Next Article in Journal
piggyBac-Based Non-Viral In Vivo Gene Delivery Useful for Production of Genetically Modified Animals and Organs
Next Article in Special Issue
Dendritic Cells Pre-Pulsed with Wilms’ Tumor 1 in Optimized Culture for Cancer Vaccination
Previous Article in Journal
Affinity Effects on the Release of Non-Conventional Antifibrotics from Polymer Depots
Previous Article in Special Issue
Chimeric Antigen Receptor-T-Cell Therapy for B-Cell Hematological Malignancies: An Update of the Pivotal Clinical Trial Data
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Erythrocytes as Carriers: From Drug Delivery to Biosensors

1
Laboratory of Biophysics, Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, Ministry of Healthcare of Russian Federation, Samory Mashela str., 1, GSP-7, Moscow 117198, Russia
2
Laboratory of Physiology and Biophysics of the Cell, Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Srednyaya Kalitnikovskaya, 30, Moscow 109029, Russia
3
Department of Physics, Lomonosov Moscow State University, Leninskie Gory, 1, build. 2, GSP-1, Moscow 119991, Russia
*
Authors to whom correspondence should be addressed.
Pharmaceutics 2020, 12(3), 276; https://doi.org/10.3390/pharmaceutics12030276
Submission received: 25 February 2020 / Revised: 16 March 2020 / Accepted: 16 March 2020 / Published: 18 March 2020
(This article belongs to the Special Issue Cell-Based Drug-Delivery Platforms)

Abstract

:
Drug delivery using natural biological carriers, especially erythrocytes, is a rapidly developing field. Such erythrocytes can act as carriers that prolong the drug’s action due to its gradual release from the carrier; as bioreactors with encapsulated enzymes performing the necessary reactions, while remaining inaccessible to the immune system and plasma proteases; or as a tool for targeted drug delivery to target organs, primarily to cells of the reticuloendothelial system, liver and spleen. To date, erythrocytes have been studied as carriers for a wide range of drugs, such as enzymes, antibiotics, anti-inflammatory, antiviral drugs, etc., and for diagnostic purposes (e.g., magnetic resonance imaging). The review focuses only on drugs loaded inside erythrocytes, defines the main lines of research for erythrocytes with bioactive substances, as well as the advantages and limitations of their application. Particular attention is paid to in vivo studies, opening-up the potential for the clinical use of drugs encapsulated into erythrocytes.

Graphical Abstract

1. Erythrocytes as Drug Carriers

Drug delivery using natural biological carriers is a fast-developing field. Due to the unique biophysical properties, erythrocytes (red blood cells, RBCs) have great potential in this area. RBCs are the largest population of blood cells in mammals. Their main function is oxygen transfer to cells and body tissues [1]. Mature RBCs do not have a cell nucleus and most organelles, but they contain a large amount of a special protein, hemoglobin (Hb), which is able to bind to oxygen. The biconcave shape provides good flexibility and allows the erythrocyte to deform and pass through narrow capillaries. The lifetime of erythrocytes in the bloodstream is 100–120 days, after which they are removed by the spleen. Erythrocytes can be used as carriers in two different ways: by incorporating the drug into the cells or by binding it (using non-specific adsorption or a specific association, involving antibodies or various chemical cross-linking compounds) on the RBCs’ surface. Our review focuses on the first of these methods. The binding of drugs on the surface of RBCs has both advantages and disadvantages. A great contribution to the development of this direction was made by the team of Muzykantov et al. [2,3,4,5,6,7,8,9].
To incorporate the drug into the RBC, the cell must undergo some external influences so that pores can be reversibly formed in its membrane, through which the drug can penetrate. This unique property of RBCs allows to load them with biologically active substances of different molecular weights. For these reasons, erythrocytes are promising biocompatible cells for drug delivery.
The methods for incorporating various substances into red blood cells differ in the way that substances penetrate the cells. The cause of permeability may be the pores’ formation in the cell membrane due to a physical exposure (high voltage electric pulse [10,11] or ultrasound [12]). Drug molecules can also enter the RBCs by endocytosis in the presence of certain chemical compounds (for example, primaquine [13], vinblastine, chlorpromazine, hydrocortisone or tetracaine [14,15]), or using the cell-penetrating peptides bounded to the compound that should be encapsulated [16]. However, the most popular are different variants of osmotic methods.
In some cases, RBCs are first exposed to a hyperosmotic pulse of a low molecular weight substance that penetrates very well through the cell membrane (for example, dimethyl sulfoxide (DMSO) [17,18] or glucose [19,20]). After washing the cells, which decreases the external concentration of these compounds and creates a gradient of their concentration between both sides of the RBC membrane, the target drug is introduced into the external volume. Water with this drug begins to enter into the cells to decrease the osmotic pressure there. The process ends when the gradient of DMSO or glucose disappears. The pores close and part of the drug remains into RBCs. Other, the most popular of the osmotic methods are hypoosmotic. These methods are based on creating a hypotonic environment around RBCs, which causes swelling of the cells and opening pores in the cellular membrane, through which therapeutic compounds can penetrate RBCs. Then, a hypertonic solution is introduced into the cell suspension. The pores close, the cells restore their original size, trapping the drug molecules inside the cell. Osmotic methods are divided into several types. Simple reversible cell lysis in a hypotonic solution by dilutiing a cell suspension with a hypotonic medium causes the formation of erythrocyte ghosts [21,22]. The method of hypotonic pre-swelling is based on the initial controlled cells swelling in a hypotonic solution and their subsequent lysis by adding small portions of an aqueous solution of the drug for encapsulation [23,24,25]. Dialysis methods are based on a reduction of osmolality around the RBCs by a process of dialysis versus hypotonic solution in a dialysis bag [26,27] or in special dialyzers with increased area of contact of RBCs with a buffer solution in the case of flow dialysis [28,29,30]. As mentioned above, hypoosmotic methods are most preferable for incorporation of enzymes into RBCs in terms of efficacy and the properties of obtained cell carriers [31,32].
The history of carrier erythrocytes begins in 1973, when Ihler demonstrated in his article the possibility of incorporating enzymes such as β-glucosidase and β-galactosidase into these cells by reversible hypoosmotic lysis [21]. The analysis of the number of publications (relating only to medications inside the RBCs) shows that interest in this topic since 1973 has not declined, but, instead, has been constantly growing. The number of published articles on the subject of carrier erythrocytes increases every year, and currently, their total number is about 400 (Figure 1).
RBCs for drug delivery have several advantages compared to the existing methods and systems for drug delivery. The erythrocyte is an ideal candidate for such delivery and meets all the requirements for such systems, namely:
-
biocompatibility (human, both autologous and donor erythrocytes are used to treat patients);
-
biodegradability (old or damaged erythrocytes are naturally removed by the reticuloendothelial system);
-
long life in the bloodstream (the drug has an extended lifetime inside the cells because RBCs protect it from the immune system and plasma proteases and the cells survive in the body for a long time; thus, the pharmacokinetics and pharmacodynamics of the drug in RBCs can significantly increase the desired therapeutic effect);
-
decreasing side effects of drugs (due to preventing allergic reactions, and the decrease in the peak concentrations of free drug in the blood to safer levels);
-
ease of cell isolation in large quantities and the ability to scale production.
Carrier erythrocytes (CEs) can be used both in therapy and the diagnosis of some diseases, for example, as carriers of contrast agents for magnetic resonance imaging (MRI) or as biosensors that respond to changes in the concentration of metabolites or pH in the blood [33,34,35]. In therapy, depending on the drug that is loaded, the erythrocytes can be used as carriers with a gradual drug release, as bioreactors or a system for targeted drug delivery, primarily to the reticuloendothelial system (RES), liver and spleen [36]. In the first case, either a drug encapsulated into RBCs can slowly pass through the erythrocyte membrane into the bloodstream, or a membrane-nonpenetrating prodrug is loaded into RBCs, where it turns into a therapeutically effective compound that is able to exit the cell. This ensures prolonged drug circulation in the bloodstream with a decrease in the toxic effects on the body. In the second case, the enzyme encapsulated in erythrocytes works with substrates penetrating the cell membrane. Thus, the enzyme does not directly enter the bloodstream, which solves the problem of its immunogenicity, premature inactivation and increases its half-life.
In this review, we analyzed and organized all existing information on CEs with encapsulated bioactive substances, starting from 1973, that we found in the literature. A summary diagram of their possible use is presented in Figure 2. The most interesting and significant studies in this area are described below. Since there are separate articles in this Issue devoted to a detailed description of the enzymes loaded in RBCs, in our review this subject is considered very briefly (section Erythrocytes-bioreactors). For the most part, only the names of the enzymes that were incorporated into RBCs are listed to ensure the integrity of the review.

2. Erythrocytes-Bioreactors

CEs can operate as bioreactors when the enzyme is incorporated into RBCs. A loaded enzyme can remove the appropriate substrate from the bloodstream, provided that this substrate is able to penetrate into RBCs from the blood. Such erythrocytes-bioreactors (EBRs) open up new possibilities in the treatment of diseases associated with inborn deficiencies of enzymes (enzyme replacement therapy), in the treatment of malignant tumor diseases and for the removal of some toxic compounds from the bloodstream.

2.1. Enzyme Replacement Therapy

Many human diseases are associated with the absence or decrease in the activity of certain enzymes. The logical method for solving this problem is therapy based on the administration of the missing enzyme into the body. However, the injection of free enzyme into the blood, as a rule, is accompanied by the body’s immune response and rapid drug removal from the bloodstream. Incorporating an enzyme into RBCs may be a good solution in this situation. An increase in the blood half-life and a decrease in the body’s immunological reactions to the drug were shown for all enzymes encapsulated in RBCs.
Lysosomal storage diseases [37] (Gaucher disease [38,39], Slay syndrome [40], Fabry disease [41,42] or Pompe disease [43]) are caused by a deficiency of lysosomals enzymes such as of β-glucocerebrosidase (β-glucosidase) [21,26,38,39,44,45], β-glucuronidase [40], α-galactosidase [21] or α-glucosidase [21], respectively. Deficiency of lysosomal enzymes results in the gradual accumulation of their substrates in lysosomes, which ultimately leads to disruption of lysosomes, dysfunction and cell death. The β-glucocerebrosidase enzyme was the first that was incorporated into RBCs for use in enzyme replacement therapy. For an enzyme loaded in RBCs the four–five-fold increase in circulation time was observed [45,46]. Moreover, it was suggested that if RBCs loaded with β-glucocerebrosidase are modified by γ-globulin, then in the body, they must be captured by macrophages and, thus, delivered directly to the focus of the disease—RES cells [45]. Currently, a number of free lysosomal enzymes are used to treat lysosomal storage diseases, but they have high immunogenicity and high cost. Loading appropriate enzymes into RBCs can overcome these limitations and decrease the total cost of treatment.
Other enzymes that have been described for use in enzyme replacement therapy are phenylalanine hydroxylase (for phenylketonuria [47,48,49,50]), adenosine deaminase (for severe combined immunodeficiency with impaired humoral and cellular immune response [27,51,52,53,54,55]) and thymidine phosphorylase (for the treatment of mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) [56,57]). For adenosine deaminase, successful long-term (9 years) use of the adenosine deaminase-loaded RBCs in the clinic has been described [58]. Thymidine phosphorylase and adenosine deaminase encapsulated into RBCs can be used as maintenance therapy before transplantation of allogeneic hematopoietic stem cells. These enzyme preparations are a less expensive alternative to the pegylated forms of the drugs used today.

2.2. Erythrocytes-Bioreactors for Low Molecular Metabolites Utilization

EBRs for removal of low molecular metabolites (ethanol, methanol, cyanide, glucose or ammonium) from bloodstream have been described. These EBRs were based on alcohol dehydrogenase [11,59,60], alcohol oxidase [61], acetaldehyde dehydrogenase [11,62] or alcohol- and acetaldehyde dehydrogenase together [63] in cases of ethanol, methanol and acetaldehyde removal.
Hexokinase and glucose oxidase (both separately and together) were used to remove of excess glucose. In the latter case, this allowed for the rate of glucose consumption in mice to be increased by almost 5.5 times and maintain its normal level for several weeks [64]. Rhodonase (a mitochondrial enzyme responsible for the transformation of cyanide into thiocyanate) was used in the presence of a sulfur donor (sodium thiosulfate or other) for cyanide detoxification [65,66,67,68,69,70]. In mice, it was shown in vivo that erythrocytes loaded with rhodanase in tandem with thiosulfate decreased the blood concentration of cyanide by 40% in 15 min [70].
Moreover, the use of EBRs loaded with asparaginase, methioninease (methionine-γ-lyase) and arginine deiminase for antitumor therapy has been described (see below).

Ammocytes

It would be interesting to go into more detail on the use of EBRs to remove excess ammonium from the bloodstream, since success in this direction has been demonstrated in the work of recent years. An immediate consequence of an ammonium excess in the blood (hyperammonemia) is encephalopathy with the possibility of a lethal outcome. Long-term low-degree hyperammonemia may be associated with neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, etc. [71]. This condition can be caused by both hereditary deficiencies in the enzymes of the uric acid cycle, for example, arginase, and chronic or acute liver diseases. Maintaining low levels of ammonium in the blood is important for treating hyperammonemia and preventing or slowing the development of neurodegenerative diseases. Modern pharmaceutical approaches to reduce the level of ammonium in the blood, unfortunately, do not provide a satisfactory solution to this problem from the point of view of effectiveness and side effects. EBRs for ammonium removal (so-called ammocytes) have been developed by various scientific groups. For enapsulation into RBCs, glutamate dehydrogenase was used, which catalyzed the formation of l-glutamic acid from α-ketoglutarate and ammonium in the presence of NADPH [59,60,72], as well as glutamine synthetase, which catalyzed the formation of l-glutamine from l-glutamic acid and ammonium in the presence of ATP [73,74]. Each of these enzymes was encapsulated into RBCs using reversible hypoosmotic dialysis. However, in vivo experiments showed that such bioreactors effectively removed ammonium from the circulation in mice only in the first 0.5–1 h [72,74]. After this time, the concentration of ammonium in the blood decreased at about the same rate in both experimental and control animals that received dialyzed erythrocytes, but without encapsulated enzymes. Thus, after 0.5–1 h, the loaded enzymes ceased to contribute to the process of ammonium consumption. Using mathematical models of EBRs created in [75], it was shown that the reason for this behavior is the depletion of the substrates inside the cell (l-glutamic acid or α-ketoglutarate), which are consumed during the utilization of ammonium by these enzymes, but are unable to enter the cell from the bloodstream. The authors of [75] proposed a new promising system to create ammonium-removing EBRs, based on the RBCs entrapment of a tandem from two enzymes—glutamate dehydrogenase and alanine aminotransferase. As a result, a new metabolic pathway was created in the erythrocytes, in which α-ketoglutarate and l-glutamic acid were produced and consumed in a cyclic process. Thus, the problem of depletion of these substrates inside the cell was solved, and the system became independent of their transport. The in vivo consumption rate of ammonium in mice for such bioreactors was 2 mmol/(h×lEBRs). Moreover, they continued to work even 2 h after the administration, which distinguished them from the bioreactors described previously in the literature [72,74]. The authors of [75] calculated that under physiological conditions transfusion of 200 mL of such EBRs to a patient will lead to a decrease in the plasma ammonium concentration by 6 mM/day, which is 10 times higher than similar values (600 μM/day) for the best drugs to reduce ammonium concentration currently available.

2.3. Enzymes Used in Antitumor Therapy

l-asparaginase, methioninase and arginine deiminase decrease the blood level of amino acids (asparagine, methionine or arginine, respectively) necessary for cells for biosynthesis during division. This depletion acts more efficiently towards some lines of tumor cells, which cannot synthesize asparagine or arginine on their own (since they do not contain asparagine synthetase [76] or do not express the enzymes necessary for the intermediate stages of arginine synthesis [77,78,79]). Moreover, tumor cells divide much faster than normal ones [80]. In all cases, the encapsulation of enzymes into RBCs may be a suitable alternative to the pegylated forms of these enzymes that are used currently in therapy to increase the half-life and decrease the immunogenicity of these proteins.
ERYTECH Pharma has patented and conducted clinical trial of asparaginase in RBCs (GRASPA) for the treatment of acute lymphoblastic leukemia (ALL) and acute myeloid leukemia [81,82,83], and is currently conducting clinical trials of asparaginase-loaded RBCs (Eryaspase) for the treatment of metastatic pancreatic cancer (trial TRYbeCA-1) [84,85,86] and of triple-negative breast cancer (trial TRYbeCA-2). Eryaspase has proven to be especially effective in pancreatic cancer treatment in combination with chemotherapy [85,86]. Phase 2 clinical trials demonstrated that chemotherapy treatment with Eryaspase reduces the risk of mortality by 40% compared with chemotherapy treatment alone. This is the first case in clinical practice where l-asparaginase therapy has proven effective in treating a solid tumor.
The use of methioninase encapsulated in erythrocytes (erymethioninase) has been demonstrated in vivo in mice with glioblastoma [87] or with breast carcinoma [88]. In both cases, there was a significant decrease in tumor volume, prolonged depletion of methionine and good tolerance of loaded methioninase. In addition, the possibility and effectiveness of a combination of erymethioninase therapy with cancer cell immunotherapy to block the immune control points of PD-1 (anti-PD-1 therapy) was first demonstrated by Sénécha et al. [88]. Significant inhibition of tumor growth was noted and the survival time was increased for erymethioninase therapy in tandem with immunotherapy, compared with each therapy separately.
In 2015, ERYTECH Pharma patented the use of RBCs containing arginine deiminase (ERY-ADI) for the treatment of, in particular, hepatocarcinoma and malignant melanoma [89]. It was shown in mice that the time of arginine depletion (5 days) with ERY-ADI treatment was increased compared to the same time for the free form of ADI (24 h) [90].

2.4. Inositol Hexaphosphate in Erythrocytes

Sickle cell anemia (sickle cell disease, SCD) is a hereditary disease in which anemia develops and RBCs are sickle-shaped. The cause of anemia is the presence in the cells of an altered form of Hb. This is HbS that has an increased tendency to polymerization in capillaries under conditions of partial deoxygenation. Such HbS can polymerize and precipitate inside cells under deoxygenation conditions, forming strands. As a result, the cells acquire a sickle shape and are destroyed [1]. This process may be partially reversible if the cell suspension is re-oxygenated. To improve the condition of patients with SDC, various research groups have proposed modifying donor RBCs for transfusion by incorporating inositol hexaphosphate (IHP) [17,91,92,93,94]. Such CEs do not contain a loaded enzyme, but contain an allosteric effector of the main erythrocyte protein—Hb; therefore, they can also be conditionally called bioreactors. This effector binds to Hb 1000 times stronger than 2,3-dysphosphoglycerate and reduces the affinity of oxygen to Hb, which leads to a two- to three-fold increase in the ability of such erythrocytes to give back bound oxygen.
Bourgeaux et al. proved in in vitro experiments that the addition of erythrocytes loaded with IHP (IHP-RBC) to the blood of patients with SCD was seven times more effective at decreasing the number of sickle cells after deoxygenation and subsequent reoxygenation of the cells compared with the addition of unmodified normal RBCs to this blood [92]. In vivo, in a transgenic mouse model (BERK) that mimics human SCD, four repeated injections of IHP-RBCs were shown to improve overall survival, prevent severe anemia and significantly reduce the risk of vascular occlusion in mice [95]. Thus, in vitro and in vivo studies indicate the therapeutic potential of IHP-RBCs in sickle cell anemia.

3. Carrier Erythrocytes with a Gradual Release of the Pharmacological Agent

An erythrocyte loaded with a pharmacological substance is not necessarily a bioreactor. In some cases, such an RBC can act as a system with a gradual release of the drug into the bloodstream. This approach can be useful when it is necessary to maintain a constant therapeutic drug concentration in the blood for a long time and decrease its peak concentration immediately after drug administration. As a rule, this principle of CEs’ action works when low-molecular-weight substances are loaded in the erythrocyte.

3.1. Cytotoxic Drugs in Erythrocytes

3.1.1. Anthracycline Antibiotics

More than 50 years ago, it was shown that anthracycline antibiotics (daunomycin, doxorubicin) have antitumor activity both for solid tumors and for acute lymphoblastic and myeloid leukemia [96]. Currently, anthracycline antibiotics are used in the complex treatment of many types of cancer. The mechanism of anthracycline antibiotics’ action is the inhibition of topoisomerase II due to the embedding of anthracycline between adjacent pairs of DNA bases, which causes the production of free hydroxyl radicals that adversely affect both the tumor and healthy tissues [97]. Myocardial tissue is particularly affected. The cardiotoxicity of the anthracyclines, which has a cumulative dose-dependent nature, has been shown in many works [98,99,100].
Doxorubicin (adriamycin) is a 14-hydroxidaunorubicin that was isolated from a mutant Streptomyces peucetius (var. Caesius), obtained from a daunorubicin-producing organism, S. Peucetius. Its preclinical therapeutic index was better than that of daunorubicin [101], but the number of side effects did not decrease [102]. Cardiotoxicity caused by anthracycline can be decreased or prevented by a regimen of administration that gives low peak plasma concentrations of the drug; therefore, the search for special carriers of anthracyclines is an urgent task.
Since the 1980s, various groups of authors from the USA, Russia and Japan loaded daunomycin and doxorubicin into RBCs by various methods. Tonetti et al. have shown that encapsulation of daunorubicin into RBCs can be achieved by simple diffusion of the drug through the erythrocyte membrane [103]. In vitro, it has been shown that treatment with glutaraldehyde of RBCs loaded with daunorubicin significantly slows the release of the drug from the cells compared with untreated RBCs. After cells’ incubation for 24 h at 37 °C, the amount of daunorubicin in the cells was 66% and 10% of the initial encapsulated concentration for the treated and untreated cells, respectively. Later, in dogs, in vivo, a significant decrease in the peak plasma concentration of doxorubicin for doxorubicin loaded into RBCs (18.2 ng/mL) compared to its free form (330 ng/mL) was shown, as well as the possibility of targeted drug delivery to the liver [104,105,106].
Pilot studies of RBCs loaded with anthracycline antibiotics in patients with leukemia and lymphomas were carried out by a group of Russian authors (Ataullakhanov et al.). They demonstrated the advantages of the administration of daunorubicin- and doxorubicin-loaded RBCs compared with the administration of the free medicines [107,108,109,110]. Both drugs in the RBCs were clinically effective and were better tolerated by patients. A decreased number of adverse reactions, a significant decrease in cardiotoxicity (with the absence of a cumulative effect), as well as an at least two-fold decrease in the peak concentration of drugs in plasma was observed. The half-life of drugs in the bloodstream was increased. The pharmacokinetics of doxorubicin demonstrates two phases—fast and slow. For the free form of doxorubicin, the concentration of the drug rapidly decreased within 10–30 min after administration, and after 12–24 h the concentration decreased to zero. A similar picture was observed for doxorubicin in erythrocytes in the fast phase, but after the plasma doxorubicin concentration decreased to 0.1 μg/mL, its level remained almost constant up to 3 days [109]. In a recent paper [111], the cardiotoxicity of doxorubicin in carrier erythrocytes obtained by electroporation was studied in healthy mice. All parameters related to cardiac function in mice treated with doxorubicin in erythrocytes were similar to those in the control group of healthy animals that were not injected with the drug, while the same parameters for mice treated with a free form of doxorubicin were significantly worse than in those of the control.
In 2006, a new synthetic anthracycline antibiotic, mitoxantrone, was encapsulated into RBCs. Its effectiveness is higher than that of doxorubicin; however, the use of mitoxantrone is limited by the high cardio- and nephrotoxicity of the drug. In that work, the optimal conditions for the entrapment of the drug in RBCs were selected and the possibility of incorporating sufficiently high doses of mitoxantrone without observing a damaging effect of the drug on the cells was shown. The encapsulation of this antibiotic in RBCs opens-up prospects for its use in clinics [112,113].

3.1.2. Terpene Indole Alkaloids

Vincristine and vinblastine are alkaloids isolated from the plant Cantharanthus roseus G. Don (Vinca rosea Linn.), which show an antitumor and hypoglycemic activity. Their antitumor activity was discovered in the 1960s [114,115,116,117]. The mechanism of the antitumor effect of vincristine and vinblastine is associated with inhibition of microtubule polymerization due to the binding of alkaloids to tubulin. This interferes with cell division (both tumor and normal). Currently, these alkaloids remain the most-used class of anticancer drugs and are important components of standard chemotherapy regimens [118,119]. However, both drugs have a number of serious side effects that limit the possible administered dose [119]. Vinblastine toxicity includes bone marrow suppression (which limits the dose), gastrointestinal toxicity and strong extravasation (leakage of a drug from a vein into surrounding tissues) with the appearance of blisters, deep ulcers and tissue necroses. The main side effects of vincristine are peripheral neuropathy, hyponatremia, leukopenia, thrombocytopenia and hair loss. In addition, both drugs are carcinogenic and mutagenic. Drug resistance to both drugs is also common, which interferes with therapy.
Halahakoon et al. suggested that encapsulating vincristine and vinblastine into RBCs could partially solve the problem of side effects by reducing the peak concentration of drugs in the bloodstream. The authors loaded the drugs into RBCs by hypoosmotic stepwise lysis (pre-swelling) [120]. In vitro experiments showed that vincristine and vinblastine are released from CEs during incubation at 37 °C (in autologous plasma or isotonic buffer) at a rate of 100 μg/h, and about 50% of the drugs are released from CEs after 6 h of incubation [121]. Unfortunately, in vivo experiments are not yet available; therefore, it is difficult to judge the pharmacokinetics and the possible effectiveness of the erythrocyte carriers of these drugs. Currently, the topic of the encapsulation of terpene indole alkaloids in RBCs remains open and little studied.

