Skip to main content

Nanomaterial-related hemoglobin-based oxygen carriers, with emphasis on liposome and nano-capsules, for biomedical applications: current status and future perspectives

Abstract

Oxygen is necessary for life and plays a key pivotal in maintaining normal physiological functions and treat of diseases. Hemoglobin-based oxygen carriers (HBOCs) have been studied and developed as a replacement for red blood cells (RBCs) in oxygen transport due to their similar oxygen-carrying capacities. However, applications of HBOCs are hindered by vasoactivity, oxidative toxicity, and a relatively short circulatory half-life. With advancements in nanotechnology, Hb encapsulation, absorption, bioconjugation, entrapment, and attachment to nanomaterials have been used to prepare nanomaterial-related HBOCs to address these challenges and pend their application in several biomedical and therapeutic contexts. This review focuses on the progress of this class of nanomaterial-related HBOCs in the fields of hemorrhagic shock, ischemic stroke, cancer, and wound healing, and speculates on future research directions. The advancements in nanomaterial-related HBOCs are expected to lead significant breakthroughs in blood substitutes, enabling their widespread use in the treatment of clinical diseases.

Graphical Abstract

Introduction

Oxygen (O2) is essential for life [1] and physiological processes such as cell growth, differentiation, metabolism, and tissue homeostasis [2, 3]. Factors such as trauma [4], blood loss [5], pernicious anemia [6] and ischemic cerebrovascular disease [7, 8] can decrease tissue oxygen content, lead to hypoxia [9]. This, in turn, can induce organ dysfunction, particularly in the brain [10], heart [11] and kidney [12] and leading to irreversible damage and/ or death [13]. Besides, the hypoxic microenvironment of tumor tissues can decrease the sensitivity of malignant tumors to antitumor drugs and enhance the invasion and metastasis abilities of tumor cells [14,15,16]. Thus, maintaining an adequate O2 supply to tissues and organs, is essential for maintaining the physiological functions of the body, and it is critical for disease treatment [17].

As natural oxygen carriers, red blood cells (RBCs) can bind to O2 in the lungs and deliver throughout to the body. This process is essential to meet the metabolic demands of the body [18, 19] and ensure normal organ function [20]. Hemoglobin (Hb), the main O2 carrier in RBCs, facilitates O2 delivery via its well-known oxygen-carrying function [21]. In adults, Hb consist of four subunits (α1, β1, α2, and β2) that form a tetrameric protein containing a central ferrous heme, enabling it to transport O2 by binding reversibly to O2 [22, 23].

Numerous factors, such as trauma, blood loss, and pernicious anemia [24, 25], can potentially cause decrease in RBCs and Hb levels in the bloodstream. Timely transfusion of RBCs, restoration of blood volume in the body, and ensuring adequate of O2 supply are essential for saving lives [26,27,28]. Indeed, these measures are widely practiced in clinical and military rescue scenarios [29]. However, the demand for natural blood often outpaces supply [30, 31]. Natural blood has a limited preservation period (< 42 d), requires special storage and transportation conditions [32], and is susceptible to damage and adverse reactions when infused [33, 34]. Natural blood transfusion also carriers risk of infectious diseases, requires associated, time-consuming, cross-matching [35, 36] and may conflict with religious beliefs [37, 38]. Therefore, blood substitutes that can fully and completely or partially replace the oxygen-carrying functions of RBCs have emerged. These substitutes can be classified into perfluorocarbons (PFCs) and hemoglobin-based oxygen carriers (HBOCs) [39, 40].

Perfluorocarbon emulsions, which physically solubilize O2 and CO2, were the first blood substitutes to be tested as oxygen carriers [41]. However, the biosafety and stability concerns of PFCs limit their potential for clinical use. The first-generation products, Fluosol-DA®, was originally approved by the United States of America Food and Drug Administration (FDA) for coronary transluminal angioplasty; however, it was withdrawn from the market due to O2 delivery capacity, poor stability, and complement activation [42]. The second-generation product, Oxygent™, was also discontinued during phase III clinical trials in the USA because of an increased incidence of instroke and heart diseases in patients undergoing coronary artery bypass surgery [42,43,44].

HBOCs are a class of blood substitutes that are based on natural hemoglobin obtained through polymerization, cross-linking, and modification with polymer. HBOCs have gradually become the primary research focus for artificial oxygen carriers because they closely resemble the natural oxygen-carrying/-releasing characteristics of Hb [40, 45]. HBOCs development progressed after decades of laboratory and clinical research. However, only a few HBOCs have received worldwide approved for clinical use. Glutaraldehyde-polymerized bovine hemoglobin (HBOC-201, BioPure company), was approved for the treatment of acute pernicious anemia in South Africa and Russia in 2001 and 2010, respectively [46, 47]. PEGylated carboxyhemoglobin bovine (Sanguinate, Prolong Pharmaceuticals, USA), was registered in 2015 under the FDA classification as an “orphan drug” for the treatment of sickle cell anemia; it is currently in phase III clinical trials [48]. Despite these advancement, HBOCs development is also complicated by issues such as vasoactivity caused by nitric oxide (NO) scavenging [49], nephrotoxicity from dissociated dimers [50], oxidative toxicity of hemoglobin [51], and short circulation time [52], resulting in many HBOCs varieties being discontinued or excluded from further research due to safety concerns. Thus far, researchers have focused on addressing the safety concerns of HBOCs by improving strategies such as the cross-linking technique, and the preparation process of Hb, as well as infusion methods. For instance, preventing the dissociation of hemoglobin tetramer and reducing nephrotoxicity [53, 54] by optimizing the molecular weight of the polymer [55]. Hb oxidation is reduced by co-cross-linking Hb with internal biological enzymes such as superoxide dismutase (SOD) and catalase (CAT) [56,57,58]. While these technical strategies have reduced the safety concerns of HBOCs to extent; they are still in the preclinical research stage.

The role of nanomaterials in drug delivery and disease treatment has received increased research attention due to the rapid development of nanotechnology [59, 60], which provides new opportunities for developing HBOCs. Nanomaterial-related HBOCs (Nano-HBOCs) are prepared by organically conjugating of nanomaterials with hemoglobin through encapsulation, self-assembly, bioconjugation, entrapment, and attachment [61,62,63]. This approach not only closely mimics the physiological structure of natural RBCs but also offers more advantages, such as reduced the vasoactivity, improved circulation time [64,65,66], and enhanced biological safety [67] (Table 1).

Table 1 Several advantages of Nano-HBOCs, classified into seven categories, with information related to their formulations, Hb source, and references

Although some excellent reviews have discussed Nano-HBOCs [35, 45, 65,66,67,68], there is a lack of detailed and systematic introductions, which, coupled with the rapid progress in the research of Nano-HBOCs applications, has resulted in a lack of reviews on the latest advances in the preparation and application of Nano-HBOCs. In this review, we introduce the specific research on Nano-HBOCs in biomedical fields over decade. We focus on the applications of Nano-HBOCs in hemorrhagic shock, ischemic stroke, cancer therapy, and wound healing (Fig. 1). Additionally, we systematically summarize the research progress, current challenges and future prospects.

Fig. 1
figure 1

Schematic representation of the application of Nano-HBOCs in hemorrhagic shock, ischemic stroke, cancer, wound healing, and other disease treatment

Nano-HBOCs for hemorrhagic shock therapy

Hemorrhagic shock (HS) is a complex condition caused by a reduction in the effective circulating blood volume [87, 88], resulting in irreversible tissue hypoxia [89], eventually triggers circulatory failure [90]. Increasing the oxygen-carrying capacity to restore O2 supply to the tissue is a necessary approach to treating HS [88, 91] and perverting circulatory failure.

Restoring oxygen supply

Liposome-encapsulated human hemoglobin (LEH) is a type of HBOCs prepared by encapsulating intact Hb molecules in liposomes, also known to as hemoglobin vesicles (HbV) [65]. LEH has been extensively researched as a potential treatment for HS in animal models [92,93,94,95]. Studies have shown that LEH has excellent biocompatibility [96], high biological safety [97,98,99], easy metabolism and excretion [100, 101], and low vasoactivity [102]. Researchers at the Terumo Research and Development Center [69] (Terumo Co., Tokyo, Japan) used a high-speed emulsification method to encapsulate purified human Hb in liposomes that mimic RBCs membranes. They modified the lipid membrane surface with PEG to form a nanoscale liposomal oxygen carrier (namely TRM-645). In a mouse hypohemoglobinemic shock model, the intravenous infusion of TRM-645 rapidly restored mean arterial pressure (MPA) from 30 to 70 mmHg; its effect on restoring and maintaining MAP was comparable to that of native RBCs. The intracavitary infusion of TRM-645 significantly improved overall survival [103] in a murine of HS induced by sustained massive hemorrhage, with a better treatment effect RBCs. TRM-645, due to its smaller size, is more likely to enter the circulatory system from the bone marrow cavity, rendering it an effective method for critical scenarios and pre-hospital emergency care. Vivek et al. [104] utilized non-phospholipid anionic lipid conjugated PEG containing hexadecyl carbamoyl methyl hexadecanoate (HDAS) to modify the surface of LEH. This modification improved the host’s tolerance to the immune response against LEH [105, 106], delayed its phagocytic clearance by monocytes, and increased the mean residence time in circulation. In a rat model of HS, LEH infusion restored blood volume, improved tissue oxygenation capacity, and ameliorated HS-induced systemic inflammation and multiple organ failure, suggesting that this modified LEH is a potential treatment for HS.

To address the critical need for a transportable, and temperature-stable blood substitute, a bio-synthetic, nano-artificial erythrocyte called Erythromer (EM) [107] has been developed. EM encapsulates human Hb within deformable, hybrid peptidic-lipid nanoparticles. This design, allows for a true physiological correlation between O2 affinity and tissue respiration. EM employs a novel shuttle, a small molecule allosteric effector, which adjusts P50 in response to pH changes, influencing Hb ~ O2 affinity. The use of EM allows for the mimicry of key physiological properties of natural RBCs, offering a promising solution to address the aforementioned critical need. Compared to the use of hydroxyethyl starch for resuscitation, EM maintains hemodynamics stability, increases arterial oxygen partial pressure, and improves acidosis in the HS rat and hemodilution mouse models [80]. Okamoto et al. [108] demonstrated that the combination of bovine hemoglobin (bHb) and human serum albumin (HSA) forms a core–shell protein called HbX-HSA3, which has high oxygen affinity. HbX-HSA3 facilitates O2 delivery to hypoxic tissues during HS. In the rat HS model, HbX-HSA3 effectively restores and maintains basic life indicators such as blood pressure, blood oxygen, respiration, and body temperature. It also reduces body lactate levels; and improve the survival rate of HS. Lu et al. [70] used methoxy polyethylene glycol-poly (D, L-lactide-co-glycolide) and mPEG-PLGA to encapsulate bHb using the double emulsion (w/o/w) technique for preparing hemoglobin-loaded nanoparticles (HbPs). This was based on the good biodegradability and biocompatibility characteristics of PLGA, which had a uniform particle size with stable oxygen-carrying function and excellent blood compatibility. In a controlled hemorrhage mouse model, HbPs maintained MAP, improved venous oxygen partial pressure, and restored tissue oxygen supply, demonstrating a promising application potential.

Polydopamine (PDA) has excellent biocompatibility and antioxidant capacity [109]. It can easily and efficiently adhere to the surface of various substrate materials [110], suggesting that PDA has the potential to serve as a universal modification platform for Hb. This can help address the issue of oxidative toxicity in HBOCs. Wang et al. [77] harnessed the unique characteristics of PDA to modify bHb and prepare Hb-PDA, which does not cause platelet aggregation or significant hemolysis. These novel Nano-HBOCs demonstrated outstanding oxygen affinity (P50 = 13.86 mmHg). Hu et al. [78] combined a template-based co-precipitation technique to develop Nano-oxygen carriers, (named Hb-PDA), which displayed uniform size and high biocompatibility. In vitro and in vivo experiments showed that Hb-PDA enhanced the antioxidant properties of Hb, alleviated the toxicity of free Hb, and maintained the O2 delivery capacity. Further, the resuscitation efficacy of Hb-PDA was investigated using in rat HS model. The results revealed that Hb-PDA maintained the stability of the blood pressure for a longer time relative to using saline alone. Baidukova et al. [111] modified bHb with PDA to synthesize morphologically homogeneous PD-HbMPs. These novel HBOCs showed antioxidant activity and enhanced Hb oxygen-binding capacity. Further, PD-HbMPs exhibited a high ability to scavenge free radical including H2O2, indicating their potential as practical HBOCs. Ethan et al. [73] proposed a photocatalytic method to synthesize the oxygen therapy agent PDA-LtEc using cell-free hemoglobin (Ec) from the PDA surface-coated worm Lumbricus terrestris (Lt). This method increased the oxidative protection of Ec during circulation, thereby making PDA-LtEc a promising oxygen therapy.

Recently, metal–organic frameworks (MOFs; ZIF-8) have been widely used as protective coatings for functional biomacromolecules [112, 113] (such as DNA, proteins, and enzymes). This is because of their flexible topology and favorable physicochemical properties that improve stability during storage and manipulation [114,115,116,117,118,119]. Peng et al. [74] used MOFs to encapsulate bHb and created an oxygen-carrier platform with nanoporous coatings, ZIF-8@Hb. In vitro studies showed that the neutral charge state of the surface of ZIF-8@Hb surface significantly enhanced stability in alkaline, oxidative, elevated temperature, or enzymatic environments, which is favorable for long-term blood circulation (as the study found that the blood circulation half-life (t1/2) of ZIF-8@Hb was 13.9 h, compared to 1.4 and 5.1 h for ZIF-8 and Hb, respectively). Animal studies showed that intravenous injection of ZIF-8@Hb significantly prolonged the survival time of mice in an HS model. This study provides a highly stable and long-circulating oxygen-carrier platform for Nano-HBOCs (Fig. 2).

Fig. 2
figure 2

Copyright 2019, American Chemical Society

A Nanoparticle preparation, stability behavior, blood circulation, and oxygen supplying of ZIF-8@Hb are shown in figure. B Survival curves of mice with hemorrhagic shock following intravenous injection of PBS, Hb, ZIF-8, and ZIF-8@Hb. C Oxygen dissociation curves of Hb and ZIF-8@Hb. D Biodistribution of Hb, ZIF-8, and ZIF-8@Hb after intravenous injection for 12 h. E Biodistribution of Hb, ZIF-8, and ZIF-8@Hb after intravenous injection for 24 h. A–E Reproduced with permission [76].

HS is an important application scenario for Nano-HBOCs. It is worth noting that HS treatment often requires large doses, and multiple intravenous infusion of Nano-HBOCs. Therefore, the safety requirements for Nano-HBOCs are extremely high. However, most current HS studies tend to focus only on the therapeutic effect and do not adequately evaluating biocompatibility, especially in the case of multiple and massive administrations. In this regard, a research group, from Denmark, developed the concept of “stealth properties” [120] of Nano-HBOCs, which may provide new ideas for the treatment of HS usig Nano-HOBCs.

