEGO-like self-assembly for efficient intracellular delivery of NO, NO released by the nanocarrier can not only improve hypoxia in the tumor by mediating vasodilation, but also increase the concentration of ROS generated by PDT through GSH depletion and generate cytotoxic peroxynitrite anions (ONOO-), resulting in a significant synergistic therapeutic effect with PDT in tumor therapy.

It is well known that the blood flow of cancer can be augmented by mild heating [109, 110]. Brizel et al. [110] created a nanomaterial that can increase blood flow in a hypoxic tumor environment by mild photothermal heating. This is achieved through a near-infrared (NIR) exposure combined with intravenous injection of DiR-hCe6-liposome and then leads to mild photothermal heating, which would increase blood flow in a hypoxic tumor environment and enhance the therapeutic effect of PDT in the hypoxic TME [32].

2.2.4 Decreasing Oxygen Consumption in the TME

Diminishing oxygen consumption is also an alternative treatment strategy to increase oxygen levels in hypoxia of the TME [111]. Therefore, strategies for inhibiting oxidative respiration to decrease tumor oxygen consumption also have been designed to solve the hypoxia issues in tumors [108] (Fig. 2ix). For example, Denko et al put forward an “O2-economizer” concept by preventing cell oxidative respiration to spare endogenous O2 and then overcome the hypoxia barrier. A NO donor and a photosensitizer were combined with poly (D, L-lactide-co-glycolide) nanovesicles to create a PDT-specific O2 economizer. When PDT-specific O2 economizer aggregated into the tumor, the NO donor released NO to restrain cellular respiration, which reserved more oxygen to support the therapeutic effect of PDT. This concept of O2-economizer provides an alternative strategy to reduce hypoxia in the TME [33, 34, 35].

2.3 Characteristics and Comparison of Four Strategies

For strategy of bringing external oxygen to tumor hypoxia microenvironment, due to the existence of membrane protein complexes that are crucial for long-term blood circulation, the RBCM-encapsulated nanoparticles have significantly prolonged blood half-life and biocompatibility. They can be used as long-circulating oxygen carriers to alleviate tumor hypoxia and enhance therapeutic effects [11, 66, 67, 68]. Studies have shown that hypoxia affects the function and phenotype of immune cells in the TME, and RBCs are closely related to immune cells in the liver, such as Kuffer cells. Therefore, RBCM-coated nanoparticles with both immune regulation and oxygen-carrying functions shows research potential for anti-tumor immunotherapy in liver cancer [112, 113]. The advantages of MOFs are high porosity, large surface area and tunable structure, various modifying groups have been introduced into the system to improve the performance and functions of this particle. In addition, single therapeutic way of MOFs doesn’t work very well. Therefore, it’s an urgent need for more combination therapy. For example, MOFs can be combined with more other therapies like immunotherapy to acquire a synergistic effect. Furthermore, the synthesis processes of these well-designed MOFs are often difficult to control. In different tumor microenvironments, different modifying component may alter the kinetic behavior of delivery and release of MOFs, which need to be further clarified in the future [114].

For strategy of Generating O2 based on nanotechnology in situ. MnO2 nanoparticles is the most widely used chemical decomposition nanoparticles, which have outstanding performance for improving the treatment of tumors. MnO2-based nanoplatforms with excellent biodegradability can be gradually broken down into Mn2+ ions, which may cause potential toxicity especially under higher concentrations. In addition, the introduction of other functional groups, including anti-cancer drugs and biomolecules will cause great troubles in the process of massive production. Simple and reliable methods to produce MnO2-based nanosystems would helpful for their applications in the future [115].

For strategy of regulating the structure of TME. Nanoparticles mainly mediate vasodilation in tumors through NO releasing and tissue heating to increase blood flow and oxygen supply in TME. In addition, the release of NO from nanoparticles can also enhance the effect of photodynamic therapy through GSH depletion and reactive nitrogen species generation [30, 31]. It is well known that intravenous injection is one of the main application methods of anti-tumor drugs. The increased blood supply in TME helps improve drug infiltration into the tumor, which offers the possibility of combining these nanoparticles with antitumor drug treatments such as chemotherapy. However, the increased blood flow may lead to adjacent dissemination and distant metastasis of the tumor, this risk requires further study and evaluation.

For strategy of decreasing oxygen consumption in the TME. The nanoparticles mainly target the respiration process of tumor cells, thereby reducing the consumption of oxygen in the tumor and enhancing the effect of PDT. Metabolic reprogramming is a key hallmark of cancer, in which aerobic glycolysis is an important feature [116], so targeting aerobic respiration to reduce oxygen consumption in tumors may have limited effect. In addition, O2-economizer also affect aerobic respiration of immune cells, stromal cells, etc. in TME. These effects may require further research.