3.2. Glucocorticoids

Glucocorticoids are steroid hormones synthesized by the adrenal cortex. Since the 1940s, natural and synthetic glucocorticoids have been widely used in various fields of medicine. They have anti-inflammatory, desensitizing, anti-allergic immunosuppressive, anti-shock and anti-toxic effects. However, prolonged use of steroids leads to serious side effects. The most commonly used systemic glucocorticoids are hydrocortisone, prednisolone, methylprednisolone and dexamethasone. These glucocorticoids have good oral bioavailability and are excreted mainly due to liver metabolism and renal excretion [122]. Frequent and high doses of glucocorticoids cause hormonal dependence and serious side effects, such as immune suppression and diabetes [123,124,125,126,127]. Since glucocorticoids are rapidly eliminated from the body (within 3–4 h), the drug should be taken several times a day to maintain the therapeutic dose [122]. A good solution to the problem of the drug’s rapid elimination is to create a carrier that gradually releases the drug into the bloodstream.
EryDel (Italy) created erythrocytes-carriers of glucocorticoids, in particular, with dexamethasone-21-phosphate. In 1997, D’Ascenzo et al. created CEs with the gradual release of dexamethasone and prednisolone by incorporating their prodrugs (dexamethasone-21-phosphate and prednisolone-21-phosphate) into RBCs by hypotonic dialysis. The encapsulation yield was 30% and 28% for dexamethasone-21-phosphate and prednisolone-21-phosphate, respectively. In the cell, dexamethasone-21-phosphate and prednisolone-21-phosphate are dephosphorylated by phosphorylases present in the erythrocyte and are gradually released from the cell by diffusion [128]. Recently, EryDel has conducted numerous clinical trials of dexamethasone-21-phosphate (Dex 21-P) in autologous RBCs (Ery-Dex), which have proven the advantages of using dexamethasone in RBCs over the free form of the drug. To investigate the safety and tolerability of the drug, patients with cystic fibrosis in the first part of the study [129] received increasing doses of ERY-Dex. To evaluate the effectiveness of long-term continuous release of low doses of dexamethasone in the bloodstream, in the second part of the study, nine patients received Ery-Dex at 4-week intervals for 15 months [129]. After repeated injections, a slow and prolonged delivery of dexamethasone into the bloodstream of up to 28 days was observed. It was shown that with prolonged use of Ery-Dex, very low doses of glucocorticoids provide a significant improvement in one of the indicators for diseases of the lungs or bronchi—FEV1. This is the maximum volume of air exhaled in 1 s after the deepest inhalation. There was also a significant reduction in infectious relapses caused by Pseudomonas aeruginosa, and the absence of side effects.
Positive results of the use of Dex 21-P loaded in autologous RBCs have also been shown for steroid-dependent patients of different age groups with Crohn’s disease. The absence of both side effects and the need to take steroid drugs, in addition to patients going into clinical remission were shown [130,131,132]. Similar efficacy (achieving remission and the absence of side effects) was observed in patients with ulcerative colitis [133]. Six-month treatment of patients with steroid-dependent ulcerative colitis using a low dose of Dex 21-P in autologous RBCs allowed for the abolition of oral steroids in most patients without steroid-related side effects, while maintaining clinical remission [133]. EryDel is currently conducting Phase III clinical trials of Ery-Dex (trials.gov NCT02770807) for patients with a rare hereditary disease—ataxia telangiectasia (AT) or Louis–Bar syndrome, for which there is currently no effective treatment. AT is a rare hereditary neurodegenerative disease caused by mutations in the ATM gene (Ataxia Telangiectasia, Mutated), which encodes a protein of the same name, whose role is to coordinate cell signaling pathways in response to double-stranded DNA breaks, oxidative stress and other genotoxic stress [134]. The disease is primarily characterized by cerebellar degeneration, telangiectasia, immunodeficiency, susceptibility to cancer and radiation sensitivity. The results of a Phase II clinical trial for 22 patients with AT who received Ery-Dex for 6 months were published in [135]. They showed good drug tolerance and the possibility of slowing the natural progression of the disease.
In parallel with the development of a new dosage form of dexamethasone, EryDel invented and patented an automatic device, the Red Cell Loader (RCL), which allows small molecules and proteins to be incorporated into RBCs by gradual hypoosmotic swelling of RBCs [136].

3.3. Insulin in Erythrocytes

Few studies have been devoted to the encapsulation of insulin into RBCs, probably because it has been shown that insulin inside RBCs loses activity [137,138]. It was shown that due to inactivation, the percentage of insulin incorporation into RBCs is only 4.8%–6% of the initial amount [138,139,140]. The amount of insulin in the cells can be stabilized, if inhibitors of its degradation are loaded in the RBCs together with insulin. Despite this, there are very little data available. In addition, the work of such cells in vivo has not been investigated [139].
In [140], the half-elimination time of insulin from rabbit bloodstream was compared in the case of intravenous and subcutaneous administration of a free form of porcine insulin or intravenous administration of this insulin loaded in rabbit RBCs. The efficiency of glucose removal using these forms of insulin from the bloodstream of normal and diabetic rabbits was also studied. The plasma half-life for the encapsulated insulin was almost two-times longer than for the free form (12 and 7 min, respectively). The difference between the initial and minimum glucose concentrations achieved during the administration of different forms of insulin was 95 ± 12, 53 ± 11 and 98 ± 19 mg/dL, for normal rabbits, and 304 ± 26, 488 ± 68 and 532 ± 57 mg/dL for rabbits with diabetes for the free form, administered intravenously and subcutaneously, and for insulin in RBCs, respectively. Judging by the data obtained, the entrapment of insulin in RBCs does not offer significant advantages over its free form, but the existing data are insufficient for final conclusions to be drawn.

3.4. Erythrocytes Containing Blood Coagulation Factors

In 1979, Goldsmith et al. studied the possibility of loading coagulation factors IX and X into RBCs [141]. These factors were encapsulated into the RBCs of healthy volunteers and patients with deficiencies of IX and X factors by simple reversible hypoosmotic lysis. Despite the fact that factors IX and X are proteins, the obtained CEs were not bioreactors, since they showed procoagulant activity only after the destruction of the cell membrane and release of coagulation factors into the external environment.
In the work of Sinauridze et al., the pharmacokinetics of free factor IX and factor IX incorporated in autologous RBCs was studied in healthy volunteers [142]. The authors suggested that CEs can maintain a significant level of incorporated factor in plasma due to the natural hemolysis of loaded cells in blood vessels at a low rate. It was shown that encapsulation of factor IX into RBCs prolongs its circulation in the bloodstream by 5–10 times compared with a free factor administered intravenously (t1/2 were 73.9 ± 16 and 8.9 ± 5.6 h, respectively). Despite the fact that erythrocytes loaded with factor IX were safe and circulated in the bloodstream for a long time, their anticoagulant activity was not investigated in this work. Thus, further study of possible therapeutic efficacy of these CEs is needed.

3.5. Morphine Encapsulation into Erythrocytes

To ensure prolonged postoperative analgesia, morphine was incorporated into the autologous RBCs of patients (using the glucose hyperosmotic pulse method). Blood was mixed with a solution of 50% glucose in a ratio of 1:0.5 and incubated for 30 min. Then the RBCs were washed and incubated with a solution of morphine [19,20,143]. In clinical trials in different patients it was found that morphine loaded into RBCs (RBC-M) was able to provide longer analgesic effects than intravenous free-form morphine (M) (24 h for RBC-M vs. 3.2 h for M). However, the observed side effects in patients receiving morphine in these two forms did not differ [144,145,146].

3.6. Nanoparticles and Erythrocytes

Inorganic nanoparticles (NPs), along with RBCs, are increasingly used in medicine, in particular, in the field of drug delivery and diagnostics. NPs have a large surface area per unit volume, and are able to bind to a large number of ligands. This increases their affinity for target molecules. In addition, NPs can have unique optical and magnetic properties that enable magnetic targeting and directional fluorescence imaging of cancer cells in the near-infrared. Artificial nanocarriers (NCs) of a new generation have potential advantages unattainable for RBCs, especially with the development of technologies for the synthesis of NCs. Layer-by-Layer (LbL) technology, which allows obtaining NPs with precisely controlled structure and size using various classes of materials, has become an active area of research [147]. The possibility of precise synthesis control allows designing carriers with the specified almost unlimited properties, functions and geometry (from films to fibers and capsules). The methods for preparing LbL-carriers are different and make it possible to encapsulate various types of molecules, such as antibiotics, growth factors and biosensor substances including hydrophobic compounds with the possibility of controlled release in intravascular and extravascular target-organs [148]. One review [148] discussed the possibility of using LbL technology to create synthetic NCs with encapsulated enzymes. However, the key issue here is the opportunity of transferring the synthesis technology of artificial NCs from the scientific laboratories to the production level for clinical application, since some methods of NCs creating are applicable only for small volumes. Scaling of the production process requires large material and time costs [147,148,149]. Further research in vivo is needed to identify the balance of efficiency/risk ratio and to create a regulatory framework for adjusting the production of artificial NCs. In addition, like other synthetic materials, NPs do not have perfect biocompatibility and biodegradability. They are often rapidly destroyed by macrophages of the immune system, and cannot reach other target organs of therapeutic interest. Injection of artificial carriers can activate the complement system, induce the formation of reactive oxygen species, autophagy, inflammation and other toxic side effects. The review by Parhiz et al. [150] discusses in detail the limitations and undesirable side effects of NCs, including biodegradability and biocompatibility ones. In contrast to synthetic capsules, RBCs are well-studied, can be readily obtained and in many ways represent ideal biocompatible and biodegradable drug carriers for intravascular delivery. A combination of these two delivery systems is a promising approach. In this case, the encapsulation of nanoparticles into RBCs creates a “camouflage” for them against the immune system [151]. One review [152] described, in detail, the types of nanoparticles that were already associated with the surface or loaded inside RBCs, as well as the prospects for their use in antitumor therapy.
Muzykantov’s et al. proposed the promising use of synthetic and natural carriers tandem for the treatment of acute critical diseases such as acute respiratory distress syndrome (ARDS), pulmonary embolism (PE) and acute ischemic stroke. They presented the concept of RBC-hitchhiking (RH), in which NCs (adsorbed on the RBC membrane) are transfered from RBCs to the first organ downstream of the intravascular injection [153]. The authors obtained impressive results: they showed that optimized RH formulations can safely and powerfully target NCs to chosen organs via select placement of intravascular catheters in animals. For example, intravenous injection of RH increases liposome uptake in the first downstream organ (lungs) by ~40-fold compared with free NCs. Injection of RH-nanogels intra-carotid artery delivers >10% injected NCs dose to the brain, approximately 10-fold higher than the best affinity component targeting the brain (transferrin), which only delivered 1% of the injected dose.
Various nanoparticles incorporated with drugs such as doxorubicin [154,155], valproate [156], fazudil [157] and pravastatin [158] and encapsulated into RBCs were tested. Entrapment of fluorescent silicone nanoparticles (SiNPs) with doxorubicin into RBCs allowed for a four-fold increase in the half-elimination time of doxorubicin from the mouse bloodstream (up to 7.31 ± 0.96 h) compared to such particles without RBCs [155]. The literature describes promising examples of the use of erythrocytes loaded with nanoparticles with unique optical properties, such as photostability and strong fluorescence, for in vivo imaging and tumor photodestruction, fluorescence imaging for tumor surgery and photoacoustic imaging [157,159,160,161,162].
Thus, the combination of artificial and natural carriers of drugs extends the application boundaries for both of them. Two drug delivery systems with unique advantages/disadvantages supplement each other, which opens up their new multifunctional capabilities. The use of RBCs for the delivery of artificial NCs significantly increases the efficiency and safety of the latter, which can lead to an increase of the benefit/risk ratio and trigger the expansion of NCs production with access to clinical practice. However, there are limitations of this concept because NPs can affect RBCs, as was shown in [163,164]. Adsorption of NPs onto RBCs can lead to an increase in the RBCs stiffening and sensitize RBCs to damage by osmotic, mechanical and oxidative stress. Therefore, it is important to optimize the composition and properties of NCs (NPs) and to perform a detailed analysis of the modified RBCs for their proper use in tandem. To date, RBCs remain the most attractive system for drug delivery due to their easy preparation, complete biocompatibility and biodegradability and the ability to circulate in the bloodstream for a long time.

4. Erythrocytes for Targeted Drug Delivery

The targeted delivery of drugs using RBCs can be carried out, firstly, to the cells of the RES (macrophages), as well as in the liver and spleen, i.e., in the body cells, that remove old and damaged RBCs. Thus, this approach may be successfully used to treat tumors of these tissues. To deliver the erythrocyte loaded with the drug into these target cells, it must be modified so that the target cells perceive it as being damaged. There are various methods of such modification. All of them lead to a modification of the erythrocyte membrane. This may be the opsonization of RBCs with antibodies to their membrane determinants (for example, by rhesus-antibodies [165]) or the binding of the complement component C3b to them, since there are receptors for the Fc fragment of IgG and for C3b on the cell surface. Treatment of RBCs with calcium ionophore leads to phosphatidylserine exposure on their surface [166], and treatment with glutaraldehyde cross-links the amino groups on the membrane surface, which makes the cell more rigid. Another method is treatment with reagents that cause clustering of the band 3 protein, for example, by a bifunctional amine–amine cross-linking agent, bisulfosuccinimidyl suberate (BS3) in ZnCl2 medium [167,168], which leads to the binding of Hb and proteins of the membrane and fixation of complement components on the cell surface [169]. Inactivation of intracellular hexokinase is also described, which leads to disruption of the cell metabolism and a decrease in the concentration of ATP necessary for cell survival [170].

4.1. Methotrexate

Methotrexate (MTX) is one of the cytostatic preparations (see above). In 1978, Zimmermann et al. were among the first to demonstrate, in mice, the advantage in the distribution of the erythrocytic form of methotrexate (MTX-RBC) in the body over the free form for intravenous administration. The authors encapsulated the drug by electroporation (i.e., created pores in the RBC membrane using an electrical impulse) through which methotrexate (MTX) penetrated the cell. When this form of the drug was administered to mice over 10 min, almost all the methotrexate that was administered in RBCs (0.75–1.0 doses) accumulated in the liver of animals, while in control experiments (with the introduction of the free form of methotrexate), only 0.25–0.3 of the administered dose accumulated [171].
DeLoach and Barton encapsulated methotrexate in erythrocytes by hypoosmotic methods and showed in dogs, in vivo, that in this case, the drug quickly leaves the RBCs. Thirty minutes after the injection of MTX-RBCs into the bloodstream, 50% of methotrexate appeared free in the plasma [172]. To slow the release of the drug from the cells, treatment of carrier erythrocytes with glutaraldehyde was proposed, which provides an additional advantage, since, as was shown in dogs, 50% of CEs treated with glutaraldehyde are rapidly detected in the liver, i.e., targeted delivery of methotrexate to RES occurs [172,173]. Another method for incorporating methotrexate into RBCs uses the pulse of a hyperosmotic glucose solution. In this case, the cells are incubated for 40 min in a 50% glucose solution. Then, they are gently washed and incubated for 30 min with a solution of methotrexate under normal tonicity. The half-life of such CEs with methotrexate was almost 3.5 times longer than for the free form of the drug (13.5 and 3.9 h, respectively) [174]. In addition, the peak plasma concentration of methotrexate after MTX-RBC administration was lower than with free MTX, but it decreased more slowly. A gradual release of the drug from RBCs was observed. In another study [175], N-hydroxysuccinimide biotin ester (NHS-biotin) was bound on the surface of CEs for targeted delivery of MTX-RBCs to the liver. In vivo experiments on rats showed that 1 h after administration of biotinylated MTX-RBCs to animals, 37.2% of biotin appears in the liver, which is almost three times more than after administration of free MTX (11.7%) and almost 1.8 times more than for non-biotinylated cells (20.4%). In an earlier work [176], the same authors modified MTX-RBCs with trypsin (Tt) or phenylhydrazine (PhT) to desialize the cell surface and induce hemichrome in cells, respectively. These two approaches were equally used for the recognition of erythrocytes by macrophages in order to deliver methotrexate for the treatment of RES tumors. Surface-modified erythrocytes loaded with MTX 1 h after administration to animals showed an increased level of methotrexate in the liver compared with the free form of the drug (approximately six times) and with unmodified cells (approximately two times). Phagocytosis by macrophages of surface-treated MTX-loaded erythrocytes was increased by three–five and five–six times for Tt- and PhT-treated CEs, respectively, compared with untreated CEs [176].
The presented examples demonstrate promising possibilities of using erythrocytes for targeted drug delivery to the liver and RES.

4.2. Erythrocytes-Carriers for Treatment of Retroviral Infection

Retroviruses are a family of RNA viruses that primarily infect vertebrates. The most famous and actively studied representative of retroviruses is the human immunodeficiency virus (HIV). Currently, nucleoside analogs, which are inhibitors of reverse transcriptase (after anabolic intracellular phosphorylation), are essential components of highly active antiretroviral therapy (HAART). The most famous of these are azidothymidine (and its analogs), dideoxycytidine and other 2′,3′-dideoxynucleosides [169,177]. Furthermore, the antiviral activity of reduced glutathione (GSH) against RNA and DNA viruses is well known. This activity is realized by interfering with protein-envelope folding and by blocking cell transcriptional factor (NF-kB) activation, which decreases the virus transcription and replication [178,179]. The nucleoside analogs protect lymphocytes, but cannot enter macrophages, while GSH inside specially modified RBCs can be captured by macrophages and protect them against viral infection.
Thus, to treat this immunodeficiency, both CEs containing antiretroviral drugs and CEs containing GSH or GSH + antiretroviral drugs can be used, since GSH-loaded RBCs has been shown to provide significant additional effects compared to monotherapy with antiretroviral drugs (nucleoside analogues) [178].
Since the 1990s, Magnani et al. has been actively developing CEs for the treatment of the human immunodeficiency virus. Since the targets and reservoirs of human immunodeficiency infection are cells of the monocyte/macrophage line, attempts have been made to deliver antiretroviral drugs directly to macrophages to prevent transmission of HIV from already infected macrophages to target lymphocytes [180]. The most popular nucleoside analogues, such as 3’-azido-2’,3’-dideoxythymidine and 2’,3’-dideoxycytidine (ddCTP), were encapsulated into RBCs.
It was shown [169] that for the manifestation of pharmacological activity, dideoxynucleosides must be phosphorylated to 5’-triphosphate by cell kinases. Different types of cells within the same species have different abilities to phosphorylate these compounds. To reduce the toxicity of nucleoside analogues, as well as to overcome the problem of the effectiveness of their phosphorylation, Magnani et al. incorporated ddCTP into RBCs in an active phosphorylated form (by the method of hypoosmotic dialysis). For targeted delivery of such RBCs to macrophages, the loaded cells were treated with a bifunctional amine–amine cross-linking agent BS3 in ZnCl2 medium. This makes the RBCs tougher and induces the binding of autologous immunoglobulin G (IgG) and complement component C3b on the cell surface. Such RBCs are recognized by macrophages and actively phagocitosed. In vitro and in vivo, it was shown that erythrocytes treated in this way loaded with phosphorylated ddCTP were able to significantly reduce typical symptoms of the disease within 3 months [169,181,182,183,184]. The ability to release 3’-azido-2′,3’-deoxythymidine (AZT) from erythrocytes loaded with the azidothymidine derivative di-(thymidine-3’-azido-2’,3’-dideoxy-d-β-riboside)-5’-5’-p1-p2-pyrophosphate (AZTp2AZT) has also been demonstrated in vitro. This prodrug is converted inside erythrocytes into the pharmacologically active AZT by sequential hydrolysis and dephosphorylation [185].
In [179,186,187], interesting results of combination therapy using oral AZT, AZT + DDI (2′,3′-dideoxyinosine) and the additional administration of erythrocytes encapsulated with GSH in each case were demonstrated. The experiments were performed on mice infected by the retrovirus complex (LP-BM5). Studies have shown a decrease in proviral DNA in the brain by about 50% with AZT + DDI treatment and 85% when GSH-loaded RBCs were added to AZT + DDI therapy. For bone marrow, this decrease was about 37% and 60%, respectively [187]. The addition of GSH-loaded RBCs to AZT monotherapy decreased proviral DNA in bone marrow by 60% [186].
RBCs encapsulated with fludarabine have become another possible approach for treating HIV-1 infection. As mentioned above, long-living macrophages in the infected body are the reservoir for the HIV-1 virus. It was shown that chronic infection of human macrophages with this virus increases the expression and phosphorylation of the protein STAT1, which is included in the regulation of many macrophage functions, including cell growth and proliferation [188]. The nucleoside analogue of 9-(β-d-arabinofuranosyl)-2-fluoroadenin-5’-monophosphate (FaraAMP, fludarabine) is active against STAT1-expressing cells and, in culture, is able to kill HIV-infected macrophages, but not uninfected cells. To direct fludarabine to macrophages, it was encapsulated into RBCs, which were then processed by the method described in [169], which causes clustering of the band 3 protein. The final concentration of fludarabine in macrophages after a single 18-h exposure with erythrocytes loaded with fludarabine was estimated at 10–20 μM. In that study, a powerful (>98%) and long-lasting (at least 4 weeks) effect of inhibiting the release of the virus from HIV-infected macrophages was obtained [189].

4.3. Drugs Loaded into RBCs for the Treatment of Hepatitis C

To enhance the effectiveness of the therapeutic effect of drugs used in the treatment of hepatitis C, and to minimize their side effects associated with an increase in the dose of drugs, Skorokhod et al. were searching for new ways to simultaneously deliver interferon (INF-α) and ribavirin (RIBA) to the liver [189]. Both drugs were loaded into human RBCs (RBCs-INF-α-RIBA) by the method of hypoosmotic reversible lysis. Cells were opsonized for targeted delivery to macrophages and liver. The entrapment efficiency was 40%. It was shown that RBCs-INF-α-RIBA were stored for up to 3 days at 4 °C without loss of antiviral activity. In vitro, monocyte activation by RBCs-INF-α-RIBA was also demonstrated, as well as the induction of surface receptors of the major histocompatibility complex type II (MHC class II) and Fc receptors that activate cell phagocytic activity. The authors argue that encapsulating INF-α and RIBA into RBCs and targeting the liver helps: (1) to release large amounts of INF-α and achieve higher therapeutically effective concentrations in the liver; (2) to induce autocrine stimulation of macrophages of the liver (and spleen) using INF-α to enhance cellular antiviral protection; (3) to control viral proliferation in macrophages. In this regard, it is advisable to further study a potentially therapeutically effective system in animals.
Forezesh and Zarrin proposed encapsulating a more modern hepatitis C drug, boceprevir, into RBCs in addition to interferon and ribavirin [190].

4.4. Macrophage Depletion

It is known that macrophages play an important role in the regulation of numerous biological processes in the body. In addition, it has been repeatedly shown that macrophages contribute to the development of pathologies such as autoimmune hemolytic anemia, immunothrombocytopenia, rheumatoid arthritis and sepsis, and play a key role in the spread of viruses in HIV infections [191]. Tumor-associated macrophages create favorable conditions for cancer progression, promoting angiogenesis and metastasis [192,193,194]. Rossi et al. studied the possibility of temporary depletion of macrophages by incorporating bisphosphonates (clodronate, zoledronate) into RBCs and the targeted delivery of such carriers to macrophages. They showed that RBCs loaded with zoledronate are able to deplete macrophages both in vitro and in vivo [160]. Balb/C mice were injected with 59 mg/mouse of zoledronate encapsulated into RBCs. For targeted delivery to macrophages, loaded erythrocytes were incubated in medium with BS3 and ZnCl2. After a single injection of encapsuled erythrocytes, macrophage depletion was 29% and 67% for liver and spleen macrophages, respectively.
Another study evaluated the effect of macrophage depletion to prevent Langerhans islet cell allograft rejection in diabetes mice [195]. Graft survival was 19–20 days for control groups of mice receiving unloaded erythrocytes or saline, 25 days for mice receiving free clodronate and 35 days for mice receiving clodronate in RBCs.

4.5. Antigens Loaded into Erythrocytes or Associated with Their Surface

4.5.1. Immunization

Binding antigens to the surface or encapsulating them inside the carrier erythrocytes opens up new possibilities for using such erythrocytes for immunization as an alternative to adjuvants (substances that adsorb antigen on their surface), namely, the possibility of delivering antigens directly to the immune system into antigen-presenting cells—macrophages or dendritic cells (DCs). Dendritic cells are believed to be most effective in initiating antigen-specific responses, but macrophages are also able to facilitate the presentation of peptides to T lymphocytes [196]. Magnani et al. has repeatedly shown that protein antigens (bovine serum albumin, porcine liver uricase, yeast hexokinase) and glycoproteins B of herpes simplex virus type 1 (HSV-I), which are associated with the surface of autologous RBCs via the biotin–avidin–biotin bridges, induce a higher immunological response (higher antibody levels) in mice than the response obtained using Freund’s adjuvant, which is often used in immunization [197,198]. Later, it was shown that the HIV-1 Tat protein, linked through the biotin–avidin–biotin bridges to the erythrocyte surface (RBC-Tat), has immunotherapeutic potential. This protein is important for virus replication and infectious activity (the presence of antibodies against Tat correlate with slower progression of the disease). Tat protein is immunogenic [199]. Erythrocytes associated with Tat (RBCs-Tat), in amounts 250 times less than the amount of soluble Tat in Freund’s adjuvant, are capable of eliciting specific responses of anti-Tat T killers. Moreover, the production of Tat neutralizing antibodies was observed in six out of six mice, in contrast to two out of six mice for Tat in Freund’s adjuvant.
In other works [200,201], using bacterial toxoids, proteins and enzymes as antigens, it was shown that immunization is also possible by encapsulating antigen in RBCs. In B6D2F1 and Balb/C mice, the total titers of specific antibodies (binding, lysing and neutralizing the antigens) and only neutralizing antibodies against introduced antigens were several times higher during immunization with antigens loaded into RBCs than after immunization with free forms of antigens [200].

4.5.2. Cancer Immunotherapy

Cancer immunotherapy is the use of the immune system to kill tumor cells that have specific tumor-associated antigens (TAA) [202]. Banz et al. proposed a strategy for using RBCs loaded with tumor-associated antigens in cancer immunotherapy. Immunization against TAA induces TAA-specific cytotoxic T lymphocytes (CTLs), which are capable of controlling tumor growth. Efficient and targeted delivery of TAA in vivo to DCs can be effective in tumor immunotherapy since it induces strong CTLs responses against the tumor [203]. It was shown in mice [204] that erythrocytes bearing an antigen (in this case, ovalbumin) in combination with polyinosine–polycytidylic acid (Poly (I:C)) introduced intravenously, can be effectively captured by antigen-presenting cells (APC). This causes antigen-specific responses of CD4+ and CD8+ T cells, which are able to induce in vivo ovalbumin-specific cell lysis even 30 days after CEs administration. Ovalbumin was loaded into RBCs by hypoosmotic dialysis (RBC-OVA). To enhance the phagocytosis of these erythrocytes with antigen-presenting cells, they were treated externally with antibodies (anti-TER119 mAb), and then were administered to C57BL/6 mice intravenously. RBC-OVA was mixed with Poly (I:C) before injection to enhance the induction of T-cell responses, as Poly (I:C) is a toll-like receptor III ligand that activates the CD4+ T cell response specific for alloantigen of RBCs [205,206]. Ninety minutes after injection of RBC-OVA + Poly (I:C) to mice, phagocytosis of the introduced RBCs by antigen-presenting macrophages and dendritic cells was observed.
The effectiveness of such a tumor-associated antigen delivery system was also demonstrated in two models of mice with melanoma [207]. The artificial ovalbumin antigen or tyrosinase 2 protein antigen (TRP-2) was encapsuled into red blood cells and tested on E.G7-OVA and B16F10 tumor models, respectively. The administration of a small amount of tumor-associated antigen (TRP-2) loaded into RBCs treated with antigen anti-TER119 in combination with Poly (I:C) caused an antigen-specific T-cell response and tumor growth control in mice, whereas the same amount of free TRP-2 did not cause a similar response.