Nano-HBOCs for ischemic stroke therapy

Ischemic stroke is a clinical syndrome that refers to the disruption of blood supply in the brain caused by different cerebrovascular diseases, which eventually lead to local brain tissue ischemia, hypoxic necrosis, and corresponding neurological deficits [121]. Ischemic strokes are typically associated with varying degrees of cerebral infarcts and, in recent years, have become one of the leading causes of human mortality [122, 123] due to their increasing incidence and lethality. Ischemia and hypoxia in brain tissue cause impaired oxygen metabolism, which rapidly triggers brain tissue edema, focal neuronal cell function defects, and even necrosis [124, 125]. Following ischemia stroke, the blood flow in the tissue is typically restored within a short period, however, an excess of reactive oxygen species (ROS) and secondary inflammatory responses are generated in the body during ischemia stroke [126, 127]. These molecules cause tissue organ reperfusion injury [128], further exacerbating the ischemic stroke condition. Currently, several studies have indicated that early improvement in the hypoxic state of the infarct site can benefit ischemia stroke treatment [129, 130].

Relieving cerebral infarction

HBOCs possess adjustable oxygen-carrying/-releasing capabilities and have a small particle size. These characteristics, which can enhance microvascular perfusion and collateral blood flow, lead to continuous improvement in oxygen supply to the ischemic area. As a result, they can effectively reduce cerebral infarct size [131, 132], providing a new choice for acute ischemic stroke treatment. Besides, Nano-HBOCs can inhibit glycolysis and lactic acid accumulation, which offers obvious advantages in reducing ischemic stroke injury and a significant protective effect on ischemic stroke tissue organs [133,134,135,136]. Komatsu et al. [137] established a rat middle cerebral artery occlusion (MCAO) model and an arachidonic acid (AA)-induced stroke models to evaluate the therapeutic effect of HbV. HbV treatment immediately after MCAO significantly reduced the infarct size (34.7%) in rats; the effect, was comparable to tthat of intravenous thrombolysis with a tissue plasminogen activator (tPA) in a thromboembolic model (34% reduction). The intravenous infusion of HbV reduced brain edema in an AA-induced thromboembolic stroke model. Kawaguchi [138, 139] et al. found that encapsulating Hb in liposomes prevented extravasation in rat stroke models; LEH significantly improved oxygen supply to the ischemic area and reduced cortical infarct size. Daiki Tomita [140] et al. covalently combined purified bHb and HSA to design a completely new oxygen-carrier platform with a core–shell structure to minimize the vascular leakage of HBOCs and increase circulation time in vivo (possibly through the effect of net surface charge). This platform, known as HbX-HSAm, exhibits a high O2 affinity (P50 = 11.3 mmHg). Gekka [141] et al. covalently conjugated one Hb molecule with three human serum albumin (Hb1-HSA3) form HemoAct with a core–shell structure. In rat model of a transient MCAO (tMCAO), a significant increase in microvascular perfusion in the cortical penumbra and tissue oxygen partial pressure in the cortical penumbra were observed. Additionally, the treatment reduced liposome peroxidation, edema, and cerebral infarct size; while exerting potent neuroprotective effects in the treatment of transient ischemic injury encephalopathy via intravenous infusion. These studies suggest that improvement in the oxygen supply at the infarct site using infused Nano-HBOCs infusion effectively attenuates stroke injury.

Nano-HBOCs can be used in multifunctional composites in combination with other functional materials boost their therapeutic efficacy. Hosaka [142] et al. synthesized covalent core–shell structural protein clusters called Hb-HSA based on purified bHb and HSA. Next, platinum nanoparticles with SOD and CAT-mimetic enzyme activities were combined with HSA on the outer layer of protein clusters to form Hb-HSA3 (PtNP) with antioxidant capacity. Based on the pathological microenvironment characteristic of stroke, Liu [81] et al. introduced PDA to noncovalently engage Hb and methoxatin (M) to form PMH, whose surface-coated ROS-sensitive linker (thioketal, termed TK linker) connects to a specific fibrin-binding peptide (CREKA, termed C-peptide), and they developed a bioinspired nanoerythrocyte (C-peptide-PRG-TK linker @PDA-Methoxatin Hb, referred to as CPTK@PMH). The half-life of free fluorescence labeled Hb was measured use a microplate reader, and the pharmacokinetic results showed that CPTK@PMH could significantly prolong Hb half-life from 12.045 ± 1.251 h to about 35 h. In a murine MCAO model, CPTK@PMH was shown to target the thrombus and ischemic site, release O2 in response to hypoxic signals, regulate the metabolic microenvironment, inhibit the oxidative stress injury, and reduce the cerebral infarct size during reperfusion, which ultimately protecting neurons from acute injury and improving neurological function after cerebral ischemia (Fig. 3).

Fig. 3
figure 3

Copyright 2019, Nano Today.

A Illustration of the CPTK@PMH nanoerythrocyte formation and metabolic microenvironment modulation in ischemic brain: nanoerythrocyte accumulation in ischemic core via microthrombus binding and uptake by neurovascular unit after blood brain barrier penetration; B hypoxia-responsive oxygen release to relieve necroptosis; C oxygen balance regulation to alleviate acute reperfusion injury via oxygen enrichment, ROS scavenging and microglia polarization; D repair promotion achieved by metabolic microenvironment modulation via energy and glucose metabolism activation and blood brain barrier protection. A–D Reproduced with permission [83].

Inhibiting an inflammatory reaction

An inflammatory response is a key factor in the development and outcome of an ischemic stroke [143]. A series of inflammatory cascades are triggered after an ischemic stroke. Among these, microglial activation in brain tissue and neutrophil infiltration from peripheral blood into brain tissue are the hallmarks underlying microglial transformation from the M1 to M2 type. In particular, these processes inhibit neutrophil infiltration, which is a potential direction for treating cerebral ischemic injury [144]. Yin et al. [145] modified MG1 and RVG29 peptide chains based on erythrocyte-derived nanovesicles to prepare engineered nanoerythrocytes (NEMR) to promote microglial reprogramming. In an MCAO model, NEMR targeted the cerebral infarct site to attenuate tissue damage by delivering/-releasing oxygen, while inducing the overexpression of heme oxygenase-1 and CO production thriugh the Hb metabolism. This promotes microglial transformation into the M2 type, ultimately improving therapeutic outcomes in ischemic stroke. Furthermore, the modifications of MG1 and RVG29 peptides effectively prevented NEMR uptake immune cells in the bloodstream (including mononuclear macrophages), and improved the immune escape ability of NEMR. In addition, Shimbo et al. [146, 147] found that LEH inhibited neutrophil infiltration, matrix metalloproteinase-9 (MMP-9) [148] expression, and oxygen-free radical production, reduced cerebral infarct size and edema volume, and improved neurological indices in a rat MCAO model.

Although few studies have reported the applications of Nano-HBOCs in ischemic stroke, current research confirms that Nano-HBOCs can serve as an effective therapeutic option by targeting drug delivery and easily passing through the damaged blood–brain barrier, et al. However, stroke is a complex disease, and the unique advantages of Nano-HBOCs include their ability to relieve the ischemic and hypoxic state of brain tissues, co-load drugs, and ultimately achieve comprehensive treatment by combining thrombolysis, anti-inflammation and neuron protection. This approach also promotes rehabilitation, providing a diversified platform for treating of ischemic stroke.

Nano-HBOCs for cancer therapy

Hypoxia is a common feature of most malignant tumors. Cancer cells proliferate in a disorderly manner and consume large amounts of oxygen [149, 150], which results in a tumor microenvironment (TME) [151,152,153] that is characterized by hypoxia accompanied by weak acids and high levels of H2O2, and it directly contributes to cancer cell metastasis, invasion, and angiogenesis [154, 155]. Hypoxic TME decreases the antitumor efficacy of chemotherapy, radiation therapy (RT), photodynamic therapy (PDT), and sonodynamic therapy (SDT) [156,157,158,159,160]. As oxygen carriers, Nano-HBOCs showed excellent stability and oxygen supply capacity, improved O2 delivery efficiency in hypoxic regions [161], alleviated the hypoxic state of tumors, and enhanced antitumor efficacy.

Increasing drug sensitivity

The increased expression of hypoxia-inducible factor-1α (HIF-1α) in TME facilitates the adaptation of tumor cells to hypoxia [162, 163]. This is attributed to the development of chemical resistance that promotes the expression of p-glycoprotein (P-gp), which leads to chemotherapy failure [164, 165]. Therefore, improving the hypoxic environment of tumors is important to enhance the efficacy of chemotherapy. Kawaguchi et al. [166] reported that liposomes encapsulating hemoglobin (h-LEH) with high O2 affinity improved the oxygenation capacity of tumor cells in a mouse rectal cancer (Colon26) model. h-LEH combined with chemotherapeutic drugs by inhibiting HIF-1α activity, which prevented angiogenesis and inhibited tumor growth and metastasis. Human organic cation transporter 2 (OCT2) abnormalities confer renal cell carcinoma (RCC) increased resistance to chemotherapeutic agents such as oxaliplatin (OXA) and decitabine (DAC) under hypoxic conditions [167, 168]. To tackle this issue, Chen [169] et al. developed a simple and efficient oxygen nanocarrier based on Hb (H-NPs) combined with DAC. The H-NPs increased the oxygen content of the RCC tissue, alleviated hypoxia-induced loss of DAC activity, and enhanced the sensitivity of RCC cells to the combined treatment with DAC and OXA.

Targeted drug releasing

A hypoxic environment causes the overexpression of glucose transporter isoform 1 (GLUT1) [170] on the surface of cancer cells, and nanocarriers modified with glucose can identify cancer cells via GLUT1. Thus, Bu et al. [171] designed Nano-HBOC with both oxygen-carrying capacity and cancer cell-specific recognition. The synthesis of the triblock copolymer poly [2-(methacrylamido) glucopyranose]-b-poly (methacrylic acid) -b-poly (butyl methacrylate) (PMAG-b-PMAA-b-PBMA) micelles indicated that Hb was bound to the micelles via a condensation reaction to form Hb-conjugated micelles (HCM). PMAG-linked GLUT1 specifically recognizes cancer cells, and HCM can bind O2. Meanwhile, the uptake of DOX-loaded HCM by tumor cells was increased in a model of human cervical cancer cells (HeLa), and Nano-HBOCs were confirmed to target cancer cell recognition and reduce tumor drug resistance, ultimately improving therapeutic efficacy. Tian et al. [83] designed and constructed camouflaged biomimetic nanocomposites (DHCNPNPs) based on PLGA encapsulated with Hb and DOX. They modified the surface of the cancer cell membrane to realize homologous recognition. The DHCNPNPs exhibited strong self-recognition ability, affinity for homologous cancer cells in a model of human breast cancer cells (MCF-7 cells), and minimal toxicity to normal tissues. DHCNPNPs achieved O2-independent conditions and high target selectivity in the TME by inhibiting HIF-1α, down-regulating the expression of multidrug resistance gene 1 (MDR1) and P-gp, and increasing chemosensitivity, which improved the overall therapeutic efficacy (Fig. 4). Yang et al. [172] designed a multifunctional liposome, DOX-Hb-lipo (DHL), which was simultaneously loaded with human hemoglobin (hHb) and DOX. They showed that DHL could precisely release DOX and deliver O2 to specific hypoxic sites, thereby alleviating hypoxia-induced chemoresistance. DHL inhibited tumor growth in mouse breast cancer models (4T1) and mouse colon cancer models (CT-26), achieving in situ O2 release and relieving the hypoxic state of the TME, thereby providing an effective alternative for cancer treatment. Wang et al. [84] engineered an endogenous Nano-HBOCs called V(Hb)@DOX, which self-assembled resulting DOX-loaded biomimetic into hollow nanovesicles. In the acidic intertumoral environment, V(Hb)@DOX targeted M2-type macrophages and released DOX, thereby effectively suppressing of tumor growth and metastasis. V(Hb)@DOX prolonged the survival of mice bearing 4T1 orthotopic breast tumors and achieved long-lasting specific immunity in a murine colon cancer CT26 model.

Fig. 4
figure 4

Copyright 2017, Advanced Functional Materials.

The design and function of DOX/Hb loaded PLGA-cancer cell membrane nanoparticles (DHCNPs) for homologous targeting and O2 interference. A Synthesis of oxy-DHCNPs. DHCNPs were prepared by extrusion with preformed DOX/Hb-PLGA NPs, DSPE-PEG, and MCF-7 cancer cell membrane, and then were oxygenated to obtain oxy-DHCNPs. B Cellular functions of DHCNPs, including homologous targeting, downregulation of predictive markers (HIF-1α, MDR1, and P-gp), and inhibited DOX export. (C): MCF-7 tumor growth curves of different treated groups (scale bar 50 µm). (D): Survival rates of tumor-bearing mice in various groups. A–D Reproduced with permission [85].

Promoting ROS generation

ROS are derived from O2 and are formed through oxidative reactions or electron excitation, they mainly consist of hydroxyl radicals (•OH), superoxide anion (•O-2) and singlet oxygen (1O2) [173]. When the levels of ROS exceed a certain threshold, they can disrupt the cellular structure and thus be used to inhibit tumor growth [174]. Nano-HBOCs can promote ROS production in three ways. Firstly, Nano-HBOCs can provides sufficient O2 as a feedstock for ROS production [175], while improving the TME. Furthermore, in the presence of ferrous ions (derived from Hb), high levels of H2O2 in the TME undergo a Fenton and Haber–Weiss reaction, which continuously produce highly toxic •OH [176]. Additionally, Hb can mimic horseradish peroxidase (HRP), which also produce •OH from H2O2 [177].

Application of Nano-HBOCs in RT

RT, a common method for cancer treatment, damages DNA using X-rays and generates ROS to inhibit tumor growth; its therapeutic effect is directly related to the O2 content in the TME [178, 179]. Sang et al. [180] developed an oxygen-enriched X-ray nanoprocessor (Hb@Hf-Ce6 NPs) based on metal-phenolic coordination to modulate the oxygen balance in the TME and revere immunosuppression for cancer eradication and metastasis inhibition. In the nanoprocessor, the radiosensitizer hafnium (Hf) coordinated with chlorin e6 (Ce6) nanocomplexes and encapsulated Hb to deliver O2. Under X-ray irradiation, Hf emitted radioluminescence to activate Ce6 for ROS generation, and Hb@Hf-Ce6 NPs released O2 at the tumor site. This promoted T cell-mediated systemic immunity to exert long-range antitumor effects on tumors and enhance RT efficacy, thereby exhibiting superior antitumor efficacy in a bilateral tumor model and a lung metastatic model of 4T1 breast cancer. (Fig. 5). Xia et al. [85] combined a Hb oxygen carrier with gold nanoparticles (Au) radiosensitizer to prepare Au-Hb NPs. Inspired by the fact that P-selectin on the surface of platelets (PLT) can specifically bind to CD44 on the surface of cancer cells [181], platelet-coated oxygen-carrying nanoparticles (Au-Hb@ PLT NPs) were synthesized by coating Au-Hb NPs with PLT membrane. Au-Hb@PLT NPs could deliver sufficient O2 deep into the tumor tissue, relieve hypoxia, and improve the effect of RT. Au acts as a sensitizer, which enhances the sensitivity of tumor cells to X-rays and their therapeutic effects, showing good tumor-therapeutic efficiency and a few side effects in the HeLa cell model. Of note, a clear boundary between the normal parenchyma and dead cancer cells was confirmed using the hematoxylin and eosin staining (H&E), suggesting that Au-Hb@PLT + RT (2 Gy) killed tumor cells while causing relatively less damage to healthy tissue.