3. Conclusions

The tumor hypoxic microenvironment is a major obstacle to oxygen-dependent cancer therapy such as radiotherapy, chemotherapy, PDT, immunotherapy, and so on. Currently, the strategies of nanotechnology for increasing the oxygen level mainly include the following aspects: (1) Bring external oxygen to tumor hypoxia microenvironment. (2) Generating O2 based on nanotechnology in situ. (3) Regulating the structure of the TME. (4) Decreasing oxygen consumption in the TME. These nanotechnologies have ameliorated the hypoxia of certain tumors and achieved enhanced anti-tumor effects, which may reduce malignancy phenotypes and biological behaviors of tumors including EMT, invasion, migration, metastasis, cancer stem cell maintenance, autophagy, pathological angiogenesis, drug resistance, and immunosuppression (Fig. 5).

Fig. 5.

Schematic illustration of the possible antitumor mechanism by alleviating hypoxia.

Although nanotechnology targeting hypoxia has been researched and developed for a long time, the clinical applications associated with it have lagged behind. A variety of nanotechnology have been tried to improve tumor hypoxia, however, there are no FDA-approved treatments to reverse tumor hypoxia. On the one hand, tumor heterogeneity is the most important factor because different tumors have phenotypic features and genetic mutations. This requires that different tumors should be treated in different ways. On the other hand, the size, shape, surface charge and mechanical properties, as well as unstable biocompatibility and safety, also negatively affect the clinical application of these nanoparticles. Further research is needed to improve these aspects. In addition, multifunctional composite nanoparticles with complementary mechanisms have shown encouraging research prospects. The existing nanoparticles targeting hypoxic TME mainly focus on radiotherapy, chemotherapy and PDT and other treatments. The immunomodulatory effects of these nanoparticles and the prospect of combination with immunotherapy deserve further attention.

Abbreviations

PDT, photodynamic therapy; PTT, photothermal therapy; MRI, magnetic resonance imaging; EMT, epithelial-mesenchymal-transition; TME, tumor microenvironment; PDAC, pancreatic ductal adenocarcinoma; HCC, hepatocellular carcinoma; HIF, hypoxia-inducible factor; HBOC, Hb-based oxygen carrier; PFOC, PFC-based oxygen carrier; MOF, Metal–organic framework; NU-125, Northwestern University-125; UMCM-152, University of Michigan Crystalline Material-152; GSH, glutathione; MONP, mesoporous organosilica nanoparticle; ROS, reactive oxygen species.

Author Contributions

JW, JSo wrote the manuscript and contributed equally. XY, JT, JZ, XW, YJ, YZ, and DC provided help and advice on the manuscript, JSh, XB, and TL reviewed the manuscript and secured the funding. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

The authors would like to thank Zhou Meisheng for administrative help.

Funding

This work was funded by Zhejiang Provincial Natural Science Foundation, grant LR22H160009, National Key Research and Development Program of China (grant number 2019YFA0803000, grant 2019YFC1316000), the National Natural Science Foundation of China, grant U20A20378, the National Natural Science Foundation of China, grant 82173078, the National Natural Science Foundation of China, grant 81871925.