4.5.3. Induction of Immune Tolerance

The opposite of immunization is the stimulation of immune tolerance, that is, the “training” of the immune system to create tolerance (resistance) to a particular antigen in order to prevent its attack. Such stimulation can be used in autoimmune diseases, when the immune system attacks its own antigens, during an allograft transplant, or in case of an allergy to a drug used in therapy. The induction of immune tolerance is often carried out using molecules that inhibit the immune system, such as rituximab (anti-CD20 monoclonal antibody), cyclophosphamide, and methotrexate, or by depleting B cells necessary for the immune response. In [208], the authors proposed the use of erythrocytes for the induction of immune tolerance. The drug, to which it was necessary to induce immune tolerance, was loaded into RBCs. That study showed that the drug inside the cell-carriers does not interact directly with antibodies, which may be present in plasma. The authors investigated the possibility of obtaining immune tolerance in mice for the enzyme alglucosidase α (AGA), a recombinant analogue of acidic α-glucosidase, which is currently used in enzyme replacement therapy for Pompe disease (glycogen storage disease caused by α-glucosidase deficiency). For targeted delivery of the drug to antigen-presenting cells of the liver and spleen, the erythrocytes loaded with the enzyme were treated with BS3/ZnCl2.
As mentioned above, therapy for Pompe disease is carried out by frequent intravenous administration of AGA, which ultimately causes a stable humoral response and leads to the need to discontinue treatment. This work showed that erythrocytes encapsulated with AGA and then BS3/ZnCl2-treated have tolerogenic properties, i.e., they are able to eliminate the humoral response to AGA and restore tolerance to replacement therapy. First, the mice were injected intravenously with AGA-loaded RBCs (three times) and then they were sensitized to AGA using different adjuvant molecules. Control animals received free AGA instead of the encapsulated molecules. A strong decrease in the specific humoral response was observed in the experimental group one-week after treatment with AGA-loaded RBCs. This effect was maintained for at least two months without affecting the overall immune response [208].
The effectiveness of the induction of immune tolerance depends on several factors, such as the route of administration and the dose of antigen (Ag), as well as the type of target antigen-presenting cells [209]. DCs and macrophages ingest foreign antigens and present fragments of these antigens on their own surface for recognition by T cells, and thereby, participate both in the induction of immunity and in the stimulation of its tolerance. After B or T cells recognize Ag on the APC surface, the choice between tolerance and immunity depends on the amount and type of Ag, type of APC and the number of co-stimulation molecules CD80 and CD86 (which bind to the CD28 receptor on the membrane of T-lymphocytes) on the DCs’ surface. The maturation status of DCs is a key factor in the development of immunity or the induction of tolerance. Mature DCs induce immunity, while immature DCs induce tolerance, since they are capable of expressing low levels of MHC class II surface antigens and costimulatory molecules, which are necessary for the antigen presentation to T-lymphocytes [210]. The presentation of antigen to T-lymphocytes, in turn, stimulates the differentiation of immature T-lymphocytes into cytotoxic CD8+ cells or CD4+ helper cells. The liver plays an important role in the induction of tolerance due to its specific composition of antigen-presenting cells. Liver DCs have an immature phenotype and, therefore, are not able to elicit an Ag-specific T-cell response, but induce the development of T-cell tolerance. Several subpopulations of DCs of the spleen are also involved in the induction of tolerance [211]. Thus, the delivery of Ag to the corresponding DCs of the liver and spleen is an attractive strategy for the induction of specific antigen tolerance.
An example is the work of Cremel et al., which demonstrated the possibility of inducing immune tolerance in mice by administration of RBCs loaded with ovalbumin (OVA) as antigen and treated with calcium ionophore or BS3 [209]. It was shown that intravenous injection of such erythrocytes into mice sensitized to ovalbumin caused a strong decrease in specific humoral and cellular immune responses (the appearance of 19%–22% of activated OVA-specific CD8+ T cells vs. 58%–64% for mice without the induction of immune tolerance). Such a response was observed during, at least, 34 days after the induction of tolerance and was antigen-specific, without causing complete suppression of the immune system.
ERYTECH Pharma has patented both methods of using erythrocytes as carriers of antigens—in cancer immunotherapy to stimulate a cytotoxic cell response directed against tumor cells expressing an antigen [212], and as a system that induces a specific immune tolerance to enzymes, which are used in enzyme-replacement therapy of diseases such as, for example, Pompe disease, Fabry disease, mucopolysaccharidosis, hemophilia A and B, rheumatoid arthritis, multiple sclerosis, etc., requiring stimulation of the immune tolerance to achieve a therapeutic effect [213].

5. Carrier Erythrocytes in the Diagnostics

5.1. Contrast Agents in Magnetic Resonance Imaging

MRI is a non-invasive method for visualizing the structure and function of tissues, which is widely used in clinical practice. Despite the fact that MRI allows high-resolution anatomical images to be obtained, the possibilities of this method can be significantly expanded with the help of contrast agents. They are used to improve the differentiation of malignant and healthy tissues [214], as well as for MR angiography, which reveals damage to blood vessels, primarily myocardial damage, atherosclerosis, thrombosis, aneurysms and other vascular diseases [215]. The localized interaction of contrasting agents with protons of water molecules in various tissues creates a contrast by decreasing the time of their longitudinal (T1) and transverse (T2) relaxation (the time during which the protons return to their equilibrium state after exposure to an electromagnetic pulse). This relaxation is different in healthy and pathological tissues, and depends on the surrounding molecules and atoms. Based on this difference, MRI images are constructed. Paramagnetic and superparamagnetic contrasting agents increase relaxation rates (1/T), thereby enhancing contrast. A measure of the sensitivity of the contrast agent is its longitudinal and transverse relaxivity (r1 and r2, respectively), which show how the corresponding relaxation rate changes as the concentration of the contrast agent changes (C) (see Equations (1) and (2)):
1/T = r × C
r = 1/(T × C)
Various metal derivatives, primarily gadolinium oxides, chelate complexes of lanthanides and gold nanoparticles, as well as superparamagnetic iron oxide nanoparticles (SPIO) and ultrafine superparamagnetic iron oxide nanoparticles (USPIO), can be used as contrasting agents. However, the use of these nanoparticles in MR angiography is limited, since the surface of nanoparticles undergoes opsonization upon intravenous administration, i.e., it adsorbs plasma proteins. This stimulates and facilitates phagocytosis of these particles, so that their half-life in blood is 1–3 h (a decrease in the size of nanoparticles increases the half-life), and the time interval for observation after bolus administration of the drug is only a few minutes [216,217,218,219,220]. To date, as a result of these reasons, many SPIO and USPIO preparations in Europe and the USA are practically not used [221,222]. On the other hand, due to the selective uptake and accumulation in RES cells, superparamagnetic iron oxide nanoparticles have become very popular for imaging the liver and spleen [223,224].
In 2008, Antonelli et al. proposed the encapsulation of magnetic nanoparticles based on iron oxide in RBCs in order to increase their lifetime in circulation [225]. This group has a large number of works devoted to the study of the properties of various magnetic nanoparticles based on iron oxide, both newly created and commercially known, such as Resovist (Bayer Schering Pharma), Sinerem and Endorem (Guerbet, France) etc., loaded into RBCs [222,225,226,227,228]. It has been shown that not all iron oxide-based nanoparticles can be successfully incorporated into erythrocytes. The result depends on properties of nanoparticles, such as their size, nature of the dispersant and surface charge, that are important to obtain monodispersed nanoparticles in suspension, as well as on the chemical properties of particle surface coating [197]. On the other hand, SPIO encapsulation in RBCs can increase the circulation time of these particles in the bloodstream by up to 12 days, which makes it possible to use them in MR angiography for long-term imaging and long-term monitoring of cardiovascular diseases [229].
It was demonstrated in [230] that the encapsulation of USPIO nanoparticles into RBCs leads to an increase in their transverse relaxivity r2 and a very high ratio of relaxivities r2/r1, which makes them promising for use as a negative contrast agent in the blood pool. Other studies have also demonstrated the advantages of using carrier erythrocytes rather than suspensions for gadolinium oxide nanoparticles [231], chelate complexes of lanthanides [232] and gold nanoparticles [233] as contrasting agents for MRI.

5.2. Blood Analyte Biosensors

For long-term non-invasive in vivo monitoring of certain blood parameters (analytes), such as glucose concentration or pH, Ritter et al. proposed the use of RBCs loaded with a fluorescent dye that responds to changes in the concentration of an analyte in the bloodstream [33,234,235,236]. In this case, autologous RBCs encapsulated with fluorescent dyes (sensors) are introduced into the patient’s circulation for analytical monitoring. The fluorescent signal of the erythrosensors can be excited and detected non-invasively through the skin when excited by an external light source in the visible wavelength range (for example, a laser diode). It was shown in [34] that erythrocytes loaded with fluorescein isothiocyanate (FITC), a pH-sensitive fluorescent dye, have an excellent ability to reversibly monitor in vitro pH in the physiological range with a resolution of up to 0.014 pH units. According to the authors, the fluorescence intensity increases with increasing extracellular pH, since RBCs quickly balance pH with the external environment through a chloride–bicarbonate exchanger. However, it turned out that for pH measurements in vivo in the physiological range, the sensitivity of such a system is too low. Thus, the next step to facilitate the use of RBCs as biosensor carriers should be the development a fluorescent sensor with higher sensitivity and optimization of RBCs loading to obtain a higher signal level [35].

6. A Novel Trend in the Use of Red Blood Cells as a Delivery System

To use erythrocytes to deliver drug compounds, these compounds must be loaded into cells. There are many different loading procedures, which have been developed for a long time and continue to improve. Most often for this the RBC membrane is subjected to certain physical influences. Despite the fact that the process is carried out under conditions which spare the cell, such procedures, of course, reduce the quality of the resulting loaded cells compared to the original erythrocytes [237]. In addition, the effectiveness of the encapsulation of a protein depends on its size and other physical properties, and is far from being always sufficient. Against this background, the newest trend of using RBCs as carriers of certain enzymes looks very interesting.
RubiusTherapeutics (Boston, USA) combined the successes of genetic engineering and the unique properties of RBCs by developing a new class of cell drugs, which they called Red Cell Therapeutics™ (RCTs) [238]. RCTs are allogeneic erythrocytes that express targeted biotherapeutic proteins (enzymes) inside or on the surface of the cell. To obtain such RCTs, allogeneic hematopoetic progenitor cells (CD34+) are first genetically modified using a gene cassette or lentiviral vector to provide expression of one or more targeted therapeutic proteins. The converted cells are placed in a bioreactor for their further maturation up to reticulocytes. The resulting cells have the same characteristics as normal RBCs and contain, inside or on their surface, the target therapeutic protein for the treatment of the suspected disease. Such RCTs can be used in enzyme-replacement and anticancer therapy (cancer immunotherapy), as well as in the treatment of autoimmune diseases. Currently, the first phase of clinical trials of RTX-134, erythrocytes carrying the AvPAL gene inside cells (the phenylalanine-ammonia lyase gene Anabaena variabilis), is being conducted to treat adult phenylketonuria (NCT04110496) [239]. At conferences in Philadelphia and Boston in 2019, Zhang [240] and Moore [241] proposed interesting ideas for creating artificial antigen-presenting cells, the genetically modified erythrocytes (RCT-aAPC), which expresses immunomodulating signals that are directed against the tumor. Such cells, on the one hand, are loaded with tumor-specific antigen and costimulatory molecules, and, on the other hand, express proteins of the main histocompatibility class I complex on the surface to create an effective tumor-specific T-cell response. Using this strategy in mice showed 60% inhibition of tumor growth on day 7 after administration of RCT-aAPC to animals.
Thus, RubiusTherapeutics technology represents a new promising approach for the delivery of therapeutic substances to patients using erythrocytes. These results are especially encouraging in light of the fact that, in 2017, a method was developed to create an “immortal” line of erythrocytes from the corresponding erythrocyte precursors [242].
If you have a culture of unipotent erythrocyte precursors, you do not need to worry about managing their differentiation. However, unlike stem cells, the number of divisions of such cells is limited; thus, they must be immortalized, i.e., modified so that their division can be endless. For this, bone marrow, cells were genetically modified by adding a human papilloma virus gene to them, which allows cells to divide unlimitedly. Then, the transition of the modified cells into erythrocyte precursor cells was induced. Thus, a new cell line, BEL-A (Bristol Erythroid Line Adult), was created. The course of these cells’ differentiation did not differ from the corresponding stages of development of pluripotent stem cells. The results obtained appear promising for the possibility of scaling the process to obtain the desired RBCs in sufficient quantities.

7. Limitations of the RBCs’ Use as Drug Carriers

Despite the fact that RBCs are very promising for use as drug carriers, their use has a number of limitations. The source of RBCs is blood; thus, the use of allogeneic blood can lead to errors in choosing the right blood type and to the transmission of various infections. However, these disadvantages are common to all transfusion of blood products. These situations are very rare, and currently they are not the principal barrier to transfusion of any blood products, including erythrocytes loaded with drugs. In addition, production of carrier erythrocytes are associated with the need for sterile work and the complexity of the large-scale production of such cells. Creating automatic devices can solves these problems. Another disadvantage is related to the fact that if any crude method was used for CEs preparation, the quality of the resulting cells may not be high enough. In this case, these CEs will rapidly degrade in the bloodstream, and the drug may be released uncontrollably. This complicates drug delivery and can lead to adverse side effects. However, the methods currently used are soft enough and do not have a strong effect on RBCs.
There are also other restrictions. The first of them is that far from any substance can be incorporated into RBCs. Some low molecular weight compounds that easily pass through the erythrocyte membrane are not only easy to enter, but also just as easy to leave the cells, which makes it impossible to create a long-term depot form of these compounds based on RBCs in the bloodstream [82,94,140]. To slow the release of such substances from RBCs, the cells may be treated with different crosslinking agents (primarily for NH2– or HS– groups on the membrane surface). This may be glutaraldehyde, BS3, etc. [166,167,168,169]. However, although this slows the release of drug compounds from the cells, the membrane of such erythrocytes changes so much that they are quickly recognized by RES cells and removed from the bloodstream. Another way to retain a therapeutically effective substance that easily passes through the erythrocyte membrane inside the cell is to encapsulate a prodrug in the erythrocytes, for example, a phosphorylated form of this compound, which cannot pass through the cell membrane but can be dephosphorylated by phosphatases of RBCs, turning it into a therapeutically active substance that gradually leaves the cells. The opposite situation is also possible when for activation, the substance must be phosphorylated inside the erythrocyte by the corresponding erythrocyte phosphokinases (as in the case of dideoxynucleotides [169]). In all these cases, the limitation of the use of RBCs as drug carriers is that the activity of the desired enzymes in the cells of different patients can vary greatly, which does not allow to obtain stable results [243].
If RBCs are supposed to be used as bioreactors, then in a number of cases a second serious limitation arises. This is due to the possible effect of the loaded enzymes on the erythrocyte metabolism, primarily glycolysis. This overlap can lead to depletion of the pools of some metabolites (for example, NAD(P) and NAD(P)H) if they are used simultaneously by glycolysis and enzymes built into RBCs. In this case, a stationary state can be lost in glycolysis, which leads to rapid cell death in the bloodstream (Protasov et al., unpublished data). A possible way to deal with this situation may be to calculate the permissible doses of the loaded enzymes, which do not yet lead to the loss of a stationary state in glycolysis (using mathematical models). Moreover, it is possible to encapsulate the necessary cofactor and the target enzyme into RBCs together (provided that cofactor cannot quickly leave the cell). Sometimes, the work of the enzyme inside the RBCs may be limited by the rate of transport for the necessary substrate of the reaction into the cells. This happened, for example, when ammocytes based on glutamate dehydrogenase [59,60,72] or glutamine synthetase [73,74] were created. In this case, the researchers proposed for incorporation into the RBCs a new enzyme system consisting of two enzymes that provided cyclic consumption and production of the necessary metabolites inside the cell. This made the process independent of the transport of these metabolites [75]. Another area of modern developments to improve the delivery of drugs that can affect the metabolism of RBCs is associated with the replacement of RBCs with artificial RBCs or hybrid nanoparticles, the surface of which contains fragments of the RBC membrane, to ensure their long lifetime in the bloodstream [9]. However, these are only scientific developments, which are far from clinical use.
Thus, there are real restrictions on the use of RBCs as drug carriers; however, they can be circumvented in many cases, both by improving experimental methods of work, and by using mathematical models of CEs to properly account for the effects of the loaded compounds on RBCs metabolism.

8. Conclusions

Drug delivery using natural biological carriers is a fast-developing field. Due to their unique biophysical properties, erythrocytes have great potential in this area. Recently, their use has been increasingly expanding both in therapy and in the diagnosis of many diseases. The use of carrier RBCs is very important to prevent unwanted immune responses after the introduction of protein molecules, especially if repeated administration of these drugs is required. RBCs are able to provide the necessary protection for the protein preparation from the immune system and plasma proteases, increasing the lifetime of the drug in the bloodstream, and thereby enhancing its therapeutic effect. In addition, special processing of the membrane of encapsulated RBCs allows targeted delivery of drug-loaded cells to macrophages, dendritic cells, liver and spleen, which is also increasingly used in various fields of medicine. In the case of a number of cytotoxic drugs, the greatest gain when loading the drug into RBCs is achieved due to the fact that, as has been proven, RBCs allow the prolongation of a drug’s therapeutical effect due to its gradual release into the bloodstream. Simultaneously, reducing the peak concentration of free drug in plasma is achieved during administration, which is associated with a decrease in negative side effects, such as cardiotoxicity, with the introduction of anthracycline antibiotics. In Table 1, we collected the drugs and substances encapsulated into RBCs since 1973.
Despite such positive properties and the widespread popularity of carrier erythrocytes in scientific research, only a few drugs loaded into RBCs have now reached clinical use. Perhaps this is due to the complexity of scaling the production of such drugs, since therapy with RBCs incorporated with drugs is more likely to be personalized medicine and requires an individual approach. However, there are two companies that have surpassed all the barriers and are actively promoting this method of drug delivery in clinical practice. These are ERYTECH Pharma (France) and EryDel (Italy). ERYTECH is conducting final clinical trials of erythrocytes loaded with asparaginase (Eryaspase) for the treatment of pancreatic cancer and triple-negative breast cancer [244]. Methionine-γ-lyase loaded into RBCs (erymethionase) for the treatment of solid tumors and the encapsulation of enzymes in RBCs for replacement enzyme therapy and of antibodies for cancer immunotherapy are under development and in preclinical trials.
EryDel, in turn, focused on clinical trials of dexamethasone (EryDex) for the treatment of ataxia telangiestasia [245]. A device developed by EdyDel was also used to prepare thymidine phosphorylase in erythrocytes (EE-TP) for the treatment of mitochondrial neurogastrointestinal encephalomyopathy [246].
Erythrocytes encapsulated with phenylalanine-ammonia lyase for the treatment of phenylketonuria, recombinant uricase for the utilization of uric acid and guanidine methyltransferase for enzyme replacement therapy are currently at the preclinical stage. The European Medical Agency has already granted the status of orphan drugs to dexamethasone phosphate for the treatment of cystic fibrosis [247] and to l-asparaginase for the treatment of pancreatic cancer [248] and acute lymphoblastic leukemia [249].
Thus, it can be expected that in the near future, the carrier erythrocytes of drugs will be widely used, particularly in enzyme replacement and antitumor therapy.