Fig. 5
figure 5

Copyright 2021, Advanced Science.

A Schematic illustration showed the Hb@Hf-Ce6 nanoparticles-mediated X-ray induced radiotherapy-radiodynamic therapy-immunotherapy for the eradication of both primary and distant tumors. Hb was encapsulated in the Hf-phenolic coordination nanoplatform for oxygen delivery through self-assembly. B The oxygen release behavior of hemoglobin (Hb) and Hb@Hf-Ce6 NPs (NP) with/without X-ray irradiation was evaluated by Ru(bpy)3Cl2 probe. Hb and NP were oxygenated previously. C Tumor volume growth curves for primary tumors. D Tumor volume growth curves for distant tumors. A–D Reproduced with permission [184].

Application of Nano-HBOCs in PDT

Photodynamic therapy (PDT), a clinically recognized and minimally invasive tumor treatment, has become an alternative to chemotherapy [182, 183]. PDT includes a photosensitizer (PS), a light source, and sufficient O2. Under PS catalysis, O2 is activated to ROS, such as •OH and 1O2, which in turn induce tumor vascular injury, thereby leading to tumor cell death [184,185,186]. However, the hypoxic state of the TME impairs PDT efficacy [187], which can be effectively reversed by increasing the O2 content of the tumor [188, 189]. Wang et al. [190] developed a new PDT therapy based on the concept of O2 “self-compensation ability,” which was synthesized by chemically conjugating Hb with polymeric micelles formed by triblock copolymers of poly (ethylene glycol)-block-poly (acrylic acid)-block-polystyrene (PEG-b-PAA-b-PS). They used Hb as a PS zinc phthalocyanine (ZnPc) carrier, which endowed the Hb-conjugated ZnPc PS carrier with good oxygen-binding capacity, antioxidant activity, and the ability to produce abundant 1O2, thereby exhibiting robust photocytotoxicity in an in vitro HeLa cell assay.

To address the low O2 levels in tumor and enhance PDT efficacy, Tang et al. [76] used ferritin, a protein-based nanocage, to tether a large amount of PS (such as ZnF16Pc) to the surface of RBCs. They utilized Hb as a PS carrier to provide O2, loaded ZnF16Pc at ferritin molar ratio (1:40) to form P-FRT, and simultaneously produced PFRT-RBCS nanoparticles by coupling RBCs. PFRT-RBCS prolonged the circulation half-life of Ps, which were labelled with IRDye800 that exhibited a high fluorescence in the blood (7.6 ± 2.1%ID g−1 at 1 h), and the signal remained strong for 24 h (3.2 ± 0.4%ID g−1 at 24 h). P-FRT-RBCs achieved continuous oxygen supply under hypoxia in the TME, thereby enhancing the ability of PDT to kill cancer cells in a U87MG tumor-bearing mouse model (human brain astroblastoma cells). Luo et al. [191] developed a biomimetic lipid-polymer nanoparticle loaded with PS (indocyanine green, ICG) and oxygen carrier (Hb). Nanoscale artificial RBCs (I-ARCs) were prepared by encapsulating Hb/ICG complexes in a PEG-modified lipid layer. In an MCF-7 human breast cancer model, the I-ARC provided sufficient O2, and ICG in the I-ARC efficiently converted O2 to ROS under near-infrared (NIR) laser irradiation, thereby triggering sustained damage to tumor cells. Furthermore, I-ARC serves as an ideal fluorescence imaging probe to dynamically monitor the biodistribution of nanoparticles and oxygen levels. The dynamic PDT process can be self-monitored through ICG, while serving as a diagnostic platform for self-monitoring therapy integration. Xu et al. [71] prepared a novel hemoglobin-polymer conjugate (HbTcMs) with high biocompatibility and stability as a carrier for both O2 and PS to provide additional O2 to enhance PDT efficacy. Compared with free Hb, HbTcMs not only retained the Hb oxygen-binding capacity but also exhibited higher stability against oxidation and trypsin digestion. In a 4T1 cell model, HbTcMs produced numerous oxygen-derived 1O2 molecules and exerted enhanced phototoxicity by utilizing the oxygen supply ability of Hb. Cao et al. [192] constructed the BP@RB-Hb nanoparticles by assembling Hb into nanocomplexes with novel two-photon species: bis(pyrene) (BP) and traditional PS (RB). Hb acts as an O2 donor to provide additional O2 resistance to the TME at the tumor site through NP targeting. Simultaneously, the resonance energy transfer effect (FRET) inside the nanoparticles indirectly stimulated PS under irradiation with a two-photon laser, thereby improving the treatment depth. In a tumor-bearing mouse model, the material showed excellent oxygen-carrying performance and penetration depth, greatly enhancing PDT efficiency. In tumor-bearing mouse models, BP@RB-Hb nanoparticles exhibited excellent penetration depth and oxygen-carrying properties, which greatly improved PDT treatment efficiency. Chen et al. [193] proposed a protein hybridization method for combining HSA and Hb to develop a bioinspired hybrid protein oxygen nanocarrier (C@HPOC) loaded with photosensitizer e6 (Ce6), which overcomes tumor hypoxia via co-delivery of tumor-targeted O2 and PS and improves PDT efficacy by generating a large amount of 1O2. C@HPOC stimulated robust systemic antitumor immunity and elicited potent antitumor metastatic and abscopal effects in a murine triple-negative breast cancer (4T1-mTNBC) model [194] (Fig. 6). Tian et al. [195] connected Hb to Ce6 to load sorafenib (SRF, an iron death promoter) and built a nanoplatform SRF@Hb-Ce6. Hb not only provides O2 and increases PDT efficacy, but it also synergizes with SPF, significantly increases the generation of lipid peroxides, downregulates GPX4 expression, and increases tumor cell iron death [196]. This platform shows promise as a nanoplatform for combination cancer therapy.

Fig. 6
figure 6

Copyright 2018, American Chemical Society Nanomaterials.

A Schematic depiction of oxygen-augmented immunogenic PDT with C@HPOC for eliciting the anti-metastatic and abscopal effect. Human serum albumin (HSA) was hybridized with oxygen carrying hemoglobin (Hb) via intermolecular disulfide bonds to form a hybrid protein oxygen nanocarrier with Ce6 loaded (C@HPOC). Under laser irradiation, oxygen self-supplied nanoparticles (C@HPOC) elevated the generation of cytotoxic 1O2 and moreover triggered immunogenic cell death (ICD). C@HPOC-mediated PDT not only destroyed the primary tumors but also inhibited the distant tumors and lung metastasis by systemic anti-tumor immune responses. B Confocal images of cellular uptake and ROS generation in 4T1 tumor cells. C Growth curves of primary tumor on mice after various treatments. D Growth curves of distant tumor on mice in different treated groups. E Immunofluorescence staining detection of CD8 T cells (red) in tumor tissues. A–E Reproduced with permission [197].

Application of Nano-HBOCs in SDT

SDT is an emerging cancer therapy that triggers ROS generation from abundant 1O2 using ultrasound (US), which leads to oxidative damage in tumor cells and enhanced therapeutic efficacy [197]. Its advantages include superior tissue penetration ability, minimal damage to surrounding healthy tissues, and lower skin sensitivity. The process of ROS generation using US irradiation of sonosensitizers during SDT requires the participation of O2, which is similarly limited by hypoxia in the TME [198]. In this context, Yin et al. [199] designed a novel intensified oxygen supply sonosensitizer system (MnPcS@HPO) by reconstituting bHb and HSA through disulfide bonds and loading the sonosensitizer Mn-phthalocyanine (MnPcs). This system combines the oxygen-carrying capacity of Hb and the tumor-targeting properties of HSA. MnPcS@HPO was enriched in the hypoxic area of the tumor, effectively alleviating the hypoxic state of the tumor. Simultaneously, MnPcS produces a large amount of 1O2 under US irradiation, effectively inhibiting tumor growth. Yuan et al. [72] constructed O2@Hb@ZIF-8 (OHZ) nanoparticles formed from ZIF-8 encapsulated Hb, which not only provided abundant O2 for US-induced ROS generation, but also showed excellent biocompatibility, as evidenced by cell viability evaluation and imagine (H&E) of major organs. ZIF-8 was used as a drug carrier for improving the Hb packaging rate, achieving pH-responsive Hb/O2 release, generating a large amount of O2 in the acidic TME, and relieving tumor hypoxia, thereby providing a source of O2 for US-triggered ROS generation. In 4T1 tumor-bearing mice, OHZ nanoparticles could not only inhibit the growth of subcutaneous tumors but also have an excellent inhibitory effect on the growth of deep-seated tumors, which helped achieve the SDT treatment of tumors at different depths. Pan et al. [200] synthesized ZIF-90 in water under mild conditions to encapsulate bHb and prepare an oxygen carrier, ZIF-90@Hb. The dynamics of oxygen consumption showed that the initial current of ZIF-90@Hb fanged from 9.15 μA to 4.18 μA, while that of free Hb was 2.78 μA and dropped to 0.55 μA within 100 s. This observation indicated that ZIF-90@Hb demonstrated better oxygen carrying/releasing abilities than free Hb. Furthermore, Cun et al. [75] similarly combined bHb and Au nanoparticles to synthesize ultrasmall protein metal clusters (MNCs) to develop a novel HBOC. They combined the complexes of Hb@AuNCs incorporated into the reported MOFs, which increased their antioxidant capacity to inhibit the formation of methemoglobin (the fluorescence intensity of hydrogen peroxide decreased sharply by 70%, with a remarkable 39% decrease in methemoglobin). Overall, these studies suggest that the utilization of the ZIF metal framework in oxygen carriers [74] provides a multifunctional platform for the development of blood substitutes in the near future. Liang et al. [201] encapsulated PtIV, a platinum (Pt) prodrug, with Hb to prepare a multimodal nanoparticle for tumor-targeted US radiation-triggered cancer therapy, harnessing the good solubility of Hb as a sonosensitizer. These nanoparticles can treat large volumes of deeply located tumors and provide new ideas for cancer treatment.

Hypoxia is a hallmark of TME in solid cancers [202]. It not only promotes tumor growth, but also reduces the effectiveness of treatment. Nano-HBOCs have been reported to have the ability to enhance synergistic therapy with stable O2 supply and can be loaded with drugs targeting tumor cells. Numerous studies have investigated the use of Nano-HBOCs in tumor therapy, and significant therapeutic effects have been observed in vitro and in vivo. By combining Nano-HBOCs with drugs and enhancing their abilities, we can potentially make a breakthrough in the clinical treatment of tumor.

Nano-HBOCs for wound healing

O2 plays a key role in wound healing [203] by promoting cell proliferation, accelerating angiogenesis, reducing infection, and increasing collagen synthesis [204, 205]. The oxygenation level in the wound microenvironment is a key rate-limiting factor for wound healing [206, 207]. Long-term hypoxic conditions lead to impaired neovascularization and limited wound healing [208], whereas a hypoxic environment promotes wound healing [209]. Therefore, in situ oxygen production, oxygen carriers, and various strategies to enhance oxygen supply have been used to relieve the hypoxic state of wounds, promote angiogenesis, and enhance collagen remodeling, ultimately accelerating wound healing [210,211,212,213].

Reducing oxidative stress

Diabetes mellitus (DM) is a chronic disease that affects millions of humans. Further, in more than 25% of patients with DM, factors such as uncontrollable bacterial infections [214], persistent inflammation [215], and hypoxia [216] can cause foot ulcers, which, in severe cases, can lead to amputation [217, 218]. According to previous studies, the long-term exposure of diabetic patients to a high glucose environment can functionally impair the of HIF-1α/ VEGF axis, resulting in reduced abilities to reverse and regulate the ischemic hypoxic state of the wound tissue [207, 219] to promote wound healing. Fukui [220] et al. suggested that LEH has the advantages of small particle size and high oxygen affinity, effectively perfusing obstructed blood vessels and improving the oxygen supply to the wound. Studies based on a diabetic mouse (dB/ dB) skin defect model have shown that h-LEH significantly increased the oxygen supply level of the wound microenvironment and inhibited the inflammatory cascade [221] while effectively improving the microcirculation and accelerating diabetic wound healing. MXene is a two-dimensional material with ultrathin layer topology and good biocompatibility [222]. Li et al. [79] used a system of H2O2/HbO2 to catalyze the oxidative coupling of dopamine-grafted hyaluronic acid and PDA with encapsulated Ti3C2MXene nanosheets to form injectable hydrogels (Ti3C2@PDA or MXene@PDA NSs). HbO2 exert HRP-like enzyme activity for catalyzing gel formation, whereas MXene scavenge excess free radicals, reduce oxidative damage, and maintain oxidative homeostasis while exerting a bacteriostatic effect [223]. Further, MXene and PDA have excellent photothermal conversion features that can generate heat to elevate temperature under 808 nm NIR irradiation, thereby achieving a Hb-reversible oxygen release in the wound microenvironment. In the dB/dB wound model, the hydrogel oxygen carrier continuously relieved the hypoxic state of the wound tissue, reduced oxidative stress, regulated macrophage polarization, promoted angiogenesis, and accelerated the healing of diabetic wounds.

Promoting tissue repair

O2 promotes tissue formation and repair, thereby accelerating wound healing [224, 225]. Currently, the development of miniature, universal, and customizable O2 carries is progressing rapidly [226, 227] and holds great potential for applications in the field of wound healing. Liu et al. [228] combined molybdenum disulfide quantum dot-blended gelatin methacryloyl (GelMa) to produce inverse opal microparticles as microcarriers for preparing NIR-triggered porous controllable oxygen carriers by coupling amide bonds with bHb for tissue repair. Based on a typical abdominal wall defect rat model, this oxygen carrier was found to improve the oxygen supply and play a role in supporting cell growth, thereby stimulating extracellular matrix secretion, promoting collagen and angiogenesis, accelerating granulation tissue formation, and promoting the repair of abdominal wall defects. The characteristics of oxygen carriers in tissue repair render them useful for wound healing and tissue engineering (Fig. 7).