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Liu J, Liu Y, Bu W, Bu J, Sun Y, Du J, et al. Ultrasensitive Nanosensors Based on Upconversion Nanoparticles for Selective Hypoxia Imaging in Vivo upon near-Infrared Excitation. Journal of the American Chemical Society. 2014; 136: 9701–9709.
[2]
Jin CS, Lovell JF, Chen J, Zheng G. Ablation of hypoxic tumors with dose-equivalent photothermal, but not photodynamic, therapy using a nanostructured porphyrin assembly. ACS Nano. 2013; 7: 2541–2550.
[3]
Li M, Xia J, Tian R, Wang J, Fan J, Du J, et al. Near-Infrared Light-Initiated Molecular Superoxide Radical Generator: Rejuvenating Photodynamic Therapy against Hypoxic Tumors. Journal of the American Chemical Society. 2018; 140: 14851–14859.
[4]
Wang B, Zhao Q, Zhang Y, Liu Z, Zheng Z, Liu S, et al. Targeting hypoxia in the tumor microenvironment: a potential strategy to improve cancer immunotherapy. Journal of Experimental & Clinical Cancer Research. 2021; 40: 24.
[5]
Fu J, Li T, Yang Y, Jiang L, Wang W, Fu L, et al. Activatable nanomedicine for overcoming hypoxia-induced resistance to chemotherapy and inhibiting tumor growth by inducing collaborative apoptosis and ferroptosis in solid tumors. Biomaterials. 2021; 268: 120537.
[6]
Emami Nejad A, Najafgholian S, Rostami A, Sistani A, Shojaeifar S, Esparvarinha M, et al. The role of hypoxia in the tumor microenvironment and development of cancer stem cell: a novel approach to developing treatment. Cancer Cell International. 2021; 21: 62.
[7]
Wigerup C, Påhlman S, Bexell D. Therapeutic targeting of hypoxia and hypoxia-inducible factors in cancer. Pharmacology & Therapeutics. 2016; 164: 152–169.
[8]
Graham K, Unger E. Overcoming tumor hypoxia as a barrier to radiotherapy, chemotherapy and immunotherapy in cancer treatment. International Journal of Nanomedicine. 2018; 13: 6049–6058.
[9]
Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nature Reviews. Cancer. 2011; 11: 393–410.
[10]
Cheng Y, Cheng H, Jiang C, Qiu X, Wang K, Huan W, et al. Perfluorocarbon nanoparticles enhance reactive oxygen levels and tumour growth inhibition in photodynamic therapy. Nature Communications. 2015; 6: 8785.
[11]
Yu Z, Zhou P, Pan W, Li N, Tang B. A biomimetic nanoreactor for synergistic chemiexcited photodynamic therapy and starvation therapy against tumor metastasis. Nature Communications. 2018; 9: 5044.
[12]
Gao M, Liang C, Song X, Chen Q, Jin Q, Wang C, et al. Erythrocyte-Membrane-Enveloped Perfluorocarbon as Nanoscale Artificial Red Blood Cells to Relieve Tumor Hypoxia and Enhance Cancer Radiotherapy. Advanced Materials. 2017. (in press)
[13]
Ren H, Liu J, Li Y, Wang H, Ge S, Yuan A, et al. Oxygen self-enriched nanoparticles functionalized with erythrocyte membranes for long circulation and enhanced phototherapy. Acta Biomaterialia. 2017; 59: 269–282.
[14]
Zhang Y, Wang F, Liu C, Wang Z, Kang L, Huang Y, et al. Nanozyme Decorated Metal-Organic Frameworks for Enhanced Photodynamic Therapy. ACS Nano. 2018; 12: 651–661.
[15]
Gao S, Zheng P, Li Z, Feng X, Yan W, Chen S, et al. Biomimetic O2-Evolving metal-organic framework nanoplatform for highly efficient photodynamic therapy against hypoxic tumor. Biomaterials. 2018; 178: 83–94.
[16]
Ma S, Zhou J, Zhang Y, Yang B, He Y, Tian C, et al. An Oxygen Self-sufficient Fluorinated Nanoplatform for Relieved Tumor Hypoxia and Enhanced Photodynamic Therapy of Cancers. ACS Applied Materials & Interfaces. 2019; 11: 7731–7742.
[17]
Ren S, Wang B, Zhu X, Zhu D, Liu M, Li S, et al. Oxygen Self-Sufficient Core-Shell Metal-Organic Framework-Based Smart Nanoplatform for Enhanced Synergistic Chemotherapy and Photodynamic Therapy. ACS Applied Materials & Interfaces. 2020; 12: 24662–24674.
[18]
Wu B, Sun Z, Wu J, Ruan J, Zhao P, Liu K, et al. Nanoparticle‐Stabilized Oxygen Microcapsules Prepared by Interfacial Polymerization for Enhanced Oxygen Delivery. Angewandte Chemie (International Ed. in English). 2021; 60: 9284–9289.