Author Contributions

L.K., E.B., F.A. and E.S. contributed to the conception and structuring of the present review, to the literature search and to the writing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The study was supported by the Ministry of Science and Higher Education of the Russian Federation (projects AAAA-A17-117112420087-0 and AAAA-A19-119111690006-9), as well as by the grants of Russian Foundation for Basic Research for graduate students of the second year, project number N 19-34-90035 and N 19-34-90036 (for L.K. and E.B., respectively) and a grant of the Presidium of the Russian Academy of Sciences, the Program for fundamental research “Fundamentals of the technology of physiological adaptations”.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ataullakhanov, F.I.; Borsakova, D.V.; Protasov, E.S.; Sinauridze, E.I.; Zeynalov, A.M. Erythrocyte: A bag with hemoglobin, or a living active cell? Pediatrics Hematol. Oncol. Immunopathol. 2018, 17, 108–116, (In Russian, abstract in English). [Google Scholar] [CrossRef]
  2. Muzykantov, V.R.; Murciano, J.C.; Taylor, R.P.; Atochina, E.N.; Herraez, A. Regulation of the complement-mediated elimination of red blood cells modified with biotin and streptavidin. Anal. Biochem. 1996, 241, 109–119. [Google Scholar] [CrossRef] [PubMed]
  3. Muzykantov, V.R.; Zaltsman, A.B.; Smirnov, M.D.; Samokhin, G.P.; Morgan, B.P. Target-sensitive immunoerythrocytes: Interaction of biotinylated red blood cells with immobilized avidin induces their lysis by complement. Biochim. Biophys. Acta (Biomembranes) 1996, 1279, 137–143. [Google Scholar] [CrossRef] [Green Version]
  4. Murciano, J.C.; Medinilla, S.; Eslin, D.; Atochina, E.; Cines, D.B.; Muzykantov, V.R. Prophylactic fibrinolysis through selective dissolution of nascent clots by tPA-carrying erythrocytes. Nat. Biotechnol. 2003, 21, 891–896. [Google Scholar] [CrossRef] [PubMed]
  5. Murciano, J.C.; Muzykantov, V.R. Coupling of anti-thrombotic agents to red blood cells offers safer and more effective management of thrombosis. Discov. Med. 2003, 3, 28–29. [Google Scholar]
  6. Ji, W.; Smith, P.N.; Koepse, R.R.; Andersen, J.D.; Baker, S.L.; Zhang, L.; Carmali, S.; Myerson, J.W.; Muzykantov, V.; Russell, A.J. Erythrocytes as carriers of immunoglobulin-based therapeutics. Acta Biomater. 2020, 101, 422–435. [Google Scholar] [CrossRef] [PubMed]
  7. Muzykantov, V.R.; Murciano, J.-C. Streptavidin-mediated coupling of therapeutic proteins to carrier erythrocytes. In Erythrocyte Engineering for Drug Delivery and Targeting. Biotechnology Intelligence; Unit 6; Magnani, M., Ed.; Landes Bioscience: Georgetown, TX, USA, 2002; Chapter 4; pp. 37–67. ISBN 1-58706-061-2. [Google Scholar]
  8. Villa, C.H.; Pan, D.C.; Zaitsev, S.; Cines, D.B.; Siegel, D.L.; Muzykantov, V.R. Delivery of drugs bound to erythrocytes: New avenues for an old intravascular carrier. Ther. Deliv. 2015, 6, 795–826. [Google Scholar] [CrossRef]
  9. Villa, C.H.; Anselmo, A.C.; Mitragotri, S.; Muzykantov, V. Red blood cells: Supercarriers for drugs, biologicals, and nanoparticles and inspiration for advanced delivery systems. Adv. Drug Deliv. Rev. 2016, 106, 88–103. [Google Scholar] [CrossRef] [Green Version]
  10. Tsong, T.Y.; Kinosita, K., Jr. Use of voltage pulses for the pore opening and drug loading and the subsequent resealing of red blood cells. In Red Blood Cells as Carriers for Drugs; Karger Publishers: Berlin, Germany, 1985; pp. 108–114. [Google Scholar] [CrossRef]
  11. Lizano, C.; Sanz, S.; Luque, J.; Pinilla, M. In vitro study of alcohol dehydrogenase and acetaldehyde dehydrogenase encapsulated into human erythrocytes by an electroporation procedure. Biochim. Biophys. Acta (Gen. Subj.) 1998, 1425, 328–336. [Google Scholar] [CrossRef]
  12. Yamagata, K.; Kawasaki, E.; Kawarai, H.; Iino, M. Encapsulation of concentrated protein into erythrocyte porated by continuous-wave ultrasound. Ultrasound. Med. Biol. 2008, 34, 1924–1933. [Google Scholar] [CrossRef]
  13. Ginn, F.L.; Hochstein, P.; Trump, B.F. Membrane alterations in hemolysis: Internalization of plasmalemma induced by primaquine. Science 1969, 164, 843–845. [Google Scholar] [CrossRef] [PubMed]
  14. Ben-Bassat, I.; Bensch, K.G.; Schrier, S.L. Drug-induced erythrocyte membrane internalization. J. Clin. Investig. 1972, 51, 1833–1844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Matovcik, L.M.; Junga, I.G.; Schrie, S.L. Drug-induced endocytosis of neonatal erythrocytes. Blood 1985, 65, 1056–1063. Available online: https://ashpublications.org/blood/article/65/5/1056/164002/Drug-induced-endocytosis-of-neonatal-erythrocytes (accessed on 10 March 2020). [CrossRef] [PubMed] [Green Version]
  16. Kwon, Y.M.; Chung, H.S.; Moon, C.; Yockman, J.; Park, Y.J.; Gitlin, S.D.; David, A.E.; Yang, V.C. l-Asparaginase encapsulated intact erythrocytes for treatment of acute lymphoblastic leukemia (ALL). J. Control. Release 2009, 139, 182–189. [Google Scholar] [CrossRef] [Green Version]
  17. Franco, R.S.; Weiner, M.; Wagner, K.; Martelo, O.J. Incorporation of inositol hexaphosphate into red blood cells mediated by dimethyl sulfoxide. Life Sci. 1983, 32, 2763–2768. [Google Scholar] [CrossRef]
  18. Mosca, A.; Paleari, R.; Russo, V.; Rosti, E.; Nano, R.; Boicelli, A.; Villa, S.; Zanella, A. IHP entrapment into human erythrocytes: Comparison between hypotonic dialysis and DMSO osmotic pulse. Adv. Exp. Med. Biol. 1992, 326, 19–26. [Google Scholar] [CrossRef]
  19. Wang, X.; Ge, W.; Xu, X.; Kang, X.; Luo, X. Investigation on the preparative method of morphine loaded in erythrocyte and encapsulating effect of carrier erythrocytes. Chin. J. Clin. Pharm. 2003, 06, 335–338, (In Chinese, Abstract in English). Available online: http://en.cnki.com.cn/Article_en/CJFDTotal-LCZZ200306003.htm (accessed on 20 February 2020).
  20. Ge, W.-h.; Lian, Y.-s.; Wang, X.-h.; Luo, X.; Xie, P.-h. Morphological observation of erythrocyte during the preparation of morphine carrier by a hyperosmotic method. Chin. Pharm. J. 2004, 04, 270–272, (In Chinese, Abstract in English). Available online: http://en.cnki.com.cn/Article_en/CJFDTOTAL-ZGYX200404011.htm (accessed on 20 February 2020).
  21. Ihler, G.M.; Glew, R.H.; Schnure, F.W. Enzyme loading of erythrocytes. Proc. Natl. Acad. Sci. USA 1973, 70, 2663–2666. [Google Scholar] [CrossRef]
  22. Updike, S.J.; Wakamiya, R.T. Infusion of red blood cell-loaded asparaginase in monkey. Immunologic, metabolic, and toxicologic consequences. J. Lab. Clin. Med. 1983, 101, 679–691. [Google Scholar]
  23. Rechsteiner, M.C. Uptake of proteins by red blood cells. Exp. Cell Res. 1975, 93, 487–492. [Google Scholar] [CrossRef]
  24. Alpar, H.O.; Lewis, D.A. Therapeutic efficacy of asparaginase encapsulated in intact erythrocytes. Biochem. Pharmacol. 1985, 34, 257–261. [Google Scholar] [CrossRef]
  25. Magnani, M.; Rossi, L.; D’ascenzo, M.; Panzani, I.; Bigi, L.; Zanella, A. Erythrocyte engineering for drug delivery and targeting. Biotechnol. Appl. Biochem. 1998, 28, 1–6. [Google Scholar] [CrossRef] [PubMed]
  26. DeLoach, J.R.; Ihler, G. A dialysis procedure for loading erythrocytes with enzymes and lipids. Biochim. Biophys. Acta (Gener. Sub.) 1977, 496, 136–145. [Google Scholar] [CrossRef]
  27. Bax, B.E.; Bain, M.D.; Fairbanks, L.D.; Webster, A.D.; Chalmers, R.A. In vitro and in vivo studies with human carrier erythrocytes loaded with polyethylene glycol-conjugated and native adenosine deaminase. Br. J. Haematol. 2000, 109, 549–554. [Google Scholar] [CrossRef]
  28. DeLoach, J.R.; Harris, R.L.; Ihler, G.M. An erythrocyte encapsulator dialyzer used in preparing large quantities of erythrocyte ghosts and encapsulation of a pesticide in erythrocyte ghosts. Anal. Biochem. 1980, 102, 220–227. [Google Scholar] [CrossRef]
  29. Ropars, C.; Nicolau, C.; Chassaigne, M. Process and Device for the Encapsulation in Erythrocytes of at Least One Biologically Active Substance, in Particular Hemoglobin Allosteric Effectors, and Erythrocytes So Obtained. EP 0101341. Data of Publication: 1 June 1983. Available online: https://europepmc.org/article/pat/ep0101341 (accessed on 10 March 2020).
  30. Godfrin, Y. Lysis/Resealing Process and Device for Incorporating an Active Ingredient, in Particular Asparaginase or Inositol Hexaphosphate, in Erythrocytes. US Patent 2008261262, 4 August 2008. Available online: https://europepmc.org/article/pat/us2008261262 (accessed on 10 March 2020).
  31. Millan, C.G.; Marinero, M.L.S.; Castaneda, A.Z.; Lanao, J.M. Drug, enzyme and peptide delivery using erythrocytes as carriers. J. Control Release 2004, 95, 27–49. [Google Scholar] [CrossRef]
  32. Pierige, F.; Serafini, S.; Rossi, L.; Magnani, M. Cell-based drug delivery. Adv. Drug Deliv. Rev. 2008, 60, 286–295. [Google Scholar] [CrossRef]
  33. Milanick, M.A.; Ritter, S.; Meissner, K. Engineering erythrocytes to be erythrosensors: First steps. Blood Cells Mol. Dis. 2011, 47, 100–106. [Google Scholar] [CrossRef]
  34. Ritter, S.C.; Milanick, M.A.; Meissner, K.E. Encapsulation of FITC to monitor extracellular pH: A step towards the development of red blood cells as circulating blood analyte biosensors. Biomed. Opt. Express 2011, 2, 2012–2021. [Google Scholar] [CrossRef]
  35. Ritter, S.C.; Shao, X.; Cooley, N.; Milanick, M.A.; Glass, T.E.; Meissner, K.E. Blood analyte sensing using fluorescent dye-loaded red blood cells. In Proceedings of the Optical Diagnostics and Sensing XIV: Toward Point-of-Care Diagnostics, San Francisco, CA, USA, 3–6 February 2014; Coté, G.L., Ed.; SPIE: San Francisco, CA, USA, ; 2014; Volume 8951. [Google Scholar] [CrossRef]
  36. Pierigè, F.; Bigini, N.; Rossi, L.; Magnani, M. Reengineering red blood cells for cellular therapeutics and diagnostics. WIREs Nanomed. Nanobiotechnol. 2017, 9, e1454:1–e1454:17. [Google Scholar] [CrossRef] [PubMed]
  37. Platt, F.M.; d’Azzo, A.; Davidson, B.L.; Neufeld, E.F.; Tifft, C.J. Lysosomal storage diseases. Nat. Rev. Dis. Prim. 2018, 4, 27:1–27:25. [Google Scholar] [CrossRef] [PubMed]
  38. Bax, B.E.; Bain, M.D.; Ward, C.P.; Fensom, A.H.; Chalmers, R.A. The entrapment of mannose-terminated glucocerebrosidase (alglucerase) in human carrier erythrocytes. Biochem. Soc. Trans. 1996, 24, 441S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Kaplan, P.; Baris, H.; De Meirleir, L.; Di Rocco, M.; El-Beshlawy, A.; Huemer, M.; Martins, A.M.; Nascu, I.; Rohrbach, M.; Steinbach, L.; et al. Revised recommendations for the management of Gaucher disease in children. Eur. J. Pediatr. 2013, 172, 447–458. [Google Scholar] [CrossRef]
  40. Genetics Home Reference. Available online: https://ghr.nlm.nih.gov/condition/mucopolysaccharidosis-type-vii (accessed on 20 February 2020).
  41. Sheppard, M.N. The heart in Fabry’s disease. Cardiovasc. Pathol. 2011, 20, 8–14. [Google Scholar] [CrossRef]
  42. Germain, D.P. Fabry disease. Orphanet J. Rare Dis. 2010, 5, 30:1–30:49. [Google Scholar] [CrossRef] [Green Version]
  43. The portal for rare diseases and orphan drugs. Available online: https://www.orpha.net/consor/cgi-bin/Disease_Search.php?lng=EN&data_id=14&Disease_Disease_Search_diseaseGroup=Pompe-disease&Disease_Disease_Search_diseaseType=Pat&Disease%28s%29/group of diseases=Glycogen-storage-disease-type-2--Pompe-disease-&title=Glyco (accessed on 20 February 2020).
  44. Dale, G.L.; Villacorte, D.G.; Beutler, E. High-yield entrapment of proteins into erythrocytes. Biochem. Med. 1977, 18, 220–225. [Google Scholar] [CrossRef]
  45. Beutler, E.; Dale, G.L.; Guinto, D.E.; Kuhl, W. Enzyme replacement therapy in Gaucher’s disease: Preliminary clinical trial of a new enzyme preparation. Proc. Natl. Acad. Sci. USA 1977, 74, 4620–4623. [Google Scholar] [CrossRef] [Green Version]
  46. Humphreys, J.D.; Ihler, G.M. Enhanced stability of erythrocyte-entrapped glucocerebrosidase activity. J. Lab. Clin. Med. 1980, 96, 682–692. [Google Scholar]
  47. Clinical Review Report: Sapropterin dihydrochloride (Kuvan). Canadian Agency for Drugs and Technologies in Health; Executive Summary; Ottawa, ON. September 2017. Available online: https://www.ncbi.nlm.nih.gov/books/NBK533800/ (accessed on 20 February 2020).
  48. Bell, S.M.; Henschell, C.; Lemontt, J.F.; Gamez, A.; Scriver, C.R.; Sarkissian, C.N.; Lambert, A.; Charbonneau, M.; Wang, L.; Zhao, B.; et al. Preclinical evaluation of multiple species of PEGylated recombinant phenylalanine ammonia lyase for the treatment of phenylketonuria. Proc. Natl. Acad. Sci. USA 2008, 105, 20894–20899. [Google Scholar] [CrossRef] [Green Version]
  49. Godfrin, Y.; Dufour, E.; Cheng, S.H.; Yew, N.S. Composition of erythrocytes encapsulating phenylalanine hydroxylase and therapeutic use thereof. US Patent 2016120956, 5 May 2016. Available online: https://pubchem.ncbi.nlm.nih.gov/patent/US2016120956 (accessed on 20 February 2020).
  50. Yew, N.S.; Dufour, E.; Przybylska, M.; Putelat, J.; Crawley, C.; Foster, M.; Gentry, S.; Reczek, D.; Kloss, A.; Meyzaud, A.; et al. Erythrocytes encapsulated with phenylalanine hydroxylase exhibit improved pharmacokinetics and lowered plasma phenylalanine levels in normal mice. Mol. Genet. Metab. 2013, 109, 339–344. [Google Scholar] [CrossRef] [PubMed]
  51. Aloj, G.; Giardino, G.; Valentino, L.; Maio, F.; Gallo, V.; Esposito, T.; Naddei, R.; Cirillo, E.; Pignata, C. Severe combined immunodeficiences: New and old scenarios. Int. Rev. Immunol. 2012, 31, 43–65. [Google Scholar] [CrossRef]
  52. Flinn, A.M.; Gennery, A.R. Adenosine deaminase deficiency: A review. Orphanet J. Rare Dis. 2018, 13, 65–72. [Google Scholar] [CrossRef] [PubMed]
  53. Blackburn, M.R.; Thompson, L.F. Adenosine deaminase deficiency: Unanticipated benefits from the study of a rare immunodeficiency. J. Immunol. 2012, 188, 933–935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Booth, C.; Gaspar, H.B. Pegademase bovine (PEG-ADA) for the treatment of infants and children with severe combined immunodeficiency (SCID). Biologics 2009, 3, 349–358. [Google Scholar] [PubMed]
  55. Bax, B.E.; Bain, M.D.; Fairbanks, L.D.; Simmonds, H.A.; Webster, A.D.; Chalmers, R.A. Carrier erythrocyte entrapped adenosine deaminase therapy in adenosine deaminase deficiency. Adv. Exp. Med. Biol. 2000, 486, 47–50. [Google Scholar] [CrossRef]
  56. Moran, N.F.; Bain, M.D.; Muqit, M.M.; Bax, B.E. Carrier erythrocyte entrapped thymidine phosphorylase therapy for MNGIE. Neurology 2008, 71, 686–688. [Google Scholar] [CrossRef]
  57. Filosto, M.; Cotti Piccinelli, S.; Caria, F.; Gallo Cassarino, S.; Baldelli, E.; Galvagni, A.; Volonghi, I.; Scarpelli, M.; Padovani, A. Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE-MTDPS1). J. Clin. Med. 2018, 7, 389. [Google Scholar] [CrossRef] [Green Version]
  58. Bax, B.E.; Bain, M.D.; Fairbanks, L.D.; Webster, A.D.; Ind, P.W.; Hershfield, M.S.; Chalmers, R.A. A 9-yr evaluation of carrier erythrocyte encapsulated adenosine deaminase (ADA) therapy in a patient with adult-type ADA deficiency. Eur. J. Haematol. 2007, 79, 338–348. [Google Scholar] [CrossRef]
  59. Sanz, S.; Lizano, C.; Garín, M.I.; Luque, J.; Pinilla, M. Biochemical properties of alcohol dehydrogenase and glutamate dehydrogenase encapsulated into human erythrocytes by a hypotonic-dialysis procedure. In Erythrocytes as Drug Carriers in Medicine; Sprandel, U., Way, J.L., Eds.; Springer Science + Business Media: New York, NY, USA, 1997; pp. 101–108. ISBN 978-1-4899-0046-3. [Google Scholar]
  60. Sanz, S.; Pinilla, M.; Garin, M.; Tipton, K.F.; Luque, J. The influence of enzyme concentration on the encapsulation of glutamate dehydrogenase and alcohol dehydrogenase in red blood cells. Biotechnol. Appl. Biochem. 1995, 22, 223–231. [Google Scholar] [CrossRef]
  61. Magnani, M.; Fazi, A.; Mangani, F.; Rossi, L.; Mancini, U. Methanol detoxification by enzyme-loaded erythrocytes. Biotechnol. Appl. Biochem. 1993, 18, 217–226. [Google Scholar] [CrossRef] [PubMed]
  62. Magnani, M.; Laguerre, M.; Rossi, L.; Bianchi, M.; Ninfali, P.; Mangani, F.; Ropars, C. In vivo accelerated acetaldehyde metabolism using acetaldehyde dehydrogenase-loaded erythrocytes. Alcohol Alcohol. 1990, 25, 627–637. [Google Scholar] [CrossRef] [PubMed]
  63. Alexandrovich, Y.G.; Kosenko, E.A.; Sinauridze, E.I.; Obydennyi, S.I.; Kireev, I.I.; Ataullakhanov, F.I.; Kaminsky, Y.G. Rapid elimination of blood alcohol using erythrocytes: Mathematical modeling and in vitro study. Biomed. Res. Int. 2017, 2017, 5849593:1–5849593:14. [Google Scholar] [CrossRef] [PubMed]
  64. Magnani, M.; Rossi, L.; Bianchi, M.; Giorgio, F.; Benatti, U.; Guida, L.; Zocchi, E.; Flora, A. De Improved metabolic properties of hexokinase-overloaded human erythrocytes. Biochim. Biophys. Acta (Bioenerg.) 1988, 972, 1–8. [Google Scholar] [CrossRef]
  65. Leung, P.; Ray, L.E.; Sander, C.; Way, J.L.; Sylvester, D.M.; Way, J.L. Encapsulation of thiosulfate: Cyanide sulfurtransferase by mouse erythrocytes. Toxicol. Appl. Pharmacol. 1986, 83, 101–107. [Google Scholar] [CrossRef]
  66. Way, J.L.; Cannon, E.P.; Leung, P.; Hawkins-Zitzer, A.; Pei, L.; Petrikovics, I. Antagonism of the lethal effects of cyanide with resealed erythrocytes containing rhodanese and thiosulfate. Adv. Exp. Med. Biol. 1992, 326, 159–163. [Google Scholar] [CrossRef]
  67. Petrikovics, I.; Cannon, E.P.; Mcguinn, W.D.; Pei, L.; Pu, L.; Lindner, L.E.; Way, J.L. Cyanide antagonism with carrier erythrocytes and organic thiosulfonates. Toxicol. Sci. 1995, 24, 86–93. [Google Scholar] [CrossRef]
  68. Leung, P.; Davis, R.W.; Yao, C.C.; Cannon, E.P.; Way, J.L. Rhodanese and sodium thiosulfate encapsulated in mouse carrier erythrocytes: II. In vivo survivability and alterations in physiologic and morphologic characteristics. Toxicol. Sci. 1991, 16, 559–566. [Google Scholar] [CrossRef]
  69. Way, J.L.; Leung, P.; Ray, L.; Sander, C. Erythrocyte encapsulated thiosulfate sulfurtransferase. In Red Blood Cells as Carriers for Drugs; Karger Publishers: Berlin, Germany, 1985; pp. 75–81. [Google Scholar] [CrossRef]
  70. Leung, P.; Cannon, E.P.; Petrikovics, I.; Hawkins, A.; Way, J.L. In vivo studies on rhodanese encapsulation in mouse carrier erythrocytes. Toxicol. Appl. Pharmacol. 1991, 110, 268–274. [Google Scholar] [CrossRef]
  71. Liu, J.; Lkhagva, E.; Chung, H.J.; Kim, H.J.; Hong, S.T. The pharmabiotic approach to treat hyperammonemia. Nutrients 2018, 10, 140:1–140:18. [Google Scholar] [CrossRef] [Green Version]
  72. Sanz, S.; Lizano, C.; Luque, J.; Pinilla, M. In vitro and in vivo study of glutamate dehydrogenase encapsulated into mouse erythrocytes by a hypotonic dialysis procedure. Life Sci. 1999, 65, 2781–2789. [Google Scholar] [CrossRef]
  73. Venediktova, N.I.; Kosenko, E.A.; Kaminsky, Y.G. Studies on ammocytes: Development, metabolic characteristics, and detoxication of ammonium. Bull. Exp. Biol. Med. 2008, 146, 730–732. [Google Scholar] [CrossRef] [PubMed]
  74. Kosenko, E.A.; Venediktova, N.I.; Kudryavtsev, A.A.; Ataullakhanov, F.I.; Kaminsky, Y.G.; Felipo, V.; Montoliu, C. Encapsulation of glutamine synthetase in mouse erythrocytes: A new procedure for ammonia detoxification. Biochem. Cell Biol. 2008, 86, 469–476. [Google Scholar] [CrossRef] [PubMed]
  75. Protasov, E.S.; Borsakova, D.V.; Alexandrovich, Y.G.; Korotkov, A.V.; Kosenko, E.A.; Butylin, A.A.; Ataullakhanov, F.I.; Sinauridze, E.I. Erythrocytes as bioreactors to decrease excess ammonium concentration in blood. Sci. Rep. 2019, 9, 1455:1–1455:16. [Google Scholar] [CrossRef]
  76. Batool, T.; Makky, E.A.; Jalal, M.; Yusoff, M.M. A Comprehensive review on l-asparaginase and its applications. Appl. Biochem. Biotechnol. 2016, 178, 900–923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Qiu, F.; Huang, J.; Sui, M. Targeting arginine metabolism pathway to treat arginine-dependent cancers. Cancer Lett. 2015, 364, 1–7. [Google Scholar] [CrossRef] [PubMed]
  78. Ni, Y.; Schwaneberg, U.; Sun, Z.H. Arginine deiminase, a potential anti-tumor drug. Cancer Lett. 2008, 261, 1–11. [Google Scholar] [CrossRef]
  79. Bobak, Y.P.; Vynnytska, B.O.; Kurlishchuk, Y.V.; Sibirny, A.A.; Stasyk, O.V. Cancer cell sensitivity to arginine deprivation in vitro is not determined by endogenous levels of arginine metabolic enzymes. Cell Biol. Int. 2010, 34, 1085–1089. [Google Scholar] [CrossRef]
  80. Fernandes, H.S.; Silva Teixeira, C.S.; Fernandes, P.A.; Ramos, M.J.; Cerqueira, N.M.F.S.A. Amino acid deprivation using enzymes as a targeted therapy for cancer and viral infections. Expert Opin. Ther. Pat. 2017, 27, 283–297. [Google Scholar] [CrossRef]
  81. Halfon-Domenech, C.; Thomas, X.; Chabaud, S.; Baruchel, A.; Gueyffier, F.; Mazingue, F.; Auvrignon, A.; Corm, S.; Dombret, H.; Chevallier, P.; et al. l-asparaginase loaded red blood cells in refractory or relapsing acute lymphoblastic leukaemia in children and adults: Results of the GRASPALL 2005-01 randomized trial. Br. J. Haematol. 2011, 153, 58–65. [Google Scholar] [CrossRef]
  82. Hunault-Berger, M.; Leguay, T.; Huguet, F.; Leprêtre, S.; Deconinck, E.; Ojeda-Uribe, M.; Bonmati, C.; Escoffre-Barbe, M.; Bories, P.; Himberlin, C.; et al. A Phase 2 study of l-asparaginase encapsulated in erythrocytes in elderly patients with Philadelphia chromosome negative acute lymphoblastic leukemia: The GRASPALL/GRAALL-SA2-2008 study. Am. J. Hematol. 2015, 90, 811–818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Thomas, X.G.; Tardy, T.E.; Guieze, R.; Chevallier, P.; Marolleau, J.P.; Orsini, F.; Hitchcock, I.; El-Hariry, I. GRASPA-AML 2012-01 study (NCT01810705): A multicenter, open, randomized phase 2b trial evaluating ERY001 (l-asparaginase encapsulated in red blood cells) plus low-dose cytarabine vs. low-dose cytarabine alone, in treatment of newly diagnosed acute myeloid. J. Clin. Oncol. 2015, 33, TPS7099. [Google Scholar] [CrossRef]
  84. Hammel, P.; Berardi, R.; Van Cutsem, E.; Feliu, J.; Greil, R.; Wasan, H.S.; Metges, J.-P.; Nygren, P.; Osterlund, P.J.; Parner, V.; et al. Trybeca-1: A randomized, phase 3 study of eryaspase in combination with chemotherapy versus chemotherapy alone as second-line treatment in patients with pancreatic adenocarcinoma (NCT03665441). J. Clin. Oncol. 2019, 37, TPS471. [Google Scholar] [CrossRef]
  85. Bachet, J.B.; Gay, F.; Maréchal, R.; Galais, M.-P.; Adenis, A.; Salaco, D.; Cros, J.; Demetter, P.; Svrcek, M.; Bardier-Dupas, A.; et al. Asparagine synthetase expression and phase I study with L -asparaginase encapsulated in red blood cells in patients with pancreatic adenocarcinoma. Pancreas 2015, 44, 1141–1147. [Google Scholar] [CrossRef] [PubMed]