Fig. 7
figure 7

Copyright 2019, small. H Schematic illustrations of wound healing using NIR responsive separable MNs which encapsulate BP QDs and oxygen-carrying Hb. I Corresponding double immunofluorescent staining of CD31 and α-SMA on day 9. The arrows indicate the vascular ducts. The scale bars are 100 μm. J Corresponding quantitative analysis of the blood vessel density on day 9. The scale bars are 50 μm. H–J Reproduced with permission

A The schematic illustration of the light-responsive MoS2 QDs integrated inverse opal microcarriers for controllable oxygen delivery and tissue repair. The H&E staining of B–D repaired samples after implantation for 2 weeks. Vessels in the samples are indicated with grey arrowheads. B Control, C Experimental I, D Experimental II. The Masson staining of E–G repaired samples after implantation for 2 weeks. Granulation tissue thickness in the samples are indicated with black arrowheads. E Control, F Experimental I, G Experimental II. B–G The scale bar is 200 µm. A–G Reproduced with permission [233]. [88]. Copyright 2020, American Chemical Society Nanomaterials.

Zhang et al. [86] constructed responsive microneedles (MNs) simultaneously loaded with black phosphorus (BP) and hHb to overcome the limitation that most oxygen carriers can only contact the surface area of the wound without reaching the inner tissue. These responsive MNs could penetrate the epidermal layer to the interior of wound tissue in a painless, noninvasive, and sterile manner [229, 230], thereby achieving controllable delivery of O2 and promoting wound healing. Further, a NIR controllable O2 releasing carrier was fabricated using GelMA as a synthetic material for responsive MNS, which helped improve the mechanical strength and penetration ability of MNs loaded with quantum dots of BP and Hb. In a full-thickness cutaneous wound type I diabetes rat model, the treatment group exhibited the smallest wound area, narrowest wound bed, thickest regenerated epithelial tissue, and densest neovascular state, indicating excellent healing capacity compared to the control groups. These results suggest that this responsive MN oxygen carrier proposed by this research group has an ideal therapeutic effect on wound healing.

Currently, there are only a limited number of studies on the application of Nano-HBOCs to wound healing, and the main forms of drug delivery primarily involve hydrogels and microneedles. By expanding the options for administration, such as through spraying, we believe that Nano-HBOCs will be able to better meet the clinical requirements of wound healing and become an effective therapeutic method.

Nano-HBOCs for other biomedical applications

Nano-HBOCs have also been used in other disease therapies, as discussed below.

Port-wine staining (PWS) is a non-neoplastic malformation caused by telangiectasia at birth [231] that mostly occurs in the head and neck region and severely affects patient penetrance and self-confidence [232, 233]. PWS grows modularly, tends to bleed [234], and forms fibrous scars on the skin surface, which become more pronounced after puberty [235]. Currently, PDT is a primary treatment strategy for PWS [236]. HbVs are smaller (250 nm) than RBCs and 1/40 the diameter of RBCs; further, they are inclined to flow in the marginal zone of microvessels in the blood [237] and increase the hemoglobin concentration in microvessels, enabling a more efficient conversion of light energy into heat energy. Rikihisa et al. [238] applied a laser as a treatment for PS in PWS (at a wavelength of 595 nm) using the oxygen-carrying capacity of HbVs (or LEHs). An oxygen-rich environment enhanced the PDT effect, generated a large amount of ROS, damaged the vessel wall, treated PWS, and achieved a better therapeutic effect in an animal model with chicken wattle as PWS.

Pre-eclampsia (PE) can lead to pre-term birth in pregnant women and is a leading cause of maternal and fetal death [239,240,241]. Studies have shown that inadequate trophoblast invasion in the placenta leads to the narrowing of the spiral arteries of the placenta, thereby causing placental ischemia/ hypoxia and limiting gas and nutrient exchange between the mother and fetus, which ultimately causes intrauterine growth restriction, maternal hypertension, and organ damage [242,243,244,245,246]. HbV, which is a type of Nano-HBOC with a nanoscale size and no blood group antigens, can pass through narrow placental spiral arteries and capillaries to provide an efficient O2 supply to the placenta [247, 248]. Ohta et al. [249] intravenously injected HbV into a rat preeclampsia model. The results showed that HbV alleviated fetal hypoxia by supplying oxygen while improving fetal growth restriction in a murine model of preeclamptic pregnancy. This study suggests that HbV is an effective treatment modality for fetal hypoxia caused by placental dysfunction during pregnancy.

Porphyromonas gingivalis, a gram-negative anaerobic bacterium, is one of the main pathogens causing oral infections (including gingivitis, periodontitis, and oral ulcers) [250]. Traditional broad-spectrum antibiotic therapies often lack selectivity and can lead to oral flora dysbiosis [251]. Previous studies have shown that P. gingivalis can bind to and acquire porphyrin molecules from Hb; therefore, Hb can be used as a specific drug delivery vehicle to treat P.gingivalis infection [252]. Based on this idea, Bai et al. [253] complexed rat oxyhemoglobin (oxyHb) with an anionic amphiphilic PS (IR820) to produce a stable nano-photosensitizer oxyHb@IR820. oxyHb@IR820 targets P.gingivalis, mediating the synergistic antibacterial effects of photothermal therapy and PDT [254,255,256] and providing a new therapeutic strategy against a P.gingivalis infection. In an oral infection model of P.gingivalis in rats, oxyHb@IR820 supplied exogenous O2 and addressed the limitation caused by the poor effectiveness of PDT on anaerobic bacterial infection in hypoxic environments. Meanwhile, the specific uptake of P.gingivalis oxyHb@IR820, which improved the photodynamic and photothermal conversion efficiencies, significantly inhibited bacterial growth and reduced the oral ulcer area.

Further, Nano-HBOCs showed great application prospects in other tissue engineering scenarios. For example, Paciello et al. [257] developed a novel smart cyclic oxygen-releasing biomaterial called G-HbOD, which demonstrated superior biocompatibility and oxygen supply capability for maintaining cell viability under hypoxia in 3D cell culture tests.

Vasosuppression [258] and oxidative stress [259] resulting from hypoxia are major obstacles to bioartificial islet (BAP) transplantation [260]. To address this issue in human islet allotransplantation, Mouré et al. [261], encapsulated marine worms (HEMOXCell) Hb [262], via alginate and silicone calcium peroxide, creating a compound that produced, carried, and released O2, increasing the O2 concentration in BPA. This strategy effectively addressed the lack of O2 within the graft in a closed environment, and promoting neovascularization. This novel technology provides innovative solutions for tissue and bioengineering O2 supply. Enzyme biofuel cells (EBFC) are widely used as implanted biomedical devices in cardiac pacemakers [263], electrical sensors [264], and biosensors for physiological parameters [265]. However, the current catalysts for EBFC have very low catalytic performance and poor adhesion on the electrode surface. Chen et al. [266] anchored Hb on exogenous Ca2+ and PO3-4 to generate hydroxyapatite (Ca10(PO4)6(OH)2, HAP) nanodots and reported a novel EBFC constructed from nanoengineered RBCs (NERBCs). Given its strong oxygen adsorption capacity, NERBCs exhibits excellent catalytic reduction capacity, biocompatibility, high selectivity, and lifespan when used as an EBFC-negative catalyst, thereby pointing to a new direction for fabricating bio-nanobatteries.

Conclusions and perspectives

O2 plays a vital role in life activities, and Hb is a natural carrier of oxygen in the body. The timely transfusion of RBCs can effectively restore blood volume and maintain tissue oxygen supply in both clinical and military treatment scenarios. HBOCs have become the main focus for artificial oxygen carriers due to their advantages of being closer to natural Hb and providing an effective method to solve the inherent problems of natural blood transfusion. The rapid development of nanotechnology and nanomaterials has opened new possibilities for HBOCs.

Nano-HBOCs are a new type of hemoglobin-based oxygen carriers developed using nanotechnology combined with nanomaterials. This combination, has helped improve the application scenarios of Hb and has shown certain advantages in enhancing the stability of the molecular structure of Hb, reducing vasoactivity, prolonging half-life, and improving biocompatibility. Furthermore, Nano-HBOCs have broad application prospects (Table 2), and have attracted increasing attention from the medical, material, chemical, biological, and engineering research attention. Despite the rapid development of Nano-HBOCs in the field of biomedical research, there are still several limitations associated Nano-HBOCs, which are listed below: Nano-HBOCs require complicated preparation processes and are associated with prohibitive costs. The development of simple, safe, and cost-effective preparation methods will be the focus of future studies.

  1. 1)

    Constructing Nano-HBOCs with characteristics and functions similar to those of natural RBCs is difficult; more in-depth systematic research and technological progress are required to achieve a complete biomimetic replacement of natural RBCs.

  2. 2)

    Clinical transformation and applications are significantly challenging. Currently, the vast majority of Nano-HBOCs are in the laboratory research stage, and relevant clinical research is yet to be conducted. Therefore, it is necessary to conduct strict and systematic safety and perform effective evaluations for diverse application scenarios to meet the regulatory requirements.

  3. 3)

    Non-functional methemoglobin poses a significant challenge in the development of Nano-HBOCs. However, existing studies have not sufficiently addressed the control of Hb oxidation and lacked molecular characterization. In our review, we emphasize the need to pay attention to methemoglobin production. Furthermore, we propose various approaches to address this issue, including low-temperature operation, reducing the reaction time, adding antioxidants, and employing carbon-bonding techniques etc. in the further, we hope to develop simpler and more efficient methods for controlling and detecting methemoglobin to explore more simple and efficient methods for the control and detection of methemoglobin.

  4. 4)

    Biosafety is crucial in development of Nano-HBOCs for use in the treatment of various diseases. This is particularly important in scenarios involving high doses and multiple infusions, where in vivo metabolism, transportation, toxicity evaluation, and clearance of by-products of Nano-HBOCs are major concerns. However, there is a lack of standardized evaluation methods for hemoglobin biocompatibility, especially in tests such as antigenicity and complementation experiments. Therefore, a comprehensive and systematic standardized evaluation of Nano-HBOCs biocompatibility is essential for complete replacement of natural erythrocytes.

Table 2 The key points of Nano-HBOCs, including approaches to fabrication, targeted fields and references

We firmly believe that further research on Nano-HBOCs holds immense potential for novel advancements and significant breakthroughs in multiple fields. This promising area of study not only exhibits great prospects for practical applications but also demonstrates the potential to revolutionize clinical practical and significantly enhance human health.

Availability of data and materials

Not applicable.

Abbreviations

AA:

Arachidonic acid

BAP:

Bioartificial islet transplantation

BP:

Black phosphorus

bHb:

Bovine hemoglobin

DHCNPNPs:

Camouflaged biomimetic nanocomposites

CAT:

Catalase

Ec:

Cell-free hemoglobin

DAC:

Decitabine

DHL:

DOX-Hb-lipo

DM:

Diabetes mellitus

EBFC:

Enzyme biofuel cells

EM:

Erythromer

FDA:

Food and drug administration

G-HbOD:

Gelatin hemoglobin depot

GelMa:

Gelatin methacryloyl

GM:

Gelatin microspheres

GLUT1:

Glucose transporter isoform 1

HCM:

Hb-conjugated micelles

Hb:

Hemoglobin

HbV:

Hemoglobin vesicles

HBOCs:

Hemoglobin-based oxygen carriers

HbPs:

Hemoglobin-loaded nanoparticles

HbTcMs:

Hemoglobin-polymer conjugate

HS:

Hemorrhagic shock

HDAS:

Hexadecyl carbamoyl methyl hexadecanoate

V(Hb):

Hollow nanovesicles

HeLa:

Human cervical cancer cells

hHb:

Human hemoglobin

HSA:

Human serum albumin

HIF-1α:

Hypoxia inducible factor-1α

HRP:

Horseradish peroxidase

ICG:

Indocyanine green

LEH:

Liposome-encapsulated human hemoglobin

h-LEH:

Liposomes encapsulating hemoglobin

RCC:

Renal cell carcinoma

Lt:

Lumbricus terrestris

MMP-9:

Matrix metalloproteinase-9

MPA:

Mean arterial pressure

MOFs:

Metal–organic frameworks

MNs:

Microneedles

MCAO:

Middle cerebral artery occlusion

MnPcs:

Mn-phthalocyanine

CT-26:

Mouse colon cancer models

MDR1:

Multidrug resistance gene 1

NERBCS:

Nanoengineered red blood cells

NEMR:

Nanoerythrocytes

Nano-HBOCs:

Nanomaterial-related HBOCs

I-ARCs:

Nanoscale artificial blood cells

NIR:

Near-infrared

OHZ:

O2@Hb@ZIF-8

OXA:

Oxaliplatin

oxyHb:

Oxyhemoglobin

PFCs:

Perfluorocarbons

P-gp:

P-glycoprotein

PDT:

Photodynamic therapy

PDT:

Photodynamic therapy

PS:

Photosensitizer

PLT:

Platelets

PDA:

Polydopamine

PWS:

Port-wine staining

PE:

Pre-eclampsia

RT:

Radiation therapy

ROS:

Reactive oxygen species

RBCs:

Red blood cells

FRET:

Resonance energy transfer effect

SDT:

Sonodynamic therapy

SOD:

Superoxide dismutase

tMCAO:

Transient MCAO

TME:

Tumor microenvironment

TAM:

Tumor-associated macrophage

ZnPc:

Zinc phthalocyanine

References

  1. Motealleh A, Kehr NS. Injectable oxygen-generating nanocomposite hydrogels with prolonged oxygen delivery for enhanced cell proliferation under hypoxic and normoxic conditions. J Mater Chem B. 2020;8(19):4195–201.

    Article  CAS  PubMed  Google Scholar 

  2. Ashammakhi N, et al. Advances in controlled oxygen generating biomaterials for tissue engineering and regenerative therapy. Biomacromol. 2020;21(1):56–72.

    Article  CAS  Google Scholar 

  3. Biro GP. Oxygen and ATP: the Energy Economy of the Cell. In: Liu H, Kaye AD, Jahr JS, editors. Blood substitutes and oxygen biotherapeutics. Cham: Springer International Publishing; 2022. p. 21–32.

    Chapter  Google Scholar 

  4. Werner M, et al. Femoral blood gas analysis, another tool to assess hemorrhage severity following trauma: an exploratory prospective study. Scand J Trauma Resusc Emerg Med. 2023;31(1):31.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Dyer WB, et al. Recovery of organ-specific tissue oxygen delivery at restrictive transfusion thresholds after fluid treatment in ovine haemorrhagic shock. Intensive Care Med Exp. 2022;10(1):12.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Chen WR, Chou CC, Wang CC. Phthalides serve as potent modulators to boost fetal hemoglobin induction therapy for β-hemoglobinopathies. Blood Adv. 2019;3(9):1493–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hönemann JN, et al. Hypoxia and cardiac function in patients with prior myocardial infarction. Circ Res. 2023;132(9):1165–7.

    Article  PubMed  Google Scholar 

  8. Kabra R, et al. Hidden mystery behind unilateral cerebellar infarction in hanging: a case report. Cureus. 2022;14(11):e31115.

    PubMed  PubMed Central  Google Scholar 

  9. Köhler D, et al. Hypoxic, anemic and cardiac hypoxemia: When does tissue hypoxia begin? Dtsch Med Wochenschr. 2023;148(8):475–82.