[19]
Liu L, Zhang Y, Qiu W, Zhang L, Gao F, Li B, et al. Dual-Stage Light Amplified Photodynamic Therapy against Hypoxic Tumor Based on an O2 Self-Sufficient Nanoplatform. Small. 2017. (in press)
[20]
Prasad P, Gordijo CR, Abbasi AZ, Maeda A, Ip A, Rauth AM, et al. Multifunctional albumin-MnO₂ nanoparticles modulate solid tumor microenvironment by attenuating hypoxia, acidosis, vascular endothelial growth factor and enhance radiation response. ACS Nano. 2014; 8: 3202–3212.
[21]
Liu J, Gao J, Zhang A, Guo Y, Fan S, He Y, et al. Carbon nanocage-based nanozyme as an endogenous H2O2-activated oxygenerator for real-time bimodal imaging and enhanced phototherapy of esophageal cancer. Nanoscale. 2020; 12: 21674–21686.
[22]
Chen Q, Feng L, Liu J, Zhu W, Dong Z, Wu Y, et al. Intelligent Albumin-MnO2 Nanoparticles as pH-/H2 O2 -Responsive Dissociable Nanocarriers to Modulate Tumor Hypoxia for Effective Combination Therapy. Advanced Materials. 2016; 28: 7129–7136.
[23]
Shin S, Jung W, Choi C, Kim S, Son A, Kim H, et al. Fucoidan-Manganese Dioxide Nanoparticles Potentiate Radiation Therapy by Co-Targeting Tumor Hypoxia and Angiogenesis. Marine Drugs. 2018; 16: 510.
[24]
Zhang R, Song X, Liang C, Yi X, Song G, Chao Y, et al. Catalase-loaded cisplatin-prodrug-constructed liposomes to overcome tumor hypoxia for enhanced chemo-radiotherapy of cancer. Biomaterials. 2017; 138: 13–21.
[25]
Phua SZF, Yang G, Lim WQ, Verma A, Chen H, Thanabalu T, et al. Catalase-Integrated Hyaluronic Acid as Nanocarriers for Enhanced Photodynamic Therapy in Solid Tumor. ACS Nano. 2019; 13: 4742–4751.
[26]
Liu C, Wang D, Zhang S, Cheng Y, Yang F, Xing Y, et al. Biodegradable Biomimic Copper/Manganese Silicate Nanospheres for Chemodynamic/Photodynamic Synergistic Therapy with Simultaneous Glutathione Depletion and Hypoxia Relief. ACS Nano. 2019; 13: 4267–4277.
[27]
Liang J, Zheng Y, Wu X, Tan C, Ji L, Mao Z. A Tailored Multifunctional Anticancer Nanodelivery System for Ruthenium‐Based Photosensitizers: Tumor Microenvironment Adaption and Remodeling. Advanced Science. 2020; 7: 1901992.
[28]
Zheng D, Li B, Li C, Fan J, Lei Q, Li C, et al. Carbon-Dot-Decorated Carbon Nitride Nanoparticles for Enhanced Photodynamic Therapy against Hypoxic Tumor via Water Splitting. ACS Nano. 2016; 10: 8715–8722.
[29]
Wang SB, Zhang C, Liu XH, Chen ZX, Peng SY, Zhong ZL, et al. A Tungsten Nitride‐Based O2 Self‐Sufficient Nanoplatform for Enhanced Photodynamic Therapy against Hypoxic Tumors. Advanced Therapeutics. 2019; 2: 1900012.
[30]
Jin Z, Wen Y, Hu Y, Chen W, Zheng X, Guo W, et al. MRI-guided and ultrasound-triggered release of no by advanced nanomedicine. Nanoscale. 2017; 9: 3637–3645.
[31]
Deng Y, Jia F, Chen S, Shen Z, Jin Q, Fu G, et al. Nitric oxide as an all-rounder for enhanced photodynamic therapy: Hypoxia relief, glutathione depletion and reactive nitrogen species generation. Biomaterials. 2018; 187: 55–65.
[32]
Feng L, Tao D, Dong Z, Chen Q, Chao Y, Liu Z, et al. Near-infrared light activation of quenched liposomal Ce6 for synergistic cancer phototherapy with effective skin protection. Biomaterials. 2017; 127: 13–24.
[33]
Yu W, Liu T, Zhang M, Wang Z, Ye J, Li CX, et al. O(2) Economizer for Inhibiting Cell Respiration To Combat the Hypoxia Obstacle in Tumor Treatments. ACS Nano. 2019; 13: 1784–1794.
[34]
Yuan P, Deng FA, Liu YB, Zheng RR, Rao XN, Qiu XZ, et al. Mitochondria Targeted O2 Economizer to Alleviate Tumor Hypoxia for Enhanced Photodynamic Therapy. Advanced Healthcare Materials. 2021; 10: e2100198.
[35]
Li M, Shao Y, Kim JH, Pu Z, Zhao X, Huang H, et al. Unimolecular Photodynamic O2-Economizer to Overcome Hypoxia Resistance in Phototherapeutics. Journal of the American Chemical Society. 2020; 142: 5380–5388.
[36]
Ye L, Zhang Q, Bai X, Pankaj P, Hu Q, Liang T. Hypoxia-inducible factor 1α expression and its clinical significance in pancreatic cancer: a meta-analysis. Pancreatology. 2014; 14: 391–397.