  86. Hammel, P.; Bachet, J.-B.; Portales, F.; Mineur, L.; Metges, J.-P.; De la Fouchardiere, C.; Louvet, C.; El Hajbi, F.; Faroux, R.; Guimbaud, R.; et al. A phase 2b of eryaspase in combination with gemcitabine or FOLFOX as second-line therapy in patients with metastatic pancreatic adenocarcinoma (NCT02195180). In Abstract Book of theProceedings of the 42nd ESMO Congress (ESMO 2017), Madrid, Spain, 8–12 September, 2017; Oxford University Press: Oxford, UK, 2017; p. 211. [Google Scholar] [CrossRef]
  87. Gay, F.; Aguera, K.; Sénéchal, K.; Tainturier, A.; Berlier, W.; Maucort-Boulch, D.; Honnorat, J.; Horand, F.; Godfrin, Y.; Bourgeaux, V. Methionine tumor starvation by erythrocyte-encapsulated methionine gamma-lyase activity controlled with per os vitamin B6. Cancer Med. 2017, 6, 1437–1452. [Google Scholar] [CrossRef] [PubMed]
  88. Sénéchal, K.; Maubant, S.; Leblanc, M.; Ciré, S.; Gallix, F.; Andrivon, A.; Duchamp, O.; Viviani, F.; Horand, F.; Scheer, A.; et al. Erymethionase (methionine-gamma-lyase encapsulated into red blood cells) potentiates anti-PD-1 therapy in TNBC syngeneic mouse model. In Proceedings of the AACR Annual Meeting, Atlanta, GA, USA, 29 March–3 April 2019; American Association for Cancer Research: Philadelphia, PA, USA, 2019; Volume 79. [Google Scholar] [CrossRef]
  89. Godfrin, Y.; Goineau, P.-O. Erythrocytes Containing Arginin Deiminase. US Patent 9,125,876 B2, 8 September 2015. [Google Scholar]
  90. Gay, F.; Aguera, K.; Senechal, K.; Bes, J.; Chevrier, A.-M.; Gallix, F.; Guicher, C.; Lorenzi, P.; Bourgeaux, V.; Berlier, W.; et al. Arginine deiminase loaded in erythrocytes: A promising formulation for l-arginine deprivation therapy in cancers. In Proceedings of the AACR 107th Annual Meeting 2016, New Orleans, LA, USA, 16–20 April 2016; American Association for Cancer Research: Philadelphia, PA, USA, 2016; Volume 76. [Google Scholar] [CrossRef]
  91. Lamarre, Y.; Bourgeaux, V.; Pichon, A.; Hardeman, M.R.; Campion, Y.; Hardeman-Zijp, M.; Martin, C.; Richalet, J.P.; Bernaudin, F.; Driss, F.; et al. Effect of inositol hexaphosphate-loaded red blood cells (RBCs) on the rheology of sickle RBCs. Transfusion 2013, 53, 627–636. [Google Scholar] [CrossRef] [PubMed]
  92. Bourgeaux, V.; Hequet, O.; Campion, Y.; Delcambre, G.; Chevrier, A.M.; Rigal, D.; Godfrin, Y. Inositol hexaphosphate-loaded red blood cells prevent in vitro sickling. Transfusion 2010, 50, 2176–2184. [Google Scholar] [CrossRef]
  93. Gersonde, K.; Nicolau, C. Improvement of the red blood cell O2 release capacity by lipid vesicle-mediated incorporation of inositol hexaphosphate. Blut 1979, 39, 1–7. [Google Scholar] [CrossRef]
  94. Teisseire, B.; Ropars, C.; Vieilledent, C.; Vallez, M.O.; Laurent, D. Encapsulation of a hemoglobin allosteric effector in erythrocytes: In vivo results. Life Support. Syst. 1984, 2, 277–280. [Google Scholar]
  95. Bourgeaux, V.; Aufradet, E.; Campion, Y.; De Souza, G.; Horand, F.; Bessaad, A.; Chevrier, A.-M.; Canet-Soulas, E.; Godfrin, Y.; Martin, C. Efficacy of homologous inositol hexaphosphate-loaded red blood cells in sickle transgenic mice. Br. J. Haematol. 2012, 157, 357–369. [Google Scholar] [CrossRef]
  96. Booser, D.J.; Hortobagyi, G.N. Anthracycline antibiotics in cancer therapy. Focus on drug resistance. Drugs 1994, 47, 223–258. [Google Scholar] [CrossRef] [PubMed]
  97. Hortobagyi, G.N. Anthracyclines in the treatment of cancer. An overview. Drugs 1997, 54 (Suppl 4), 1–7. [Google Scholar] [CrossRef]
  98. Iarussi, D.; Indolfi, P.; Galderisi, M.; Bossone, E. Cardiac toxicity after anthracycline chemotherapy in childhood. Herz 2000, 25, 676–688. [Google Scholar] [CrossRef] [PubMed]
  99. Iarussi, D.; Indolfi, P.; Casale, F.; Martino, V.; Di Tullio, M.T.; Calabro, R. Anthracycline-induced cardiotoxicity in children with cancer: Strategies for prevention and management. Paediatr. Drugs 2005, 7, 67–76. [Google Scholar] [CrossRef] [PubMed]
  100. Sawyer, D.B.; Peng, X.; Chen, B.; Pentassuglia, L.; Lim, C.C. Mechanisms of anthracycline cardiac injury: Can we identify strategies for cardioprotection? Prog. Cardiovasc. Dis. 2010, 53, 105–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Di Marco, A.; Gaetani, M.; Scarpinato, B. Adriamycin (NSC-123,127): A new antibiotic with antitumor activity. Cancer Chemother. Reports 1969, 53, 33–37. [Google Scholar]
  102. Bonadonna, G.; Silvio, M.; De Lena, M.; Fossati-Bellani, F. Clinical evaluation of adriamycin, a new antitumour antibiotic. Br. MedicalJournal 1969, 3, 503–506. [Google Scholar] [CrossRef] [Green Version]
  103. Tonetti, M.; Astroff, B.; Satterfield, W.; De Flora, A.; Benatti, U.; DeLoach, J.R. Construction and characterization of adriamycin-loaded canine red blood cells as a potential slow delivery system. Biotechnol. Appl. Biochem. 1990, 12, 621–629. [Google Scholar] [CrossRef]
  104. Matherne, C.M.; Satterfield, W.C.; Gasparini, A.; Tonetti, M.; Astroff, A.B.; Schmidt, R.D.; Rowe, L.D.; DeLoach, J.R. Clinical efficacy and toxicity of doxorubicin encapsulated in glutaraldehyde-treated erythrocytes administered to dogs with lymphosarcoma. Am. J. Vet. Res. 1994, 55, 847–853. [Google Scholar]
  105. Gasparini, A.; Tonetti, M.; Astroff, B.; Rowe, L.; Satterfield, W.; Schmidt, R.; DeLoach, J.R. Pharmacokinetics of doxorubicin loaded and glutaraldehyde treated erythrocytes in healthy and lymphoma bearing dogs. Adv. Exp. Med. Biol. 1992, 326, 299–304. [Google Scholar] [CrossRef]
  106. Tonetti, M.; Zocchi, E.; Guida, L.; Polvani, C.; Benatti, U.; Biassoni, P.; Romei, F.; Guglielmi, A.; Aschele, C.; Sobrero, A.; et al. Use of glutaraldehyde treated autologous human erythrocytes for hepatic targeting of doxorubicin. Adv. Exp. Med. Biol. 1992, 326, 307–317. [Google Scholar] [CrossRef] [PubMed]
  107. Isaev, V.G.; Garmaeva, T.T.; Skorokhod, A.A.; Parovichnikova, E.N.; Tiurina, N.G.; Kucher, R.A.; Vitvitskiĭ, V.M.; Ataullakhanov, F.I.; Savchenko, V.G. Immobilized forms of daunorubicin in patients with acute leukemia. Ter. Arkh. 1999, 71, 32–37, (in Russian, abstract in English). [Google Scholar] [PubMed]
  108. Skorokhod, O.A.; Kulikova, E.V.; Galkina, N.M.; Medvedev, P.V.; Zybunova, E.E.; Vitvitsky, V.M.; Pivnik, A.V.; Ataullakhanov, F.I. Doxorubicin pharmacokinetics in lymphoma patients treated with doxorubicin-loaded eythrocytes. Haematologica 2007, 92, 570–571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Ataullakhanov, F.I.; Isaev, V.G.; Kohno, A.V.; Kulikova, E.V.; Parovichnikova, E.N.; Savchenko, V.G.; Vitvitsky, V.M. Pharmacokinetics of doxorubicin in patients with lymphoproliferative disorders after infusion of doxorubicin-loaded erythrocytes. In Erythrocytes as Drug Carriers in Medicine; Sprandel, U., Way, J.L., Eds.; Springer: Boston, MA, USA, 1997; pp. 137–142. ISBN 978-1-4899-0044-9. [Google Scholar]
  110. Skorokhod, O.A.; Garmaeva, T.; Vitvitsky, V.M.; Isaev, V.G.; Parovichnikova, E.N.; Savchenko, V.G.; Ataullakhanov, F.I. Pharmacokinetics of erythrocyte-bound daunorubicin in patients with acute leukemia. Med. Sci Monit 2004, 10, 55–64. [Google Scholar]
  111. Lucas, A.; Lam, D.; Cabrales, P. Doxorubicin-loaded red blood cells reduced cardiac toxicity and preserved anticancer activity. Drug Deliv. 2019, 26, 433–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Vuimo, T.A.; Kulikoba, E.V.; Sinauridze, E.I.; Yurkevich, A.M.; Kravchenko, S.K.; Ataullakhanov, F.I. Erythrocyte as a potential vehicle for mitoxantrone. In New Research on Biotechnology in Biology and Medicine; Egorov, A.M., Zaikov, G., Eds.; Nova Science Publishers, Inc.: New York, NY, USA, 2006; Chapter 9; pp. 87–95. ISBN 1-60021-092-9. [Google Scholar]
  113. Vuimo, T.A.; Kulikova, E.V.; Sinauridze, E.I.; Alexandrovich, Y.G.; Lisovskaya, I.L.; Yurkevich, A.M.; Ataullakhanov, F.I. Creating a new dosage form of the anthracycline antibiotic mitoxantrone by incorporating it into red blood cells. Mol Medicine (Moscow) 2008, 2, 37–43, (In Russian, abstract in English). [Google Scholar]
  114. Coufal, N.; Farnaes, L. The vinca alkaloids. In Cancer Management in Man: Chemotherapy, Biological Therapy, Hyperthermia and Supporting Measures; Minev, B.R., Ed.; Springer: Dordrecht, The Netherlands, 2011; Chapter 2; pp. 25–37. ISBN 978-90-481-9703-3. [Google Scholar]
  115. Cutts, J.H.; Beer, C.T.; Noble, R.L. Biological properties of Vincaleukoblastine, an alkaloid in Vinca rosea Linn, with reference to its antitumor action. Cancer Res. 1960, 20, 1023–1031. [Google Scholar]
  116. Johnson, I.S.; Wright, H.F.; Svoboda, G.H.; Vlantis, S.J. Antitumor principles derived from Vinca rosea Linn. I. Vincaleukoblastine and leurosine. Cancer Res. 1960, 20, 1016–1022. [Google Scholar]
  117. Leveque, D.; Wihlm, J.; Jehl, F. Pharmacology of catharanthus alkaloids. Bull. Cancer 1996, 83, 176–186. [Google Scholar]
  118. Moudi, M.; Go, R.; Yong, C.; Nazre, M. Vinca alkaloids. Int. J. Prev. Med. 2013, 4, 1231–1235. [Google Scholar] [CrossRef]
  119. Arora, R.; Malhotra, P.; Mathur, A.; Mathur, A.; Govil, C.M.; Ahuja, P.S. Anticancer alkaloids of Catharanthus roseus: Transition from traditional to modern medicine. In Herbal Medicine: A Cancer Chemopreventive and Therapeutic Perspective, 1st ed.; Arora, R., Ed.; Jaypee Brothers Medical Publishers: New Delhi, India, 2010; Chapter 21; pp. 292–309. ISBN 9788184488418. [Google Scholar]
  120. Halahakoon, A.D.; Slivkin, A.I. The terpene-indole alkaloids loaded erythrocytes as a drug carrier: Besign and assessment. Russ. Open Med. J. 2018, 7, e0406:1–e0406:17. [Google Scholar] [CrossRef] [Green Version]
  121. Trineeva, O.V.; Khalahakun, A.D. Study of desorbtion and exemption of terpeno-indole alkaloids of vinkristin and vinblastin from erythrocitary cell carriers. Drug Dev. Regist. (Russia) 2019, 8, 16–21, (In Russian, abstract in English). [Google Scholar] [CrossRef] [Green Version]
  122. Czock, D.; Keller, F.; Rasche, F.M.; Häussler, U. Pharmacokinetics and pharmacodynamics of systemically administered glucocorticoids. Clin. Pharmacokinet. 2005, 44, 61–98. [Google Scholar] [CrossRef] [PubMed]
  123. Rossi, G.A.; Cerasoli, F.; Cazzola, M. Safety of inhaled corticosteroids: Room for improvement. Pulm. Pharmacol. Ther. 2007, 20, 23–35. [Google Scholar] [CrossRef]
  124. Gerber, A.N. Measuring safety of inhaled corticosteroids in asthma. Ann. Allergy. Asthma Immunol. 2016, 117, 577–581. [Google Scholar] [CrossRef] [PubMed]
  125. Dahl, R. Systemic side effects of inhaled corticosteroids in patients with asthma. Respir. Med. 2006, 100, 1307–1317. [Google Scholar] [CrossRef] [Green Version]
  126. Faubion, W.A.J.; Loftus, E.V.J.; Harmsen, W.S.; Zinsmeister, A.R.; Sandborn, W.J. The natural history of corticosteroid therapy for inflammatory bowel disease: A population-based study. Gastroenterology 2001, 121, 255–260. [Google Scholar] [CrossRef] [Green Version]
  127. Umland, S.P.; Schleimer, R.P.; Johnston, S.L. Review of the molecular and cellular mechanisms of action of glucocorticoids for use in asthma. Pulm. Pharmacol. Ther. 2002, 15, 35–50. [Google Scholar] [CrossRef]
  128. D’Ascenzo, M.; Antonelli, A.; Chiarantini, L.; Mancini, U.; Magnani, M. Red blood cells as a glucocorticoids delivery system. In Erythrocytes as Drug Carriers in Medicine; Sprandel, U., Way, J.L., Eds.; Springer: Boston, MA, USA, 1997; pp. 81–88. ISBN 978-1-4899-0046-3. [Google Scholar]
  129. Rossi, L.; Castro, M.; D’Orio, F.; Damonte, G.; Serafini, S.; Bigi, L.; Panzani, I.; Novelli, G.; Dallapiccola, B.; Panunzi, S.; et al. Low doses of dexamethasone constantly delivered by autologous erythrocytes slow the progression of lung disease in cystic fibrosis patients. Blood Cells, Mol. Dis. 2004, 33, 57–63. [Google Scholar] [CrossRef]
  130. Annese, V.; Latiano, A.; Rossi, L.; Lombardi, G.; Dallapiccola, B.; Serafini, S.; Damonte, G.; Andriulli, A.; Magnani, M. Erythrocytes-mediated delivery of dexamethasone in steroid-dependent IBD patients-a pilot uncontrolled study. Am. J. Gastroenterol. 2005, 100, 1370–1375. [Google Scholar] [CrossRef]
  131. Castro, M.; Rossi, L.; Papadatou, B.; Bracci, F.; Knafelz, D.; Ambrosini, M.; Calce, A.; Serafini, S.; Isacchi, G.; D’Orio, F.; et al. Long-term treatment with autologous red blood cells loaded with dexamethasone 21–phosphate in pediatric patients affected by steroid-dependent Crohn disease. J. Pediatr. Gastroenterol. Nutr. 2007, 44, 423–426. [Google Scholar] [CrossRef] [PubMed]
  132. Castro, M.; Knafelz, D.; Rossi, L.; Ambrosini, M.I.; Papadatou, B.; Mambrini, G.; Magnani, M. Periodic treatment with autologous erythrocytes loaded with dexamethasone 21-phosphate for fistulizing pediatric Crohn’s disease: Case report. J. Pediatr. Gastroenterol. Nutr. 2006, 42, 313–315. [Google Scholar] [CrossRef] [PubMed]
  133. Bossa, F.; Annese, V.; Valvano, M.R.; Latiano, A.; Martino, G.; Rossi, L.; Magnani, M.; Palmieri, O.; Serafini, S.; Damonte, G.; et al. Erythrocytes-mediated delivery of dexamethasone 21-phosphate in steroid-dependent ulcerative colitis: A randomized, double-blind sham-controlled study. Inflamm. Bowel Dis. 2013, 19, 1872–1879. [Google Scholar] [CrossRef] [PubMed]
  134. Rothblum-Oviatt, C.; Wright, J.; Lefton-Greif, M.A.; McGrath-Morrow, S.A.; Crawford, T.O.; Lederman, H.M. Ataxia telangiectasia: A review. Orphanet J. Rare Dis. 2016, 11, 159:1–159:21. [Google Scholar] [CrossRef] [Green Version]
  135. Chessa, L.; Leuzzi, V.; Plebani, A.; Soresina, A.; Micheli, R.; Agnano, D.D.; Venturi, T.; Molinaro, A.; Fazzi, E.; Marini, M.; et al. Intra-Erythrocyte Infusion of Dexamethasone Reduces Neurological Symptoms in Ataxia Teleangiectasia Patients: Results of a Phase 2 Trial. Orphanet J. Rare Dis. 2014, 9, 5:1–5:8. [Google Scholar] [CrossRef] [Green Version]
  136. EryDel. Our Thechnology. Available online: https://www.erydel.com/technology.php (accessed on 20 February 2020).
  137. Tschesche, H.; Dietl, T.; Kolb, H.J.; Standl, E. An insulin degrading proteinase from human erythrocytes and its inhibition by proteinase inhibitors. In Proteinase Inhibitors, Proceedings of the 2nd International Research Conference, Bayer Symposium V, Grosseledder, Germany, 16–20 October 1973; Fritz, H., Tschesche, H., Greene, L.J., Truscheit, E., Fritz, H., Tschesche, H., Greene, L.J., Truscheit, E., Eds.; Springer-Verlag: Berlin, Heidelberg, Germany, 1974; pp. 586–593. [Google Scholar]
  138. Pitt, E.; Johnson, C.M.; Lewis, D.A.; Jenner, D.A.; Offord, R.E. Encapsulation of drugs in intact erythrocytes: An intravenous delivery system. Biochem. Pharmacol. 1983, 32, 3359–3368. [Google Scholar] [CrossRef]
  139. Bird, J.; Best, R.; Lewis, D.A. The encapsulation of insulin in erythrocytes. J. Pharm. Pharmacol. 1983, 35, 246–247. [Google Scholar] [CrossRef]
  140. Ito, Y.; Ogiso, T.; Iwaki, M.; Kitaike, M. Encapsulation of porcine insulin in rabbit erythrocytes and its disposition in the circulation system in normal and diabetic rabbits. J. Pharmacobio-Dyn. 1989, 12, 193–200. [Google Scholar] [CrossRef] [Green Version]
  141. Goldsmith, J.C.; Roer, M.E.S.; Orringer, E.P. A new treatment strategy for hemophilia B: Incorporation of factor IX into red cell ghosts. Am. J. Hematol. 1979, 7, 119–125. [Google Scholar] [CrossRef]
  142. Sinauridze, E.I.; Vuimo, T.A.; Kulikova, E.V.; Shmyrev, I.I.; Ataullakhanov, F.I. A new drug form of blood coagulation factor IX: Red blood cell-entrapped factor IX. Med. Sci. Monit. 2010, 16, PI19–PI26. [Google Scholar]
  143. Luo, X.; Xu, X.; Wang, X.-h.; Zhu, S.-j.; Ge, W.-h. Study of erythrocyte as carrier to prolong action duration of morphine. J. Nanjing Univ. (Natural Sci.) 2003, 3, 547–553, (In Chinese, Abstract in English). Available online: http://en.cnki.com.cn/Article_en/CJFDTotal-NJDZ200305013.htm (accessed on 20 February 2020).
  144. Luo, X.; Wang, X.-H.; Xu, Z.; Cui, S.; Xu, F. Feasibility of using erythrocytes as morphine carrier for postoperative analgesia after coronary artery bypass grafting. Chin. J. Anesthesiol. 2005, 06, 410–413, (In Chinese, Abstract in English). Available online: http://en.cnki.com.cn/Article_en/CJFDTOTAL-ZHMZ200506003.htm (accessed on 20 February 2020).
  145. Xia, X.-p.; Wang, X.-h.; Luo, X. Attempts to use morphine encapsulated in erythrocytes as postoperative analgesia in the replacement of total hip of senile patients. Acta Acad. Med. Xuzhou 2002. Available online: http://en.cnki.com.cn/Article_en/CJFDTOTAL-XZYX200201010.htm (accessed on 20 February 2020).
  146. Wang, X.; Luo, X.; Zhu, S. A clinical study on morphine encapsulated in erythrocytes for postoperative analgesia. J. Clin. Anastesiol. (Chin.) 2001, 2. Available online: http://en.cnki.com.cn/Article_en/CJFDTOTAL-LCMZ200102011.htm (accessed on 17 March 2020).
  147. Costa, R.R.; Alatorre-Meda, M.; Mano, J.F. Drug nano-reservoirs synthesized using layer-by-layer technologies. Biotechnol. Adv. 2015, 33, 1310–1326. [Google Scholar] [CrossRef] [PubMed]
  148. Sakr, O.S.; Borchard, G. Encapsulation of enzymes in layer-by-layer (LbL) structures: Latest advances and applications. Biomacromolecules 2013, 14, 2117–2135. [Google Scholar] [CrossRef] [PubMed]
  149. Muzykantov, V.R. Drug delivery carriers on the fringes: Natural red blood cells versus synthetic multilayered capsules. Expert Opin. Drug Deliv 2013, 10, 1–4. [Google Scholar] [CrossRef]
  150. Parhiz, H.; Khoshnejad, M.; Myerson, J.W.; Hood, E.; Patel, P.N.; Brenner, J.S.; Muzykantov, V.R. Unintended effects of drug carriers: Big issues of small particles. Adv. Drug Deliv. Rev. 2018, 130, 90–112. [Google Scholar] [CrossRef]
  151. Zhang, H. Erythrocytes in nanomedicine: An optimal blend of natural and synthetic materials. Biomater. Sci. 2016, 4, 1024–1031. [Google Scholar] [CrossRef]
  152. Xia, Q.; Zhang, Y.; Li, Z.; Hou, X.; Feng, N. Red blood cell membrane-camouflaged nanoparticles: A novel drug delivery system for antitumor application. Acta Pharm. Sin. B 2019, 9, 675–689. [Google Scholar] [CrossRef]
  153. Brenner, J.S.; Pan, D.C.; Myerson, J.W.; Marcos-Contreras, O.A.; Villa, C.H.; Patel, P.; Hekierski, H.; Chatterjee, S.; Tao, J.-Q.; Parhiz, H.; et al. Red blood cell-hitchhiking boosts delivery of nanocarriers to chosen organs by orders of magnitude. Nat. Commun. 2018, 9, 2684:1–2684:14. [Google Scholar] [CrossRef]
  154. Wang, Y.; Chen, X.; He, D.; Zhou, Y.; Qin, L. Surface-modified nanoerythrocyte loading DOX for targeted liver cancer chemotherapy. Mol. Pharm. 2018, 15, 5728–5740. [Google Scholar] [CrossRef]
  155. Jiang, A.; Song, B.; Ji, X.; Peng, F.; Wang, H.; Su, Y.; He, Y. Doxorubicin-loaded silicon nanoparticles impregnated into red blood cells featuring bright fluorescence, strong photostability, and lengthened blood residency. Nano Res. 2018, 11, 2285–2294. [Google Scholar] [CrossRef]
  156. Hamidi, M.; Rafiei, P.; Azadi, A.; Mohammadi-Samani, S. Encapsulation of valproate-loaded hydrogel nanoparticles in intact human erythrocytes: A novel nano-cell composite for drug delivery. J. Pharm Sci. 2011, 100, 1702–1711. [Google Scholar] [CrossRef] [PubMed]
  157. Chen, J.L.; Dhanaliwala, A.H.; Dixon, A.J.; Farry, J.M.; Hossack, J.A.; Klibanov, A.L. Acoustically active red blood cell carriers for ultrasound-triggered drug delivery with photoacoustic tracking. In Proceedings of the 2015 IEEE International Ultrasonics Symposium, Taipei, Taiwan, 21–24 October 2015; IEEE: Piscataway, NJ, USA, 2015; pp. 234–237. [Google Scholar] [CrossRef]
  158. Harisa, G.I.; Badran, M.M.; AlQahtani, S.A.; Alanazi, F.K.; Attia, S.M. Pravastatin chitosan nanogels-loaded erythrocytes as a new delivery strategy for targeting liver cancer. Saudi Pharm. J. 2016, 24, 74–81. [Google Scholar] [CrossRef]
  159. Burns, J.M.; Vankayala, R.; Mac, J.T.; Anvari, B. Erythrocyte-derived theranostic nanoplatforms for near infrared fluorescence imaging and photodestruction of tumors. ACS Appl. Mater. Interfaces 2018, 10, 27621–27630. [Google Scholar] [CrossRef] [PubMed]
  160. Mac, J.T.; Nuñez, V.; Burns, J.M.; Guerrero, Y.A.; Vullev, V.I.; Anvari, B. Erythrocyte-derived nano-probes functionalized with antibodies for targeted near infrared fluorescence imaging of cancer cells. Biomed. Opt. Express 2016, 7, 1311–1322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Wang, P.; Wang, X.; Luo, Q.; Li, Y.; Lin, X.; Fan, L.; Zhang, Y.; Liu, J.; Liu, X. Fabrication of red blood cell-based multimodal theranostic probes for second near-infrared window fluorescence imaging-guided tumor surgery and photodynamic therapy. Theranostics 2019, 9, 369–380. [Google Scholar] [CrossRef] [PubMed]
  162. Dixon, A.; Farry, J.; Chen, J.; Dhanaliwala, A.H.; Hossack, J.A.; Klibanov, A. Photoacoustic imaging of stimuli-responsive red blood cell drug delivery agents. In Proceedings of the 2016 IEEE International Ultrasonics Symposium (IUS), Tours, France, 18–21 September 2016; IEEE: Piscataway, NJ, USA, 2016; pp. 1–4. [Google Scholar] [CrossRef]
  163. Pan, D.C.; Myerson, J.W.; Brenner, J.S.; Patel, P.N.; Anselmo, A.C.; Mitragotri, S.; Muzykantov, V. Nanoparticle properties modulate their attachment and effect on carrier red blood cells. Sci. Rep. 2018, 8, 1615:1–1615:12. [Google Scholar] [CrossRef] [Green Version]
  164. Pan, D.; Vargas-Morales, O.; Zern, B.; Anselmo, A.C.; Gupta, V.; Zakrewsky, M.; Mitragotri, S.; Muzykantov, V. The effect of polymeric nanoparticles on biocompatibility of carrier red blood cells. PLoS ONE 2016, 11, e0152074:1–e0152074:17. [Google Scholar] [CrossRef] [Green Version]
  165. Eichler, H.G.; Gasic, S.; Bauer, K.; Korn, A.; Bacher, S. In vivo clearance of antibody-sensitized human drug carrier erythrocytes. Clin. Pharmacol. Ther. 1986, 40, 300–303. [Google Scholar] [CrossRef]
  166. Delaby, C.; Pilard, N.; Hetet, G.; Driss, F.; Grandchamp, B.; Beaumont, C.; Canonne-Hergaux, F. A physiological model to study iron recycling in macrophages. Exp. Cell Res. 2005, 310, 43–53. [Google Scholar] [CrossRef]
  167. Chiarantini, L.; Rossi, L.; Fraternale, A.; Magnani, M. Modulated red blood cell survival by membrane protein clustering. Mol. Cell. Biochem. 1995, 144, 53–59. [Google Scholar] [CrossRef]
  168. Bratosin, D.; Mazurier, J.; Tissier, J.P.; Slomianny, C.; Estaquier, J.; Russo-Marie, F.; Huart, J.J.; Freyssinet, J.M.; Aminoff, D.; Ameisen, J.C.; et al. Molecular mechanisms of erythrophagocytosis. Characterization of the senescent erythrocytes that are phagocytized by macrophages. C. R. Acad. Sci. III. Sciences de la vie/Life Sciences 1997, 320, 811–818. [Google Scholar] [CrossRef]