    PubMed  Google Scholar 

  10. Godoy DA, et al. Avoiding brain hypoxia in severe traumatic brain injury in settings with limited resources—a pathophysiological guide. J Crit Care. 2023;75:154260.

    Article  CAS  PubMed  Google Scholar 

  11. Haddon A, et al. Cardiorespiratory responses to voluntary hyperventilation during normobaric hypoxia. Aerosp Med Hum Perform. 2023;94(2):59–65.

    Article  PubMed  Google Scholar 

  12. Wang L, et al. Shen Shuai II recipe inhibits hypoxia-induced glycolysis by preserving mitochondrial dynamics to attenuate kidney fibrosis. J Ethnopharmacol. 2023;308:116271.

    Article  CAS  PubMed  Google Scholar 

  13. Biro GP. Erythrocyte transfusion: brief history and current practice. In: Liu H, Kaye AD, Jahr JS, editors. Blood substitutes and oxygen biotherapeutics. Cham: Springer International Publishing; 2022. p. 3–19.

    Chapter  Google Scholar 

  14. Lee P, Chandel NS, Simon MC. Cellular adaptation to hypoxia through hypoxia inducible factors and beyond. Nat Rev Mol Cell Biol. 2020;21(5):268–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Chen Z, et al. Hypoxic microenvironment in cancer: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther. 2023;8(1):70.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Li B, et al. PEG-conjugated bovine haemoglobin enhances efficiency of chemotherapeutic agent doxorubicin with alleviating DOX-induced splenocardiac toxicity in the breast cancer. Artif Cells Nanomed Biotechnol. 2023;51(1):120–30.

    Article  CAS  PubMed  Google Scholar 

  17. Luo Z, et al. Hypoxia signaling in human health and diseases: implications and prospects for therapeutics. Signal Transduct Target Ther. 2022;7(1):218.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Cherian VT. Physiological functions of blood. In: Liu H, Kaye AD, Jahr JS, editors. Blood substitutes and oxygen biotherapeutics. Cham: Springer International Publishing; 2022. p. 33–43.

    Chapter  Google Scholar 

  19. Cohn EJ. Blood: a brief survey of its chemical components and of their natural functions and clinical uses. Blood. 2015;126(24):2531.

    PubMed  Google Scholar 

  20. Mohanto N, Park YJ, Jee JP. Current perspectives of artificial oxygen carriers as red blood cell substitutes: a review of old to cutting-edge technologies using in vitro and in vivo assessments. J Pharm Investig. 2023;53(1):153–90.

    Article  CAS  PubMed  Google Scholar 

  21. Ahmed MH, Ghatge MS, Safo MK. Hemoglobin: structure, function and allostery. In: Hoeger U, Harris JR, editors. Vertebrate and invertebrate respiratory proteins, lipoproteins and other body fluid proteins. Cham: Springer International Publishing; 2020. p. 345–82.

    Chapter  Google Scholar 

  22. Shu P, et al. Cefmetazole sodium as an allosteric effector that regulates the oxygen supply efficiency of adult hemoglobin. J Biomol Struct Dyn. 2023. https://doi.org/10.1080/07391102.2023.2245043.

    Article  PubMed  Google Scholar 

  23. Yu S. Hemoglobin: physiology and hemoglobinopathy. In: Liu H, Kaye AD, Jahr JS, editors. Blood substitutes and oxygen biotherapeutics. Cham: Springer International Publishing; 2022. p. 45–51.

    Chapter  Google Scholar 

  24. Liu WL, et al. Aggressive man-made red blood cells for hypoxia-resistant photodynamic therapy. Adv Mater. 2018;30(35):e1802006.

    Article  PubMed  Google Scholar 

  25. Zujalovic B, Hafner S. Damage control resuscitation: identification and treatment of life-threatening hemorrhage. Anesth Analg. 2020;130(5):e141–2.

    Article  Google Scholar 

  26. Carreau A, et al. Why is the partial oxygen pressure of human tissues a crucial parameter? Small molecules and hypoxia. J Cell Mol Med. 2011;15(6):1239–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Haque N, et al. Hypoxic culture conditions as a solution for mesenchymal stem cell based regenerative therapy. Sci World J. 2013;2013:632972.

    Article  Google Scholar 

  28. Khan F, Singh K, Friedman MT. Artificial blood: the history and current perspectives of blood substitutes. Discoveries (Craiova). 2020;8(1):e104.

    Article  PubMed  Google Scholar 

  29. Bialas C, Moser C, Sims CA. Artificial oxygen carriers and red blood cell substitutes: A historic overview and recent developments toward military and clinical relevance. J Trauma Acute Care Surg. 2019;87(1S):S48-s58.

    Article  PubMed  Google Scholar 

  30. Stanworth SJ, et al. Effects of the COVID-19 pandemic on supply and use of blood for transfusion. Lancet Haematol. 2020;7(10):e756–64.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Coll-Satue C, et al. Stepping stones to the future of haemoglobin-based blood products: clinical, preclinical and innovative examples. Biomater Sci. 2021;9(4):1135–52.

    Article  CAS  PubMed  Google Scholar 

  32. Dumont LJ, et al. Overnight, room temperature hold of whole blood followed by 42-day storage of red blood cells in additive solution-7. Transfusion. 2015;55(3):485–90.

    Article  CAS  PubMed  Google Scholar 

  33. Brand A. Immunological complications of blood transfusions. Presse Med. 2016;45(7–8 Pt 2):e313–24.

    Article  PubMed  Google Scholar 

  34. Xia S, et al. Addition of sodium pyruvate to stored red blood cells attenuates liver injury in a murine transfusion model. Mediators Inflamm. 2016;2016:3549207.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Charbe NB, et al. A new era in oxygen therapeutics? From perfluorocarbon systems to haemoglobin-based oxygen carriers. Blood Rev. 2022;54:100927.

    Article  CAS  PubMed  Google Scholar 

  36. Rangayasami A, et al. Influence of nanotechnology to combat against COVID-19 for global health emergency: a review. Sensors International. 2021;2:100079.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Berg L, et al. Obstetric outcomes in Jehovah’s Witnesses: case series over nine years in a London teaching hospital. Arch Gynecol Obstet. 2023. https://doi.org/10.1007/s00404-023-06940-x.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Naicker YD, Ahmed N, Davids R. Damage control surgery of the critical Jehovah’s Witness patient—a narrative review. S Afr J Surg. 2023;61(1):39–44.

    Article  CAS  PubMed  Google Scholar 

  39. Liu H, et al. Classifications of blood substitutes. In: Liu H, Kaye AD, Jahr JS, editors., et al., Blood substitutes and oxygen biotherapeutics. Cham: Springer International Publishing; 2022. p. 119–29.

    Chapter  Google Scholar 

  40. Jahr JS, et al. Blood substitutes and oxygen therapeutics: a review. Anesth Analg. 2021;132(1):119–29.

    Article  CAS  PubMed  Google Scholar 

  41. Wu L, et al. Perfluorocarbons-based (19)F magnetic resonance imaging in biomedicine. Int J Nanomed. 2020;15:7377–95.

    Article  CAS  Google Scholar 

  42. Castro CI, Briceno JC. Perfluorocarbon-based oxygen carriers: review of products and trials. Artif Organs. 2010;34(8):622–34.

    Article  PubMed  Google Scholar 

  43. Park KM, Gerecht S. Hypoxia-inducible hydrogels. Nat Commun. 2014;5:4075.

    Article  CAS  PubMed  Google Scholar 

  44. Spahn DR, Kocian R. Artificial O2 carriers: status in 2005. Curr Pharm Des. 2005;11(31):4099–114.

    Article  CAS  PubMed  Google Scholar 

  45. Chen L, Yang Z, Liu H. Hemoglobin-based oxygen carriers: where are we now in 2023? Med (Kaunas). 2023. https://doi.org/10.3390/medicina59020396.

    Article  Google Scholar 

  46. Mer M, et al. Hemoglobin glutamer-250 (bovine) in South Africa: consensus usage guidelines from clinician experts who have treated patients. Transfusion. 2016;56(10):2631–6.

    Article  PubMed  Google Scholar 

  47. Cao M, et al. New applications of HBOC-201: a 25-year review of the literature. Front Med (Lausanne). 2021;8:794561.

    Article  PubMed  Google Scholar 

  48. Romito BT, et al. The effect of SANGUINATE(®) (PEGylated carboxyhemoglobin bovine) on cardiopulmonary bypass functionality using a bovine whole blood model of normovolemic hemodilution. Perfusion. 2020;35(1):19–25.

    Article  PubMed  Google Scholar 

  49. Marrazzo F, et al. Inhaled nitric oxide prevents systemic and pulmonary vasoconstriction due to hemoglobin-based oxygen carrier infusion: A case report. J Crit Care. 2019;51:213–6.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Wang Y, et al. Structural, functional and physiochemical properties of dextran-bovine hemoglobin conjugate as a hemoglobin-based oxygen carrier. Process Biochem. 2017;60:67–73.

    Article  CAS  Google Scholar 

  51. Huo S, et al. Ferrous hemoglobin and hemoglobin-based oxygen carriers acting as a peroxidase can inhibit oxidative damage to endothelial cells caused by hydrogen peroxide. Artif Organs. 2021;45(10):1229–39.

    Article  CAS  PubMed  Google Scholar 

  52. Li T, Jing X, Huang Y. Polymer/hemoglobin assemblies: biodegradable oxygen carriers for artificial red blood cells. Macromol Biosci. 2011;11(7):865–75.

    Article  CAS  PubMed  Google Scholar 

  53. Moradi S, Jahanian-Najafabadi A, Roudkenar MH. Artificial blood substitutes: first steps on the long route to clinical utility. Clin Med Insights Blood Disord. 2016;9:33–41.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Sen Gupta A. Hemoglobin-based oxygen carriers: current state-of-the-art and novel molecules. Shock. 2019;52(1S):70–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Wang Y, et al. A PEGylated bovine hemoglobin as a potent hemoglobin-based oxygen carrier. Biotechnol Prog. 2017;33(1):252–60.

    Article  PubMed  Google Scholar 

  56. Bian Y, Chang TM. A novel nanobiotherapeutic poly-[hemoglobin-superoxide dismutase-catalase-carbonic anhydrase] with no cardiac toxicity for the resuscitation of a rat model with 90 minutes of sustained severe hemorrhagic shock with loss of 2/3 blood volume. Artif Cells Nanomed Biotechnol. 2015;43(1):1–9.

    Article  PubMed  Google Scholar 

  57. Chang TM. Blood replacement with nanobiotechnologically engineered hemoglobin and hemoglobin nanocapsules. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2010;2(4):418–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Bian Y, Chang TMS. Nanobiotechnological basis of an oxygen carrier with enhanced carbonic anhydrase for CO(2) transport and enhanced catalase and superoxide dismutase for antioxidant function. Front Bioeng Biotechnol. 2023;11:1188399.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Mitchell MJ, et al. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov. 2021;20(2):101–24.

    Article  CAS  PubMed  Google Scholar 

  60. Bäumler H, Xiong Y, Georgieva R. Nanotechnology-based oxygen and drug carriers. In: Liu H, Kaye AD, Jahr JS, editors. Blood substitutes and oxygen biotherapeutics. Cham: Springer International Publishing; 2022. p. 169–73.

    Chapter  Google Scholar 

  61. Modery-Pawlowski CL, et al. Synthetic approaches to RBC mimicry and oxygen carrier systems. Biomacromol. 2013;14(4):939–48.

    Article  CAS  Google Scholar 

  62. Liu X, et al. Low-fouling electrosprayed hemoglobin nanoparticles with antioxidant protection as promising oxygen carriers. Macromol Biosci. 2020;20(2):e1900293.

    Article  PubMed  Google Scholar 

  63. Nadimifar M, et al. Synthesis of bioactive hemoglobin-based oxygen carrier nanoparticles via metal-phenolic complexation. Biomater Adv. 2024;156:213698.

    Article  CAS  PubMed  Google Scholar 

  64. Chang TMS. A brief history of the development of nanobiotechnology-based blood substitutes. In: Liu H, Kaye AD, Jahr JS, editors. Blood substitutes and oxygen biotherapeutics. Cham: Springer International Publishing; 2022. p. 99–115.

    Chapter  Google Scholar 

  65. Matsuhira T, Sakai H. Artificial oxygen carriers, from nanometer- to micrometer-sized particles, made of hemoglobin composites substituting for red blood cells. Particuology. 2022;64:43–55.

    Article  CAS  Google Scholar 

  66. Jia Y, Duan L, Li J. Hemoglobin-based nanoarchitectonic assemblies as oxygen carriers. Adv Mater. 2016;28(6):1312–8.

    Article  CAS  PubMed  Google Scholar 

  67. Tao Z, Ghoroghchian PP. Microparticle, nanoparticle, and stem cell-based oxygen carriers as advanced blood substitutes. Trends Biotechnol. 2014;32(9):466–73.

    Article  CAS  PubMed  Google Scholar 

  68. Jansman MMT, Hosta-Rigau L. Recent and prominent examples of nano- and microarchitectures as hemoglobin-based oxygen carriers. Adv Colloid Interface Sci. 2018;260:65–84.

    Article  CAS  PubMed  Google Scholar 

  69. Kaneda S, et al. Liposome-encapsulated hemoglobin, TRM-645: current status of the development and important issues for clinical application. Artif Organs. 2009;33(2):146–52.

    Article  CAS  PubMed  Google Scholar 

  70. Lu M, et al. Preparation, characterization and in vivo investigation of blood-compatible hemoglobin-loaded nanoparticles as oxygen carriers. Colloid Surf B Biointerfaces. 2016;139:171–9.

    Article  CAS  PubMed  Google Scholar 

  71. Xu X, et al. A photosensitizer loaded hemoglobin-polymer conjugate as a nanocarrier for enhanced photodynamic therapy. J Mater Chem B. 2018;6(12):1825–33.

    Article  CAS  PubMed  Google Scholar 

  72. Yuan M, et al. A robust oxygen-carrying hemoglobin-based natural sonosensitizer for sonodynamic cancer therapy. Nano Lett. 2021;21(14):6042–50.

    Article  CAS  PubMed  Google Scholar 

  73. Pozy E, Savla C, Palmer AF. Photocatalytic synthesis of a polydopamine-coated acellular mega-hemoglobin as a potential oxygen therapeutic with antioxidant properties. Biomacromol. 2023;24(5):2022–9.

    Article  CAS  Google Scholar 

  74. Peng S, et al. Metal-organic framework encapsulating hemoglobin as a high-stable and long-circulating oxygen carriers to treat hemorrhagic shock. ACS Appl Mater Interfaces. 2019;11(39):35604–12.