[37]
Shah VM, Sheppard BC, Sears RC, Alani AW. Hypoxia: Friend or Foe for drug delivery in Pancreatic Cancer. Cancer Letters. 2020; 492: 63–70.
[38]
Zhao X, Gao S, Ren H, Sun W, Zhang H, Sun J, et al. Hypoxia-Inducible Factor-1 Promotes Pancreatic Ductal Adenocarcinoma Invasion and Metastasis by Activating Transcription of the Actin-Bundling Protein Fascin. Cancer Research. 2014; 74: 2455–2464.
[39]
Saxena K, Jolly MK, Balamurugan K. Hypoxia, partial EMT and collective migration: Emerging culprits in metastasis. Translational Oncology. 2020; 13: 100845.
[40]
Kao S, Wu K, Lee W. Hypoxia, Epithelial-Mesenchymal Transition, and TET-Mediated Epigenetic Changes. Journal of Clinical Medicine. 2016; 5: 24.
[41]
Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, et al. Distinct Populations of Cancer Stem Cells Determine Tumor Growth and Metastatic Activity in Human Pancreatic Cancer. Cell Stem Cell. 2007; 1: 313–323.
[42]
Hashimoto O, Shimizu K, Semba S, Chiba S, Ku Y, Yokozaki H, et al. Hypoxia Induces Tumor Aggressiveness and the Expansion of CD133-Positive Cells in a Hypoxia-Inducible Factor-1α-Dependent Manner in Pancreatic Cancer Cells. Pathobiology. 2011; 78: 181–192.
[43]
Ropolo A, Catrinacio C, Renna FJ, Boggio V, Orquera T, Gonzalez CD, et al. A Novel E2F1-EP300-VMP1 Pathway Mediates Gemcitabine-Induced Autophagy in Pancreatic Cancer Cells Carrying Oncogenic KRAS. Frontiers in Endocrinology. 2020; 11: 411.
[44]
Sarcar B, Li X, Fleming JB. Hypoxia-Induced Autophagy Degrades Stromal Lumican into Tumor Microenvironment of Pancreatic Ductal Adenocarcinoma: a Mini-Review. Journal of Cancer Treatment & Diagnosis. 2019; 3: 22–27.
[45]
New M, Tooze S. The Role of Autophagy in Pancreatic Cancer-Recent Advances. Biology. 2019; 9: 7.
[46]
Su H, Yang F, Fu R, Li X, French R, Mose E, et al. Cancer cells escape autophagy inhibition via NRF2-induced macropinocytosis. Cancer Cell. 2021; 39: 678–693.e11.
[47]
Garcea G, Doucas H, Steward WP, Dennison AR, Berry DP. Hypoxia and angiogenesis in pancreatic cancer. ANZ Journal of Surgery. 2006; 76: 830–842.
[48]
Tung K, Lin C, Kuo C, Li L, Kuo Y, Lin C, et al. CHC promotes tumor growth and angiogenesis through regulation of HIF-1α and VEGF signaling. Cancer Letters. 2013; 331: 58–67.
[49]
Thomas D, Radhakrishnan P. Tumor-stromal crosstalk in pancreatic cancer and tissue fibrosis. Molecular Cancer. 2019; 18: 14.
[50]
McCormick KA, Coveler AL, Rossi GR, Vahanian NN, Link C, Chiorean EG. Pancreatic cancer: Update on immunotherapies and algenpantucel-L. Human Vaccines & Immunotherapeutics. 2016; 12: 563–575.
[51]
Ahn DH, Ramanathan RK, Bekaii-Saab T. Emerging Therapies and Future Directions in Targeting the Tumor Stroma and Immune System in the Treatment of Pancreatic Adenocarcinoma. Cancers. 2018; 10: 193.
[52]
Pillarisetty VG. The pancreatic cancer microenvironment: an immunologic battleground. Oncoimmunology. 2014; 3: e950171.
[53]
Ying H, Dey P, Yao W, Kimmelman AC, Draetta GF, Maitra A, et al. Genetics and biology of pancreatic ductal adenocarcinoma. Genes & Development. 2016; 30: 355–385.
[54]
Ruan C, Su K, Zhao D, Lu A, Zhong C. Nanomaterials for Tumor Hypoxia Relief to Improve the Efficacy of ROS-Generated Cancer Therapy. Frontiers in Chemistry. 2021; 9: 649158.
[55]
Hess JR, MacDonald VW, Brinkley WW. Systemic and pulmonary hypertension after resuscitation with cell-free hemoglobin. Journal of Applied Physiology. 1993; 74: 1769–1778.
[56]
Rioux F, Drapeau G, Marceau F. Recombinant human hemoglobin (rHb1.1) selectively inhibits vasorelaxation elicited by nitric oxide donors in rabbit isolated aortic rings. Journal of Cardiovascular Pharmacology. 1995; 25: 587–594.
[57]
Sakai H, Sou K, Horinouchi H, Kobayashi K, Tsuchida E. Hemoglobin-Vesicle, a Cellular Artificial Oxygen Carrier that Fulfils the Physiological Roles of the Red Blood Cell Structure. Advances in Experimental Medicine and Biology. 2010; 263: 433–438.