  169. Magnani, M.; Rossi, L.; Brandit, G.; Schiavano, G.F.; Montroni, M.; Piedimonte, G. Targeting antiretroviral nucleoside analogues in phosphorylated form to macrophages : In vitro and in vivo studies. Proc. Nati. Acad. Sci. USA 1992, 89, 6477–6481. [Google Scholar] [CrossRef] [Green Version]
  170. Chiarantini, L.; Antonelli, A.; Rossi, L.; Fraternale, A.; Magnani, M. Red blood cell phagocytosis following hexokinase inactivation. CELL Biochem. Funct. 1994, 12, 217–220. [Google Scholar] [CrossRef] [PubMed]
  171. Zimmermann, U.; Pilwat, G.; Esser, B. The effect of encapsulation in red blood cells on the distribution of methotrexate in mice. Clin. Chem. Lab. Med. 1978, 16, 135–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. DeLoach, J.R.; Barton, C. Glutaraldehyde-treated carrier erythrocytes for organ targeting of methotrexate in dogs. Am. J. Vet. Res. 1981, 42, 1971–1974. [Google Scholar] [PubMed]
  173. DeLoach, J.R.; Tangner, C.H.; Barton, C. Hepatic pharmacokinetics of glutaraldehyde-treated methotrexate-loaded carrier erythrocytes in dogs. Res. Exp. Med. 1983, 183, 167–175. [Google Scholar] [CrossRef]
  174. Yuan, S.-H.; Ge, W.-H.; Huo, J.; Wang, X.-H. Slow release properties and liver-targeting characteristics of methotrexate erythrocyte carriers. Fundam. Clin. Pharmacol. 2009, 23, 189–196. [Google Scholar] [CrossRef]
  175. Mishra, P.R.; Jain, N.K. Biotinylated methotrexate loaded erythrocytes for enhanced liver uptake. ‘A study on the rat’. Int. J. Pharm. 2002, 231, 145–153. [Google Scholar] [CrossRef]
  176. Mishra, P.R.; Jain, N.K. Surface modified methotrexate loaded erythrocytes for enhanced macrophage uptake. J. Drug Target. 2000, 8, 217–224. [Google Scholar] [CrossRef]
  177. Perno, C.F.; Yarcoan, R.; Cooney, D.A.; Hartman, N.R.; Gartner, G.; Popovich, M.; Hao, Z.; Gerrard, T.L.; Wilson, Y.A. Inhibition of human immunodeficiency virus (HIV-1/HTLV-IIIBa-L) replication in fresh and cultured human peripheral blood monocytes/macrophages by azidothymidine and related 2’,3’-dideoxynucleosides. J. Exp. Med. 1988, 168, 1111–1125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Fraternale, A.; Tonelli, A.; Casabianca, A.; Vallanti, G.; Chiarantini, L.; Schiavano, G.F.; Benatti, U.; De Flora, A.; Magnani, M. Role of macrophage protection in the development of murine AIDS. J. Acquir. Immune Defic. Syndr. 1999, 21, 81–89. [Google Scholar] [PubMed]
  179. Fraternale, A.; Casabianca, A.; Rossi, L.; Chiarantini, L.; Schiavano, G.F.; Palamara, A.T.; Garaci, E.; Magnani, M. Erythrocytes as carriers of reduced glutathione (GSH) in the treatment of retroviral infections. J. Antimicrob. Chemother. 2003, 52, 551–554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Rossi, L.; Casabianca, A.; Fraternale, A.; Schiavano, G.F.; Brandi, G.; Antonelli, A.; Magnani, M. Macrophage protection by nucleoside and nucleotide analogue administration. In Erythrocytes as Drug Carriers in Medicine; Sprandel, U., Way, J.L., Eds.; Springer: Boston, MA, USA, 1997; pp. 63–71. ISBN 978-1-4899-0046-3. [Google Scholar]
  181. Magnani, M.; Rossi, L.; Fraternale, A.; Casabianca, A.; Brandi, G.; Benatti, U.; De Flora, A. Targeting antiviral nucleotide analogues to macrophages. J. Leukoc. Biol. 1997, 62, 133–137. [Google Scholar] [CrossRef] [Green Version]
  182. Magnani, M.; Rossi, L.; Fraternale, A.; Silvotti, L.; Quintavalla, F.; Piedimonte, G.; Matteucci, D.; Baldinotti, F.; Bendinelli, M. Feline immunodeficiency virus infection of macrophages: In vitro and in vivo inhibition by dideoxycytidine-5′-triphosphate-loaded erythrocytes. AIDS Res. Hum. Retrovir. 1994, 10, 1179–1186. [Google Scholar] [CrossRef]
  183. Rossi, L.; Brandi, G.; Fraternale, A.; Schiavano, G.F.; Chiarantini, L.; Magnani, M. Inhibition of murine retrovirus-induced immunodeficiency disease by dideoxycytidine and dideoxycytidine 5′-triphosphate. J. Acquir. Immune Defic. Syndr. 1993, 6, 1179–1186. [Google Scholar]
  184. Magnani, M.; Rossi, L.; Fraternale, A.; Silvotti, L.; Quintavalla, F.; Piedimonte, G.; Matteucci, D.; Baldinotti, F.; Bendinelli, M. FIV infection of macrophages: In vitro and in vivo inhibition by dideoxycytidine 5′-triphosphate. Vet. Immunol. Immunopathol. 1995, 46, 151–158. [Google Scholar] [CrossRef]
  185. Benatti, U.; Giovine, M.; Damonte, G.; Gasparini, A.; Scarfi, S.; De Flora, A.; Fraternale, A.; Rossi, L.; Magnani, M. Azidothymidine homodinucleotide-loaded erythrocytes as bioreactors for slow delivery of the antiretroviral drug azidothymidine. Biochem. Biophys. Res. Commun. 1996, 220, 20–25. [Google Scholar] [CrossRef]
  186. Magnani, M.; Fraternale, A.; Casabianca, A.; Schiavano, G.F.; Chiarantini, L.; Palamara, A.T.; Chiriolo, M.R.; Rotilio, G.; Garaci, E. Antiretroviral effect of combined zidovudine and reduced glutathione therapy in murine AIDS. AIDS Res. Hum. Retroviruses 1997, 13, 1093–1099. [Google Scholar] [CrossRef]
  187. Fraternale, A.; Casabianca, A.; Orlandi, C.; Cerasi, A.; Chiarantini, L.; Brandi, G.; Magnani, M. Macrophage protection by addition of glutathione (GSH)-loaded erythrocytes to AZT and DDI in a murine AIDS model. Antiviral Res. 2002, 56, 263–272. [Google Scholar] [CrossRef]
  188. Magnani, M.; Balestra, E.; Fraternale, A.; Aquaro, S.; Paiardini, M.; Cervasi, B.; Casabianca, A.; Garaci, E.; Perno, C.-F. Drug-loaded red blood cell-mediated clearance of HIV-1 macrophage reservoir by selective inhibition of STAT1 expression. J. Leukoc. Biol. 2003, 74, 764–771. [Google Scholar] [CrossRef] [PubMed]
  189. Franco, R.; Dufour, E.; Kosenko, E.; Bax, B.E.; Banz, A.; Skorokhod, O.A.; Lanao, M.; Vitvitsky, V.; Sinauridze, E.; Bourgeaux, V.; et al. International seminar on the red blood cells as vehicles for drugs. Expert Opin. Biol. Ther. 2012, 12, 127–133. [Google Scholar] [CrossRef] [Green Version]
  190. Foroozesh, M.; Zarrin, A. A novel combinatory paradigm for chronic hepatitis C treatment using liver-targeted carrier erythrocytes co-encapsulated with inter-feron alpha-2b, ribavirin and boceprevir. Irn. J. Med. Hypotheses Ideas 2010, 4, 10:1–10:8. Available online: http://citeseerx.ist.psu.edu/viewdoc/download?doi=10.1.1.921.5785&rep=rep1&type=pdf (accessed on 20 February 2020).
  191. Sabatino, R.; Antonelli, A.; Battistelli, S.; Schwendener, R.; Magnani, M.; Rossi, L. Macrophage depletion by free bisphosphonates and zoledronate-loaded red blood cells. PLoS ONE 2014, 9, e101260:1–e101260:12. [Google Scholar] [CrossRef]
  192. Shih, J.-Y.; Yuan, A.; Chen, J.J.-W.; Yang, P.-C. Tumor-associated macrophage : Its role in cancer invasion and metastasis. J. Cancer Mol. 2006, 2, 101–106. Available online: http://www.oalib.com/paper/2766208#.Xk6P9kpn2Ul (accessed on 20 February 2020).
  193. Solinas, G.; Germano, G.; Mantovani, A.; Allavena, P. Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation. J. Leukoc. Biol. 2009, 86, 1065–1073. [Google Scholar] [CrossRef] [Green Version]
  194. Fukuda, K.; Kobayashi, A.; Watabe, K. The role of tumor-associated macrophage in tumor progression. Front. Biosci. (Schol. Ed.). 2012, 4, 787–798. [Google Scholar] [CrossRef] [Green Version]
  195. Rossi, L.; Migliavacca, B.; Pierigé, F.; Serafini, S.; Sanvito, F.; Olivieri, S.; Nano, R.; Antonioli, B.; Magnani, M.; Bertuzzi, F. Prolonged islet allograft survival in diabetic mice upon macrophage depletion by clodronate-loaded erythrocytes. Transplantation 2008, 85, 648–650. [Google Scholar] [CrossRef]
  196. Pozzi, L.-A.M.; Maciaszek, J.W.; Rock, K.L. Both dendritic cells and macrophages can stimulate naive CD8 T cells in vivo to proliferate, develop effector function, and differentiate into memory cells. J. Immunol. 2005, 175, 2071–2081. [Google Scholar] [CrossRef]
  197. Magnani, M.; Chiarantini, L.; Vittoria, E.; Mancini, U.; Rossi, L.; Fazi, A. Red blood cells as an antigen-delivery system. Biotechnol. Appl. Biochem. 1992, 16, 188–194. [Google Scholar] [CrossRef]
  198. Chiarantini, L.; Argnanit, R.; Zucchinit, S.; Stevanatot, L.; Grossi, M.P.; Magnani, M.; Manservigi, R. Red blood cells as delivery system for recombinant HSV-1 glycoprotein B: Immunogenicity and protection in mice. Vaccine 1997, 15, 276–280. [Google Scholar] [CrossRef]
  199. Dominici, S.; Laguardia, M.E.; Serafini, G.; Chiarantini, L.; Fortini, C.; Tripiciano, A.; Scoglio, A.; Caputo, A.; Fiorelli, V.; Gavioli, R.; et al. Red blood cell-mediated delivery of recombinant HIV-1 Tat protein in mice induces anti-Tat neutralizing antibodies and CTL. Vaccine 2003, 21, 2073–2081. [Google Scholar] [CrossRef]
  200. Polvani, C.; Gasparini, A.; Benatti, U.; DeFlora, A.; Silvestri, S.; Volpini, G.; Nencioni, L. Murine red blood cells as efficient carriers of three bacterial antigens for the production of specific and neutralizing antibodies. Biotechnol. Appl. Biochem. 1991, 14, 347–356. [Google Scholar] [PubMed]
  201. Murray, A.M.; Pearson, I.F.S.; Fairbanks, L.D.; Chalmers, R.A.; Bain, M.D.; Bax, B.E. The mouse immune response to carrier erythrocyte entrapped antigens. Vaccine 2006, 24, 6129–6139. [Google Scholar] [CrossRef] [PubMed]
  202. Renno, T.; Lebecque, S.; Renard, N.; Saeland, S.; Vicari, A. What’s new in the field of cancer vaccines? Cell. Mol. Life Sci. 2003, 60, 1296–1310. [Google Scholar] [CrossRef]
  203. Melief, C.J. Cancer immunotherapy by dendritic cells. Immunity 2008, 29, 372–383. [Google Scholar] [CrossRef] [Green Version]
  204. Banz, A.; Cremel, M.; Rembert, A.; Godfrin, Y. In situ targeting of dendritic cells by antigen-loaded red blood cells: A novel approach to cancer immunotherapy. Vaccine 2010, 28, 2965–2972. [Google Scholar] [CrossRef]
  205. Hendrickson, J.E.; Chadwick, T.E.; Roback, J.D.; Hillyer, C.D.; Zimring, J.C. Inflammation enhances consumption and presentation of transfused RBC antigens by dendritic cells. Blood 2007, 110, 2736–2743. [Google Scholar] [CrossRef]
  206. Hendrickson, J.E.; Roback, J.D.; Hillyer, C.D.; Easley, K.A.; Zimring, J.C. Discrete Toll-like receptor agonists have differential effects on alloimmunization to transfused red blood cells. Transfusion 2008, 48, 1869–1877. [Google Scholar] [CrossRef]
  207. Banz, A.; Cremel, M.; Mouvant, A.; Guerin, N.; Horand, F.; Godfrin, Y. Tumor growth control using red blood cells as the antigen delivery system and poly(I:C). J. Immunother. 2012, 35, 409–417. [Google Scholar] [CrossRef]
  208. Cremel, M.; Guerin, N.; Campello, G.; Barthe, Q.; Berlier, W.; Horand, F.; Godfrin, Y. Innovative approach in Pompe disease therapy: Induction of immune tolerance by antigen-encapsulated red blood cells. Int. J. Pharm. 2015, 491, 69–77. [Google Scholar] [CrossRef] [PubMed]
  209. Cremel, M.; Guérin, N.; Horand, F.; Banz, A.; Godfrin, Y. Red blood cells as innovative antigen carrier to induce specific immune tolerance. Int. J. Pharm. 2013, 443, 39–49. [Google Scholar] [CrossRef] [PubMed]
  210. Khubutiya, M.S.; Gulyaev, V.A.; Khvatov, V.B.; Lemenev, V.L.; Kabanova, S.A.; Novruzbekov, M.S.; Lutsyk, K.N.; Olisov, O.D.; Zhuravel’, S.V.; Bulava, G.V.; et al. Immunological tolerance in organ transplantation. Transplantologiya (Russia) 2017, 9, 211–225. [Google Scholar] [CrossRef]
  211. Yamazaki, S.; Dudziak, D.; Heidkamp, G.F.; Fiorese, C.; Bonito, A.J.; Inaba, K.; Nussenzweig, M.C.; Steinman, R.M. CD8+ CD205+ splenic dendritic cells are specialized to induce Foxp3+ regulatory T cells. J. Immunol. 2008, 181, 6923–6933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Godfrin, Y.; Banz, A. Compostition and Therapeutic Anti-Tumiour Vaccine. US Patent 9,364,504 B2, 14 July 2016. Available online: https://patentimages.storage.googleapis.com/bb/3d/8f/d19346ec676e91/US9364504.pdf (accessed on 20 February 2020).
  213. Godfrin, Y.; Banz, A. Composition to Induce Specific Immune Tolerance. Canadian Patent Application CA2778669 A1, 5 May 2011. Available online: https://patentimages.storage.googleapis.com/f3/8c/8f/40cdf840aaaba0/CA2778669A1.pdf (accessed on 20 February 2020).
  214. Reimer, P.; Tombach, B. Hepatic MRI with SPIO: Detection and characterization of focal liver lesions. Eur. Radiol. 1998, 8, 1198–1204. [Google Scholar] [CrossRef] [PubMed]
  215. Waters, E.A.; Wickline, S.A. Contrast agents for MRI. Basic Res. Cardiol. 2008, 103, 114–121. [Google Scholar] [CrossRef]
  216. Sun, C.; Lee, J.S.H.; Zhang, M. Magnetic nanoparticles in MR imaging and drug delivery. Adv. Drug Deliv. Rev. 2008, 60, 1252–1265. [Google Scholar] [CrossRef] [Green Version]
  217. Veiseh, O.; Gunn, J.W.; Zhang, M. Design and fabrication of magnetic nanoparticles for targeted drug delivery and imaging. Adv. Drug Deliv. Rev. 2010, 62, 284–304. [Google Scholar] [CrossRef] [Green Version]
  218. Berry, C.C.; Curtis, A.S.G. Functionalisation of magnetic nanoparticles for applications in biomedicine. J. Phys. D. Appl. Phys. 2003, 36, R198–R206. [Google Scholar] [CrossRef]
  219. Corot, C.; Robert, P.; Idée, J.-M.; Port, M. Recent advances in iron oxide nanocrystal technology for medical imaging. Adv. Drug Deliv. Rev. 2006, 58, 1471–1504. [Google Scholar] [CrossRef]
  220. Weissleder, R.; Stark, D.D.; Engelstad, B.L.; Bacon, B.R.; Compton, C.C.; White, D.L.; Jacobs, P.; Lewis, J. Superparamagnetic iron oxide: Pharmacokinetics and toxicity. AJR Am. J. Roentgenol. 1989, 152, 167–173. [Google Scholar] [CrossRef] [PubMed]
  221. Wang, Y.-X.J. Current status of superparamagnetic iron oxide contrast agents for liver magnetic resonance imaging. World J. Gastroenterol. 2015, 21, 13400–13402. [Google Scholar] [CrossRef] [PubMed]
  222. Antonelli, A.; Pacifico, S.; Sfara, C.; Tamma, M.; Magnani, M. Ferucarbotran-loaded red blood cells as long circulating MRI contrast agents: First in vivo results in mice. Nanomedicine (Lond.) 2018, 13, 675–687. [Google Scholar] [CrossRef] [PubMed]
  223. Na, H.B.; Song, I.C.; Hyeon, T. Inorganic nanoparticles for MRI contrast agents. Adv. Mater. 2009, 21, 2133–2148. [Google Scholar] [CrossRef]
  224. Reimer, P.; Balzer, T. Ferucarbotran (Resovist): A new clinically approved RES-specific contrast agent for contrast-enhanced MRI of the liver: Properties, clinical development, and applications. Eur. Radiol. 2003, 13, 1266–1276. [Google Scholar] [CrossRef]
  225. Antonelli, A.; Sfara, C.; Mosca, L.; Manuali, E.; Magnani, M. New biomimetic constructs for improved in vivo circulation of superparamagnetic nanoparticles. J. Nanosci. Nanotechnol. 2008, 8, 2270–2278. [Google Scholar] [CrossRef]
  226. Antonelli, A.; Sfara, C.; Weber, O.; Pison, U.; Manuali, E.; Salamida, S.; Magnani, M. Characterization of ferucarbotran-loaded RBCs as long circulating magnetic contrast agents. Nanomedicine (Lond.) 2016, 21, 2781–2795. [Google Scholar] [CrossRef]
  227. Antonelli, A.; Magnani, M. Red blood cells as carriers of iron oxide-based contrast agents for diagnostic applications. J. Biomed. Nanotechnol. 2014, 10, 1732–1750. [Google Scholar] [CrossRef]
  228. Antonelli, A.; Sfara, C.; Manuali, E.; Bruce, I.J.; Magnani, M. Encapsulation of superparamagnetic nanoparticles into red blood cells as new carriers of MRI contrast agents. Nanomedicine (Lond.) 2011, 6, 211–223. [Google Scholar] [CrossRef]
  229. Antonelli, A.; Sfara, C.; Battistelli, S.; Canonico, B.; Arcangeletti, M.; Manuali, E.; Salamida, S.; Papa, S.; Magnani, M. New strategies to prolong the in vivo life span of iron-based contrast agents for MRI. PLoS ONE 2013, 8, e78542:1–e78542:17. [Google Scholar] [CrossRef] [Green Version]
  230. Boni, A.; Ceratti, D.; Antonelli, A.; Sfara, C.; Magnani, M.; Manuali, E.; Salamida, S.; Gozzi, A.; Bifone, A. USPIO-loaded red blood cells as a biomimetic MR contrast agent: A relaxometric study. Contrast Media Mol. Imaging 2014, 9, 229–236. [Google Scholar] [CrossRef] [PubMed]
  231. Zhang, K.; Cao, Y.; Kuang, Y.; Liu, M.; Chen, Y.; Wang, Z.; Hong, S.; Wang, J.; Pei, R. Gd2O3 and GH combined with red blood cells to improve the sensitivity of contrast agents for cancer targeting MR imaging. Biomater. Sci. 2016, 5, 46–49. [Google Scholar] [CrossRef] [PubMed]
  232. Ferrauto, G.; Delli Castelli, D.; Di Gregorio, E.; Langereis, S.; Burdinski, D.; Grüll, H.; Terreno, E.; Aime, S. Lanthanide-loaded erythrocytes as highly sensitive chemical exchange saturation transfer MRI contrast agents. J. Am. Chem. Soc. 2014, 136, 638–641. [Google Scholar] [CrossRef] [PubMed]
  233. Aryal, S.; Nguyen, T.D.T.; Pitchaimani, A.; Shrestha, T.B.; Biller, D.; Troyer, D. Membrane fusion-mediated gold nanoplating of red blood cell: A bioengineered CT-contrast agent. ACS Biomater. Sci. Eng. 2017, 3, 36–41. [Google Scholar] [CrossRef]
  234. Ritter, S.C.; Meissner, K.E. Loading of red blood cells with an analyte-sensitive dye for development of a long-term monitoring technique. In Proceedings of the Optical Diagnostics and Sensing XII: Toward Point-of-Care Diagnostics; and Design and Performance Validation of Phantoms Used in Conjunction with Optical Measurement of Tissue IV, San Francisco, CA, USA, 21–26 January 2012; Nordstrom, R.J., Coté, G.L., Eds.; SPIE BiOS: San Francisco, CA, USA, 2012; Volume 8229. [Google Scholar] [CrossRef] [Green Version]
  235. Bustamante Lopez, S.C.; Ritter, S.C.; Meissner, K.E. Developing strategies to enhance loading efficiency of erythrosensors. In Proceedings of the Optical Diagnostics and Sensing XIV: Toward Point-of-Care Diagnostics, SPIE BiOS, San Francisco, CA, USA, 3–6 February 2014; Coté, G.L., Ed.; SPIE: San Francisco, CA, USA, 2014; Volume 8951, pp. 895114:1–895114:6. [Google Scholar] [CrossRef]
  236. Bustamante Lopez, S.C.; Meissner, K.E. Characterization of carrier erythrocytes for biosensing applications. J. Biomed. Opt. 2017, 22, 091510:1–091510:8. [Google Scholar] [CrossRef] [Green Version]
  237. Borsakova, D.V.; Plakhotnik, M.E.; Koleva, L.D.; Bovt, E.A.; Alexandrovich, Y.G.; Ataullakhanov, F.I.; Sinauridze, E.I. Comparative methodological studies of l-asparaginase encapsulation into erythrocytes. Oncohematology 2018, 13, 91–101, (In Russian, abstract in English). [Google Scholar] [CrossRef]
  238. Rubius Therapeutics. Red Cell TherapeuticsTM. Available online: https://www.rubiustx.com/our-science/#red-cell-therapeutics (accessed on 20 February 2020).
  239. US National Library of Medicine. ClinicalTrials.gov. Safety and Tolerability of RTX-134 in Adults with Phenylketonuria. Available online: https://clinicaltrials.gov/ct2/show/NCT04110496 (accessed on 20 February 2020).
  240. Zhang, X.; Dastagir, S.R.; Subbiah, N.; Luo, M.; Soman, V.; Pawar, S.; McLaughlin, D.C.; Bayhi, N.; Amin, V.; Nissen, T.S.; et al. Engineered red-cell therapeutics (RCT) as artificial antigen presenting cells promote in vivo expansion and anti-tumor activity of antigen specific T cells. In Proceedings of the American Association for Cancer Research Annual Meeting 2019, Bioinformatics, Convergence Science, and Systems Biology, Atlanta, GA, USA, 29 March–3 April 2019; Volume 79 (suppl. 13). Abstract 3260. [Google Scholar] [CrossRef]
  241. Moore, C.L.; Pawar, S.; Nixon, M.; Lyford, T.J.; McLaughlin, D.C.; Dastagir, S.R.; Bracha, A.; Melancon, L.; Carpenter, C.L.; Wickham, T.J.; et al. Enabling the rapid generation of allogeneic artificial antigen presenting cell (aAPC) Red Cell Therapeutics with a loadable MHC system. In Proceedings of the Abstracts of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics, Boston, MA, USA, 26–30 October 2019; Volume 18 (suppl. 12). Abstract B062. [Google Scholar] [CrossRef]
  242. Trakarnsanga, K.; Griffiths, R.E.; Wilson, M.C.; Blair, A.; Satchwel, T.J.; Meinders, M.; Cogan, N.; Kupzig, S.; Kurita, R.; Nakamura, Y.; et al. An immortalized adult human erythroid linefacilitates sustainable and scalable generation of functional red cells. Nat. Commun. 2017, 8, 14750:1–14750:7. [Google Scholar] [CrossRef] [Green Version]
  243. Balzarini, J.; Pauwels, R.; Baba, M.; Herdewijn, P.; de Clercq, E.; Broder, S.; Johns, D.G. The in vitro and in vivo anti-retrovirus activity, and intracellular metabolism of 3’-azido-2’,3’-dideoxythymidine and 2’,3’-dideoxycytidine are highly dependent on the cell species. Biochem. Pharmacol. 1988, 37, 897–903. [Google Scholar] [CrossRef]
  244. Erytech. Pipeline. Available online: https://erytech.com/pipeline/ (accessed on 20 February 2020).
  245. EryDel. A Late Stage and Broad Pipeline. Available online: https://www.erydel.com/pipeline.php (accessed on 20 February 2020).
  246. Bax, B.E.; Levene, M.; Bain, M.D.; Fairbanks, L.D.; Filosto, M.; Kalkan Uçar, S.; Klopstock, T.; Kornblum, C.; Mandel, H.; Rahman, S.; et al. Erythrocyte encapsulated thymidine phosphorylase for the treatment of patients with mitochondrial neurogastrointestinal encephalomyopathy: Study protocol for a multi-centre, multiple dose, open label trial. J. Clin. Med. 2019, 8, 1096:1–1096:18. [Google Scholar] [CrossRef] [Green Version]
  247. European Medicines Agency. Science Medicines Health. EU/3/04/230. Available online: https://www.ema.europa.eu/en/medicines/human/orphan-designations/eu304230 (accessed on 20 February 2020).
  248. European Medicines Agency. Science Medicines Health. EU/3/09/633. Available online: https://www.ema.europa.eu/en/medicines/human/orphan-designations/eu309633 (accessed on 20 February 2020).
  249. European Medicines Agency. Science Medicines Health. EU/3/06/409. Available online: https://www.ema.europa.eu/en/medicines/human/orphan-designations/eu306409 (accessed on 20 February 2020).
  250. Dale, G.L.; Beutler, E. Enzyme replacement therapy in Gaucher’s disease: A rapid, high-yield method for purification of glucocerebrosidase. Proc. Natl. Acad. Sci. USA 1976, 73, 4672–4674. [Google Scholar] [CrossRef] [Green Version]
  251. Thorpe, S.R.; Fiddler, M.B.; Desnick, R.J. Enzyme therapy. V. In vivo fate of erythrocyte-entrapped β-glucuronidase in β-glucuronidase-deficient mice. Pediatr. Res. 1975, 9, 918–923. [Google Scholar] [CrossRef] [PubMed]
  252. Rossi, L.; Pierigè, F.; Carducci, C.; Gabucci, C.; Pascucci, T.; Canonico, B.; Bell, S.M.; Fitzpatrick, P.A.; Leuzzi, V.; Magnani, M. Erythrocyte-mediated delivery of phenylalanine ammonia lyase for the treatment of phenylketonuria in BTBR-Pah(enu2) mice. J. Control. Release 2014, 194, 37–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Bell, S.M.; Wendt, D.J.; Zhang, Y.; Taylor, T.W.; Long, S.; Tsuruda, L.; Zhao, B.; Laipis, P.; Fitzpatrick, P.A. Formulation and PEGylation optimization of the therapeutic PEGylated phenylalanine ammonia lyase for the treatment of phenylketonuria. PLoS ONE 2017, 12, e0173269:1–e0173269:17. [Google Scholar] [CrossRef] [PubMed]