    Article  CAS  PubMed  Google Scholar 

  75. Cun X, et al. Hemoglobin-stabilized gold nanoclusters displaying oxygen transport ability, self-antioxidation, auto-fluorescence properties and long-term storage potential. RSC Adv. 2023;13(23):15540–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Tang W, et al. Red blood cell-facilitated photodynamic therapy for cancer treatment. Adv Funct Mater. 2016;26(11):1757–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Wang Q, et al. Bioinspired polydopamine-coated hemoglobin as potential oxygen carrier with antioxidant properties. Biomacromol. 2017;18(4):1333–41.

    Article  Google Scholar 

  78. Hu J, et al. Polydopamine-based surface modification of hemoglobin particles for stability enhancement of oxygen carriers. J Colloid Interface Sci. 2020;571:326–36.

    Article  CAS  PubMed  Google Scholar 

  79. Li Y, et al. Artificial nonenzymatic antioxidant mxene nanosheet-anchored injectable hydrogel as a mild photothermal-controlled oxygen release platform for diabetic wound healing. ACS Nano. 2022;16(5):7486–502.

    Article  CAS  PubMed  Google Scholar 

  80. Pan D, et al. Erythromer (EM), a nanoscale bio-synthetic artificial red cell: proof of concept and in vivo efficacy results. Blood. 2016;128(22):1027–1027.

    Article  Google Scholar 

  81. Liu P, et al. Bioinspired nanoerythrocytes for metabolic microenvironment remodeling and long-term prognosis promoting of acute ischemic stroke. Nano Today. 2023;49:101806.

    Article  CAS  Google Scholar 

  82. Bu H, et al. Synthesis of a hemoglobin-conjugated triblock copolymer for oxygen carrying and specific recognition of cancer cells. RSC Adv. 2017;7(76):48166–75.

    Article  CAS  Google Scholar 

  83. Tian H, et al. Cancer cell membrane-biomimetic oxygen nanocarrier for breaking hypoxia-induced chemoresistance. Adv Func Mater. 2017;27(38):1703197.

    Article  Google Scholar 

  84. Wang Y, et al. Engineering endogenous tumor-associated macrophage-targeted biomimetic nano-RBC to reprogram tumor immunosuppressive microenvironment for enhanced chemo-immunotherapy. Adv Mater. 2021;33(39):e2103497.

    Article  PubMed  Google Scholar 

  85. Xia D, et al. Au-hemoglobin loaded platelet alleviating tumor hypoxia and enhancing the radiotherapy effect with low-dose X-ray. ACS Nano. 2020;14(11):15654–68.

    Article  CAS  PubMed  Google Scholar 

  86. Zhang X, et al. Black phosphorus-loaded separable microneedles as responsive oxygen delivery carriers for wound healing. ACS Nano. 2020;14(5):5901–8.

    Article  CAS  PubMed  Google Scholar 

  87. Munoz C, et al. Resuscitation after hemorrhagic shock in the microcirculation: targeting optimal oxygen delivery in the design of artificial blood substitutes. Front Med (Lausanne). 2020;7:585638.

    Article  PubMed  Google Scholar 

  88. Chen G, et al. Effects of synthetic colloids on oxidative stress and inflammatory response in hemorrhagic shock: comparison of hydroxyethyl starch 130/0.4, hydroxyethyl starch 200/0.5, and succinylated gelatin. Crit Care. 2013;17(4):R141.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Bragin DE, et al. Addition of drag-reducing polymers to colloid resuscitation fluid enhances cerebral microcirculation and tissue oxygenation after traumatic brain injury complicated by hemorrhagic shock. Adv Exp Med Biol. 2021;1269:283–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Dyer WB, et al. An ovine model of hemorrhagic shock and resuscitation, to assess recovery of tissue oxygen delivery and oxygen debt, and inform patient blood management. Shock. 2021;56(6):1080–91.

    Article  CAS  PubMed  Google Scholar 

  91. Hallisey SD, Greenwood JC. Beyond mean arterial pressure and lactate: perfusion end points for managing the shocked patient. Emerg Med Clin North Am. 2019;37(3):395–408.

    Article  PubMed  Google Scholar 

  92. Yamamoto M, et al. Fluid resuscitation of hemorrhagic shock with hemoglobin vesicles in Beagle dogs: pilot study. Artif Cells Blood Substit Immobil Biotechnol. 2012;40(1–2):179–95.

    Article  CAS  PubMed  Google Scholar 

  93. Yuki Y, et al. Efficacy of resuscitative infusion with hemoglobin vesicles in rabbits with massive obstetric hemorrhage. Am J Obstet Gynecol. 2021;224(4):398.e1-398.e11.

    Article  CAS  PubMed  Google Scholar 

  94. Takase B, et al. Liposome-encapsulated hemoglobin (HbV) transfusion rescues rats undergoing progressive lethal 85% hemorrhage as a result of an anti-arrhythmogenic effect on the myocardium. Artif Organs. 2021;45(11):1391–404.

    Article  CAS  PubMed  Google Scholar 

  95. Sakai H. Overview of potential clinical applications of hemoglobin vesicles (HbV) as artificial red cells, evidenced by preclinical studies of the academic research consortium. J Funct Biomater. 2017. https://doi.org/10.3390/jfb8010010.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Fujihara M, et al. Primary and secondary immune responses to keyhole limpet hemocyanin in rats after infusion of hemoglobin vesicle, an artificial oxygen carrier. Artif Organs. 2014;38(3):234–8.

    Article  CAS  PubMed  Google Scholar 

  97. Sakai H, et al. One-year observation of Wistar rats after intravenous infusion of hemoglobin-vesicles (artificial oxygen carriers). Artif Cells Blood Substit Immobil Biotechnol. 2007;35(1):81–91.

    Article  CAS  PubMed  Google Scholar 

  98. Sakai H, et al. Metabolism of hemoglobin-vesicles (artificial oxygen carriers) and their influence on organ functions in a rat model. Biomaterials. 2004;25(18):4317–25.

    Article  CAS  PubMed  Google Scholar 

  99. Sakai H, et al. Hemoglobin-vesicles as oxygen carriers: influence on phagocytic activity and histopathological changes in reticuloendothelial system. Am J Pathol. 2001;159(3):1079–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Taguchi K, et al. Hepatically-metabolized and -excreted artificial oxygen carrier, hemoglobin vesicles, can be safely used under conditions of hepatic impairment. Toxicol Appl Pharmacol. 2010;248(3):234–41.

    Article  CAS  PubMed  Google Scholar 

  101. Taguchi K, et al. Biological responsiveness and metabolic performance of liposome-encapsulated hemoglobin (Hemoglobin-Vesicles) in apolipoprotein E-deficient mice after massive intravenous injection. Biol Pharm Bull. 2015;38(10):1606–16.

    Article  CAS  PubMed  Google Scholar 

  102. Shono S, et al. Intraosseous transfusion with liposome-encapsulated hemoglobin improves mouse survival after hypohemoglobinemic shock without scavenging nitric oxide. Shock. 2011;35(1):45–52.

    Article  CAS  PubMed  Google Scholar 

  103. Eriksson M, et al. The effect of hemorrhagic shock and intraosseous adrenaline injection on the delivery of a subsequently administered drug - an experimental study. Scand J Trauma Resusc Emerg Med. 2019;27(1):29.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Yadav VR, et al. Nanovesicular liposome-encapsulated hemoglobin (LEH) prevents multi-organ injuries in a rat model of hemorrhagic shock. Eur J Pharm Sci. 2016;93:97–106.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Nag OK, et al. Post-modification of preformed liposomes with novel non-phospholipid poly(ethylene glycol)-conjugated hexadecylcarbamoylmethyl hexadecanoic acid for enhanced circulation persistence in vivo. Int J Pharm. 2013;446(1–2):119–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Agashe H, et al. Improved formulation of liposome-encapsulated hemoglobin with an anionic non-phospholipid. Colloids Surf B Biointerfaces. 2010;75(2):573–83.

    Article  CAS  PubMed  Google Scholar 

  107. Mittal N, et al. Erythromer (EM), a Nanoscale Bio-Synthetic Artificial Red Cell. In: Liu H, Kaye AD, Jahr JS, editors., et al., Blood substitutes and oxygen biotherapeutics. Cham: Springer International Publishing; 2022. p. 253–65.

    Chapter  Google Scholar 

  108. Okamoto W, et al. Hemoglobin–albumin clusters as an artificial O2 carrier: Physicochemical properties and resuscitation from hemorrhagic shock in rats. J Biomed Mater Res B Appl Biomater. 2022;110(8):1827–38.

    Article  CAS  PubMed  Google Scholar 

  109. Liu Y, Ai K, Lu L. Polydopamine and its derivative materials: synthesis and promising applications in energy, environmental, and biomedical fields. Chem Rev. 2014;114(9):5057–115.

    Article  CAS  PubMed  Google Scholar 

  110. Zhao Y, et al. Hollow PDA-Au nanoparticles-enabled signal amplification for sensitive nonenzymatic colorimetric immunodetection of carbohydrate antigen 125. Biosens Bioelectron. 2015;71:200–6.

    Article  CAS  PubMed  Google Scholar 

  111. Baidukova O, et al. Antioxidative protection of haemoglobin microparticles (HbMPs) by PolyDopamine. Artif Cells Nanomed Biotechnol. 2018;46(sup3):S693-s701.

    Article  CAS  PubMed  Google Scholar 

  112. Jansman MMT, et al. Hemoglobin-based oxygen carriers incorporating nanozymes for the depletion of reactive oxygen species. ACS Appl Mater Interfaces. 2020;12(45):50275–86.

    Article  CAS  PubMed  Google Scholar 

  113. Liu X, et al. Metal-organic framework-based oxygen carriers with antioxidant activity resulting from the incorporation of gold nanozymes. Biomater Sci. 2023;11(7):2551–65.

    Article  CAS  PubMed  Google Scholar 

  114. Lian X, et al. Enzyme-MOF (metal-organic framework) composites. Chem Soc Rev. 2017;46(11):3386–401.

    Article  CAS  PubMed  Google Scholar 

  115. Chen Y, et al. Acid-resistant mesoporous metal-organic framework toward oral insulin delivery: protein encapsulation, protection, and release. J Am Chem Soc. 2018;140(17):5678–81.

    Article  CAS  PubMed  Google Scholar 

  116. Wang S, et al. DNA-functionalized metal-organic framework nanoparticles for intracellular delivery of proteins. J Am Chem Soc. 2019;141(6):2215–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Liang K, et al. Biomimetic mineralization of metal-organic frameworks as protective coatings for biomacromolecules. Nat Commun. 2015;6:7240.

    Article  CAS  PubMed  Google Scholar 

  118. Chen G, et al. A convenient and versatile amino-acid-boosted biomimetic strategy for the nondestructive encapsulation of biomacromolecules within metal-organic frameworks. Angew Chem Int Ed Engl. 2019;58(5):1463–7.

    Article  CAS  PubMed  Google Scholar 

  119. Poddar A, et al. Encapsulation, visualization and expression of genes with biomimetically mineralized zeolitic imidazolate framework-8 (ZIF-8). Small. 2019;15(36):e1902268.

    Article  PubMed  Google Scholar 

  120. Jansman MMT, et al. Hemoglobin-based oxygen carriers camouflaged with membranes extracted from red blood cells: optimization and assessment of functionality. Biomater Adv. 2022;134:112691.

    Article  CAS  PubMed  Google Scholar 

  121. Sun H, et al. Real-time model-based cerebral perfusion calculation for ischemic stroke. Comput Methods Programs Biomed. 2024;243: 107916.

    Article  PubMed  Google Scholar 

  122. Demyanenko S, Dzreyan V, Sharifulina S. Histone deacetylases and their isoform-specific inhibitors in ischemic stroke. Biomedicines. 2021;9(10):1445.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Hurd MD, et al. Current status of ischemic stroke treatment: from thrombolysis to potential regenerative medicine. Regenerat Ther. 2021;18:408–17.

    Article  CAS  Google Scholar 

  124. Wu DM, et al. Immune pathway activation in neurons triggers neural damage after stroke. Cell Rep. 2023;42(11):113368.

    Article  CAS  PubMed  Google Scholar 

  125. Jun-Long H, et al. Necroptosis signaling pathways in stroke: from mechanisms to therapies. Curr Neuropharmacol. 2018;16(9):1327–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Lin W, Powers WJ. Oxygen metabolism in acute ischemic stroke. J Cereb Blood Flow Metab. 2018;38(9):1481–99.

    Article  CAS  PubMed  Google Scholar 

  127. Tirapelli CR. Oxidative stress and vascular disease. Curr Hypertens Rev. 2020;16(3):162.

    Article  PubMed  Google Scholar 

  128. Zuluaga Tamayo M, et al. Astaxanthin complexes to attenuate muscle damage after in vivo femoral ischemia-reperfusion. Mar Drugs. 2019. https://doi.org/10.3390/md17060354.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Deuchar GA, et al. Preclinical validation of the therapeutic potential of glasgow oxygen level dependent (GOLD) technology: a theranostic for acute stroke. Transl Stroke Res. 2019;10(5):583–95.

    Article  CAS  PubMed  Google Scholar 

  130. Klaus JA, et al. Early treatment of transient focal cerebral ischemia with bovine PEGylated carboxy hemoglobin transfusion. Artificial Cell Blood Substitutes Biotechnol. 2010;38(5):223–9.

    Article  CAS  Google Scholar 

  131. Kakehata J, et al. Therapeutic potentials of an artificial oxygen-carrier, liposome-encapsulated hemoglobin, for ischemia/reperfusion-induced cerebral dysfunction in rats. J Pharmacol Sci. 2010;114(2):189–97.

    Article  CAS  PubMed  Google Scholar 

  132. Kaneda S, et al. Efficacy of liposome-encapsulated hemoglobin in a rat model of cerebral ischemia. Artif Organs. 2014;38(8):650–5.

    Article  CAS  PubMed  Google Scholar 

  133. Cipolla MJ, et al. Pharmacologically increasing collateral perfusion during acute stroke using a carboxyhemoglobin gas transfer agent (Sanguinate™) in spontaneously hypertensive rats. J Cereb Blood Flow Metab. 2018;38(5):755–66.

    Article  CAS  PubMed  Google Scholar 

  134. Powanda DD, Chang TMS. Cross-linked polyhemoglobin-superoxide dismutase-catalase supplies oxygen without causing blood brain barrier disruption or brain edema in a rat model of transient global brain ischemia-reperfusion. Artificial Cell Blood Substitutes Biotechnol. 2002;30(1):23–37.

    Article  CAS  Google Scholar 

  135. Xie Z, et al. The protective effect of polymerized porcine hemoglobin (pPolyHb) on transient focal cerebral ischemia/reperfusion injury. Artif Cells Nanomed Biotechnol. 2015;43(3):180–5.

    Article  CAS  PubMed  Google Scholar 

  136. Caswell JE, et al. A novel hemoglobin-based blood substitute protects against myocardial reperfusion injury. Am J Physiol-Heart Circulatory Physiol. 2005;288(4):H1796–801.