[58]
Tsuchida E, Sakai H, Horinouchi H, Kobayashi K. Hemoglobin-vesicles as a transfusion alternative. Artificial Cells, Blood Substitutes, and Immobilization Biotechnology. 2006; 34: 581–588.
[59]
Kaneda S, Ishizuka T, Goto H, Kimura T, Inaba K, Kasukawa H. Liposome-Encapsulated Hemoglobin, TRM-645: Current Status of the Development and Important Issues for Clinical Application. Artificial Organs. 2009; 33: 146–152.
[60]
Pimenova T, Pereira CP, Schaer DJ, Zenobi R. Characterization of high molecular weight multimeric states of human haptoglobin and hemoglobin-based oxygen carriers by high-mass MALDI MS. Journal of Separation Science. 2009; 32: 1224–1230.
[61]
Buehler PW, Alayash AI. All hemoglobin-based oxygen carriers are not created equally. Biochimica Et Biophysica Acta (BBA) - Proteins and Proteomics. 2008; 1784: 1378–1381.
[62]
Paciello A, Amalfitano G, Garziano A, Urciuolo F, Netti PA. Hemoglobin-Conjugated Gelatin Microsphere as a Smart Oxygen Releasing Biomaterial. Advanced Healthcare Materials. 2016; 5: 2655–2666.
[63]
Cao H, Wang L, Yang Y, Li J, Qi Y, Li Y, et al. An Assembled Nanocomplex for Improving both Therapeutic Efficiency and Treatment Depth in Photodynamic Therapy. Angewandte Chemie (International Ed. in English). 2018; 57: 7759–7763.
[64]
Jansman MMT, Hosta-Rigau L. Recent and prominent examples of nano- and microarchitectures as hemoglobin-based oxygen carriers. Advances in Colloid and Interface Science. 2018; 260: 65–84.
[65]
Yu C, Huang X, Qian D, Han F, Xu L, Tang Y, et al. Fabrication and evaluation of hemoglobin-based polydopamine microcapsules as oxygen carriers. Chemical Communications. 2018; 54: 4136–4139.
[66]
Hu J, Wang Q, Wang Y, You G, Li P, Zhao L, et al. Polydopamine-based surface modification of hemoglobin particles for stability enhancement of oxygen carriers. Journal of Colloid and Interface Science. 2020; 571: 326–336.
[67]
Jia Y, Duan L, Li J. Hemoglobin-Based Nanoarchitectonic Assemblies as Oxygen Carriers. Advanced Materials. 2016; 28: 1312–1318.
[68]
Gu J, Chang TMS. Extraction of Erythrocyte Enzymes for the Preparation of Polyhemoglobin-catalase-superoxide Dismutase. Artificial Cells, Blood Substitutes, and Biotechnology. 2009; 37: 69–77.
[69]
Freire MG, Dias AMA, Coelho MAZ, Coutinho JAP, Marrucho IM. Aging mechanisms of perfluorocarbon emulsions using image analysis. Journal of Colloid and Interface Science. 2005; 286: 224–232.
[70]
Glass SB, Gonzalez-Fajardo L, Beringhs AO, Lu X. Redox Potential and ROS-Mediated Nanomedicines for Improving Cancer Therapy. Antioxidants & Redox Signaling. 2019; 30: 747–761.
[71]
Moulder JE, Rockwell S. Tumor hypoxia: its impact on cancer therapy. Cancer Metastasis Reviews. 1987; 5: 313–341.
[72]
Song G, Ji C, Liang C, Song X, Yi X, Dong Z, et al. TaOx decorated perfluorocarbon nanodroplets as oxygen reservoirs to overcome tumor hypoxia and enhance cancer radiotherapy. Biomaterials. 2017; 112: 257–263.
[73]
Xiang Y, Bernards N, Hoang B, Zheng J, Matsuura N. Perfluorocarbon nanodroplets can reoxygenate hypoxic tumors in vivo without carbogen breathing. Nanotheranostics. 2019; 3: 135–144.
[74]
Tsuchida E, Sou K, Nakagawa A, Sakai H, Komatsu T, Kobayashi K. Artificial Oxygen Carriers, Hemoglobin Vesicles and Albumin-Hemes, Based on Bioconjugate Chemistry. Bioconjugate Chemistry. 2009; 20: 1419–1440.
[75]
Zhou Z, Song J, Nie L, Chen X. Reactive oxygen species generating systems meeting challenges of photodynamic cancer therapy. Chemical Society Reviews. 2016; 45: 6597–6626.
[76]
Modery-Pawlowski CL, Tian LL, Pan V, Sen Gupta A. Synthetic Approaches to RBC Mimicry and Oxygen Carrier Systems. Biomacromolecules. 2013; 14: 939–948.
[77]
Yang Q, Xu Q, Jiang H. Metal-organic frameworks meet metal nanoparticles: synergistic effect for enhanced catalysis. Chemical Society Reviews. 2017; 46: 4774–4808.
[78]
Kaur R, Vellingiri K, Kim K, Paul AK, Deep A. Efficient photocatalytic degradation of rhodamine 6G with a quantum dot-metal organic framework nanocomposite. Chemosphere. 2016; 154: 620–627.
[79]
DeCoste JB, Weston MH, Fuller PE, Tovar TM, Peterson GW, LeVan MD, et al. Metal-Organic Frameworks for Oxygen Storage. Angewandte Chemie (International Ed. in English). 2014; 53: 14092–14095.
[80]
Moghadam PZ, Islamoglu T, Goswami S, Exley J, Fantham M, Kaminski CF, et al. Computer-aided discovery of a metal-organic framework with superior oxygen uptake. Nature Communications. 2018; 9: 1378.
[81]
Dai X, Ruan J, Guo Y, Sun Z, Liu J, Bao X, et al. Enhanced radiotherapy efficacy and induced anti-tumor immunity in HCC by improving hypoxia microenvironment using oxygen microcapsules. Chemical Engineering Journal. 2021; 422: 130109.
[82]
Hu L, Wang P, Zhao M, Liu L, Zhou L, Li B, et al. Near-infrared rechargeable “optical battery” implant for irradiation-free photodynamic therapy. Biomaterials. 2018; 163: 154–162.
[83]
Zou Y, Zhang W, Chen N, Chen S, Xu W, Cai R, et al. Generating Oxygen Vacancies in MnO Hexagonal Sheets for Ultralong Life Lithium Storage with High Capacity. ACS Nano. 2019; 13: 2062–2071.
[84]
Kim J, Heo JN, Do JY, Chava RK, Kang M. Electrochemical Synergies of Heterostructured Fe2O3-MnO Catalyst for Oxygen Evolution Reaction in Alkaline Water Splitting. Nanomaterial. 2019; 9: 1486.
[85]
Wang X, Liu T, Yu J. A limiting Current Oxygen Sensor Constituted of(CeO2)0.95(Y2O3)0.05 as Solid Electrolyte Layer and(CeO2)0.75(ZrO2)0.25 as Dense Diffusion Barrier Layer. Sensors. 2019; 19: 3511.
[86]
Tian D, Li K, Wei Y, Zhu X, Zeng C, Cheng X, et al. DFT insights into oxygen vacancy formation and CH4 activation over CeO2 surfaces modified by transition metals (Fe, Co and Ni). Physical Chemistry Chemical Physics. 2018; 20: 11912–11929.
[87]
Mock SA, Sharp SE, Stoner TR, Radetic MJ, Zell ET, Wang R. CeO2 nanorods-supported transition metal catalysts for CO oxidation. Journal of Colloid and Interface Science. 2016; 466: 261–267.
[88]
Hu Y, Wang X, Zhao P, Wang H, Gu W, Ye L. Nanozyme-catalyzed oxygen release from calcium peroxide nanoparticles for accelerated hypoxia relief and image-guided super-efficient photodynamic therapy. Biomaterials Science. 2020; 8: 2931–2938.
[89]
Zhao L, Fu C, Tan L, Li T, Zhong H, Meng X. Advanced nanotechnology for hypoxia-associated antitumor therapy. Nanoscale. 2020; 12: 2855–2874.
[90]
He Z, Xiao Y, Zhang J, Zhang P, Zhu J. In situ formation of large pore silica-MnO2 nanocomposites with H+/H2O2 sensitivity for O2-elevated photodynamic therapy and potential MR imaging. Chemical Communications. 2018; 54: 2962–2965.
[91]
Zhu P, Chen Y, Shi J. Nanoenzyme-Augmented Cancer Sonodynamic Therapy by Catalytic Tumor Oxygenation. ACS Nano. 2018; 12: 3780–3795.
[92]
Rich LJ, Damasco JA, Bulmahn JC, Kutscher HL, Prasad PN, Seshadri M. Photoacoustic and Magnetic Resonance Imaging of Hybrid Manganese Dioxide-Coated Ultra-small NaGdF(4) Nanoparticles for Spatiotemporal Modulation of Hypoxia in Head and Neck Cancer. Cancers. 2020; 12: 3294.
[93]
Abbasi AZ, Gordijo CR, Amini MA, Maeda A, Rauth AM, DaCosta RS, et al. Hybrid Manganese Dioxide Nanoparticles Potentiate Radiation Therapy by Modulating Tumor Hypoxia. Cancer Research. 2016; 76: 6643–6656.
[94]
Yang C, Liu Y, Su S, Gao N, Jing J, Zhang X. A multifunctional oxygen-producing MnO2-based nanoplatform for tumor microenvironment-activated imaging and combination therapy in vitro. Journal of Materials Chemistry B. 2020; 8: 9943–9950.
[95]
Tseng S-, Kempson IM, Huang K, Li H, Fa Y, Ho Y, et al. Targeting Tumor Microenvironment by Bioreduction-Activated Nanoparticles for Light-Triggered Virotherapy. ACS Nano. 2018; 12: 9894–9902.
[96]
Li X, Kwon N, Guo T, Liu Z, Yoon J. Innovative Strategies for Hypoxic-Tumor Photodynamic Therapy. Angewandte Chemie (International Ed. in English). 2018; 57: 11522–11531.
[97]
López-Lázaro M. Dual role of hydrogen peroxide in cancer: possible relevance to cancer chemoprevention and therapy. Cancer Letters. 2007; 252: 1–8.
[98]
Lippert AR, Van de Bittner GC, Chang CJ. Boronate oxidation as a bioorthogonal reaction approach for studying the chemistry of hydrogen peroxide in living systems. Accounts of Chemical Research. 2011; 44: 793–804.
[99]
Chen H, Tian J, He W, Guo Z. H2O2-activatable and O2-evolving nanoparticles for highly efficient and selective photodynamic therapy against hypoxic tumor cells. Journal of the American Chemical Society. 2015; 137: 1539–1547.
[100]
Li S, Cheng H, Xie B, Qiu W, Zeng J, Li C, et al. Cancer Cell Membrane Camouflaged Cascade Bioreactor for Cancer Targeted Starvation and Photodynamic Therapy. ACS Nano. 2017; 11: 7006–7018.
[101]
Liu J, Liu T, Du P, Zhang L, Lei J. Metal-Organic Framework (MOF) Hybrid as a Tandem Catalyst for Enhanced Therapy against Hypoxic Tumor Cells. Angewandte Chemie (International Ed. in English). 2019; 58: 7808–7812.
[102]
Jia Q, Ge J, Liu W, Zheng X, Chen S, Wen Y, et al. A Magnetofluorescent Carbon Dot Assembly as an Acidic H2 O2 -Driven Oxygenerator to Regulate Tumor Hypoxia for Simultaneous Bimodal Imaging and Enhanced Photodynamic Therapy. Advanced Materials. 2018; 30: e1706090.
[103]
Zhang L, Wan S, Li C, Xu L, Cheng H, Zhang X. An Adenosine Triphosphate-Responsive Autocatalytic Fenton Nanoparticle for Tumor Ablation with Self-Supplied H2O2 and Acceleration of Fe(III)/Fe(II) Conversion. Nano Letters. 2018; 18: 7609–7618.
[104]
Chen Q, Chen J, Yang Z, Xu J, Xu L, Liang C, et al. Nanoparticle-Enhanced Radiotherapy to Trigger Robust Cancer Immunotherapy. Advanced Materials. 2019; 31: e1802228.
[105]
Walter MG, Warren EL, McKone JR, Boettcher SW, Mi Q, Santori EA, et al. Solar water splitting cells. Chemical Reviews. 2010; 110: 6446–6473.
[106]
Zhao C, Chen Z, Shi R, Yang X, Zhang T. Recent Advances in Conjugated Polymers for Visible‐Light‐Driven Water Splitting. Advanced Materials. 2020; 32: 1907296.
[107]
Perović K, Dela Rosa FM, Kovačić M, Kušić H, Lavrenčič Štangar U, Fresno F, et al. Recent Achievements in Development of TiO(2)-Based Composite Photocatalytic Materials for Solar Driven Water Purification and Water Splitting. Materials. 2020; 13: 1338.
[108]
Shih CY, Wang PT, Su WC, Teng H, Huang WL. Nanomedicine-Based Strategies Assisting Photodynamic Therapy for Hypoxic Tumors: State-of-the-Art Approaches and Emerging Trends. Biomedicines. 2021; 9: 137.
[109]
Song CW, Rhee JG, Levitt SH. Effect of hyperthermia on hypoxic cell fraction in tumor. International Journal of Radiation Oncology, Biology, Physics. 1982; 8: 851–856.
[110]
Brizel DM, Scully SP, Harrelson JM, Layfield LJ, Dodge RK, Charles HC, et al. Radiation therapy and hyperthermia improve the oxygenation of human soft tissue sarcomas. Cancer Research. 1996; 56: 5347–5350.
[111]
Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg Effect: the Metabolic Requirements of Cell Proliferation. Science. 2009; 324: 1029–1033.
[112]
NELSON RA. The immune-adherence phenomenon; an immunologically specific reaction between microorganisms and erythrocytes leading to enhanced phagocytosis. Science. 1953; 118: 733–737.
[113]
Glassman PM, Hood ED, Ferguson LT, Zhao Z, Siegel DL, Mitragotri S, et al. Red blood cells: the metamorphosis of a neglected carrier into the natural mothership for artificial nanocarriers. Advanced Drug Delivery Reviews. 2021; 178: 113992.
[114]
Gao D, Gao Y, Shen J, Wang Q. Modified nanoscale metal organic framework-based nanoplatforms in photodynamic therapy and further applications. Photodiagnosis and Photodynamic Therapy. 2020; 32: 102026.
[115]
Yang G, Ji J, Liu Z. Multifunctional MnO2 nanoparticles for tumor microenvironment modulation and cancer therapy. Wiley Interdisciplinary Reviews - Nanobiotechnology. 2021; 13: e1720.
[116]
Ganapathy-Kanniappan S, Geschwind JF. Tumor glycolysis as a target for cancer therapy: progress and prospects. Molecular Cancer. 2013; 12: 152.
Share
Back to top