  254. Gámez, A.; Wang, L.; Straub, M.; Patch, M.G.; Stevens, R.C. Toward PKU enzyme replacement therapy: PEGylation with activity retention for three forms of recombinant phenylalanine hydroxylase. Mol. Ther. 2004, 9, 124–129. [Google Scholar] [CrossRef]
  255. Magnani, M.; Mancini, U.; Bianchi, M.; Fazi, A. Comparison of uricase-bound and uricase-loaded erythrocytes as bioreactors for uric acid degradation. Adv. Exp. Med. Biol. 1992, 326, 189–194. [Google Scholar] [CrossRef]
  256. Ihler, G.; Lantzy, A.; Purpura, J.; Glew, R.H. Enzymatic degradation of uric acid by uricase-loaded human erythrocytes. J. Clin. Investig. 1975, 56, 595–602. [Google Scholar] [CrossRef]
  257. Hamarat Baysal, S.; Uslan, A.H. In vitro study of urease/AlaDH enzyme system encapsulated into human erythrocytes and research into its medical applications. Artif. Cells Blood Substit. Immobil. Biotechnol. 2002, 30, 71–77. [Google Scholar] [CrossRef]
  258. Hamarat Baysal, S.; Uslan, A.H. Encapsulation of urease and PEG-urease in erythrocyte. Artif. Cells Blood Substit. Immobil. Biotechnol. 2000, 28, 263–271. [Google Scholar] [CrossRef] [Green Version]
  259. Hamarat Baysal, S.; Uslan, A.H.; Pala, H.H.; Tunçoku, Ö. Encapsulation of PEG-urease/PEG-AlaDH within sheep erythrocytes and determination of the system’s activity in lowering blood levels of urea in animal models. Artif. Cells Blood Substit. Immobil. Biotechnol. 2007, 35, 391–403. [Google Scholar] [CrossRef]
  260. Bax, B.E.; Fairbanks, L.D.; Bain, M.D.; Simmonds, H.A.; Chalmers, R.A. The entrapment of polyethylene glycol-bound adenosine deaminase (Pegademase) in human carrier erythrocytes. Biochem. Soc. Trans. 1996, 24, 442S. [Google Scholar] [CrossRef] [Green Version]
  261. Levene, M.; Bain, M.; Moran, N.; Nirmalananthan, N.; Poulton, J.; Scarpelli, M.; Filosto, M.; Mandel, H.; MacKinnon, A.; Fairbanks, L.; et al. Safety and efficacy of erythrocyte encapsulated thymidine phosphorylase in mitochondrial neurogastrointestinal encephalomyopathy. J. Clin. Med. 2019, 8, 457:1–457:22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Levene, M.; Coleman, D.G.; Kilpatrick, H.C.; Fairbanks, L.D.; Gangadharan, B.; Gasson, C.; Bax, B.E. Preclinical toxicity evaluation of erythrocyte-encapsulated thymidine phosphorylase in BALB/c mice and beagle dogs: An enzyme-replacement therapy for mitochondrial neurogastrointestinal encephalomyopathy. Toxicol. Sci. 2013, 131, 311–324. [Google Scholar] [CrossRef] [PubMed]
  263. Adriaenssens, K.; Karcher, D.; Lowenthal, A.; Terheggen, H.G. Use of enzyme-loaded erythrocytes in in-vitro correction of arginase-deficient erythrocytes in familial hyperargininemia. Clin. Chem. 1976, 22, 323–326. [Google Scholar] [CrossRef] [PubMed]
  264. Muthuvel, A.; Rajamani, R.; Manikandan, S.; Sheeladevi, R. Detoxification of formate by formate dehydrogenase-loaded erythrocytes and carbicarb in folate-deficient methanol-intoxicated rats. Clin. Chim. Acta 2006, 367, 162–169. [Google Scholar] [CrossRef] [PubMed]
  265. Cannon, E.P.; Leung, P.; Hawkins, A.; Petrikovics, I.; Deloach, J.; Way, J.L. Antagonism of cyanide intoxication with murine carrier erythrocytes containing bovine rhodanese and sodium thiosulfate. J. Toxicol. Environ. Health 1994, 41, 267–274. [Google Scholar] [CrossRef] [PubMed]
  266. Petrikovics, I.; Pei, L.; McGuinn, W.D.; Cannon, E.P.; Way, J.L. Encapsulation of rhodanese and organic thiosulfonates by mouse erythrocytes. Toxicol. Sci. 1994, 23, 70–75. [Google Scholar] [CrossRef]
  267. Hamarat Baysal, S.; Uslan, A.H. Encapsulation of catalase and PEG-catalase in erythrocyte. Artif. Cells Blood Substit. Immobil. Biotechnol. 2001, 29, 359–366. [Google Scholar] [CrossRef]
  268. Plisson, C.; Hunault, M.; Thomas, X.; Legay, T.; Bertrand, Y.; Andre, T.; Godfrin, Y. l-Asparaginase loaded inside red cells has an acceptable tolerability profile on bilirubin value. Blood 2013, 122, 2642. [Google Scholar] [CrossRef]
  269. Sinauridze, E.I.; Vitvitsky, V.M.; Pichugin, A.V. A new chemotherapeutic agent: l-asparaginase entrapped in red blood cells. Adv. Exp. Med. Biol. 1992, 326, 203–206. [Google Scholar] [CrossRef]
  270. Kravtzoff, R.; Desbois, I.; Lamagnere, J.P.; Muh, J.P.; Valat, C.; Chassaigne, M.; Colombat, P.; Ropars, C. Improved pharmacodynamics of l-asparaginase-loaded in human red blood cells. Eur. J. Clin. Pharmacol. 1996, 49, 465–470. [Google Scholar] [CrossRef]
  271. Kravtzoff, R.; Ropars, C.; Laguerre, M.; Muh, J.P.; Chassaigne, M. Erythrocytes as carriers for l-asparaginase. Methodological and mouse in-vivo studies. J. Pharm. Pharmacol. 1990, 42, 473–476. [Google Scholar] [CrossRef] [PubMed]
  272. Lorenzi, P.L.; Horvath, T.D.; Martin, L.A.; Chan, W.K.; Du, D.; Hawke, D.H.; Weinstein, J.N.; Swart, K.J.; El-Hariry, I. Red blood cell-encapsulation of l-asparaginase favorably modulates target selectivity and pharmacodynamics. Blood 2016, 128, 1266. [Google Scholar] [CrossRef]
  273. Baruchel, A.; Bertrand, Y.; Thomas, X.; Blin, N.; Tavernier, E.; Ducassou, S.; Vey, N.; Gandemer, V.; Cacheux, V.; Mazingue, F.; et al. Updated clinical activity of Graspa versus native l-asparaginase in combination with cooprall regimen in Phase 3 randomized trial in patients with relapsed acute lymphoblastic leukemia (NCT01518517). Blood 2015, 126, 3723. [Google Scholar] [CrossRef]
  274. Updike, S.J. Entrapment of l-asparaginase in red blood cells. A strategy to improve treatment of acute lymphoblastic leukemia. Bibl. Haematol. 1985, 51, 65–74. [Google Scholar] [CrossRef]
  275. Naqi, A.; DeLoach, J.R.; Andrews, K.; Satterfield, W.; Keeling, M. Determination of parameters for enzyme therapy using l-asparaginase entrapped in canine erythrocytes. Biotechnol. Appl. Biochem. 1988, 10, 365–372. [Google Scholar] [CrossRef] [PubMed]
  276. DeLoach, J.R.; Andrews, K.; Satterfield, W.; Keeling, M. Intraperitoneal administration of carrier erythrocytes in dogs: An improved method for delivery of l-asparaginase. Biotechnol. Appl. Biochem. 1990, 12, 331–335. [Google Scholar] [CrossRef] [PubMed]
  277. Ktavtzoff, R.; Desbois, I.; Doinel, C.; Colombat, P.; Lamagnere, J.P.; Chassaigne, M.; Ropars, C. Immunological response to l-asparaginase loaded into red blood cells. Adv. Exp. Med. Biol. 1992, 326, 175–182. [Google Scholar] [CrossRef]
  278. Garin, M.I.; Kravtzoff, R.; Chestier, N.; Sanz, S.; Pinilla, M.; Luque, J.; Ropars, C. Density gradient separation of l-asparaginase-loaded human erythrocytes. Biochem. Mol. Biol. Int. 1994, 33, 807–814. [Google Scholar]
  279. Kravtzoff, R.; Colombat, P.H.; Desbois, I.; Linassier, C.; Muh, J.P.; Philip, T.; Blay, J.Y.; Gardenbas, M.; Poumier-Gaschard, P.; Lamagnere, J.P.; et al. Tolerance evaluation of l-asparaginase loaded in red blood cells. Eur. J. Clin. Pharmacol. 1996, 51, 221–225. [Google Scholar] [CrossRef]
  280. Hunault-Berger, M.; Leguay, T.; Huguet, F.; Leprêtre, S.; Deconinck, E.; Uribe, M.O.; Bonmati, C.; Bories, P.; Himberlin, C.; Chevallier, P.; et al. Two years follow-up results of Graspall/Graall-SA2–2008 study: l-asparaginase-loaded red blood cell combined with standard EWALL chemotherapy in older patients with newly diagnosed Philadelphia chromosome-negative acute lymphoblastic leukemia (Ph-ALL). Blood 2012, 120, 1473. [Google Scholar] [CrossRef]
  281. Agrawal, V.; Woo, J.H.; Borthakur, G.; Kantarjian, H.; Frankel, A.E. Red blood cell-encapsulated l-asparaginase: Potential therapy of patients with asparagine synthetase deficient acute myeloid leukemia. Protein Pept. Lett. 2013, 20, 392–402. [Google Scholar] [CrossRef] [PubMed]
  282. Bertrand, Y.; Baruchel, A.; Thomas, X.; Blin, N.; Tavernier, E.; Ducassou, S.; Vey, N.; Gandemer, V.; Cacheux, V.; Mazingue, F.; et al. Evaluation of the impact of the presence of neutralizing l-asparaginase antibodies on the efficacy and safety of Graspa in Phase 3 randomized trial versus native l-asparaginase in patients with relapsed acute lymphoblastic leukemia (NCT01518517). Blood 2015, 126, 3734. [Google Scholar] [CrossRef]
  283. Bertrand, Y.; Dombret, H.; Quesnel, B.; Stephan, J.-L.; Schmitt, C.; Lissandre, S.; Poiree, M.; Recher, C.; Plouvier, E.; Dumesnil de Maricourt, C.; et al. Expanded access program of Graspa for treatment of patients with acute lymphoblastic leukemia unable to receive other form of l-asparaginase—A status update (NCT02197650). Blood 2015, 126, 4877. [Google Scholar] [CrossRef]
  284. Thomas, X.; Le Jeune, C. Erythrocyte encapsulated l-asparaginase (GRASPA) in acute leukemia. Int. J. Hematol. Oncol. 2016, 5, 11–25. [Google Scholar] [CrossRef] [PubMed]
  285. Machover, D.; Rossi, L.; Hamelin, J.; Desterke, C.; Goldschmidt, E.; Chadefaux-Vekemans, B.; Bonnarme, P.; Briozzo, P.; Kopečný, D.; Pierigè, F.; et al. Effects in cancer cells of the recombinant l-methionine gamma-lyase from Brevibacterium aurantiacum. Encapsulation in human erythrocytes for sustained l-methionine elimination. J. Pharmacol. Exp. Ther. 2019, 369, 489–502. [Google Scholar] [CrossRef]
  286. Gay, F.J.; Bourgeaux, V.; Godfrin, Y. Methioninase-loaded erythrocytes: A promising drug for l-methionine restriction therapy in cancer. In Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; Philadelphia, PA, USA, 18–22 April 2015, Volume 75, (Suppl. 15), Abstract nr 5330. [CrossRef]
  287. Rossi, L.; Bianchi, M.; Magnani, M. Increased glucose metabolism by enzyme-loaded erythrocytes in vitro and in vivo normalization of hyperglycemia in diabetic mice. Biotechnol. Appl. Biochem. 1992, 15, 207–216. [Google Scholar] [CrossRef]
  288. Rossi, L.; Bianchi, M.; Fraternale, A.; Magnani, M. Normalization of hyperglycemia in diabetic mice by enzyme-loaded erythrocytes. Adv. Exp. Med. Biol. 1992, 326, 183–188. [Google Scholar] [CrossRef]
  289. Xia, D.; He, H.; Wang, Y.; Wang, K.; Zuo, H.; Gu, H.; Xu, P.; Hu, Y. Ultrafast glucose-responsive, high loading capacity erythrocyte to self-regulate the release of insulin. Acta Biomater. 2018, 69, 301–312. [Google Scholar] [CrossRef]
  290. Al-Achi, A.; Greenwood, R. Human insulin binding to erythrocyte-membrane. Drug Dev. Ind. Pharm. 1993, 19, 673–684. [Google Scholar] [CrossRef]
  291. Villereal, M.C.; Ropars, C.; Hurel, C.; Teisseire, B.; Chassaigne, M.; Itti, R.; Casset, D.; Nicolau, C. Oxygen transport to tissue modified by entrapment of an allosteric effector of haemoglobin in erythrocytes. Folia Haematol. Int. Mag. Klin. Morphol. Blutforsch. 1987, 114, 488–492. [Google Scholar]
  292. Teisseire, B.; Ropars, C.; Villereal, M.C.; Nicolau, C. Long-term physiological effects of enhanced O2 release by inositol hexaphosphate-loaded erythrocytes. Proc. Natl. Acad. Sci. USA 1987, 84, 6894–6898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  293. Bailleul, C.; Borrelly-Villereal, M.C.; Chassaigne, M.; Ropars, C. Modification of partial pressure of oxygen (P50) in mammalian red blood cells by incorporation of an allosteric effector of hemoglobin. Biotechnol. Appl. Biochem. 1989, 11, 31–40. [Google Scholar] [CrossRef] [PubMed]
  294. Mouneimne, Y.; Barhoumi, R.; Myers, T.; Slogoff, S.; Nicolau, C. Stable rightward shifts of the oxyhemoglobin dissociation curve induced by encapsulation of inositol hexaphosphate in red blood cells using electroporation. FEBS Lett. 1990, 275, 117–120. [Google Scholar] [CrossRef] [Green Version]
  295. Bourget, G.; Boucher, L.; Ropars, C. Density gradient separation of inositol hexaphosphate loaded red blood cells in various preparation conditions. Adv. Exp. Med. Biol. 1992, 326, 27–33. [Google Scholar] [CrossRef]
  296. Villa, S.; Rossi, F.; Biondi, P.A.; Russo, V.; Crimella, T.; Fiorelli, G.; Zanella, A. Determination of inositol hexaphosphate (IHP) in human IHP-loaded red blood cells by a simple high performance liquid chromatography method. Adv. Exp. Med. Biol. 1992, 326, 41–49. [Google Scholar] [CrossRef]
  297. Boucher, L.; Chassaigne, M.; Ropars, C. Internalization and distribution of inositol hexakisphosphate in red blood cells. Biotechnol. Appl. Biochem. 1996, 24, 73–78. [Google Scholar] [CrossRef]
  298. Ropars, C.; Chassaigne, M.; Avenard, G. Engineered erythrocytes: Influence of P50 rightward shift and oxemia on oxygen transport to tissues. Med. Biol. Eng. Comput. 1998, 36, 508–512. [Google Scholar] [CrossRef]
  299. Nicolau, C.; Gersonde, K. Incorporation of inositol hexaphosphate into intact red blood cells. I. Fusion of effector-containing lipid vesicles with erythrocytes. Naturwissenschaften 1979, 66, 563–566. [Google Scholar] [CrossRef]
  300. Gersonde, K.; Nicolau, C. Incorporation of inositol hexaphosphate into intact red blood cells. II. Enhancement of gas transport in inositol hexaphosphate-loaded red blood cells. Naturwissenschaften 1979, 66, 567–570. [Google Scholar] [CrossRef]
  301. Teisseire, B.; Ropars, C.; Nicolau, C.; Vallez, M.O.; Chassaigne, M. Enhancement of P50 by inositol hexa phosphate entrapped in resealed erythrocytes in piglets. Adv. Exp. Med. Biol. 1984, 180, 673–677. [Google Scholar] [CrossRef]
  302. Kruse, C.A.; Freehauf, C.L.; Patel, K.R.; Baldeschwieler, J.D. Mouse erythrocyte carriers osmotically loaded with methotrexate. Biotechnol. Appl. Biochem. 1987, 9, 123–140. [Google Scholar] [CrossRef] [PubMed]
  303. Kruse, C.A.; Mierau, G.W.; James, G.T. Methotrexate loading of red cell carriers by osmotic stress and electric-pulse methods: Ultrastructural observations. Biotechnol. Appl. Biochem. 1989, 11, 571–580. [Google Scholar] [CrossRef] [PubMed]
  304. Tyrrell, D.A.; Ryman, B.E. The Entrapment of therapeutic agents in resealed erythrocyte ‘ghosts’ and their fate in vivo. Biochem. Soc. Trans. 1976, 4, 677–680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  305. Kitao, T.; Hattori, K.; Takeshita, M. Agglutination of leukemic cells and daunomycin entrapped erythrocytes with lectin in vitro and in vivo. Experientia 1978, 34, 94–95. [Google Scholar] [CrossRef] [PubMed]
  306. Kitao, T.; Hattori, K. Erythrocyte entrapment of daunomycin by amphotericin B without hemolysis. Cancer Res. 1980, 40, 1351–1353. [Google Scholar] [PubMed]
  307. De Flora, A.; Benatti, U.; Guida, L.; Zocchi, E. Encapsulation of adriamycin in human erythrocytes. Proc. Natl. Acad. Sci. USA 1986, 83, 7029–7033. [Google Scholar] [CrossRef] [Green Version]
  308. Zocchi, E.; Tonetti, M.; Polvani, C.; Guida, L.; Benatti, U.; De Flora, A. In vivo liver and lung targeting of adriamycin encapsulated in glutaraldehyde-treated murine erythrocytes. Biotechnol. Appl. Biochem. 1988, 10, 555–562. [Google Scholar] [CrossRef]
  309. Zocchi, E.; Tonetti, M.; Polvani, C.; Guida, L.; Benatti, U.; De Flora, A. Encapsulation of doxorubicin in liver-targeted erythrocytes increases the therapeutic index of the drug in a murine metastatic model. Proc. Natl. Acad. Sci. USA 1989, 86, 2040–2044. [Google Scholar] [CrossRef] [Green Version]
  310. Gaudreault, R.C.; Bellemare, B.; Lacroix, J. Erythrocyte membrane-bound daunorubicin as a delivery system in anticancer treatment. Anticancer Res. 1989, 9, 1201–1205. [Google Scholar]
  311. Tonetti, M.; Astroff, A.B.; Satterfield, W.; De Flora, A.; Benatti, U.; DeLoach, J.R. Pharmacokinetic properties of doxorubicin encapsulated in glutaraldehyde-treated canine erythrocytes. Am. J. Vet. Res. 1991, 52, 1630–1635. [Google Scholar]
  312. Tonetti, M.; Polvani, C.; Zocchi, E.; Guida, L.; Benatti, U.; Biassoni, P.; Romei, F.; Guglielmi, A.; Aschele, C.; Sobrero, A.; et al. Liver targeting of autologous erythmocytes loaded with doxorubicin. Eur J. Cancer 1991, 27, 947–948. [Google Scholar] [CrossRef]
  313. Gasparini, A.; Chiarantini, L.; Kirch, H.; DeLoach, J.R. In vitro targeting of doxorubicin loaded canine erythrocytes to cytotoxic T-lymphocytes (CTLL). Adv. Exp. Med. Biol. 1992, 326, 291–297. [Google Scholar] [CrossRef] [PubMed]
  314. Ataullakhanov, F.I.; Vitvitsky, V.M.; Kovaleva, V.L.; Mironova, S.B. Rubomycin loaded erythrocytes in the treatment of mouse tumor P388. Adv. Exp. Med. Biol. 1992, 326, 209–212. [Google Scholar] [CrossRef] [PubMed]
  315. Ataullakhanov, F.I.; Batasheva, T.V.; Bukhman, V.M.; Komarova, S.V.; Oreshkina, T.D.; Vitvitsky, V.M. Treatment of Rausher virus induced murine erythroblastic leukemia with rubomycin loaded erythrocytes. Adv. Biosci. 1994, 92, 177–183. [Google Scholar]
  316. Ataullakhanov, F.I.; Kulikova, E.V.; Vitvitsky, V.M. Doxorubicin binding by human erythrocytes. Adv. Biosci. 1994, 92, 163–168. [Google Scholar]
  317. Tikhonova, A.G.; Aleksandrovich, I.G.; Vuĭmo, T.A.; Sinauridze, E.I.; Ataullakhanov, F.I. Erythrocytes as carriers of anthracycline antibiotics. Ter. Arkh. 2008, 80, 91–94, (In Russian, abstract in English). [Google Scholar]
  318. Benatti, U.; Zocchi, E.; Tonetti, M.; Guida, L.; Polvani, C.; De Flora, A. Enhanced antitumor activity of adriamycin by encapsulation in mouse erythrocytes targeted to liver and lungs. Pharmacol. Res. 1989, 21 (Suppl. 2), 27–33. [Google Scholar] [CrossRef]
  319. Briones, E.; Colino, C.I.; Millán, C.G.; Lanao, J.M. Increasing the selectivity of amikacin in rat peritoneal macrophages using carrier erythrocytes. Eur. J. Pharm. Sci. 2009, 38, 320–324. [Google Scholar] [CrossRef]
  320. Millan, C.G.; Castaneda, A.Z.; Lopez, F.G.; Marinero, M.L.S.; Lanao, J.M. Pharmacokinetics and biodistribution of amikacin encapsulated in carrier erythrocytes. J. Antimicrob. Chemother. 2008, 61, 375–381. [Google Scholar] [CrossRef]
  321. Millan, C.G.; Bax, B.E.; Castañeda, A.Z.; Marinero, M.L.S.; Lanao, J.M. In vitro studies of amikacin-loaded human carrier erythrocytes. Transl. Res. 2008, 152, 59–66. [Google Scholar] [CrossRef]
  322. Millán, C.G.; Castañeda, A.Z.; López, F.G.; Marinero, M.L.S.; Lanao, J.M.; Arévalo, M. Encapsulation and In Vitro Evaluation of Amikacin-Loaded Erythrocytes. Drug Deliv. 2005, 12, 409–416. [Google Scholar] [CrossRef] [PubMed]
  323. Eichler, H.G.; Rameis, H.; Bauer, K.; Korn, A.; Bacher, S.; Gasic, S. Survival of gentamicin-loaded carrier erythrocytes in healthy human volunteers. Eur. J. Clin. Investig. 1986, 16, 39–42. [Google Scholar] [CrossRef] [PubMed]
  324. DeLoach, J.R.; Wagner, G.G. Pharmacokinetics of tetracycline encapsulated in bovine carrier erythrocytes. Am. J. Vet. Res. 1984, 45, 640–642. [Google Scholar] [PubMed]
  325. DeLoach, J.R.; Barton, C. Circulating carrier erythrocytes: Slow-release vehicle for an antileukemic drug, cytosine arabinoside. Am. J. Vet. Res. 1982, 43, 2210–2212. [Google Scholar]
  326. Tonetti, M.; Gasparini, A.; Giovine, M.; Benatti, U.; De Flora, A. Interactions of carboplatin with human erythrocytes and murine erythroleukemic cells. Adv. Exp. Med. Biol. 1992, 326, 223–232. [Google Scholar] [CrossRef]
  327. Gasparini, A.; Giovine, M.; Damonte, G.; Tonetti, M.; Grandi, T.; Mazzei, M.; Balbi, A.; Silvestro, L.; Benatti, U.; De Flora, A. A novel dimeric fluoropyrimidine molecule behaves as a remote precursor of 5-fluoro-2′-deoxyuridine in human erythrocytes. Biochem. Pharmacol. 1994, 48, 1121–1128. [Google Scholar] [CrossRef]
  328. De Flora, A.; Zocchi, E.; Guida, L.; Polvani, C.; Benatti, U. Conversion of encapsulated 5-fluoro-2′-deoxyuridine 5′-monophosphate to the antineoplastic drug 5-fluoro-2′-deoxyuridine in human erythrocytes. Proc. Natl. Acad. Sci. USA 1988, 85, 3145–3149. [Google Scholar] [CrossRef] [Green Version]
  329. Lynch, W.E.; Sartiano, G.P.; Ghaffar, A. Erythrocytes as carriers of chemotherapeutic agents for targeting the reticuloendothelial system. Am. J. Hematol. 1980, 9, 249–259. [Google Scholar] [CrossRef]
  330. Trineeva, O.V.; Halahakoon, A.D.; Slivkin, A.I.; Chupandina, E.E. Morphological and physico-chemical properies of erythrocyte carriers incapsulated by terpineindolic alkaloids. Drug Dev. Regist. (Russia) 2018, 1, 146–150, (In Russian, abstract in English). Available online: https://www.pharmjournal.ru/jour/article/view/557?locale=ru_RU (accessed on 20 February 2020).
  331. Harisa, G.I.; Ibrahim, M.F.; Alanazi, F.; Shazly, G.A. Engineering erythrocytes as a novel carrier for the targeted delivery of the anticancer drug paclitaxel. Saudi Pharm. J. 2014, 22, 223–230. [Google Scholar] [CrossRef] [Green Version]
  332. Pierige, F.; De Marco, C.; Orlotti, N.; Dominici, S.; Biagiotti, S.; Serafini, S.; Zaffaroni, N.; Magnani, M.; Rossi, L. Cytotoxic activity of 2-Fluoro-ara-AMP and 2-Fluoro-ara-AMP-loaded erythrocytes against human breast carcinoma cell lines. Int. J. Oncol. 2010, 37, 133–142. [Google Scholar] [CrossRef] [PubMed]
  333. Fraternale, A.; Rossi, L.; Magnani, M. Encapsulation, metabolism and release of 2-fluoro-ara-AMP from human erythrocytes. Biochim. Biophys. Acta (Gener. Sub.) 1996, 1291, 149–154. [Google Scholar] [CrossRef]
  334. Cervasi, B.; Paiardini, M.; Serafini, S.; Fraternale, A.; Menotta, M.; Engram, J.; Lawson, B.; Staprans, S.I.; Piedimonte, G.; Perno, C.F.; et al. Administration of fludarabine-loaded autologous red blood cells in simian immunodeficiency virus-infected sooty mangabeys depletes pSTAT-1-expressing macrophages and delays the rebound of viremia after suspension of antiretroviral therapy. J. Virol. 2006, 80, 10335–10345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  335. Rossi, L.; Pierigè, F.; Andriulli, A.; Magnani, M. Erythrocytes as pharmacological carriers for corticosteroids and other drugs in patients with inflammatory bowel diseases. In Drug Delivery; Popesku, M.A., Ed.; Nova Science Publishers Inc.: Hauppauge, NY, USA, 2011; Chapter 5; pp. 135–145. ISBN 978-1-61324-538-5. Available online: http://www.novapublishers.org/catalog/product_info.php?products_id=30120 (accessed on 20 February 2020).
  336. Coker, S.; Szczepiorkowski, Z.M.; Seigel, A.H.; Ferrari, A.; Benatti, L.; Mambrini, G.; Anand, R.; Hartman, R.D.; Dumont, L.J. The in vivo recovery/survival and pharmacokinetic properties of dexamethasone sodium phosphate encapsulated in autologous erythrocytes. Blood 2016, 128, 2629. [Google Scholar] [CrossRef]
  337. Mambrini, G.; Mandolini, M.; Rossi, L.; Pierige, F.; Capogrossi, G.; Salvati, P.; Serafini, S.; Benatti, L.; Magnani, M. Ex vivo encapsulation of dexamethasone sodium phosphate into human autologous erythrocytes using fully automated biomedical equipment. Int. J. Pharm. 2017, 517, 175–184. [Google Scholar] [CrossRef] [PubMed]