    Article  CAS  Google Scholar 

  137. Komatsu H, et al. Effect of hemoglobin vesicle, a cellular-type artificial oxygen carrier, on middle cerebral artery occlusion- and arachidonic acid-induced stroke models in rats. Neurosci Lett. 2007;421(2):121–5.

    Article  CAS  PubMed  Google Scholar 

  138. Fukumoto D, et al. Liposome-encapsulated hemoglobin reduces the size of cerebral infarction in rats: effect of oxygen affinity. Artif Organs. 2009;33(2):159–63.

    Article  CAS  PubMed  Google Scholar 

  139. Kawaguchi AT, et al. Effect of oxygen affinity of liposome-encapsulated hemoglobin on cerebral ischemia and reperfusion as detected by positron emission tomography in nonhuman primates. Artif Organs. 2017;41(4):336–45.

    Article  CAS  PubMed  Google Scholar 

  140. Tomita D, et al. Covalent core-shell architecture of hemoglobin and human serum albumin as an artificial O2 carrier. Biomacromol. 2013;14(6):1816–25.

    Article  CAS  Google Scholar 

  141. Gekka M, et al. Novel hemoglobin-based oxygen carrier bound with albumin shows neuroprotection with possible antioxidant effects. Stroke. 2018;49(8):1960–8.

    Article  CAS  PubMed  Google Scholar 

  142. Hosaka H, et al. Hemoglobin-albumin cluster incorporating a Pt nanoparticle: artificial O2 carrier with antioxidant activities. PLoS ONE. 2014;9(10):e110541.

    Article  PubMed  PubMed Central  Google Scholar 

  143. Wang S, et al. Pan-immune-inflammatory value predicts the 3 month outcome in acute ischemic stroke patients after intravenous thrombolysis. Curr Neurovasc Res. 2023. https://doi.org/10.2174/0115672026276427231024045957.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Haupt M, et al. Preconditioning concepts for the therapeutic use of extracellular vesicles against stroke. Stem Cells Transl Med. 2023. https://doi.org/10.1093/stcltm/szad055.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Yin N, et al. Engineered nanoerythrocytes alleviate central nervous system inflammation by regulating the polarization of inflammatory microglia. Adv Mater. 2022;34(27):e2201322.

    Article  PubMed  Google Scholar 

  146. Shimbo D, et al. Post-ischemic intra-arterial infusion of liposome-encapsulated hemoglobin can reduce ischemia reperfusion injury. Brain Res. 2014;1554:59–66.

    Article  CAS  PubMed  Google Scholar 

  147. Shimbo D, et al. Superior microvascular perfusion of infused liposome-encapsulated hemoglobin prior to reductions in infarctions after transient focal cerebral ischemia. J Stroke Cerebrovasc Dis. 2017;26(12):2994–3003.

    Article  PubMed  Google Scholar 

  148. Choudhari OK, et al. Matrix metalloproteinase-9 gene polymorphism and its methylation in stroke patients. Malays J Med Sci. 2021;28(6):32–41.

    PubMed  PubMed Central  Google Scholar 

  149. Viallard C, Larrivée B. Tumor angiogenesis and vascular normalization: alternative therapeutic targets. Angiogenesis. 2017;20(4):409–26.

    Article  CAS  PubMed  Google Scholar 

  150. Liao C, et al. Tumor hypoxia: from basic knowledge to therapeutic implications. Semin Cancer Biol. 2023;88:172–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Dai Y, et al. Nanoparticle design strategies for enhanced anticancer therapy by exploiting the tumour microenvironment. Chem Soc Rev. 2017;46(12):3830–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Kwon S, et al. Nanomedicines for reactive oxygen species mediated approach: an emerging paradigm for cancer treatment. Acc Chem Res. 2019;52(7):1771–82.

    Article  CAS  PubMed  Google Scholar 

  153. Shi R, Tang YQ, Miao H. Metabolism in tumor microenvironment: Implications for cancer immunotherapy. MedComm. 2020;1(1):47–68.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Semenza GL. Oxygen sensing, hypoxia-inducible factors, and disease pathophysiology. Annu Rev Pathol. 2014;9:47–71.

    Article  CAS  PubMed  Google Scholar 

  155. Gilkes DM, Semenza GL, Wirtz D. Hypoxia and the extracellular matrix: drivers of tumour metastasis. Nat Rev Cancer. 2014;14(6):430–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Yang S, et al. Boosting the anti-tumor performance of disulfiram against glioblastoma by using ultrasmall nanoparticles and HIF-1α inhibitor. Compos B Eng. 2022;243:110117.

    Article  CAS  Google Scholar 

  157. Liu Y, et al. Modulation of hypoxia and redox in the solid tumor microenvironment with a catalytic nanoplatform to enhance combinational chemodynamic/sonodynamic therapy. Biomater Sci. 2023;11(5):1739–53.

    Article  CAS  PubMed  Google Scholar 

  158. Huang J, et al. Oxygen-carrying nanoplatform to reprogram tumor immunosuppressive microenvironment and enhance photothermal-immunotherapy. Mater Today Bio. 2023;19:100555.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Yuan CS, et al. Hypoxia-modulatory nanomaterials to relieve tumor hypoxic microenvironment and enhance immunotherapy: where do we stand? Acta Biomater. 2021;125:1–28.

    Article  CAS  PubMed  Google Scholar 

  160. Telarovic I, Wenger RH, Pruschy M. Interfering with tumor hypoxia for radiotherapy optimization. J Exp Clin Cancer Res. 2021;40(1):197.

    Article  PubMed  PubMed Central  Google Scholar 

  161. Zhang J, et al. Nanotechnological strategies to increase the oxygen content of the tumor. Front Pharmacol. 2023;14:1140362.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Vaupel P, Multhoff G. Fatal alliance of hypoxia-/HIF-1α-Driven microenvironmental traits promoting cancer progression. Adv Exp Med Biol. 2020;1232:169–76.

    Article  CAS  PubMed  Google Scholar 

  163. Mortezaee K, Majidpoor J. The impact of hypoxia on immune state in cancer. Life Sci. 2021;286:120057.

    Article  CAS  PubMed  Google Scholar 

  164. Doublier S, et al. HIF-1 activation induces doxorubicin resistance in MCF7 3-D spheroids via P-glycoprotein expression: a potential model of the chemo-resistance of invasive micropapillary carcinoma of the breast. BMC Cancer. 2012;12:4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Samanta D, et al. Hypoxia-inducible factors are required for chemotherapy resistance of breast cancer stem cells. Proc Natl Acad Sci U S A. 2014;111(50):E5429–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Kawaguchi F, et al. Liposome-encapsulated hemoglobin improves tumor oxygenation as detected by near-infrared spectroscopy in colon carcinoma in mice. Artif Organs. 2017;41(4):327–35.

    Article  CAS  PubMed  Google Scholar 

  167. Liu Y, et al. Epigenetic activation of the drug transporter OCT2 sensitizes renal cell carcinoma to oxaliplatin. Sci Trans Med. 2016;8(348):348ra97-348ra97.

    Article  Google Scholar 

  168. Zheng X, et al. Response to Comment on “epigenetic activation of the drug transporter OCT2 sensitizes renal cell carcinoma to oxaliplatin.” Sci Trans Med. 2017;9(391):eaam6298.

    Article  Google Scholar 

  169. Chen L, et al. The failure of DAC to induce OCT2 expression and its remission by hemoglobin-based nanocarriers under hypoxia in renal cell carcinoma. Theranostics. 2020;10(8):3562–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Guo Y, et al. Cell microenvironment-controlled antitumor drug releasing-nanomicelles for GLUT1-targeting hepatocellular carcinoma therapy. ACS Appl Mater Interfaces. 2015;7(9):5444–53.

    Article  CAS  PubMed  Google Scholar 

  171. Synthesis of a hemoglobin-conjugated triblock copolymer for oxygen carrying and specific recognition of cancer cells. RSC Adv., 2017; 7(76):48166–48175.

  172. Yang, J., et al., Hypoxic tumor therapy by hemoglobin-mediated drug delivery and reversal of hypoxia-induced chemoresistance. Biomaterials, 2018. 182: p. 145–156.

    Article  CAS  PubMed  Google Scholar 

  173. Sies H, Jones DP. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat Rev Mol Cell Biol. 2020;21(7):363–83.

    Article  CAS  PubMed  Google Scholar 

  174. Jia S, et al. Promoting reactive oxygen species generation: a key strategy in nanosensitizer-mediated radiotherapy. Nanomedicine (Lond). 2021;16(9):759–78.

    Article  CAS  PubMed  Google Scholar 

  175. Huang L, et al. Photodynamic therapy for hypoxic tumors: advances and perspectives. Coord Chem Rev. 2021;438:213888.

    Article  CAS  Google Scholar 

  176. Li M, et al. Near-infrared light-initiated molecular superoxide radical generator: rejuvenating photodynamic therapy against hypoxic tumors. J Am Chem Soc. 2018;140(44):14851–9.

    Article  CAS  PubMed  Google Scholar 

  177. Wang S, et al. Hemoglobins from scapharca subcrenata (Bivalvia: Arcidae) likely play an bactericidal role through their peroxidase activity. Comp Biochem Physiol B: Biochem Mol Biol. 2021;253:110545.

    Article  CAS  PubMed  Google Scholar 

  178. Wang R, et al. PEGylated hollow gold nanoparticles for combined X-ray radiation and photothermal therapy in vitro and enhanced CT imaging in vivo. Nanomed Nanotechnol Biol Med. 2019;16:195–205.

    Article  CAS  Google Scholar 

  179. Goel S, Ni D, Cai W. Harnessing the power of nanotechnology for enhanced radiation therapy. ACS Nano. 2017;11(6):5233–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Sang W, et al. Oxygen-enriched metal-phenolic X-ray nanoprocessor for cancer radio-radiodynamic therapy in combination with checkpoint blockade immunotherapy. Adv Sci. 2021;8(4):2003338.

    Article  CAS  Google Scholar 

  181. Hu CM, et al. Nanoparticle biointerfacing by platelet membrane cloaking. Nature. 2015;526(7571):118–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Yu M, et al. Development of “smart” drug delivery systems for chemo/PDT synergistic treatment. J Mater Chem B. 2023;11(7):1416–33.

    Article  CAS  PubMed  Google Scholar 

  183. Mossakowska BJ, et al. Possible mechanisms of resistance development to photodynamic therapy (PDT) in vulvar cancer cells. Int J Mol Sci. 2022. https://doi.org/10.3390/ijms232314689.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Dang J, et al. Manipulating tumor hypoxia toward enhanced photodynamic therapy (PDT). Biomater Sci. 2017;5(8):1500–11.

    Article  CAS  PubMed  Google Scholar 

  185. Zhou Z, et al. Reactive oxygen species generating systems meeting challenges of photodynamic cancer therapy. Chem Soc Rev. 2016;45(23):6597–626.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Feng L, et al. Multifunctional UCNPs@MnSiO(3)@g-C(3)N(4) nanoplatform: improved ROS generation and reduced glutathione levels for highly efficient photodynamic therapy. Biomater Sci. 2017;5(12):2456–67.

    Article  CAS  PubMed  Google Scholar 

  187. Chae YC, et al. Mitochondrial akt regulation of hypoxic tumor reprogramming. Cancer Cell. 2016;30(2):257–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Liang T, et al. Adapting and remolding: orchestrating tumor microenvironment normalization with photodynamic therapy by size transformable nanoframeworks. Angew Chem Int Ed Engl. 2021;60(20):11464–73.

    Article  CAS  PubMed  Google Scholar 

  189. Shi X, et al. Hemoglobin-mediated biomimetic synthesis of paramagnetic O(2)-evolving theranostic nanoprobes for MR imaging-guided enhanced photodynamic therapy of tumor. Theranostics. 2020;10(25):11607–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Wang S, et al. Synthesis of hemoglobin conjugated polymeric micelle: a ZnPc carrier with oxygen self-compensating ability for photodynamic therapy. Biomacromol. 2015;16(9):2693–700.

    Article  CAS  Google Scholar 

  191. Luo Z, et al. Self-monitoring artificial red cells with sufficient oxygen supply for enhanced photodynamic therapy. Sci Rep. 2016;6:23393.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Cao H, et al. An assembled nanocomplex for improving both therapeutic efficiency and treatment depth in photodynamic therapy. Angew Chem Int Ed Engl. 2018;57(26):7759–63.

    Article  CAS  PubMed  Google Scholar 

  193. Chen Z, et al. Bioinspired hybrid protein oxygen nanocarrier amplified photodynamic therapy for eliciting anti-tumor immunity and abscopal effect. ACS Nano. 2018;12(8):8633–45.

    Article  CAS  PubMed  Google Scholar 

  194. Steeg PS. Tumor metastasis: mechanistic insights and clinical challenges. Nat Med. 2006;12(8):895–904.

    Article  CAS  PubMed  Google Scholar 

  195. Xu T, et al. Enhanced ferroptosis by oxygen-boosted phototherapy based on a 2-in-1 nanoplatform of ferrous hemoglobin for tumor synergistic therapy. ACS Nano. 2020;14(3):3414–25.

    Article  CAS  PubMed  Google Scholar 

  196. Wang H, et al. Discovery of ML210-Based glutathione peroxidase 4 (GPX4) degrader inducing ferroptosis of human cancer cells. Eur J Med Chem. 2023;254:115343.

    Article  CAS  PubMed  Google Scholar 

  197. Hu H, et al. Emerging nanomedicine-enabled/enhanced nanodynamic therapies beyond traditional photodynamics. Adv Mater. 2021;33(12):e2005062.

    Article  PubMed  Google Scholar 

  198. Jiang Q, et al. A hydrogen peroxide economizer for on-demand oxygen production-assisted robust sonodynamic immunotherapy. Theranostics. 2022;12(1):59–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Yin T, et al. Hypoxia-alleviated sonodynamic therapy based on a hybrid protein oxygen carrier to enhance tumor inhibition. Biomater Sci. 2021;10(1):294–305.

    Article  PubMed  Google Scholar 

  200. Pan ZQ, et al. Construction and evaluation of zeolitic imidazolate framework-encapsulated hemoglobin microparticles as oxygen carriers. ACS Appl Bio Mater. 2023;6(4):1471–8.

    Article  CAS  PubMed  Google Scholar 

  201. Liang G, et al. Reduction of platinum(IV) prodrug hemoglobin nanoparticles with deeply penetrating ultrasound radiation for tumor-targeted therapeutically enhanced anticancer therapy. Angew Chem Int Ed. 2023;62(22):e202301074.

    Article  CAS  Google Scholar 

  202. Mortezaee K, Majidpoor J. The impact of hypoxia on extracellular vesicle secretome profile of cancer. Med Oncol. 2023;40(5):128.

    Article  PubMed  Google Scholar 

  203. Wijekoon A, Fountas-Davis N, Leipzig ND. Fluorinated methacrylamide chitosan hydrogel systems as adaptable oxygen carriers for wound healing. Acta Biomater. 2013;9(3):5653–64.

    Article  CAS  PubMed  Google Scholar 

  204. Younis I. Role of oxygen in wound healing. J Wound Care. 2020;29(Sup5b):S4-s10.

    Article  PubMed  Google Scholar 

  205. Gupta S, et al. Dynamic role of oxygen in wound healing: a microbial, immunological, and biochemical perspective. Arch Razi Inst. 2022;77(2):513–23.

    CAS  PubMed  PubMed Central  Google Scholar 

  206. Sen CK. Wound healing essentials: let there be oxygen. Wound Repair Regenerat. 2009;17(1):1–18.

    Article  Google Scholar 

  207. Zhu Y, et al. Roxadustat promotes angiogenesis through HIF-1α/VEGF/VEGFR2 signaling and accelerates cutaneous wound healing in diabetic rats. Wound Repair Regen. 2019;27(4):324–34.

    Article  PubMed  Google Scholar 

  208. Thangarajah H, et al. HIF-1α dysfunction in diabetes. Cell Cycle. 2010;9(1):75–9.

    Article  CAS  PubMed  Google Scholar 

  209. Fosen KM, Thom SR. Hyperbaric oxygen, vasculogenic stem cells, and wound healing. Antioxid Redox Signal. 2014;21(11):1634–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Wang X, et al. In situ 3D bioprinting living photosynthetic scaffolds for autotrophic wound healing. Research. 2022. https://doi.org/10.34133/2022/9794745.

    Article  PubMed  PubMed Central  Google Scholar 

  211. Kang JI, Park KM. Oxygen-supplying syringe to create hyperoxia-inducible hydrogels for in situ tissue regeneration. Biomaterials. 2023;293:121943.

    Article  CAS  PubMed  Google Scholar 

  212. Zhao E, et al. Separable microneedles with photosynthesis-driven oxygen manufactory for diabetic wound healing. ACS Appl Mater Interfaces. 2023;15(6):7725–34.

    Article  CAS  PubMed  Google Scholar 

  213. Chen H, et al. Dissolved oxygen from microalgae-gel patch promotes chronic wound healing in diabetes. Sci Adv. 2020;6(20):eaba4311.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Wang T, et al. Combined antioxidant-antibiotic treatment for effectively healing infected diabetic wounds based on polymer vesicles. ACS Nano. 2021;15(5):9027–38.

    Article  CAS  PubMed  Google Scholar 

  215. Hauck S, et al. Collagen/hyaluronan based hydrogels releasing sulfated hyaluronan improve dermal wound healing in diabetic mice via reducing inflammatory macrophage activity. Bioact Mater. 2021;6(12):4342–59.

    CAS  PubMed  PubMed Central  Google Scholar 

  216. Wang S, et al. Nanoenzyme-reinforced injectable hydrogel for healing diabetic wounds infected with multidrug resistant bacteria. Nano Lett. 2020;20(7):5149–58.

    Article  CAS  PubMed  Google Scholar 

  217. Jia Z, et al. Bioinspired conductive silk microfiber integrated bioelectronic for diagnosis and wound healing in diabetes. Adv Func Mater. 2021;31(19):2010461.

    Article  CAS  Google Scholar 

  218. Chen Y, et al. Research advances in smart responsive-hydrogel dressings with potential clinical diabetic wound healing properties. Mil Med Res. 2023;10(1):37.

    PubMed  PubMed Central  Google Scholar 

  219. Thangarajah H, et al. The molecular basis for impaired hypoxia-induced VEGF expression in diabetic tissues. Proc Natl Acad Sci U S A. 2009;106(32):13505–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Fukui T, et al. Liposome-encapsulated hemoglobin accelerates skin wound healing in diabetic dB/dB mice. Artif Organs. 2017;41(4):319–26.

    Article  CAS  PubMed  Google Scholar 

  221. Rius J, et al. NF-kappaB links innate immunity to the hypoxic response through transcriptional regulation of HIF-1alpha. Nature. 2008;453(7196):807–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Jin L, et al. An NIR photothermal-responsive hybrid hydrogel for enhanced wound healing. Bioact Mater. 2022;16:162–72.

    CAS  PubMed  PubMed Central  Google Scholar 

  223. Peng Y, et al. Construction of heparin-based hydrogel incorporated with Cu5.4O ultrasmall nanozymes for wound healing and inflammation inhibition. Bioact Mater. 2021;6(10):3109–24.

    CAS  PubMed  PubMed Central  Google Scholar 

  224. Waris TS, et al. Chitosan-sodium percarbonate-based hydrogels with sustained oxygen release potential stimulated angiogenesis and accelerated wound healing. J Biomed Mater Res B Appl Biomater. 2023. https://doi.org/10.1002/jbm.b.35344.

    Article  PubMed  Google Scholar 

  225. Yang Z, et al. Oxygen-generating hydrogels as oxygenation therapy for accelerated chronic wound healing. Adv Healthc Mater. 2023. https://doi.org/10.1002/adhm.202302391.

    Article  PubMed  PubMed Central  Google Scholar 

  226. Shiekh PA, Singh A, Kumar A. Oxygen-releasing antioxidant cryogel scaffolds with sustained oxygen delivery for tissue engineering applications. ACS Appl Mater Interfaces. 2018;10(22):18458–69.

    Article  CAS  PubMed  Google Scholar 

  227. Frykberg R, et al. Use of Topical Oxygen Therapy in Wound Healing. J Wound Care. 2023;32(Sup8b):S1-s32.

    Article  PubMed  Google Scholar 

  228. Liu Y, et al. Responsive porous microcarriers with controllable oxygen delivery for wound healing. Small. 2019;15(21):e1901254.

    Article  PubMed  Google Scholar 

  229. Zhang X, et al. Encoded microneedle arrays for detection of skin interstitial fluid biomarkers. Adv Mater. 2019;31(37):e1902825.

    Article  PubMed  Google Scholar 

  230. Li W, et al. Rapidly separable microneedle patch for the sustained release of a contraceptive. Nat Biomed Eng. 2019;3(3):220–9.

    Article  CAS  PubMed  Google Scholar 

  231. Priya S, et al. Concurrent occurrence of lobular capillary haemangioma and port-wine stain: a case report and literature review. Cureus. 2023;15(5):e38642.

    PubMed  PubMed Central  Google Scholar 

  232. Demirel Öğüt N. Acquired plantar port-wine stain (Fegeler syndrome) following prolonged standing. J Cosmet Dermatol. 2022;21(10):5234–5.

    Article  PubMed  Google Scholar 

  233. Jiang F, et al. Influence of port-wine stains on quality of life of children and their parents. Acta Derm Venereol. 2021;101(8):adv00516.

    Article  PubMed  Google Scholar 

  234. Artzi O, et al. Treatment of port wine stain with Tixel-induced rapamycin delivery following pulsed dye laser application. Dermatol Ther. 2020;33(1):e13172.

    Article  CAS  PubMed  Google Scholar 

  235. Sabeti S, et al. Consensus statement for the management and treatment of port-wine birthmarks in sturge-weber syndrome. JAMA Dermatol. 2021;157(1):98–104.

    Article  PubMed  PubMed Central  Google Scholar 

  236. Yang C, et al. Mapping port wine stain in vivo by optical coherence tomography angiography and multi-metric characterization. Opt Express. 2023;31(9):13613–26.

    Article  PubMed  Google Scholar 

  237. Sakai H, et al. O2 release from Hb vesicles evaluated using an artificial, narrow O2-permeable tube: comparison with RBCs and acellular Hbs. Am J Physiol-Heart Circulatory Physiol. 2003;285(6):H2543–51.

    Article  CAS  Google Scholar 

  238. Rikihisa N, et al. Artificial red blood cells as potential photosensitizers in dye laser treatment against port-wine stains. J Funct Biomater. 2017;8(2):14.

    Article  PubMed  PubMed Central  Google Scholar 

  239. Ranjbar A, et al. Machine learning models for predicting pre-eclampsia: a systematic review protocol. BMJ Open. 2023;13(9):e074705.

    Article  PubMed  PubMed Central  Google Scholar 

  240. Davidson KW, et al. Screening for gestational diabetes: us preventive services task force recommendation statement. JAMA. 2021;326(6):531–8.

    Article  PubMed  Google Scholar 

  241. Henderson JT, et al. Preeclampsia screening: evidence report and systematic review for the US preventive services task force. JAMA. 2017;317(16):1668–83.

    Article  PubMed  Google Scholar 

  242. Burton GJ, et al. Rheological and physiological consequences of conversion of the maternal spiral arteries for uteroplacental blood flow during human pregnancy. Placenta. 2009;30(6):473–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Fruci S, et al. Pravastatin for severe preeclampsia with growth restriction: Placental findings and infant follow-up. Eur J Obstet Gynecol Reprod Biol. 2023;283:37–42.

    Article  CAS  PubMed  Google Scholar 

  244. Myatt L, Webster RP. Vascular biology of preeclampsia. J Thromb Haemost. 2009;7(3):375–84.

    Article  CAS  PubMed  Google Scholar 

  245. Powe CE, Levine RJ, Karumanchi SA. Preeclampsia, a disease of the maternal endothelium: the role of antiangiogenic factors and implications for later cardiovascular disease. Circulation. 2011;123(24):2856–69.

    Article  PubMed  Google Scholar 

  246. Cotechini T, et al. Inflammation in rat pregnancy inhibits spiral artery remodeling leading to fetal growth restriction and features of preeclampsia. J Exp Med. 2014;211(1):165–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  247. Kawaguchi AT, et al. Liposome-encapsulated hemoglobin reduces the size of cerebral infarction in the rat. Stroke. 2007;38(5):1626–32.

    Article  CAS  PubMed  Google Scholar 

  248. Chang TMS. Therapeutic applications of polymeric artificial cells. Nat Rev Drug Discovery. 2005;4(3):221–35.

    Article  CAS  PubMed  Google Scholar 

  249. Ohta H, et al. Potential new non-invasive therapy using artificial oxygen carriers for pre-eclampsia. J Funct Biomater. 2017. https://doi.org/10.3390/jfb8030032.

    Article  PubMed  PubMed Central  Google Scholar 

  250. Mysak J, et al. Porphyromonas gingivalis: major periodontopathic pathogen overview. J Immunol Res. 2014;2014:476068.

    Article  PubMed  PubMed Central  Google Scholar 

  251. Yang J, et al. Porphyromonas gingivalis oral infection promote T helper 17/Treg imbalance in the development of atherosclerosis. J Dent Sci. 2017;12(1):60–9.

    Article  PubMed  Google Scholar 

  252. Paramaesvaran M, et al. Porphyrin-Mediated Cell Surface Heme Capture from Hemoglobin by <i>Porphyromonas gingivalis</i>. J Bacteriol. 2003;185(8):2528–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  253. Bai L, et al. Oxyhemoglobin-based nanophotosensitizer for specific and synergistic photothermal and photodynamic therapies against porphyromonas gingivalis oral infection. ACS Biomater Sci Eng. 2023;9(1):485–97.

    Article  CAS  PubMed  Google Scholar 

  254. Mao C, et al. Local photothermal/photodynamic synergistic therapy by disrupting bacterial membrane to accelerate reactive oxygen species permeation and protein leakage. ACS Appl Mater Interfaces. 2019;11(19):17902–14.

    Article  CAS  PubMed  Google Scholar 

  255. Zhang H, et al. Dual-mode antibacterial conjugated polymer nanoparticles for photothermal and photodynamic therapy. Macromol Biosci. 2020;20(2):e1900301.

    Article  PubMed  Google Scholar 

  256. Hu X, et al. Synergistic antibacterial strategy based on photodynamic therapy: Progress and perspectives. Chem Eng J. 2022;450:138129.

    Article  CAS  Google Scholar 

  257. Paciello A, et al. Hemoglobin-conjugated gelatin microsphere as a smart oxygen releasing biomaterial. Adv Healthc Mater. 2016;5(20):2655–66.

    Article  CAS  PubMed  Google Scholar 

  258. Jones GL, et al. Time course and quantification of pancreatic islet revasculariztion following intraportal transplantation. Cell Transplant. 2007;16(5):505–16.

    Article  PubMed  Google Scholar 

  259. Rodriguez-Brotons A, et al. Impact of pancreatic rat islet density on cell survival during hypoxia. J Diabetes Res. 2016;2016:3615286.

    Article  CAS  PubMed  Google Scholar 

  260. Okere B, et al. Cell therapies for pancreatic beta-cell replenishment. Ital J Pediatr. 2016;42(1):62.

    Article  PubMed  PubMed Central  Google Scholar 

  261. Mouré A, et al. Extracellular hemoglobin combined with an O(2) -generating material overcomes O(2) limitation in the bioartificial pancreas. Biotechnol Bioeng. 2019;116(5):1176–89.

    Article  PubMed  Google Scholar 

  262. Le Pape F, et al. Advancement in recombinant protein production using a marine oxygen carrier to enhance oxygen transfer in a CHO-S cell line. Artif Cells Nanomed Biotechnol. 2015;43(3):186–95.

    Article  PubMed  Google Scholar 

  263. Sue C-Y, Tsai N-C. Human powered MEMS-based energy harvest devices. Appl Energy. 2012;93:390–403.

    Article  CAS  Google Scholar 

  264. Zebda A, et al. Mediatorless high-power glucose biofuel cells based on compressed carbon nanotube-enzyme electrodes. Nat Commun. 2011;2:370.

    Article  PubMed  Google Scholar 

  265. Weltin A, et al. Polymer-based, flexible glutamate and lactate microsensors for in vivo applications. Biosens Bioelectron. 2014;61:192–9.

    Article  CAS  PubMed  Google Scholar 

  266. Chen H, et al. In Situ Engineering of Intracellular Hemoglobin for Implantable High-Performance Biofuel Cells. Angew Chem Int Ed Engl. 2019;58(20):6663–8.

    Article  CAS  PubMed  Google Scholar 

  267. Rikihisa N, et al. Artificial red blood cells as potential photosensitizers in dye laser treatment against port-wine stains. J Funct Biomater. 2017. https://doi.org/10.3390/jfb8020014.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are all thankful to all the researchers who are dedicated to Hemoglobin-based oxygen carriers and nano-material research.

Funding

Financial support was supported by the National Natural Science Foundation of China (No. 82170229).

Author information

Authors and Affiliations

Authors

Contributions

K.Z. and L.J.W. analyzed the data and drafted the manuscript. Y.X., X.Y.Z., G.X.Y., Y.Z.C. and Q.W. contributed to the data collection and interpretation. L.Z., H.Z., and G.C. designed this work and critically commented on the manuscript. All authors contributed to the development of the manuscript and approved the final draft.

Corresponding authors

Correspondence to Lian Zhao, Hong Zhou or Gan Chen.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

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://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhu, K., Wang, L., Xiao, Y. et al. Nanomaterial-related hemoglobin-based oxygen carriers, with emphasis on liposome and nano-capsules, for biomedical applications: current status and future perspectives. J Nanobiotechnol 22, 336 (2024). https://doi.org/10.1186/s12951-024-02606-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12951-024-02606-1

Keywords