  338. Coker, S.A.; Szczepiorkowski, Z.M.; Siegel, A.H.; Ferrari, A.; Mambrini, G.; Anand, R.; Hartman, R.D.; Benatti, L.; Dumont, L.J. A Study of the pharmacokinetic properties and the in vivo kinetics of erythrocytes loaded with dexamethasone sodium phosphate in healthy volunteers. Transfus. Med. Rev. 2018, 32, 102–110. [Google Scholar] [CrossRef] [PubMed]
  339. Annese, V.; Latiano, A.; Rossi, L.; Bossa, F.; Damonte, G.; Dallapiccola, B.; Serafini, S.; Pierigé, F.; Andriulli, A.; Magnani, M. The polymorphism of multi-drug resistance 1 gene (MDR1) does not influence the pharmacokinetics of Dexamethasone loaded into autologous erythrocytes of patients with inflammatory bowel disease. Eur. Rev. Med. Pharmacol. Sci. 2006, 10, 27–31. [Google Scholar]
  340. Ogiso, T.; Iwaki, M.; Ohtori, A. Encapsulation of dexametasone in rabit erythrocytes, the disposition in circulation and anti-inflamatory effect. J. Pharmacobio-Dyn 1985, 8, 1032–1040. [Google Scholar] [CrossRef] [Green Version]
  341. Crinelli, R.; Antonelli, A.; Bianchi, M.; Gentilini, L.; Scaramucci, S.; Magnani, M. Selective inhibition of NF-kB activation and TNF-alpha production in macrophages by red blood cell-mediated delivery of dexamethasone. Blood Cells. Mol. Dis. 2000, 26, 211–222. [Google Scholar] [CrossRef]
  342. Rossi, L.; Serafini, S.; Cenerini, L.; Picardi, F.; Bigi, L.; Panzani, I.; Magnani, M. Erythrocyte-mediated delivery of dexamethasone in patients with chronic obstructive pulmonary disease. Biotechnol. Appl. Biochem. 2001, 33, 85–89. [Google Scholar] [CrossRef]
  343. Lucidi, V.; Tozzi, A.E.; Bella, S.; Turchetta, A. A pilot trial on safety and efficacy of erythrocyte-mediated steroid treatment in CF patients. BMC Pediatr. 2006, 6, 17:1–17:6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  344. Zhang, X.; Qiu, M.; Guo, P.; Lian, Y.; Xu, E.; Su, J. Autologous red blood cell delivery of betamethasone phosphate sodium for long anti-inflammation. Pharmaceutics 2018, 10, 286:1–286:12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  345. Shavi, G.V.; Doijad, R.C.; Deshpande, P.B.; Manvi, F.V.; Meka, S.R.; Udupa, N.; Omprakash, R.; Dhirendra, K. Erythrocytes as carrier for prednisolone: In vitro and in vivo evaluation. Pak. J. Pharm. Sci. 2010, 23, 194–200. [Google Scholar] [PubMed]
  346. Bukara, K.; Drvenica, I.; Ilic, V.; Stancic, A.; Misic, D.; Vasic, B.; Gajic, R.; Vucetic, D.; Kiekens, F.; Bugarski, B. Comparative studies on osmosis based encapsulation of sodium diclofenac in porcine and outdated human erythrocyte ghosts. J. Biotechnol. 2016, 240, 14–22. [Google Scholar] [CrossRef] [PubMed]
  347. Franchetti, P.; Cappellacci, L.; Petrelli, R.; Vita, P.; Grifantini, M.; Rossi, L.; Pierigé, F.; Serafini, S.; Magnani, M.; Balestra, E.; et al. Inhibition of HIV-1 replication in macrophages by red blood cell-mediated delivery of a heterodinucleotide of azidothymidine and 9-(R)-2-(phosphono methoxypropyl)adenine. Antivir. Chem. Chemother. 2001, 12, 151–159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  348. Magnani, M.; Casabianca, A.; Rossi, L.; Fraternale, A.; Brandi, G.; Silvotti, L.; Pledimonte, G. Inhibition of HIV-1 and LP-BM5 replication in macrophages by dideoxycytidine and dideoxycytidine 5′-triphosphate. Antivir. Chem. Chemother. 1995, 6, 312–319. [Google Scholar] [CrossRef]
  349. Briones, E.; Colino, C.I.; Lanao, J.M. Study of the factors influencing the encapsulation of zidovudine in rat erythrocytes. Int. J. Pharm. 2010, 401, 41–46. [Google Scholar] [CrossRef]
  350. Fraternale, A.; Casabianca, A.; Rossi, L.; Chiarantini, L.; Brandi, G.; Aluigi, G.; Schiavano, G.F.; Magnani, M. Inhibition of Murine AIDS by Combination of AZT and Dideoxycytidine 5′-Triphosphate. J. Acquir. Immune Defic. Syndr. 1996, 12, 164–173. [Google Scholar] [CrossRef]
  351. Magnani, M.; Rossi, L.; Casabianca, A.; Fraternale, A.; Schiavano, G.; Brandi, G.; Mannello, F.; Piedimonte, G. Red blood cells as advanced drug delivery systems for antiviral nucleoside analogues. Adv. Exp. Med. Biol. 1992, 326, 239–245. [Google Scholar] [CrossRef]
  352. Magnani, M.; Rossi, L.; Chiarantini, L.; Fraternale, A.; Casabianca, A. Red blood cells as carriers of drugs against retroviruses. In Targeting of Drugs 4: Advances in System Constructs; NATO ASI Series (Series A: Life Sciences); Gregoriadis, G., McCormack, B., Poste, G., Eds.; Springer: Boston, MA, USA, 1994; Volume 273, pp. 147–152. ISBN 978-1-4899-1209-1. [Google Scholar]
  353. Fraternale, A.; Casabianca, A.; Tonelli, A.; Chiarantini, L.; Brandi, G.; Magnani, M. New drug combinations for the treatment of murine AIDS and macrophage protection. Eur. J. Clin. Investig. 2001, 31, 248–252. [Google Scholar] [CrossRef]
  354. Fraternale, A.; Paoletti, M.F.; Casabianca, A.; Orlandi, C.; Millo, E.; Balestra, E.; Damonte, G.; Perno, C.F.; Magnani, M. Erythrocytes as carriers of antisense PNA addressed against HIV-1 gag-pol transframe domain. J. Drug Target. 2009, 17, 278–285. [Google Scholar] [CrossRef] [PubMed]
  355. Walid, A.A.-R.; Salama, A.; Afouna, M.I.; Samy, A.M. Ribavirin loaded erythrocytes by endocytosis as targeted drug carrier system. Univers. J. Pharm. Res. 2016, 1, 38–53. [Google Scholar] [CrossRef]
  356. Corinti, S.; Chiarantini, L.; Dominici, S.; Laguardia, M.; Magnani, M.; Girolomoni, G. Erythrocytes deliver Tat to interferon-γ-treated human dendritic cells for efficient initiation of specific type 1 immune responses in vitro. J. Leukoc. Biol. 2002, 71, 652–658. [Google Scholar] [CrossRef] [PubMed]
  357. Chiarantini, L.; Matteucci, D.; Pistello, M.; Mancini, U.; Mazzetti, P.; Massi, C.; Giannecchini, S.; Lonetti, I.; Magnani, M.; Bendinelli, M. AIDS vaccination studies using an ex vivo feline immunodeficiency virus model: Homologous erythrocytes as a delivery system for preferential immunization with putative protective antigens. Clin. Diagn. Lab. Immunol. 1998, 5, 235–241. [Google Scholar] [CrossRef] [Green Version]
  358. Godfrin, Y.; Horand, F.; Cremel, M. Can red blood cells prove to be a useful tool in tumor immunotherapy? Immunotherapy 2012, 4, 871–873. [Google Scholar] [CrossRef]
  359. Hamidi, M.; Tajerzadeh, H.; Dehpour, A.R.; Rouini, M.R.; Ejtemaee-Mehr, S. In vitro characterization of human intact erythrocytes loaded by enalaprilat. Drug Deliv. 2001, 8, 223–230. [Google Scholar] [CrossRef]
  360. Tajerzadeh, H.; Hamidi, M. Evaluation of hypotonic preswelling method for encapsulation of enalaprilat in intact human erythrocytes. Drug Dev. Ind. Pharm. 2000, 26, 1247–1257. [Google Scholar] [CrossRef]
  361. Hamidi, M.; Tajerzadeh, H.; Dehpour, A.R.; Ejtemaee-Mehr, S. Inhibition of serum angiotensin-converting enzyme in rabbits after intravenous administration of enalaprilat-loaded intact erythrocytes. J. Pharm. Pharmacol. 2001, 53, 1281–1286. [Google Scholar] [CrossRef]
  362. Foroozesh, M.; Hamidi, M.; Zarrin, A.; Mohammadi-Samani, S.; Montaseri, H. Preparation and in-vitro characterization of tramadol-loaded carrier erythrocytes for long-term intravenous delivery. J. Pharm. Pharmacol. 2011, 63, 322–332. [Google Scholar] [CrossRef]
  363. DeLoach, J.R.; Andrews, K.; Sheffield, C.L. Encapsulation of interleukin-2 in murine erythrocytes and subsequent deposition in mice receiving a subcutaneous injection. Biotechnol. Appl. Biochem. 1988, 10, 183–190. [Google Scholar] [CrossRef]
  364. Moyes, R.B.; Kirch, H.; DeLoach, J.R. Enhanced biological activity of human recombinant interleukin 2 coupled to mouse red blood cells as evaluated using the mouse Meth A sarcoma model. Biotechnol. Appl. Biochem. 1996, 23, 29–36. [Google Scholar] [PubMed]
  365. Kirch, H.J.; Moyes, R.B.; Chiarantini, L.; DeLoach, J.R. Effect of targeted erythrocytes coated with recombinant human interleukin 2 on T-lymphocyte proliferation in vitro. Biotechnol. Appl. Biochem. 1994, 19, 331–340. [Google Scholar] [CrossRef] [PubMed]
  366. Mitchell, D.H.; James, G.T.; Kruse, C.A. Bioactivity of electric field-pulsed human recombinant interleukin-2 and its encapsulation into erythrocyte carriers. Biotechnol. Appl. Biochem. 1990, 12, 264–275. [Google Scholar] [CrossRef] [PubMed]
  367. Olmos, G.; Lotero, L.A.; Tejedor, M.C.; Diez, J.C. Delivery to macrophages of interleukin 3 loaded in mouse erythrocytes. Biosci. Rep. 2000, 20, 399–410. [Google Scholar] [CrossRef] [PubMed]
  368. Naiyang, F.; Erxian, Z.; Daben, P.; Xusheng, C.; Zhefu, L.; Lijun, Y. [Rat erythrocytes as a new kind of carrier for superoxide dismutase (SOD) at cellular level]. Chinese J. Biochem. Mol. Biol. 2000, 16, 275–280, (In Chinese, Abstract in English). Available online: https://europepmc.org/article/cba/334132 (assesed on 25 February 2020).
  369. Wang, X.; Yang, J.; Liu, F. Superoxide dismutase encapsulated erythrocytes used in the study of cerebral ischemia-reperfusion. Zhongguo Yi Xue Ke Xue Yuan Xue Bao 1996, 18, 392–396, (In Chinese, Abstract in English). [Google Scholar]
  370. Wang, X.; Zhou, H.; Yang, J. GSH. Px or SOD encapsulated erythrocytes in the study of cerebral ischemia-reperfusion. Zhonghua Yi Xue Za Zhi 1997, 77, 43–46, (In Chinese, Abstract in English). [Google Scholar]
  371. Wang, L.; Zhang, Y.; Xu, Y. Red blood cell as a carrier of superoxide dismutase. Space Med. Med. Eng. (Beijing) 1997, 10, 430–433, (In Chinese, Abstract in English). [Google Scholar]
  372. Larson, G.; Pieterse, A.; Quick, G.; van der Bijl, P.; van Zyl, J.; Hawtrey, A. Development of a reproducible procedure for plasmid DNA encapsulation by red blood cell ghosts. BioDrugs 2004, 18, 189–198. [Google Scholar] [CrossRef]
  373. Byun, H.M.; Suh, D.; Yoon, H.; Kim, J.M.; Choi, H.G.; Kim, W.K.; Ko, J.J.; Oh, Y.K. Erythrocyte ghost-mediated gene delivery for prolonged and blood-targeted expression. Gene Ther. 2004, 11, 492–496. [Google Scholar] [CrossRef] [Green Version]
  374. Liu, X.; Li, Y.-p.; Zhong, Z.-m.; Tan, H.-q.; Lin, H.-p.; Chen, S.-j.; Fu, Y.-c.; Xu, W.-c.; Wei, C.-j. Incorporation of viral glycoprotein VSV-G improves the delivery of DNA by erythrocyte ghost into cells refractory to conventional transfection. Appl. Biochem. Biotechnol. 2017, 181, 748–761. [Google Scholar] [CrossRef] [PubMed]
  375. Rossi, L.; Brandi, G.; Schiavano, G.F.; Scarfi, S.; Millo, E.; Damonte, G.; Benatti, U.; De Flora, A.; Magnani, M. Heterodimer-loaded erythrocytes as bioreactors for slow delivery of the antiviral drug azidothymidine and the antimycobacterial drug ethambutol. AIDS Res. Hum. Retroviruses 1999, 15, 345–353. [Google Scholar] [CrossRef] [PubMed]
  376. Rossi, L.; Serafini, S.; Antonelli, A.; Pierigé, F.; Carnevali, A.; Battistelli, V.; Malatesta, M.; Balestra, E.; Caliò, R.; Perno, C.F.; et al. Macrophage depletion induced by clodronate-loaded erythrocytes. J. Drug Target. 2005, 13, 99–111. [Google Scholar] [CrossRef] [PubMed]
  377. Hamidi, M.; Azimi, K.; Mohammadi-Samani, S. Co-encapsulation of a drug with a protein in erythrocytes for improved drug loading and release: Phenytoin and bovine serum albumin. J. Pharm. Pharm. Sci. 2011, 14, 46–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  378. Talwar, N.; Jaind, N.K. Erythrocyte based delivery system of primaquine: In vitro characterization. J. Microencapsul. 1992, 9, 357–364. [Google Scholar] [CrossRef] [PubMed]
  379. Alanazi, F.K.; Harisa, G.E.-d.I.; Maqboul, A.; Abdel-Hamid, M.; Neau, S.H.; Alsarra, I.A. Biochemically altered human erythrocytes as a carrier for targeted delivery of primaquine: An in vitro study. Arch. Pharm. Res. 2011, 34, 563–571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  380. Harisa, G.E.-d.I.; Ibrahim, M.F.; Alanazi, F.K. Characterization of human erythrocytes as potential carrier for pravastatin: An in vitro study. Int. J. Med. Sci. 2011, 8, 222–230. [Google Scholar] [CrossRef] [Green Version]
  381. Harisa, G.I.; Ibrahim, M.F.; Alanazi, F.K. Erythrocyte-mediated delivery of pravastatin: In vitro study of effect of hypotonic lysis on biochemical parameters and loading efficiency. Arch. Pharm. Res. 2012, 35, 1431–1439. [Google Scholar] [CrossRef]
  382. Favretto, M.E.; Cluitmans, J.C.; Bosman, G.J.; Brock, R. Human erythrocytes as drug carriers: Loading efficiency and side effects of hypotonic dialysis, chlorpromazine treatment and fusion with liposomes. J. Control. Release 2013, 170, 343–351. [Google Scholar] [CrossRef]
  383. Noel-Hocquet, S.; Jabbouri, S.; Lazar, S.; Maunier, J.C.; Guillaumet, G.; Ropars, C. Erythrocytes as carriers of new anti-opioid prodrugs: In vitro studies. Adv. Exp. Med. Biol. 1992, 326, 215–221. [Google Scholar] [CrossRef]
  384. Dey, P.; Banerjee, S.; Mandal, S.; Chattopadhyay, P. Design and evaluation of anti-fibrosis drug engineered resealed erythrocytes for targeted delivery. Drug Deliv. Transl. Res. 2019, 9, 997–1007. [Google Scholar] [CrossRef] [PubMed]
  385. Aryal, S.; Stigliano, C.; Key, J.; Ramirez, M.; Anderson, J.; Karmonik, C.; Fung, S.; Decuzzi, P. Paramagnetic Gd3+ labeled red blood cells for magnetic resonance angiography. Biomaterials 2016, 98, 163–170. [Google Scholar] [CrossRef] [PubMed]
  386. Takeuchi, Y.; Suzuki, H.; Sasahara, H.; Ueda, J.; Yabata, I.; Itagaki, K.; Saito, S.; Murase, K. Encapsulation of iron oxide nanoparticles into red blood cells as a potential contrast agent for magnetic particle imaging. Adv. Biomed. Eng. 2014, 3, 37–43. [Google Scholar] [CrossRef] [Green Version]
  387. Antonelli, A.; Sfara, C.; Manuali, E.; Salamida, S.; Louin, G.; Magnani, M. Magnetic red blood cells as new contrast agents for MRI applications. In Proceedings of the SPIE Medical Imaging 2013: Biomedical Applications in Molecular, Structural, and Functional Imaging, Lake Buena Vista (Orlando Area), FL, USA, 29 March 2013; Volume 8672. [Google Scholar] [CrossRef]
  388. Rahmer, J.; Antonelli, A.; Sfara, C.; Tiemann, B.; Gleich, B.; Magnani, M.; Weizenecker, J.; Borgert, J. Nanoparticle encapsulation in red blood cells enables blood-pool magnetic particle imaging hours after injection. Phys. Med. Biol. 2013, 58, 3965–3977. [Google Scholar] [CrossRef] [Green Version]
  389. Markov, D.E.; Boeve, H.; Gleich, B.; Borgert, J.; Antonelli, A.; Sfara, C.; Magnani, M. Human erythrocytes as nanoparticle carriers for magnetic particle imaging. Phys. Med. Biol. 2010, 55, 6461–6473. [Google Scholar] [CrossRef]
  390. Brähler, M.; Georgieva, R.; Buske, N.; Müller, A.; Müller, S.; Pinkernelle, J.; Teichgräber, U.; Voigt, A.; Bäumler, H. Magnetite-loaded carrier erythrocytes as contrast agents for magnetic resonance imaging. Nano Lett. 2006, 6, 2505–2509. [Google Scholar] [CrossRef]
  391. Du, B.; Yan, X.; Ding, X.; Wang, Q.; Du, Q.; Xu, T.; Shen, G.; Yao, H.; Zhou, J. Oxygen self-production red blood cell carrier system for MRI mediated cancer therapy: Ferryl-Hb, sonodynamic and chemical therapy. ACS Biomater. Sci. Eng. 2018, 4, 4132–4143. [Google Scholar] [CrossRef]
  392. Tang, J.C.; Partono, A.; Anvari, B. Near-infrared-fluorescent erythrocyte-mimicking particles: Physical and optical characteristics. IEEE Trans. Biomed. Eng. 2019, 66, 1034–1044. [Google Scholar] [CrossRef]
  393. Chen, Z.A.; Wu, S.H.; Chen, P.; Chen, Y.P.; Mou, C.Y. Critical features for mesoporous silica nanoparticles encapsulated into erythrocytes. ACS Appl. Mater. Interfaces 2019, 11, 4790–4798. [Google Scholar] [CrossRef]
  394. Wu, Y.W.; Goubran, H.; Seghatchian, J.; Burnouf, T. Smart blood cell and microvesicle-based Trojan horse drug delivery: Merging expertise in blood transfusion and biomedical engineering in the field of nanomedicine. Transfus. Apher. Sci. 2016, 54, 309–318. [Google Scholar] [CrossRef]
  395. Atukorale, P.U.; Yang, Y.S.; Bekdemir, A.; Carney, R.P.; Silva, P.J.; Watson, N.; Stellacci, F.; Irvine, D.J. Influence of the glycocalyx and plasma membrane composition on amphiphilic gold nanoparticle association with erythrocytes. Nanoscale 2015, 7, 11420–11432. [Google Scholar] [CrossRef] [PubMed]
  396. Dong, X.; Niu, Y.; Ding, Y.; Wang, Y.; Zhao, J.; Leng, W.; Qin, L. Formulation and drug loading features of nano-erythrocytes. Nanoscale Res. Lett. 2017, 12, 202:1–202:13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  397. Gupta, N.; Patel, B.; Ahsan, F. Nano-engineered erythrocyte ghosts as inhalational carriers for delivery of fasudil: Preparation and characterization. Pharm. Res. 2014, 31, 1553–1565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  398. Zelepukin, I.V.; Yaremenko, A.V.; Shipunova, V.O.; Babenyshev, A.V.; Balalaeva, I.V.; Nikitin, P.I.; Deyev, S.M.; Nikitin, M.P. Nanoparticle-based drug delivery: Via RBC-hitchhiking for the inhibition of lung metastases growth. Nanoscale 2019, 11, 1636–1646. [Google Scholar] [CrossRef]
Figure 1. Change in the total number of articles published in the world on the subject of erythrocyte carriers of biologically active substances, since 1973.
Figure 1. Change in the total number of articles published in the world on the subject of erythrocyte carriers of biologically active substances, since 1973.
Pharmaceutics 12 00276 g001
Figure 2. Possible modes of use of RBCs loaded with biologically active compounds and nanoparticles. MRI—magnetic resonance imaging; IHP—inositol hexaphosphate; MPH—macrophages; RES—reticuloendothelial system.
Figure 2. Possible modes of use of RBCs loaded with biologically active compounds and nanoparticles. MRI—magnetic resonance imaging; IHP—inositol hexaphosphate; MPH—macrophages; RES—reticuloendothelial system.
Pharmaceutics 12 00276 g002
Table 1. Substances that were loaded into erythrocyte.
Table 1. Substances that were loaded into erythrocyte.
Active SubstanceApplicationReferences
β-Galactosidase-[21]
β-Glucocerebrosidase (β-glucosidase)Gaucher disease[21,37,38,44,45,46,250]
β-GlucuronidaseSyndrome Slaya[251]
l-Phenylalanine ammonia lyasePhenylketonuria[48,252,253]
Phenylalanine hydroxylase[50,254]
Uricase (uratoxidase)Uric acid removal[255,256]
Urease, urease + alanine dehydrogenaseUrea utilization[257,258,259]
Adenosine deaminaseSevere combined immunodeficiency caused by deaminase deficiency[27,55,56,57,58,260]
Thymidine phosphorylaseMitochondrial neurogastrointestinal encephalomyopathy (MNGIE)[56,246,261,262]
Glutamate dehydrogenaseHyperammonemia[59,60,72]
Glutamine synthetase[73,74]
Glutamate dehydrogenase + alanine aminotransferase[11,63]
ArginaseHyperammonemia due to arginase deficiency[263]
Alcohol dehydrogenaseAlcohol and methanol intoxication[59,60]
Alcohol oxidase[61]
Acetaldehyde dehydrogenase[62]
Alcohol dehydrogenase + acetaldehyde dehydrogenase[11,63]
Formate dehydrogenaseMethanol intoxication[264]
Cyanide sulfurtransferase (rhodanase)Cyanide intoxication[65,66,67,68,69,70,265,266]
Catalase, PEG-catalaseAntioxidant[267]
l-AsparaginaseAntitumor therapy[16,22,24,83,84,85,86,237,268,269,270,271,272,273,274,275,276,277,278,279,280,281,282,283,284]
l-Methioninase[87,88,285,286]
Arginine deiminase[89,90]
Hexokinase, glucose oxidaseTo decrease blood glucose (diabetes)[287,288]
Insulin[138,139,140,289,290]
Inositol hexaphosphate (IHP)Sickle cell anemia[17,18,91,92,93,94,95,291,292,293,294,295,296,297,298,299,300,301]
MethotrexateCytotoxic drugs (antitumor antibiotics)[138,171,172,173,174,175,176,302,303,304]
Mitaxantrone[112,113]
Doxorubicin, daunomycin[103,104,105,106,107,108,109,110,111,305,306,307,308,309,310,311,312,313,314,315,316,317,318]
AmikacinBroad-spectrum antibiotics[319,320,321,322]
Gentamicin[323]
Tetracycline[324]
Penicillin G[10]
Actinomycin DCytotoxic drugs (antitumor antibiotics)[10]
Cytosine β-d-arabinoside[10,325]
Carboplatin[326]
Fluorouracil (5-fluoro-2-deoxyuridine)[327,328]
Bleomycin[329]
Vincristine, vinblastine[120,121,330]
Paclitaxel[331]
Fludarabine phosphate (2-Fluoro-ara-AMP)Cytostatic drug (antitumor therapy, HIV)[118,332,333,334]
DexamethasoneGlucocorticosteroids (anti-inflammatory drugs)[10]
Dexamethasone-21-phosphate[129,130,131,132,133,135]
[335,336,337,338,339,340,341,342,343]
Betamethasone phosphate[344]
Prednisolone-21-phosphate[25,128,345]
DiclofenacNonsteroidal anti-inflammatory drug[346]
Nucleoside reverse transcriptase inhibitors (2,3-dideoxytidine-5-triphosphate (ddCTP), zidovudine (AZT), (AZTp2AZT), didanosine (DDI)) in combination with reduced glutathione (GSH)Therapy of HIV, retroviral infections[169,181,182,183,184,185,187,347,348,349,350,351,352]
Fludarabine + AZT + GSH[353]
Nucleoside protease inhibitors (PNAPR2)[354]
Interferon + ribavirinHepatitis C therapy[189,190]
Ribavirin[355]
AntigensImmunization[197,198,199,200,201,356,357]
Cancer immunotherapy[204,207,358]
Induction of immune tolerance[208,209,213]
EnalaprilatAngiotensin-converting enzyme (ACE) inhibitor (arterial hypertension)[359,360,361]
MorphineOpioid analgesia[19,20,143,144,145,146]
Tramadol[362]
Factors IX and XHemophilia[141,142]
Interleukins 2 and 3Immunomodulators, antitumor therapy[363,364,365,366,367,368]
Superoxide dismutaseAntioxidant[368,369,370,371]
DNAGene therapy (gene delivery)[372,373,374]
ClodronateMacrophage depletion[195,375,376]
Zoledronate[191]
ValproateEpilepsy[156]
Phenytoin[377]
PrimaquineMalaria[378,379]
PravastatinCardiovascular disease prevention, treatment of abnormal lipids[158,380,381]
Cyclosporin A, tacrolimusImmunosuppressants[36]
Aminazine (Chlorpromazine)Antipsychotic (in psychiatric practice)[382]
NaloxoneOpioid receptor antagonist (opioid overdose)[383]
AmbroxolRespiratory diseases (fibrosis)[384]
Superparamagnetic nanoparticlesContrast agents in MRI[222,226,227,228,229,230,231,232,233,385,386,387,388,389,390,391]
NanoparticlesFrom drug delivery to fluorescence or photoacoustic imaging[151,152,154,155,156,158,159,160,161,162,392,393,394,395,396,397,398]
Fluorescent dyes (FITC)Blood analyte biosensors (glucose, pH)[33,34,35,234,235,236]

Share and Cite

MDPI and ACS Style

Koleva, L.; Bovt, E.; Ataullakhanov, F.; Sinauridze, E. Erythrocytes as Carriers: From Drug Delivery to Biosensors. Pharmaceutics 2020, 12, 276. https://doi.org/10.3390/pharmaceutics12030276

AMA Style

Koleva L, Bovt E, Ataullakhanov F, Sinauridze E. Erythrocytes as Carriers: From Drug Delivery to Biosensors. Pharmaceutics. 2020; 12(3):276. https://doi.org/10.3390/pharmaceutics12030276

Chicago/Turabian Style

Koleva, Larisa, Elizaveta Bovt, Fazoil Ataullakhanov, and Elena Sinauridze. 2020. "Erythrocytes as Carriers: From Drug Delivery to Biosensors" Pharmaceutics 12, no. 3: 276. https://doi.org/10.3390/pharmaceutics12030276

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop