Chapter 12

Micro/Nano-Engineering of Cells for Delivery of Therapeutics

Oren Levy, Edward Han, Jessica Ngai, Priya Anandakumaran, Zhixiang Tong, Kelvin S. Ng and Jeffrey M. Karp,    Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women’s Hospital, Boston, MA; Harvard Medical School, Harvard University, Boston, MA; Harvard Stem Cell Institute, Cambridge, MA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA

Cell-based therapies hold a great promise for treatment of a wide array of tragic diseases. Specifically, the use of cellular vehicle platforms emerges as a powerful strategy for targeted delivery of therapeutics to sites of disease. In this chapter, we first review the success and current challenges of different cell therapies, focusing on pancreatic islet transplantation as well as multiple types of stem cell therapies. We then shift our discussion to cellular delivery systems. Efficient cell targeting to desired sites is a crucial aspect towards effective use of such platforms. Accordingly, we thoroughly discuss a variety of cell engineering strategies to improve cell targeting to sites of interest. We then discuss key aspects of cellular delivery, including the types and characteristics of different nanosystems, candidate therapeutic agents and cell types suitable for this approach. Finally, we conclude with a case study of Trojan Horse cell-based cancer therapy.

Keywords

Stem Cells; Cell Therapy; Cell-based delivery platforms; Cell engineering

In this chapter, we discuss micro/nano-engineering strategies to enable efficient targeting of transplanted cells to diseased/damaged tissues that should improve the efficacy of cell therapy and permit the use of cells as drug delivery vehicles. To introduce the tremendous clinical potential and the challenges of cell therapy, we begin by describing a selection of current cell therapies and the importance of cell targeting. We will then discuss bioengineering approaches to modify the cell surface to improve cell targeting to desired sites. This will be followed by in-depth discussion of targeted cell-based drug delivery and its clinical potential.

12.1 Cell Therapy—Success and Current Challenges

12.1.1 Hematopoietic Stem Cell Transplantation

Over 10,000 bone marrow transplants are performed every year in the United States to treat patients with severe blood disorders [1]. Hematopoietic stem cells (HSCs), capable of giving rise to the entire hematopoietic system [2], are the key therapeutic component of the transplanted bone marrow units and are therefore widely used to treat many blood malignancies, including myeloma, leukemia, and lymphoma, as well as immune deficiencies and other hematological disorders [36]. In most cases, the patient’s own diseased hematopoietic system is first ablated and then reconstituted with healthy cells using HSC transplantation (HSCT). Following systemic transplantation, the first key cellular process that affects HSCT efficacy is the homing of HSCs to the bone marrow, where an optimal niche is provided for hematopoiesis [7]. Akin to leukocytes, HSCs roll on, adhere to, and migrate across the vascular endothelium at specific tissues, guided by chemotactic gradients [8,9]. Lack of homing capacity has been shown to drastically reduce HSCT efficacy [10]. After homing, engraftment is required to retain and maintain HSCs in a conductive microenvironment for long-term survival and proper function (e.g., self-renewal, differentiation). Enhancing homing and engraftment will likely improve efficacy and reduce the dosage of HSC infusion, allowing more patients to be treated with the same donor pool. This is important since HSCs are difficult to expand in vitro without inducing undesirable differentiation and cell death [11,12].

Because HSCT is typically allogeneic, a main challenge in HSCT is significant immune response following transplantation. Not only may the host immune response compromise the engraftment and survival of transplanted HSCs [13], patients are also at risk for graft-versus-host disease (GvHD), which can lead to death [14]. The risk of an immune response can be lessened via prophylactic immune suppression [15]. Haplotype matching based on the human leukocyte antigen (HLA) further ameliorates the risk, although this limits the number of potential donors [6]. Nowadays, HSCs can be efficiently harvested from multiple tissue sources, including cord blood, substantially increasing the number of patients who can benefit from HSCT [16,17]. Importantly, genetic modification enables the potential use of autologous HSCs: for some diseases, patients’ cells can be corrected ex vivo and returned to the same patient, providing a promising platform when matched donors are unavailable [18].

However, significant challenges still remain for HSCT. In addition to GvHD and poor engraftment of transplanted cells, disease relapse often occurs after allogeneic (unmatched), haploidentical (matched), or autologous transplantations [19]. Although continuous efforts are made to engineer cells to improve therapy, successful implementation into the clinic is challenging due to safety issues and preservation of engineered properties post-transplantation [20]. Genetic modification entails potential risks such as abnormal splicing, inactivation of tumor suppressor genes, or accidental oncogene activation, that may result in development of leukemia [21]. Nongenetic modifications may circumvent some risks while conferring better control over cell fate post-transplantation, as we will discuss in detail later.

12.1.2 Pancreatic Islet Transplantation

Another cell therapy in growing demand is pancreatic islet transplantation for treatment of type 1 diabetes (T1D) [22]. In T1D, the pancreas does not produce sufficient levels of insulin, mostly due to autoimmune destruction of beta cells, resulting in impaired glucose metabolism [23]. Patients with T1D need to constantly monitor their blood glucose and receive insulin injections to reduce hyperglycemia when necessary. Poor control of blood glucose may lead to hypoglycemia, which can be fatal, or chronic hyperglycemia, which may result in complications such as heart disease, kidney disease, and nerve damage [24,25]. Successful islet transplantation would restore autonomous, precise control of blood glucose, freeing patients from constant monitoring, and adjustment of blood glucose. Allogeneic islets are required for T1D [25], since the patient’s own islets are typically dysfunctional. Unfortunately, islets are composed of terminally differentiated cells and cannot be expanded ex vivo, while donor morbidity is clinically unjustifiable. Elaborate procedures for quickly obtaining fresh islets from cadavers are therefore required, especially given the fact that islets from at least two deceased donors are required to treat a single patient [26].

Like HSCT, allogeneic islet transplantation faces immune rejection and donor shortage. But unlike HSCs, islets are large multicellular aggregates containing multiple cell types and have to be administered locally instead of systemically (i.e., via the bloodstream) [27]. This greatly affects the repertoire of strategies for cell engineering. For example, genetic modification becomes more difficult since transfection efficiency will vary according to the location of the cell within the islet, whereas tissue engineered islet with defined compositional and structural features becomes instrumental for controlling cell fate and function following local transplantation [28]. However, while homing may be less relevant, engraftment and survival remain crucial for the success of islet transplantation: islets have to be vascularized quickly post-transplantation to maintain sufficient viability for long-term function [29].

Although the host immune response may be reduced by genetically downregulating foreign antigens [3032], encapsulation inside biocompatible scaffolds such as alginate or porous pouches can sufficiently shield islets from both cellular and humoral components of the host immune system and thereby offer stronger and longer term protection [3335]. Native islets deliver insulin directly into the hepatic portal vein; hence the gold standard has been intraportal implantation [36], which requires invasive surgery. Scaffold technologies may potentially enable minimally invasive [37] islet transplantation, such as subcutaneous implantation, given that a variety of problems will be addressed, such as capability of islets to detect changes in blood glucose, delivery of relevant amounts of insulin into the bloodstream, scaffold degradation over time, foreign body response against the scaffold, and nutrient transport to encapsulated islets [38]. Supplementing scaffold technologies with genetic strategies, such as accelerating vascularization by ectopically expressing pro-angiogenic factors [39], or promoting islet viability and functionality by ectopically expressing hormones [40] may be necessary to achieve sufficient efficacy of islet transplantation in the future [26]. An emerging paradigm is to derive pancreatic cells from precursors in vitro before transplantation [41,42]. Cells may then be administered systemically as single cells instead of aggregates, allowing them to efficiently home to and engraft at sites (e.g., hepatic or pancreatic capillary beds) where optimal islet physiology may be achieved.

12.1.3 Mesenchymal Stem Cell Therapy

Mesenchymal stromal cells, otherwise known as mesenchymal stem cells (MSCs), have emerged during the past two decades as promising candidates for cell therapy [4345]. Unlike pancreatic islets, MSCs are readily isolated from bone marrow and other adult tissues without significant donor morbidity. Unlike HSCs, MSCs can be easily expanded ex vivo to obtain a sufficient quantity for cell transplantation [46]. Moreover, their immune-evasive and potent immunomodulatory properties following systemic infusion permit allogeneic transplantation and have prompted their use in over 380 clinical trials for treating multiple diseases, including lupus nephritis, GvHD, multiple sclerosis, and cardiovascular diseases [47]. While results from pre-clinical animal studies have been encouraging and hundreds of millions of allogeneic MSCs have been safely administered, clinical trials have produced only mixed results for efficacy endpoints and MSC translational potential has not yet been fully realized [4851]. This could be attributed to the innate heterogeneity of MSC population, significant variability in their donor/tissue sources, manufacturing processes, as well as their diverse characterization methods [52]. One of the major challenges for MSC therapy is poor homing to diseased or damaged tissues, potentially due to minimal expression of key homing ligands on MSC surface [49,53,54]. Since the MSC secretome serves a crucial role in their therapeutic impact, increasing the number of transplanted MSCs at disease sites at the same total dosage will likely significantly improve the efficacy of MSC therapy [55].

12.1.4 Embryonic Stem Cells and Induced Pluripotent Stem Cells

Embryonic stem cells (ESCs) are perhaps the most promising yet most controversial cell type being investigated for therapy. Unlike adult stem cells, ESCs have an unlimited capacity for self-renewal, can differentiate into all three germ layers (ectoderm, endoderm, and mesoderm), and thus can generate almost any cell type [56]. However, adverse effects, such as formation of teratomas, and the many ethical issues that plague the use of ESCs, significantly hinder their translation into the clinic [57]. Numerous studies demonstrated the therapeutic potential of ESCs. For example, Min et al. [58] were able to significantly improve left ventricular function in post-infarcted rats after ESC transplantation. Brustle et al. [59] demonstrated that ESCs differentiate into oligodendrocyte and astrocyte precursors and induce axon myelination in a rat model of dysmyelinating Pelizaeus–Merzbacher disease. Besides cardiac and neural applications, ESC studies have spanned virtually every discipline of the biomedical field [60]. Better understanding of the mechanisms governing ESC differentiation and transformation may advance the development of safe and effective ESC therapies in the future.

Induced pluripotent stem cells (IPSCs) bypass ethical issues of ESCs and are also starting to be tested in clinical trials [61]. IPSCs are generated by reprogramming somatic cells to regain pluripotency [61,62], first demonstrated when Takahashi and Yamanaka [63] successfully reprogrammed adult fibroblasts into an embryonic-like state. Theoretically, this would enable a virtually limitless quantity of highly desirable ESC-like cells, while avoiding any ethical issues. One of the main questions is whether IPSCs are truly as pluripotent as ESCs [62]. Different groups have shown significant differences between IPSCs and ESCs. For example, the differentiation potential of IPSCs appears intrinsically lower than that of ESCs [64]. In addition, IPSCs seem to have an increased propensity to differentiate back into the cells they were formed from [65]. Despite these differences, IPSCs have been used in ample studies spanning a multitude of biomedical applications. For example, Wernig et al. [66] were able to demonstrate that neurons derived from IPSCs were able to functionally integrate into the brains of rats with Parkinson’s disease. Much of the potential of IPSCs remains untapped, however, making this field an exciting source of potential therapies.

Among the challenges discussed above, poor cell targeting to disease sites following transplantation seems to be a common drawback shared by all the aforementioned cell therapies. We will now discuss micro/nano-engineering approaches to control cell homing/targeting. These approaches mainly involve the cell surface, the key interface where the cell interacts with its environment during the homing process.

12.2 Cell Surface Engineering to Improve Cell Targeting

One of the major challenges in cell therapy is poor control over cell homing to disease sites following transplantation—to maximize therapeutic impact, delivering a sufficient amount of functional cells to target organs is crucial. Improving cell homing would also reduce the number of transplanted cells needed to achieve clinical improvement. This has enormous significance since cell expansion ex vivo has both biological and financial costs [48], and not every cell type can be expanded. For MSCs, which exert their immunomodulatory effects primarily via their secretome [55,67], increasing their bioavailability at the disease site is likely to significantly improve their therapeutic impact [68].

12.2.1 Affinity-Based Cell Targeting

Different bioengineering approaches have been exploited to modify the cell surface to enhance cell targeting to desired sites. A generalized concept is to trigger receptor–ligand interactions between the surface of transplanted cells and the cells at the target site. Cells can be engineered to overexpress biological or artificial receptors or antibodies to specifically target cells expressing the corresponding ligands or antigens [69] (Figure 12.1).

image
Figure 12.1 Bioengineering strategies to improve cell targeting to disease sites.

Whole and fragmented antibodies have been incorporated into cell surfaces to enhance cell localization to target sites. For instance, coating MSCs with whole antibodies against MAdCAM increased their homing to inflamed bowel and improved therapeutic efficacy on inflammatory bowel disease [70]. Likewise, in adoptive immunotherapy, autologous T cells may be modified with antibodies ex vivo and redirected to target specific cells, such as cancer cells. Whole antibodies have been engineered to create chimeric T-cell receptors (TCRs), which signal the T cell to launch a killing response upon binding to malignant cells [71,72].

Fragmented antibodies, such as single chain variable fragments (scFv) antibodies which do not contain the Fc domain but retain the high specificity and affinity of whole antibodies, have also been used to modify the cell surface [73]. Omitting the Fc domain eliminates potential binding of Fc receptors onto the cell surface and facilitates vector design for genetic engineering, but removes the signaling ability intrinsic to the Fc region. Fragmented antibodies have been engineered to create bispecific adaptors (diabodies) that link the specificity of two antibodies into a single agent and act as a bridge between two cell types. Cells have been genetically modified to secrete diabodies which bind tumor cells at one end and bind/activate T cells on the other, directing T-cell cytotoxic activity specifically toward tumors and significantly inhibiting tumor growth [7476]. Fragmented antibodies, instead of whole antibodies, are preferred for this approach since they penetrate tissues better due to their reduced size [77].

These two approaches represent how both membrane-anchored and secreted factors may enhance cell targeting via the cell surface. The same concept extends to receptors beyond antibodies. We will next discuss how such factors may be imparted to cells that natively underexpress them.

12.2.2 Bioengineering Approaches to Accomplish Affinity-Based Cell Targeting

12.2.2.1 Genetic Modification

Genetic modification has been employed to introduce new genes or to knock out specific genes to confer added functionalities to cells. The stability and specificity of genetic modifications have made it an attractive technique to achieve enhanced cell homing through cell surface modifications. The use of viral vectors such as adenoviruses and lentiviruses, as well as nonviral reagents such as lipid and polymeric micro/nanoparticles, has been widely explored to deliver DNA/RNA to cells to overexpress specific receptors to achieve improved homing to target sites.

12.2.2.1.1 Viral Vectors

Viral vectors have been utilized as gene delivery vehicles to overexpress receptors on cell surfaces due to their high transfection efficiencies. Yu et al. [78] found that transducing MSCs with lentiviral vectors encoding the gene for the chemokine receptor CXCR4 resulted in enhanced engraftment and neuroprotection in rats subjected to middle cerebral artery occlusion. This was facilitated via interactions between CXCR4 and, its ligand, SDF-1, which is expressed along the ischemic boundary of the brain, and was shown to promote the migration of CXCR4-MSCs toward the ischemic zone [78]. In another study, increased homing of MSCs to the brain in a malignant glioma model was induced by transducing cells with retroviruses encoding EGFR [79]. Ectopic expression of EGFR on the surface of MSCs resulted in increased targeting toward its ligands EGF, HB-EGF, and TGF-alpha, all of which are present on glioma cells [79]. Retrovirally induced CXCR4 overexpression was also demonstrated to improve MSC homing to infarcted myocardium, also resulting in improved cardiac function following cell treatment [80]. In another study, retroviral delivery to increase CCR1 expression on MSC surface prior to transplantation increased cell viability, migration, and engraftment to infarcted myocardium [81]. Furthermore, viral-induced expression of integrin alpha 4 on MSC surface was shown to target MSCs to bone in immunocompetent mice, where MSCs exhibited differentiation into osteoblasts and osteocytes in the growth plate of recipient mouse limb bones [82]. Other studies have used genetically modified affinity-based targeting to increase delivery of systemically administered MSCs to tumor sites. For example, MSCs have been genetically modified with adenoviral vectors to express artificial receptors that can bind to the tumor cell marker erbB2 [69]. These erbB2-receptor-modified MSCs were retained in high concentrations in the lungs of erbB2-expressing mice, and they were targeted toward ovarian tumors in ovarian xenograft tumor models [69]. In another study, monocytes were adenovirally transduced to express a chimeric CD64 receptor to target CEA-expressing tumor cells, resulting in significantly reduced in vivo tumor growth rates in xenograft studies [83].

T cells which have been genetically modified with viral vectors to express chimeric TCRs that recognize tumor markers have also been evaluated. For instance, peripheral blood-derived human T cells were transduced with retroviruses encoding chimeric TCR specific to CD30 and were found to specifically target CD30+ lymphoma cells. Following engraftment with CD30+ lymphoma cells, the TCR-modified T cells secreted high levels of IFN-γ to induce killing of specific tumor cells [84]. Furthermore, human lymphocytes which have been genetically engineered to express tumor-specific chimeric receptors have been translated to phase I clinical trials [85] due to their high potency against tumors in animal models, which has resulted in partial or complete remission of cancer [72,86]. Despite their high transfection efficiency, viral vectors are challenged by several safety issues such as immunogenicity and potential oncogene activation, resulting in the search for and development of nonviral vectors to genetically engineer cells [87].

12.2.2.1.2 Nonviral Vectors

A primary challenge in nonviral transfection is overcoming the electrostatic repulsion between negatively charged nucleic acids and the negatively charged cell membrane to facilitate uptake of DNA/RNA by cells. Common transfection methods include reagent-based methods such as cationic lipids or polymers, and device-based methods such as electroporation. Despite the lower transfection efficiency of nonviral vectors in comparison to viral vectors, the reduced immune response and toxicity of nonviral vectors have made them attractive gene delivery vehicles [8894].

Kim et al. demonstrated that MSCs transfected with plasmids encoding the gene for CXCR1 through electroporation resulted in superior migration of MSCs toward gliomas. The increase in migration was attributed to the binding interaction between CXCR1 and IL-8, which is released by glioma cells [95]. MSCs have also been genetically modified through electroporation to express scFv antibodies on their surface to target EGFRvIII, which is highly expressed in many glioblastoma (GB) tumors. The modified MSCs were found to bind to U87-EGFRvIII cells at high levels in vitro, and they experienced a sevenfold increase in retention in human U87-EGFRvIII expressing tumors in vivo [96].

Finally, peripheral blood human T cells, which were transfected via electroporation with chimeric TCRs specific to CD20, were found to preferentially migrate towards CD20+ lymphoma cells. Following coculture with CD20+ lymphoma cells, the TCR-modified T cells exhibited cytotoxic characteristics through an increased secretion of IFN-γ [97].

Recently, we have demonstrated the use of lipofectamine to deliver in vitro transcribed mRNA to control MSC phenotype following transplantation [98]. Unlike delivery of DNA, which could result in stable transfection but risks integration of foreign DNA into the genome, mRNA transfection elicits a transient, yet functionally significant, expression of genes of interest. Via transfection with pseudouridine-modified mRNA (to increase translation efficiency), we have expressed sialyl Lewis X (SLeX) and PSGL-1 on MSC surface, resulting in improved homing of systemically transplanted MSCs to healthy and irradiated bone marrow as well as to distant sites of local inflammation [98]. The transient effect of mRNA transfection may not be desired for all clinical applications. However, this may be partially addressed by administering multiple infusions to patients and further research is needed to fully elucidate the translatability of this approach into the clinic.

Genetic engineering has the potential to stably and specifically modify cell surfaces to achieve increased cell targeting to different disease sites. However, this approach raises potential safety risks, mostly due to potential integration of transfected DNA into the genome of the host cell and the use of viruses for delivery. An alternative approach, which is safer and may result in more transient cell surface alteration, is chemical or enzymatic modification of the cell surface prior to transplantation.

12.2.2.2 Chemical Modification

Another technique to modify the cell surface is to chemically conjugate molecules or nanoparticles (NPs) onto existing functional groups on cell surface proteins, polysaccharides, or lipids. It is also possible to metabolically or genetically introduce new, reactive functional groups onto the surface of cells to act as anchors for other molecules to remodel the cell surface [99]. However, chemical conjugation is a simpler and more transient method to modify the cell surface.

12.2.2.2.1 Surface Functional Groups

Functional groups such as thiols and amines are naturally present on the cell surface and are readily available for covalent conjugation. Other functional groups such as aldehydes and ketones have been used as anchors for surface-modifying molecules; however, they must first be exposed through chemical or enzymatic treatment of cell surface carbohydrates. Primary amine groups have been utilized as sites for covalent conjugation through biotinylation, in which the amine groups are reacted with amine-reactive biotin, such as N-hydroxy-succinimide biotin derivatives. The cell surface can then be functionalized with other biotinylated molecules or NPs through a streptavidin bridge [100,101]. We previously found that covalently coupling SLeX to the surface of MSCs through a streptavidin–biotin bridge resulted in a robust cell rolling on P-selectin-coated substrates in vitro, as well as an increased homing efficiency to inflamed tissue in vivo, without compromising the intrinsic properties of MSCs [102,103].

12.2.2.2.2 Lipid Insertion

Another approach involving the chemical conjugation of molecules to the cell surface takes advantage of the hydrophobic portion of the cell membrane to immobilize antibodies onto the cell surface. This two-step method consists of first coating cells with palmitate-conjugated proteins which are stabilized onto the cell membrane through the insertion of the hydrophobic palmitate moiety into the phospholipid bilayer. The palmitated proteins, which have high binding affinity for the Fc region of antibodies, allow cell surfaces to be functionalized with a variety of antibodies. These antibodies then act as artificial receptors for soluble or cell surface antigens [104]. Kim et al. [104] showed that cells coated with palmitate-conjugated protein A followed by rabbit anti-mouse IgG demonstrated enhanced adhesiveness to surface-bound IgG-positive mouse B cells in vitro. Others investigated MSCs coated with palmitated protein G (PPG) followed by antibodies against ICAM-1 and found an increased binding of anti-ICAM-1 MSCs to activated endothelial cells in flow chamber studies [105]. Similarly, Lo et al. [106] recently attached PSGL-1 (via PPG) to MSC surface, resulting in MSC tethering and rolling on stimulated endotheilal cell monolayers. Finally, Dennis et al. [107] focused on enhancing the engraftment of pre-chondrocytes to cartilage defects by coating the cell surface with lipidated protein G and antibodies which are specific to cartilage matrix antigens. Painted cells which were injected into rabbit cartilage explants with a partial thickness defect preferentially bound to the exposed cartilage material within the defect.

Another example of cell painting to improve cell homing involves binding antibodies utilizing the interaction between glycosylphosphatidylinositol (GPI) and cell surface proteins [108]. Sheep erythrocytes surface loaded with GPI-anchored scFv antibodies against CD-40 were found to specifically target and form rosettes with CD20-positive cancer cells [109]. Finally, Kean et al. [110] improved cell homing of MSCs to sites of myocardial infarction (MI) by coating MSCs with MI-specific peptides which have palmitic acid tails to facilitate integration into the cell membrane. Peptide-coated MSCs which were injected into mice with MI were localized at the heart in higher numbers than native MSCs [110].

12.2.2.3 Enzymatic Modification

It is also possible to directly modify existing cell surface molecules through enzymatic transformations. This method was employed to improve HSC homing and engraftment by ex vivo fucosylation of cord blood in order to achieve an increase in HSC tethering to activated endothelial cells expressing P-selectin and E-selectin [111]. This concept was further applied to human MSCs, where enzymatically modifying native CD44 glycoforms into hematopoietic cell E-selectin/L-selectin ligand resulted in targeting of modified MSCs to bone marrow in a murine model [112,113].

12.2.2.4 Physical Targeting

Physical targeting strategies utilize appropriate devices or procedures to enhance cell targeting to a specific site and may involve cell surface modifications. Although the concept of using devices or procedures to enhance cell targeting appears simple, a number of issues associated with gaining physical and direct access to organs severely complicate this method. While guiding devices exist, such as catheters, which can directly introduce cells into the brain [114,115], heart [116], and into wounds [117] with positive therapeutic outcomes, other physical methods exist like loading cells with magnetic compounds to redirect cell localization via application of a magnetic field to the target site. The initial purpose of using magnetic compounds was to monitor cell migration in vivo with magnetic resonance imaging; however, it has since evolved to be used to redirect cell localization both in vitro and in vivo. Nakashima et al. [118] showed that magnetically labeled NK cells targeted human osteosarcoma cells under a magnetic field in vitro. Other groups managed to magnetically target MSCs in animal models. For example, iron oxide-labeled MSCs were shown to specifically concentrate in the liver when external magnets were placed over it [119]. The same group showed that bone marrow stromal cells injected into the cerebrospinal fluid were able to aggregate on the surface of the spinal cord nearest to the magnetic field [119]. Another study focused on delivering magnetic NP-loaded endothelial cells to the steel surfaces of intra-arterial stents in a rat carotid artery stenting model [120]. Loading cells with nano/microparticles can be used beyond cell localization, especially for drug delivery applications.

12.3 Cell-Based Drug Delivery

The ability of cells to target specific tissues presents new opportunities for drug delivery. Harnessing cells as vehicles for drug delivery potentially overcomes challenges faced by current treatments. Local administration of therapeutics is undesirable when the disease is systemic or requires invasive access to target sites. Meanwhile, challenges of systemic administration include host toxicity, uncontrolled drug levels at desired sites, premature inactivation of the drug [121], and fast clearance of the agent from the blood, consequently requiring frequent repeat dosing. The usage of drug carriers is critical in the field of drug delivery and has been researched intensively [121]. Encapsulation of therapeutic agents in micro/nanoparticles increases safety by reducing toxicity and increases efficacy by protecting the drug from early degradation, permitting sustained therapeutic effects of systemically administered drugs. However, targeted delivery, which maximizes efficacy by releasing drugs only at disease sites, remains challenging. Particles passively target tissues where the vasculature is leaky (e.g., liver, kidney, and tumor). Although immobilizing ligands on the particle surface can actively target particles to specific tissues and counter passive targeting, excessive active targeting limits the depth of tissue penetration, and may also compromise the drug release kinetics [122]. In contrast, cells have homing and migratory properties; they can target and penetrate specific tissues even without modification. Therefore, cells with known or controllable tissue targets following transplantation emerge as a promising platform for targeted drug delivery. Like a Trojan horse, a cellular carrier protects the drug, infiltrates into the target tissue, and locally releases therapeutic cargo. Sustained drug release may be achieved by loading cells with drug-containing particles [123]. Not only does this arm the cell with additional therapeutic capabilities, its fate and phenotype may also be controlled by the released drug, like in the case of MSCs [55,98]. Herein, we will discuss several nanosystems, cell types, delivery methods, and disease models that utilize this strategy (Figure 12.2).

image
Figure 12.2 Multiple cell types can be used as drug delivery vehicles to treat a wide array of tragic diseases.

12.3.1 Nanosystem Types and Key Characteristics

The largest group of particulate carriers is NPs which encompass nanospheres, nanocapsules, micelles, dendrimers, nanocrystals, and nanogolds. NPs are chosen for their small size, high surface-to-volume ratio, and surface functionality. The most commonly used NP materials are polymers and lipids, which typically encapsulate hydrophobic drugs. Polymer-based NPs are made from synthetic polymers such as poly(lactic acid) (PLA) and poly(lactide-co-glycolide) (PLGA) or natural polymers such as gelatin, alginate, collagen, and chitosan [124126]. PLA and PLGA NPs are especially attractive since their degradation time can be tuned from days to months based on copolymer configuration and molecular weight [127]. However, without special treatments to render their surfaces hydrophilic, they are unstable in the aqueous biological environment and are unsuitable for systemic delivery. Lipid-based NPs typically have an amphiphilic coating to stabilize their hydrophobic cores in an aqueous environment. There are two common types of lipid-based NPs: solid lipid NPs [128] which consist of a solid lipid core and lipid nanocapsules [129] which have an oily (liquid) center [130]. Although lipid-based NPs may be more biologically stable, tuning their biodegradation in vivo is challenging. Other lipid-based NPs include micelles and liposomes, with liposomes being particularly attractive for their ability to encapsulate hydrophilic molecules [131].

Nucleic acids represent an important class of therapeutic hydrophilic molecules. Safety concerns over viral vectors have shifted research focus toward nonviral vectors [132]. Cationic liposomes and cationic polymer-based vectors interact with negatively charged molecules such as DNA while maintaining an overall positive charge on the surface, enabling interaction with the negatively charged cell surface and subsequent endocytosis, thus preventing DNA from systemic enzymatic degradation. Cargo is released into the cytoplasm via several mechanisms of endosomal escape such as membrane disruption/reassembly [133]. Similarly, cationized polysaccharides such as spermine-dextran [134] and spermine-pullulan [135] can be recognized by cell surface receptors to promote internalization, with biodegradability serving as another advantage. Although recent in vivo results have shown toxicity and low transfection efficiency, charged polysaccharides still demonstrated remarkable potential for use in tissue regeneration and cancer target therapy [91,136,137].

Another promising nanosystem is carbon nanotubes (CNTs) [138,139], which can immobilize and intracellularly deliver small molecules, nucleic acids, and proteins as long as the cargo can adsorb onto the CNT surface, typically by pi–pi stacking interactions [140,141]. Unfortunately, studies have also associated CNTs with cell toxicity [142], reducing its potential for clinical translation.

Protein adsorption on the surface of particles was originally viewed as undesirable as it promotes clearance by phagocytes. Efforts have since shifted toward using cellular binding and internalization properties to their advantage, such as to target specific cell types or to develop cellular drug carriers [143]. Internalization efficiency of particles by cells depends on cell type, particle size, surface chemistry, surface topography, shape, and mechanical properties [89]. Additional ligands can be conjugated to the particle surface to promote internalization via specific endocytic pathways [144].

12.3.2 Candidate Therapeutic Agents and Cell Types for Cell-Based Delivery

The types of therapeutic agents to be encapsulated in particles or cells depend not only on the disease of interest but also their stability during encapsulation and release kinetics after encapsulation. Encapsulation in cells warrants extra attention because the released drug may affect the cell and vice versa. For example, cell-based delivery of cytotoxic chemotherapeutics requires careful tuning of release kinetics, since excessive burst release may kill the cellular carrier before it reaches the target tissue [145]. An appealing strategy is to deliver prodrugs—drugs that are administered as inactive molecules, but are converted into their active form after specific chemical or enzymatic activity intracellularly or extracellularly. For instance, one can use the resident enzymes present in cells to convert prodrug into diffusible drug that can be released into the circulation [146]. Many therapeutic cargos, including nucleoside/nucleotide analogs [147], glucocorticoid analogs [148151], enzymes [152154], toxins [155,156], mRNA [98], peptides [157167], and antisense peptide nucleic acids [168171], are available as prodrugs.

An ideal cellular carrier must not only withstand its therapeutic cargo but also home to target tissues for efficient drug delivery. The most obvious, yet valuable, approach is to harness the innate homing properties of specific cells. Examples include leukocytes and stem cells that are known to home to tumors and sites of inflammation [172175]. However, the innate homing properties may be suboptimal; hence cell surface engineering can be applied to maximize cell delivery to specific sites, as discussed in the previous section. The next section reviews a selection of cell types and their tropism. Discussion of cancer applications will be deferred to the following section, where we provide a case study on how multiple cell studies have been used as Trojan horses to target tumors.

12.3.2.1 Erythrocytes

One of the deficiencies of using pure drugs or drug delivery nanosystems is their short life spans—they are quickly cleared by immune cells, liver, and kidneys. Frequent injections of the drug or nanosystem are required to resupply the effective dosage. Known for their long circulating life spans (up to 120 days), erythrocytes or red blood cells (RBCs) have been harnessed for sustained drug delivery [176]. Drugs or drug-loaded particles may be conjugated as “backpacks” onto RBC surfaces where immune-evasive molecules reside [177] or encapsulated within RBC membranes [178]. Both strategies “camouflage” the drugs from clearance pathways. RBCs also have other advantages such as abundance (~5.4 million cells/mm3 blood), size and shape uniformity [176], as well as long life span in circulation [146].

Particularly, RBC modification can be used in blood disorders. Fibrinolysis, the process of breaking down blood clots, is used to treat conditions due to hypercoagulation such as stroke and brain ischemia. Coupling fibrinolytic agents such as tissue-type plasminogen activator (tPA), streptokinase, and urokinase onto RBCs have been shown to sustain fibrinolytic activity without affecting RBC circulation [179181]. Furthermore, when an antibody against collagen was attached on the RBC membrane with streptokinase, RBCs were able to attach to immobilized collagen and lyse the overlaying fibrin clots [182]. This approach may potentially be used to target sites where integrity of the vascular endothelium is compromised, since coagulation can be triggered by exposure of basement membrane proteins (e.g., collagen) to blood.

Dexamethasone is a potent glucocorticoid drug that has anti-inflammatory and immunosuppressant properties. Unfortunately, dexamethasone treatment has severe adverse effects, including redistribution of fat, muscle wasting, acne, bruising, thinning of skin, osteoporosis, exacerbation of diabetes mellitus, suppression of growth in children, and cataract. In light of these systemic effects, Pierige et al. developed a dexamethasone prodrug that utilizes enzymes in RBCs for its activation. The prodrug is encapsulated into autologous RBCs. When applied as a therapy, it is dephosphorylated by RBC enzymes and releases active dexamethasone. Owing to the slow, yet sustained, release of effective doses of dexamethasone, patients fighting chronic obstructive pulmonary disease [149], cystic fibrosis [150], and bowel inflammatory disease [183186] were able to see benefits from treatments without adverse effects [147].

Although utilizing RBCs as drug carriers may achieve sustained systemic delivery, circulation times of “camouflaged” drugs were not as long as expected, typically lasting less than a week in stark contrast with the 120-day life span of unmodified RBCs [178]. Biomolecules on the RBC membranes that are responsible for prolonged circulation may be irreversibly damaged during RBC modification. In particular, RBCs lack tissue penetration properties and are unsuitable for delivering drugs deep into solid tissues. Consequently, targeted cell-based drug delivery strategies favor immune and stem cells, mostly for their innate tropism toward inflamed, hypoxic, and cancerous tissues [68].

12.3.2.2 Stem Cells

12.3.2.2.1 Neural Stem Cells

Neural stem cells (NSCs) exhibit tropism toward intracranial gliomas and medullablastomas in animal models [187189]. However, NSCs are only found in the brain [190] which poses significant donor morbidity. The challenge of harvesting enough autologous NSCs for clinical applications is a downfall of using NSCs. Other cell types—fetal brain, adult allogeneic brain, and ESCs—are being investigated as potential alternatives for autologous NSCs [191].

12.3.2.2.2 Mesenchymal Stem Cells

MSCs exhibit tropism toward sites of injury [192] and tumors [174,175], and their homing efficiency can be enhanced by genetic, chemical, enzymatic, and other modifications as previously described. Unlike NSCs and other stem cells, MSCs can be feasibly obtained at sufficient numbers for clinical applications since they can be isolated from many adult tissues [193202] at significant yields using minimally invasive methods and, more importantly, can be readily expanded ex vivo in large scales. Recently, our group has explored the potential of using MSC as cellular carriers via several bioengineering approaches. For instance, MSCs carrying intracellular dexamethasone-loaded PLGA microparticles could program the phenotypes of the MSC carrier as well as neighboring cells [103]. We have also demonstrated the use of mRNA modification on MSCs to suppress local inflammation via targeted delivery of the anti-inflammatory cytokine interleukin-10 following systemic MSC administration [98]. Moreover, MSCs can also be genetically modified to promote wound repair [203,204].

12.3.2.3 Leukocytes

Leukocytes, also known as white blood cells, possess natural homing properties toward tumors as well as other disease sites due to their major role in the immune response. Isolation of leukocytes is minimally invasive (via blood) and relatively straightforward. Consequently, multiple types of leukocytes have been implicated for Trojan horse delivery of therapeutics [205].

12.3.2.3.1 Monocytes

Monocytes are one of the first cells recruited by damaged, infected, or diseased tissues. After arriving at the site, monocytes differentiate into macrophages that phagocytose pathogens and recruit additional immune cells to the disease site [206]. At tumor sites, monocytes may become tumor-associated macrophages (TAMs), which constitute up to 70% of the tumor mass in breast cancer [206]. As drug carriers, monocytes may infiltrate into the hypoxic core of tumors and force surrounding TAMs to prevent proliferation [200,207210], tumor neoangiogenesis [158160,201], invasion [161,164,167,207], and metastasis [207,211218] of malignant epithelial cells. An advantage of using monocytes or macrophages as transport vehicles is their innate phagocytic capability which provides high efficiency in particle loading. The use of macrophages as cellular carriers for NPs arose from the observation that systemically administered of NPs for imaging were ingested by endogenous macrophages that migrated and accumulated in and around tumors [219221]. Another advantage of macrophages as cellular carriers is their ability to cross the blood brain barrier (BBB). For instance, macrophages preincubated with NPs containing a retroviral drug and administered in a murine model of HIV-1 encephalitis were observed to cross the BBB, increase local drug concentration for 14 days, and thereby suppress HIV-1 replication [222,223].

12.3.2.3.2 T Cells

T cells play a key role in adaptive immunity, for instance by mediating cellular responses against infected or cancerous cells [224]. T cells have potential as chaperones of surface-attached therapeutic cargo, supported by their previous use in clinical trials for adoptive T-cell therapy in cancer, ease of harvest, and well-established protocols for their genetic modifications and expansion [225]. They have also shown promising infiltration into tumor lesions by crossing the barriers imposed by endothelial and stromal tissues [226].

12.3.2.3.3 Dendritic Cells

Dendritic cells (DCs) are antigen-presenting cells, mostly found in the lymph nodes. After a local infection, DCs process and present antigens on their surfaces in order to activate naïve T cells and B cells [227]. Cell–cell interactions between T cells and DCs occur through direct contact [228] and paracrine signaling. There have only been a limited number of studies employing DCs as cellular vehicles. For example, DCs have been employed to systemically deliver oncolytic reovirus for killing melanoma cells and shown effective protection for the virus against the preexisting antiviral immunity [229]. Interestingly, the principle of “antigen presentation” on DCs was mimicked by immobilizing receptors and co-stimulatory ligands onto synthetic particles to activate adaptive immunity [230].

12.3.3 Case Study: “Trojan Horse” Cell Therapy for Cancers

Tumors are characterized by chaotic vasculature, high interstitial pressure, and dense extracellular matrices, presenting major obstacles for treatment via systemically administered drugs or drug-loaded particles [126]. Often, drugs and particles are unable to penetrate into the tumor and, as a result, are rapidly cleared. An example is GBs, which are extremely malignant brain tumors, known for their rapid growth and vascularization [231]. Current treatments revolve around surgery, radiation, and chemotherapy with DNA alkylating agents such as temozolomide (TMZ). However, there are several limitations with current treatments such as the poor effectiveness of TMZ for TMZ-sensitive GB patients as well as GB resistance against chemotherapy and radiotherapy. In addition, glioblastoma stem cells (GSCs) possess infiltrative, proliferative, and progressive characteristics, rendering them more resistant to hypoxic and acidotic tumor microenvironments [232]. Although GBs are highly vascularized, penetrating the BBB is yet another challenge toward effectively delivering therapeutic agents to the tumor. In fact, the BBB prevents uptake of large molecules and of over 98% of small-molecule drugs available [233]. Likewise, NP transport efficiency across the BBB is poor. To deliver antitumor agents across the BBB and deep into GBs, cellular carriers—namely monocytes and stem cells—are attractive candidates due to their ability to infiltrate into gliomas [234238].

Known for their tropism toward intracranial tumors, NSCs have been used to transport toxic molecules [239244], proliferation inhibitors [245], anti-angiogenic agents [246,247], and cytokines with toxic molecules [240]. Additionally, NSCs have been genetically modified to yield cytokines such as IL-18, IL-2 [248250], IFN-γ [251], and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) [252254] for modifying increased immune response at gliomas. NSCs have also been engineered to secrete cytosine deaminase (CD) that activates the prodrug 5-fluorocytosine (5-FC), so that cytotoxic effects of the systemically administered drug are localized within the tumor [189]. Off-target homing of antitumor NSCs is potentially dangerous as antitumor agents can harm healthy cells. To reduce off-target actions, overexpression of a pH-sensitive viral fusogen on the NSC surface has been used to fuse NSCs specifically with tumor cells, since tissue pH is lower at tumor sites [255]. These engineered NSCs carried a suicide gene, allowing selective killing of any syncytium formed by the NSCs and residual, unfused NSCs.

Monocytes or macrophages can also cross the BBB, but unlike NSCs, monocytes are isolated with much less invasive procedures. Choi et al. envisioned loading monocytes with gold nanoshells such that they home to the tumor site, differentiate into TAMs, migrate and deliver the nanoshells into hypoxic regions of the tumor. Once in place, near-infrared illumination would be used to heat the nanoshells and destroy the TAMs and adjacent tumor cells [205]. Nanoshell-based photothermal ablation therapy has shown promising results toward reducing tumor in mice, with over 90% remission rate [256]. Combining these concepts, monocyte carriers of gold nanoshells were successfully used to ablate human breast carcinoma tumor spheroids [172,205,219]. In another study employing a murine glioma model, cyclodextrin-based NPs were internalized by TAMs. Due to their tumor-tropic properties, the TAMs migrated within and around the tumor, demonstrating their capability to distribute therapeutic agents across the tumor [257]. T cells have also been used for nanoshell-based photothermal ablation [258], demonstrating the general effectiveness of leukocytes as tumor-homing cellular carriers.

Another use of T cells as a cellular vehicle followed from observations that viral particles can “hitchhike” on the T-cell surface, be taken up by tumor cells in vivo, and be released at the tumor site [259]. Extending the success of this strategy, numerous groups have used tumor antigen-specific T cells to carry oncolytic viruses to tumor deposits [260262]. The same strategy can be applied to deliver NPs for cancer diagnostics and therapeutics, where the use of viruses is undesirable. Such agents include small-molecule drugs, antibody–drug conjugates, aptamers, and magnetic imaging agents [263,264]. The Irvine group used thio-reactive maleimide groups to crosslink nanocarriers, surface proteins, or lipids to the T-cell surface for up to a week. Their in vivo results showed successful biodistribution of T-cell-coupled NPs in tumor models, while systemic injection without cellular carriers resulted in fast clearance of the particles in liver and spleen [265].

MSCs are yet another cell type that exhibits tumor-tropic properties. Systemically administered MSCs carrying intracellular PLA and lipid nanocapsules demonstrated successful delivery of nanocapsules to gliomas [175]. NPs have also been conjugated onto the MSC surface using biotin–streptavidin bridges without attenuating MSC tropism toward tumor spheroids [266]. In multiple studies, MSCs were genetically engineered using viral and nonviral vectors to secrete immunostimulators such as IL-2, IL-12, IFN-beta, IL-18, and IL-7 to suppress tumor growth in glioma and other cancer models [203,250,251,267276]. MSCs are also being used to deliver oncolytic adenoviruses to tumor sites [277,278]. More complex systems combining gene modification and prodrugs such as the CD/5-FC system (also applied on NSCs) have been tested on MSCs [279,280]. Another system uses herpes simplex virus thymidine kinase (HSVtk), which activates the chemotherapeutic prodrug ganciclovir [281]. Retrovirally transfected MSCs carried HSVtk to the tumor site, such that systemically administered ganciclovir was preferentially activated in the tumor, resulting in tumor suppression in GB patients [281]. The same system appeared to be less efficacious when NSCs are used instead [241,282284]. Yet another combination involves rabbit carboxylesterase and CPT-11, in which genetically modified adipose-derived MSCs were used to deliver carboxylesterase that enzymatically cleaves the CPT-11 prodrug to release the active drug SN-38 to combat brainstem gliomas [285]. Despite these advances, thorough investigation is required to fully elucidate the interaction between MSCs and tumor environment, especially when MSCs have been suggested to promote tumor growth [286], and clinical trials have not shown long-term engraftment of MSCs in tumors [287].

While most cellular drug delivery vehicles are still in early stages of research, one cell type has been successfully translated into FDA-approved therapy. DCs have become a part of FDA-approved therapeutic cancer vaccine (against prostate cancer), in which antigen-loaded autologous DCs are injected into patients [288]. In addition, another group has proposed a method to track cells that have been loaded with NPs. Noh et al. encapsulated near-infrared fluorophores and iron oxide NPs together with model antigens such that cells carrying the NPs could be tracked in real time. Injection of those DCs resulted in tumor suppression [289]. DCs also show tropism toward lymph nodes which enables the use of DCs to carry immunosuppressive drugs to lymphoid tissues in order to suppress T-cell proliferation [290].

Apart from solid tumors, blood cancers may also be treated with cellular carrier. In a pilot clinical trial, autologous RBCs were loaded with asparaginase so they could remove asparagine—a nonessential amino acid—from blood. Asparagine has been shown to increase lymphoblastic proliferation which is crucial for the progression of lymphoblastic leukemia [291].

12.4 Concluding Remarks

Cell therapy provides hope for treating many tragic diseases. Harnessing the innate therapeutic properties of certain cell types, such as MSCs, and using cells as vehicles for targeted delivery of therapeutics are promising strategies to further advance cell-based therapy. However, for cell therapy to fully realize its clinical potential, better control over cell fate following transplantation must be achieved. Bioengineering approaches, such as genetic, enzymatic, or chemical surface modifications, may enable specific delivery of systemically infused cells to desired organs, although this has yet to be tested in humans. Cells loaded with micro/nanoparticles containing specific drugs can then be directed to diseased organs, generating high local levels of therapeutics in the desired location, minimizing systemic toxicity, and significantly improving treatment efficacy. Tailoring drug release from cells has potential to provide a highly regulated and sustained therapeutic impact, and multiple cell infusions can further maximize the effect. Furthermore, mRNA engineering can be utilized to simultaneously but transiently control multiple cell properties, enabling improved cell homing and delivery of soluble biologics to distant diseased sites, while avoiding potential risks associated with DNA transfection. Targeted delivery of biologics is a major challenge that potentially could be overcome by use of cell-based approaches.

To circumnavigate current challenges in cell therapy, a concerted effort from clinicians, cell processing professionals, cell biologists, and bioprocess engineers is required [292]. Developing large quantities of cells that are immune evasive and that can be readily engineered to accurately deliver therapeutics including biologics to distant desired sites would have immense clinical potential.

References

1. BeTheMatch.org. Homepage. Available at: <http://bethematch.org/>.

2. Osawa M, Hanada K, Hamada H, Nakauchi H. Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science. 1996;273(5272):242–245.

3. Guimaraes FA, Oliveira-Cardoso EA, Mastropietro AP, Voltarelli JC, Santos MA. Impact of autologous hematopoetic stem cell transplantation on the quality of life of patients with multiple sclerosis. Arq Neuropsiquiatr. 2010;68(4):522–527.

4. Buckley RH, Schiff SE, Schiff RI, et al. Hematopoietic stem-cell transplantation for the treatment of severe combined immunodeficiency. N Engl J Med. 1999;340(7):508–516.

5. Pfender N, Saccardi R, Martin R. Autologous hematopoietic stem cell transplantation as a treatment option for aggressive multiple sclerosis. Curr Treat Options Neurol. 2013;15(3):270–280.

6. Van Zant G, Liang Y. Concise review: hematopoietic stem cell aging, life span, and transplantation. Stem Cells Transl Med. 2012;1(9):651–657.

7. Lymperi S, Ferraro F, Scadden DT. The HSC niche concept has turned 31 Has our knowledge matured? Ann N Y Acad Sci. 2010;1192:12–18.

8. Lapidot T, Dar A, Kollet O. How do stem cells find their way home? Blood. 2005;106(6):1901–1910.

9. Kavanagh DP, Kalia N. Hematopoietic stem cell homing to injured tissues. Stem Cell Rev. 2011;7(3):672–682.

10. Frenette PS, Subbarao S, Mazo IB, von Andrian UH, Wagner DD. Endothelial selectins and vascular cell adhesion molecule-1 promote hematopoietic progenitor homing to bone marrow. Proc Natl Acad Sci USA. 1998;95(24):14423–14428.

11. Magnusson M, Sierra MI, Sasidharan R, et al. Expansion on stromal cells preserves the undifferentiated state of human hematopoietic stem cells despite compromised reconstitution ability. PLoS ONE. 2013;8(1):e53912.

12. Dmitrieva RI, Anisimov SV. Optional protocols of hematopoietic stem cell expansion in vitro. Tsitologiia. 2013;55(1):11–15.

13. Huang Y, Rezzoug F, Chilton PM, Grimes HL, Cramer DE, Ildstad ST. Matching at the MHC class I K locus is essential for long-term engraftment of purified hematopoietic stem cells: a role for host NK cells in regulating HSC engraftment. Blood. 2004;104(3):873–880.

14. Ratanatharathorn V, Ayash L, Lazarus HM, Fu J, Uberti JP. Chronic graft-versus-host disease: clinical manifestation and therapy. Bone Marrow Transplant. 2001;28(2):121–129.

15. Marmont AM, Horowitz MM, Gale RP, et al. T-cell depletion of HLA-identical transplants in leukemia. Blood. 1991;78(8):2120–2130.

16. Xu F, Deeg HJ. Current status of allogeneic hematopoietic cell transplantation for MDS. Curr Pharm Des. 2012;18(22):3215–3221.

17. Paun O, Lazarus HM. Allogeneic hematopoietic cell transplantation for acute myeloid leukemia in first complete remission: have the indications changed? Curr Opin Hematol. 2012;19(2):95–101.

18. Aiuti A, Biasco L, Scaramuzza S, et al. Lentiviral hematopoietic stem cell gene therapy in patients with Wiskott–Aldrich syndrome. Science. 2013;341 1233151.

19. Phillips GL. Allogeneic hematopoietic stem cell transplantation (HSCT) for high-risk acute myeloid leukemia (AML)/myelodysplastic syndrome (MDS): how can we improve outcomes in the near future? Leukemia Res. 2012;36(12):1490–1495.

20. Riviere I, Dunbar CE, Sadelain M. Hematopoietic stem cell engineering at a crossroads. Blood. 2012;119(5):1107–1116.

21. Watts KL, Adair J, Kiem HP. Hematopoietic stem cell expansion and gene therapy. Cytotherapy. 2011;13(10):1164–1171.

22. Hesse UJ, Weyer J, Meyer G, Isselhard W, Pichlmaier H. Long-term results after porcine islet transplantation. Transplant Proc. 1989;21(1 Pt 3):2763–2764.

23. Acharjee S, Ghosh B, Al-Dhubiab BE, Nair AB. Understanding type 1 diabetes: etiology and models. Can J Diab. 2013;37(4):269–276.

24. Sachedina S, Toth C. Association of comorbidities with increasing severity of peripheral neuropathy in diabetes mellitus. World J Diab. 2013;4(4):135–144.

25. Schneider DA, Kretowicz AM, von Herrath MG. Emerging immune therapies in type 1 diabetes and pancreatic islet transplantation. Diab Obes Metab. 2013;15(7):581–592.

26. van der Windt DJ, Bottino R, Kumar G, et al. Clinical islet xenotransplantation: how close are we? Diabetes. 2012;61(12):3046–3055.

27. Merani S, Toso C, Emamaullee J, Shapiro AM. Optimal implantation site for pancreatic islet transplantation. Br J Surg. 2008;95(12):1449–1461.

28. Kodama S, Kojima K, Furuta S, Chambers M, Paz AC, Vacanti CA. Engineering functional islets from cultured cells. Tissue Eng Part A. 2009;15(11):3321–3329.

29. Korsgren O, Lundgren T, Felldin M, et al. Optimising islet engraftment is critical for successful clinical islet transplantation. Diabetologia. 2008;51(2):227–232.

30. O’Connell PJ, Cowan PJ, Hawthorne WJ, Yi S, Lew AM. Transplantation of xenogeneic islets: are we there yet? Curr Diab Rep. 2013;13(5):687–694.

31. Komoda H, Miyagawa S, Kubo T, et al. A study of the xenoantigenicity of adult pig islets cells. Xenotransplantation. 2004;11(3):237–246.

32. Thompson P, Badell IR, Lowe M, et al. Islet xenotransplantation using gal-deficient neonatal donors improves engraftment and function. Am J Transplant. 2011;11(12):2593–2602.

33. Jeong JH, Yook S, Lee H, Ahn CH, Lee DY, Byun Y. Effects of surface camouflaged islet transplantation on pathophysiological progression in a db/db type 2 diabetic mouse model. Biochem Biophys Res Commun. 2013;433(4):513–518.

34. Elliott RB, Escobar L, Calafiore R, et al. Transplantation of micro- and macroencapsulated piglet islets into mice and monkeys. Transplant Proc. 2005;37(1):466–469.

35. Kumagai-Braesch M, Jacobson S, Mori H, et al. The TheraCyte device protects against islet allograft rejection in immunized hosts. Cell Transplant. 2013;22(7):1137–1146.

36. Shapiro AM, Ricordi C, Hering BJ, et al. International trial of the Edmonton protocol for islet transplantation. N Engl J Med. 2006;355(13):1318–1330.

37. O’Cearbhaill ED, Ng KS, Karp JM. Emerging medical devices for minimally invasive cell therapy. Mayo Clinic Proceed. 2014;89(2):259–273.

38. Vaithilingam V, Fung C, Ratnapala S, et al. Characterisation of the xenogeneic immune response to microencapsulated fetal pig islet-like cell clusters transplanted into immunocompetent C57BL/6 mice. PLoS ONE. 2013;8(3):e59120.

39. Narang AS, Cheng K, Henry J, et al. Vascular endothelial growth factor gene delivery for revascularization in transplanted human islets. Pharm Res. 2004;21(1):15–25.

40. Park JB, Jeong JH, Lee M, Lee DY, Byun Y. Xenotransplantation of exendin-4 gene transduced pancreatic islets using multi-component (alginate, poly-L-lysine, and polyethylene glycol) microcapsules for the treatment of type 1 diabetes mellitus. J Biomater Sci Polym Ed. 2013;24(18):2045–2057.

41. Lumelsky N, Blondel O, Laeng P, Velasco I, Ravin R, McKay R. Differentiation of embryonic stem cells to insulin-secreting structures similar to pancreatic islets. Science. 2001;292(5520):1389–1394.

42. Bonner-Weir S, Weir GC. New sources of pancreatic beta-cells. Nat Biotechnol. 2005;23(7):857–861.

43. Singer NG, Caplan AI. Mesenchymal stem cells: mechanisms of inflammation. Annu Rev Pathol. 2011;6:457–478.

44. Hoogduijn MJ, Popp F, Verbeek R, et al. The immunomodulatory properties of mesenchymal stem cells and their use for immunotherapy. Int Immunopharmacol. 2010;10(12):1496–1500.

45. Liang J, Zhang H, Hua B, et al. Allogeneic mesenchymal stem cells transplantation in treatment of multiple sclerosis. Mult Scler. 2009;15(5):644–646.

46. Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells The international society for cellular therapy position statement. Cytotherapy. 2006;8(4):315–317.

47. Ankrum JA, Ong JF, Karp JM. Mesenchymal stem cells: immune evasive, not immune privileged. Nat Biotechnol. 2014;32(3):252–260.

48. Francois M, Galipeau J. New insights on translational development of mesenchymal stromal cells for suppressor therapy. J Cell Physiol. 2012;227(11):3535–3538.

49. Ankrum J, Karp JM. Mesenchymal stem cell therapy: two steps forward, one step back. Trends Mol Med. 2010;16(5):203–209.

50. Bernardo ME, Ball LM, Cometa AM, et al. Co-infusion of ex vivo-expanded, parental MSCs prevents life-threatening acute GVHD, but does not reduce the risk of graft failure in pediatric patients undergoing allogeneic umbilical cord blood transplantation. Bone Marrow Transplant. 2011;46(2):200–207.

51. Kuzmina LA, Petinati NA, Parovichnikova EN, et al. Multipotent mesenchymal stromal cells for the prophylaxis of acute graft-versus-host disease—a phase II study. Stem Cells Int. 2012;2012 968213.

52. Mendicino M, Bailey AM, Wonnacott K, Puri RK, Bauer SR. MSC-based product characterization for clinical trials: an FDA perspective. Cell Stem Cell. 2014;14(2):141–145.

53. Sarkar D, Spencer JA, Phillips JA, et al. Engineered cell homing. Blood. 2011;118(25):e184–e191.

54. Rombouts WJ, Ploemacher RE. Primary murine MSC show highly efficient homing to the bone marrow but lose homing ability following culture. Leukemia. 2003;17(1):160–170.

55. Ranganath SH, Levy O, Inamdar MS, Karp JM. Harnessing the mesenchymal stem cell secretome for the treatment of cardiovascular disease. Cell Stem Cell. 2012;10(3):244–258.

56. Wobus AM, Boheler KR. Embryonic stem cells: prospects for developmental biology and cell therapy. Physiol Rev. 2005;85(2):635–678.

57. Knoepfler PS. Deconstructing stem cell tumorigenicity: a roadmap to safe regenerative medicine. Stem cells (Dayton, OH). 2009;27(5):1050–1056.

58. Min JY, Yang Y, Converso KL, et al. Transplantation of embryonic stem cells improves cardiac function in postinfarcted rats. J Appl Physiol. 2002;92(1):288–296.

59. Brustle O, Jones KN, Learish RD, et al. Embryonic stem cell-derived glial precursors: a source of myelinating transplants. Science. 1999;285(5428):754–756.

60. Brignier AC, Gewirtz AM. Embryonic and adult stem cell therapy. J Allergy Clin Immunol. 2010;125(Suppl. 2):S336–S344.

61. Yamanaka S. Induced pluripotent stem cells: past, present, and future. Cell Stem Cell. 2012;10(6):678–684.

62. Robinton DA, Daley GQ. The promise of induced pluripotent stem cells in research and therapy. Nature. 2012;481(7381):295–305.

63. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–676.

64. Feng Q, Lu SJ, Klimanskaya I, et al. Hemangioblastic derivatives from human induced pluripotent stem cells exhibit limited expansion and early senescence. Stem Cells. 2010;28(4):704–712.

65. Hu Q, Friedrich AM, Johnson LV, Clegg DO. Memory in induced pluripotent stem cells: reprogrammed human retinal-pigmented epithelial cells show tendency for spontaneous redifferentiation. Stem Cells. 2010;28(11):1981–1991.

66. Wernig M, Zhao J-P, Pruszak J, et al. Neurons derived from reprogrammed fibroblasts functionally integrate into the fetal brain and improve symptoms of rats with Parkinson’s disease. Proc Nat Acad Sci USA. 2008;105(15):5856–5861.

67. Lai RC, Arslan F, Lee MM, et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010;4(3):214–222.

68. Karp JM, Leng Teo GS. Mesenchymal stem cell homing: the devil is in the details. Cell Stem Cell. 2009;4(3):206–216.

69. Komarova S, Roth J, Alvarez R, Curiel DT, Pereboeva L. Targeting of mesenchymal stem cells to ovarian tumors via an artificial receptor. J Ovarian Res. 2010;3:12.

70. Ko IK, Kim BG, Awadallah A, et al. Targeting improves MSC treatment of inflammatory bowel disease. Mol Ther. 2010;18(7):1365–1372.

71. Willemsen RA, Debets R, Chames P, Bolhuis RL. Genetic engineering of T cell specificity for immunotherapy of cancer. Hum Immunol. 2003;64(1):56–68.

72. Brentjens RJ, Santos E, Nikhamin Y, et al. Genetically targeted T cells eradicate systemic acute lymphoblastic leukemia xenografts. Clin Cancer Res. 2007;13(18 Pt 1):5426–5435.

73. Hudson PJ, Souriau C. Engineered antibodies. Nat Med. 2003;9(1):129–134.

74. Blanco B, Holliger P, Vile RG, Alvarez-Vallina L. Induction of human T lymphocyte cytotoxicity and inhibition of tumor growth by tumor-specific diabody-based molecules secreted from gene-modified bystander cells. J Immunol. 2003;171(2):1070–1077.

75. Compte M, Blanco B, Serrano F, et al. Inhibition of tumor growth in vivo by in situ secretion of bispecific anti-CEA x anti-CD3 diabodies from lentivirally transduced human lymphocytes. Cancer Gene Ther. 2007;14(4):380–388.

76. Chan JK, Hamilton CA, Cheung MK, et al. Enhanced killing of primary ovarian cancer by retargeting autologous cytokine-induced killer cells with bispecific antibodies: a preclinical study. Clin Cancer Res. 2006;12(6):1859–1867.

77. Chames P, Van Regenmortel M, Weiss E, Baty D. Therapeutic antibodies: successes, limitations and hopes for the future. Br J Pharmacol. 2009;157(2):220–233.

78. Yu X, Chen D, Zhang Y, et al. Overexpression of CXCR4 in mesenchymal stem cells promotes migration, neuroprotection and angiogenesis in a rat model of stroke. J Neurol Sci. 2012;316(1–2):141–149.

79. Kuwashima N, Nishimura F, Eguchi J, et al. Delivery of dendritic cells engineered to secrete IFN-alpha into central nervous system tumors enhances the efficacy of peripheral tumor cell vaccines: dependence on apoptotic pathways. J Immunol. 2005;175(4):2730–2740.

80. Cheng Z, Ou L, Zhou X, et al. Targeted migration of mesenchymal stem cells modified with CXCR4 gene to infarcted myocardium improves cardiac performance. Mol Ther. 2008;16(3):571–579.

81. Huang J, Zhang Z, Guo J, et al. Genetic modification of mesenchymal stem cells overexpressing CCR1 increases cell viability, migration, engraftment, and capillary density in the injured myocardium. Circ Res. 2010;106(11):1753–1762.

82. Kumar S, Ponnazhagan S. Bone homing of mesenchymal stem cells by ectopic alpha 4 integrin expression. FASEB J. 2007;21(14):3917–3927.

83. Biglari A, Southgate TD, Fairbairn LJ, Gilham DE. Human monocytes expressing a CEA-specific chimeric CD64 receptor specifically target CEA-expressing tumour cells in vitro and in vivo. Gene Ther. 2006;13(7):602–610.

84. Hombach A, Muche JM, Gerken M, et al. T cells engrafted with a recombinant anti-CD30+ receptor target autologous CD30+ cutaneous lymphoma cells. Gene Ther. 2001;8(11):891–895.

85. Carroll RG, June CH. Programming the next generation of dendritic cells. Mol Ther. 2007;15(5):846–848.

86. Gade TP, Hassen W, Santos E, et al. Targeted elimination of prostate cancer by genetically directed human T lymphocytes. Cancer Res. 2005;65(19):9080–9088.

87. Walther W, Stein U. Viral vectors for gene transfer: a review of their use in the treatment of human diseases. Drugs. 2000;60(2):249–271.

88. De Smedt SC, Demeester J, Hennink WE. Cationic polymer based gene delivery systems. Pharm Res. 2000;17(2):113–126.

89. Green JJ, Zhou BY, Mitalipova MM, et al. Nanoparticles for gene transfer to human embryonic stem cell colonies. Nano Lett. 2008;8(10):3126–3130.

90. McBain SC, Yiu HH, Dobson J. Magnetic nanoparticles for gene and drug delivery. Int J Nanomed. 2008;3(2):169–180.

91. Okazaki A, Jo J-I, Tabata Y. A reverse transfection technology to genetically engineer adult stem cells. Tissue Eng. 2007;13(2):245–251.

92. Prabha S, Labhasetwar V. Critical determinants in PLGA/PLA nanoparticle-mediated gene expression. Pharm Res. 2004;21(2):354–364.

93. Salem AK, Searson PC, Leong KW. Multifunctional nanorods for gene delivery. Nat Mater. 2003;2(10):668–671.

94. Vonarbourg A, Passirani C, Desigaux L, et al. The encapsulation of DNA molecules within biomimetic lipid nanocapsules. Biomaterials. 2009;30(18):3197–3204.

95. Cho NH, Cheong TC, Min JH, et al. A multifunctional core–shell nanoparticle for dendritic cell-based cancer immunotherapy. Nat Nanotechnol. 2011;6(10):675–682.

96. Balyasnikova IV, Franco-Gou R, Mathis JM, Lesniak MS. Genetic modification of mesenchymal stem cells to express a single-chain antibody against EGFRvIII on the cell surface. J Tissue Eng Regen Med. 2010;4(4):247–258.

97. Jensen MC, Cooper LJ, Wu AM, Forman SJ, Raubitschek A. Engineered CD20-specific primary human cytotoxic T lymphocytes for targeting B-cell malignancy. Cytotherapy. 2003;5(2):131–138.

98. Levy O, Zhao W, Mortensen LJ, et al. mRNA-engineered mesenchymal stem cells for targeted delivery of interleukin-10 to sites of inflammation. Blood. 2013;122(14):e23–32.

99. Kayser H, Geilen CC, Paul C, Zeitler R, Reutter W. Incorporation of N-acyl-2-amino-2-deoxy-hexoses into glycosphingolipids of the pheochromocytoma cell line PC 12. FEBS Lett. 1992;301(2):137–140.

100. Chakraborty K, Bose A, Pal S, et al. Neem leaf glycoprotein restores the impaired chemotactic activity of peripheral blood mononuclear cells from head and neck squamous cell carcinoma patients by maintaining CXCR3/CXCL10 balance. Int Immunopharmacol. 2008;8(2):330–340.

101. Krishnamachari Y, Pearce ME, Salem AK. Self-assembly of cell–microparticle hybrids. Adv Mater. 2008;20(5):989–993.

102. Sarkar D, Vemula PK, Teo GS, et al. Chemical engineering of mesenchymal stem cells to induce a cell rolling response. Bioconjug Chem. 2008;19(11):2105–2109.

103. Sarkar D, Ankrum JA, Teo GS, Carman CV, Karp JM. Cellular and extracellular programming of cell fate through engineered intracrine-, paracrine-, and endocrine-like mechanisms. Biomaterials. 2011;32(11):3053–3061.

104. Kim SA, Peacock JS. The use of palmitate-conjugated protein a for coating cells with artificial receptors which facilitate intercellular interactions. J Immunol Methods. 1993;158(1):57–65.

105. Ko IK, Kean TJ, Dennis JE. Targeting mesenchymal stem cells to activated endothelial cells. Biomaterials. 2009;30(22):3702–3710.

106. Lo CY, Antonopoulos A, Dell A, Haslam SM, Lee T, Neelamegham S. The use of surface immobilization of P-selectin glycoprotein ligand-1 on mesenchymal stem cells to facilitate selectin mediated cell tethering and rolling. Biomaterials. 2013;34(33):8213–8222.

107. Dennis JE, Cohen N, Goldberg VM, Caplan AI. Targeted delivery of progenitor cells for cartilage repair. J Orthop Res. 2004;22(4):735–741.

108. Englund PT. The structure and biosynthesis of glycosyl phosphatidylinositol protein anchors. Annu Rev Biochem. 1993;62:121–138.

109. Hamdy N, Goustin AS, Desaulniers JP, Li M, Chow CS, Al-Katib A. Sheep red blood cells armed with anti-CD20 single-chain variable fragments (scFvs) fused to a glycosylphosphatidylinositol (GPI) anchor: a strategy to target CD20-positive tumor cells. J Immunol Methods. 2005;297(1–2):109–124.

110. Kean TJ, Duesler L, Young RG, et al. Development of a peptide-targeted, myocardial ischemia-homing, mesenchymal stem cell. J Drug Target. 2012;20(1):23–32.

111. Xia L, McDaniel JM, Yago T, Doeden A, McEver RP. Surface fucosylation of human cord blood cells augments binding to P-selectin and E-selectin and enhances engraftment in bone marrow. Blood. 2004;104(10):3091–3096.

112. Sackstein R, Merzaban JS, Cain DW, et al. Ex vivo glycan engineering of CD44 programs human multipotent mesenchymal stromal cell trafficking to bone. Nat Med. 2008;14(2):181–187.

113. Sackstein R. Directing stem cell trafficking via GPS. Methods Enzymol. 2010;479:93–105.

114. Kawabata K, Migita M, Mochizuki H, et al. Ex vivo cell-mediated gene therapy for metachromatic leukodystrophy using neurospheres. Brain Res. 2006;1094(1):13–23.

115. Parr AM, Kulbatski I, Tator CH. Transplantation of adult rat spinal cord stem/progenitor cells for spinal cord injury. J Neurotrauma. 2007;24(5):835–845.

116. Poh KK, Sperry E, Young RG, Freyman T, Barringhaus KG, Thompson CA. Repeated direct endomyocardial transplantation of allogeneic mesenchymal stem cells: safety of a high dose, “off-the-shelf”, cellular cardiomyoplasty strategy. Int J Cardiol. 2007;117(3):360–364.

117. Mi Q, Riviere B, Clermont G, Steed DL, Vodovotz Y. Agent-based model of inflammation and wound healing: insights into diabetic foot ulcer pathology and the role of transforming growth factor-beta1. Wound Repair Regen. 2007;15(5):671–682.

118. Nakashima Y, Deie M, Yanada S, Sharman P, Ochi M. Magnetically labeled human natural killer cells, accumulated in vitro by an external magnetic force, are effective against HOS osteosarcoma cells. Int J Oncol. 2005;27(4):965–971.

119. Arbab AS, Jordan EK, Wilson LB, Yocum GT, Lewis BK, Frank JA. In vivo trafficking and targeted delivery of magnetically labeled stem cells. Hum Gene Ther. 2004;15(4):351–360.

120. Polyak B, Fishbein I, Chorny M, et al. High field gradient targeting of magnetic nanoparticle-loaded endothelial cells to the surfaces of steel stents. Proc Natl Acad Sci USA. 2008;105(2):698–703.

121. Roth JC, Curiel DT, Pereboeva L. Cell vehicle targeting strategies. Gene Therapy. 2008;15(10):716–729.

122. Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol. 2007;2(12):751–760.

123. Ferreira L, Karp JM, Nobre L, Langer R. New opportunities: the use of nanotechnologies to manipulate and track stem cells. Cell Stem Cell. 2008;3(2):136–146.

124. Jain RA. The manufacturing techniques of various drug loaded biodegradable poly(lactide-co-glycolide) (PLGA) devices. Biomaterials. 2000;21(23):2475–2490.

125. Jain RA, Rhodes CT, Railkar AM, Malick AW, Shah NH. Comparison of various injectable protein-loaded biodegradable poly(lactide-co-glycolide) (PLGA) devices: in situ-formed implant versus in situ-formed microspheres versus isolated microspheres. Pharm Dev Technol. 2000;5(2):201–207.

126. Jain RK, Stylianopoulos T. Delivering nanomedicine to solid tumors. Nat Rev Clin Oncol. 2010;7(11):653–664.

127. Lin SY, Chen KS, Teng HH, Li MJ. In vitro degradation and dissolution behaviours of microspheres prepared by three low molecular weight polyesters. J Microencapsul. 2000;17(5):577–586.

128. Wissing SA, Kayser O, Muller RH. Solid lipid nanoparticles for parenteral drug delivery. Adv Drug Deliv Rev. 2004;56(9):1257–1272.

129. Heurtault B, Saulnier P, Pech B, Proust JE, Benoit JP. A novel phase inversion-based process for the preparation of lipid nanocarriers. Pharm Res. 2002;19(6):875–880.

130. Beduneau A, Saulnier P, Anton N, et al. Pegylated nanocapsules produced by an organic solvent-free method: evaluation of their stealth properties. Pharm Res. 2006;23(9):2190–2199.

131. Collnot EM, Ali H, Lehr CM. Nano- and microparticulate drug carriers for targeting of the inflamed intestinal mucosa. J Control Release. 2012;161(2):235–246.

132. Lechardeur D, Verkman AS, Lukacs GL. Intracellular routing of plasmid DNA during non-viral gene transfer. Adv Drug Deliv Rev. 2005;57(5):755–767.

133. Liang W, Lam J. Endosomal escape pathways for non-viral nucleic acid delivery systems. Molecular regulation of endocytosis; Dr. Brian Ceresa (editor), InTech (Rijeka, Croatia); 2012: [Chapter 17].

134. Chen Y-Z, Yao X-L, Tabata Y, Nakagawa S, Gao J-Q. Gene carriers and transfection systems used in the recombination of dendritic cells for effective cancer immunotherapy. Clin Dev Immunol. 2010;2010 565643.

135. Nagane K, Jo J-i, Tabata Y. Promoted adipogenesis of rat mesenchymal stem cells by transfection of small Interfering RNA complexed with a cationized dextran. Tissue Eng Part A. 2010;16(1):21–31.

136. Jo J-I, Nagaya N, Miyahara Y, et al. Transplantation of genetically engineered mesenchymal stem cells improves cardiac function in rats with myocardial infarction: benefit of a novel nonviral vector, cationized dextran. Tissue Eng. 2007;13(2):313–322.

137. Thakor DK, Teng YD, Obata H, Nagane K, Saito S, Tabata Y. Nontoxic genetic engineering of mesenchymal stem cells using serum-compatible pullulan-spermine/DNA anioplexes. Tissue Eng C Methods. 2011;17(2):131–144.

138. Zhu L, Chang DW, Dai L, Hong Y. DNA damage induced by multiwalled carbon nanotubes in mouse embryonic stem cells. Nano Lett. 2007;7(12):3592–3597.

139. Mooney E, Dockery P, Greiser U, Murphy M, Barron V. Carbon nanotubes and mesenchymal stem cells: biocompatibility, proliferation and differentiation. Nano Lett. 2008;8(8):2137–2143.

140. Iijima S. Helical microtubules of graphitic carbon. Nature. 1991;354(6348):56–58.

141. Zhang W, Zhang Z, Zhang Y. The application of carbon nanotubes in target drug delivery systems for cancer therapies. Nanoscale Res Lett. 2011;6:555.

142. Magrez A, Kasas S, Salicio V, et al. Cellular toxicity of carbon-based nanomaterials. Nano Lett. 2006;6(6):1121–1125.

143. Kocbek P, Obermajer N, Cegnar M, Kos J, Kristl J. Targeting cancer cells using PLGA nanoparticles surface modified with monoclonal antibody. J Control Release. 2007;120(1–2):18–26.

144. Bareford LM, Swaan PW. Endocytic mechanisms for targeted drug delivery. Adv Drug Deliv Rev. 2007;59(8):748–758.

145. Gao Z, Zhang L, Hu J, Sun Y. Mesenchymal stem cells: a potential targeted-delivery vehicle for anti-cancer drug, loaded nanoparticles. Nanomedicine. 2013;9(2):174–184.

146. Biagiotti S, Paoletti MF, Fraternale A, Rossi L, Magnani M. Drug delivery by red blood cells. IUBMB Life. 2011;63(8):621–631.

147. Pierigè F, Serafini S, Rossi L, Magnani M. Cell-based drug delivery. Adv Drug Deliv Rev. 2008;60(2):286–295.

148. 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(3):211–222.

149. Rossi L, Serafini S, Cenerini L, et al. Erythrocyte-mediated delivery of dexamethasone in patients with chronic obstructive pulmonary disease. Biotechnol Appl Biochem. 2001;33(Pt 2):85–89.

150. Rossi L, Castro M, D’Orio F, 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(1):57–63.

151. Sprandel U, Way JL. International society for the use of resealed erythrocytes Meeting Irsee G erythrocytes as drug carriers in medicine New York, NY: Plenum Press; 1997.

152. Magnani M, Fazi A, Mangani F, Rossi L, Mancini U. Methanol detoxification by enzyme-loaded erythrocytes. Biotechnol Appl Biochem. 1993;18:217–226 Pt 3.

153. Magnani M, Laguerre M, Rossi L, et al. Acetaldehyde dehydrogenase-loaded erythrocytes as bioreactors for the removal of blood acetaldehyde. Alcohol Clin Exp Res. 1989;13(6):849.

154. 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(2):207.

155. de Chastellier C, Lang T, Thilo L. Phagocytic processing of the macrophage endoparasite, Mycobacterium avium, in comparison to phagosomes which contain Bacillus subtilis or latex beads. Eur J Cell Biol. 1995;68(2):167–182.

156. Rossi L, Brandi G, Malatesta M, et al. Effect of listeriolysin O-loaded erythrocytes on Mycobacterium avium replication within macrophages. J Antimicrob Chemother. 2004;53(5):863–866.

157. Antonelli A, Crinelli R, Bianchi M, et al. Efficient inhibition of macrophage TNF-alpha production upon targeted delivery of K48R ubiquitin. Br J Haematol. 1999;104(3):475–481.

158. Fraternale A, Casabianca A, Orlandi C, et al. Macrophage protection by addition of glutathione (GSH)-loaded erythrocytes to AZT and DDI in a murine AIDS model. Antiviral Res. 2002;56(3):263–272.

159. Fraternale A, Casabianca A, Rossi L, et al. Erythrocytes as carriers of reduced glutathione (GSH) in the treatment of retroviral infections. J Antimicrob Chemother. 2003;52(4):551–554.

160. Palamara AT, Perno CF, Aquaro S, Bue MC, Dini L, Garaci E. Glutathione inhibits HIV replication by acting at late stages of the virus life cycle. AIDS Res Hum Retroviruses. 1996;12(16):1537–1541.

161. Buhl R, Jaffe HA, Holroyd KJ, et al. Systemic glutathione deficiency in symptom-free HIV-seropositive individuals. Lancet. 1989;2(8675):1294–1298.

162. Garaci E, Palamara AT, Di Francesco P, Favalli C, Ciriolo MR, Rotilio G. Glutathione inhibits replication and expression of viral proteins in cultured cells infected with Sendai virus. Biochem Biophys Res Commun. 1992;188(3):1090–1096.

163. Magnani M, Fraternale A, Casabianca A, et al. Antiretroviral effect of combined zidovudine and reduced glutathione therapy in murine AIDS. AIDS Res Hum Retroviruses. 1997;13(13):1093–1099.

164. Mihm S, Galter D, Droge W. Modulation of transcription factor NF kappa B activity by intracellular glutathione levels and by variations of the extracellular cysteine supply. FASEB J. 1995;9(2):246–252.

165. Murata Y, Shimamura T, Hamuro J. The polarization of T(h)1/T(h)2 balance is dependent on the intracellular thiol redox status of macrophages due to the distinctive cytokine production. Int Immunol. 2002;14(2):201–212.

166. Palamara AT, Perno CF, Ciriolo MR, et al. Evidence for antiviral activity of glutathione: in vitro inhibition of herpes simplex virus type 1 replication. Antiviral Res. 1995;27(3):237–253.

167. Staal FJ, Roederer M, Israelski DM, et al. Intracellular glutathione levels in T cell subsets decrease in HIV-infected individuals. AIDS Res Hum Retroviruses. 1992;8(2):305–311.

168. Arima H, Sakamoto T, Aramaki Y, Ishidate K, Tsuchiya S. Specific inhibition of nitric oxide production in macrophages by phosphorothioate antisense oligonucleotides. J Pharm Sci. 1997;86(10):1079–1084.

169. Chiarantini L, Cerasi A, Fraternale A, et al. Inhibition of macrophage iNOS by selective targeting of antisense PNA. Biochemistry. 2002;41(26):8471–8477.

170. Egholm M, Buchardt O, Christensen L, et al. PNA hybridizes to complementary oligonucleotides obeying the Watson–Crick hydrogen-bonding rules. Nature. 1993;365(6446):566–568.

171. Wittung P, Kajanus J, Edwards K, Nielsen P, Norden B, Malmstrom BG. Phospholipid membrane permeability of peptide nucleic acid. FEBS Lett. 1995;365(1):27–29.

172. Choi M-R, Stanton-Maxey KJ, Stanley JK, et al. A cellular Trojan horse for delivery of therapeutic nanoparticles into tumors. Nano Lett. 2007;7(12):3759–3765.

173. Chen X, Lin X, Zhao J, et al. A tumor-selective biotherapy with prolonged impact on established metastases based on cytokine gene-engineered MSCs. Mol Ther. 2008;16(4):749–756.

174. Brennen WN, Chen S, Denmeade SR, Isaacs JT. Quantification of mesenchymal stem cells (MSCs) at sites of human prostate cancer. Oncotarget. 2013;4(1):106–117.

175. Roger M, Clavreul A, Venier-Julienne M-C, et al. Mesenchymal stem cells as cellular vehicles for delivery of nanoparticles to brain tumors. Biomaterials. 2010;31(32):8393–8401.

176. Hamidi M, Tajerzadeh H. Carrier erythrocytes: an overview. Drug Deliv. 2003;10(1):9–20.

177. Beck Z, Brown BK, Wieczorek L, et al. Human erythrocytes selectively bind and enrich infectious HIV-1 virions. PLoS ONE. 2009;4(12):e8297.

178. Hu CM, Zhang L, Aryal S, Cheung C, Fang RH. Erythrocyte membrane-camouflaged polymeric nanoparticles as a biomimetic delivery platform. Proc Natl Acad Sci USA. 2011;108(27):10980–10985.

179. Murciano JC, Medinilla S, Eslin D, Atochina E, Cines DB, Muzykantov VR. Prophylactic fibrinolysis through selective dissolution of nascent clots by tPA-carrying erythrocytes. Nat Biotechnol. 2003;21(8):891–896.

180. Ganguly K, Krasik T, Medinilla S, et al. Blood clearance and activity of erythrocyte-coupled fibrinolytics. J Pharmacol Exp Ther. 2005;312(3):1106–1113.

181. Armstead WM, Ganguly K, Riley J, et al. RBC-coupled tPA prevents whereas tPA aggravates JNK MAPK-mediated impairment of ATP- and Ca-sensitive K channel-mediated cerebrovasodilation after cerebral photothrombosis. Transl Stroke Res. 2012;3(1):114–121.

182. Muzykantov VR, Sakharov DV, Smirnov MD, Samokhin GP, Smirnov VN. Immunotargeting of erythrocyte-bound streptokinase provides local lysis of a fibrin clot. Biochim Biophys Acta. 1986;884(2):355–362.

183. Castro M, Rossi L, Papadatou B, 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(4):423–426.

184. Annese V, Latiano A, Rossi L, et al. Erythrocytes-mediated delivery of dexamethasone in steroid-dependent IBD patients—a pilot uncontrolled study. Am J Gastroenterol. 2005;100(6):1370–1375.

185. Annese V, Latiano A, Rossi L, et al. 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(1):27–31.

186. Castro M, Knafelz D, Rossi L, et al. Periodic treatment with autologous erythrocytes loaded with dexamethasone 21-phosphate for fistulizing pediatric Crohn’s disease: case report. J Pediatr Gastroenterol Nutr. 2006;42(3):313–315.

187. Aboody KS, Brown A, Rainov NG, et al. Neural stem cells display extensive tropism for pathology in adult brain: evidence from intracranial gliomas. Proc Natl Acad Sci USA. 2000;97(23):12846–12851.

188. Benedetti S, Pirola B, Pollo B, et al. Gene therapy of experimental brain tumors using neural progenitor cells. Nat Med. 2000;6(4):447–450.

189. Kim SK, Kim SU, Park IH, et al. Human neural stem cells target experimental intracranial medulloblastoma and deliver a therapeutic gene leading to tumor regression. Clin Cancer Res. 2006;12(18):5550–5556.

190. Ming GL, Song H. Adult neurogenesis in the mammalian central nervous system. Annu Rev Neurosci. 2005;28:223–250.

191. Singh G. Sources of neuronal material for implantation. Neuropathology. 2001;21(2):110–114.

192. Chen L, Tredget EE, Wu PYG, Wu Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS ONE. 2008;3(4):e1886.

193. Friedenstein AJ, Piatetzky II S, Petrakova KV. Osteogenesis in transplants of bone marrow cells. J Embryol Exp Morphol. 1966;16(3):381–390.

194. Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143–147.

195. Zuk PA, Zhu M, Mizuno H, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7(2):211–228.

196. Bieback K, Kern S, Kluter H, Eichler H. Critical parameters for the isolation of mesenchymal stem cells from umbilical cord blood. Stem Cells. 2004;22(4):625–634.

197. Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol. 2000;109(1):235–242.

198. Goodwin HS, Bicknese AR, Chien SN, Bogucki BD, Quinn CO, Wall DA. Multilineage differentiation activity by cells isolated from umbilical cord blood: expression of bone, fat, and neural markers. Biol Blood Marrow Transplant. 2001;7(11):581–588.

199. Kogler G, Sensken S, Airey JA, et al. A new human somatic stem cell from placental cord blood with intrinsic pluripotent differentiation potential. J Exp Med. 2004;200(2):123–135.

200. van Netten JP, George EJ, Ashmead BJ, Fletcher C, Thornton IG, Coy P. Macrophage–tumour cell associations in breast cancer. Lancet. 1993;342(8875):872–873.

201. Lewis CE, Pollard JW. Distinct role of macrophages in different tumor microenvironments. Cancer Res. 2006;66(2):605–612.

202. Jiang Y, Vaessen B, Lenvik T, Blackstad M, Reyes M, Verfaillie CM. Multipotent progenitor cells can be isolated from postnatal murine bone marrow, muscle, and brain. Exp Hematol. 2002;30(8):896–904.

203. Peng LH, Tsang SY, Tabata Y, Gao JQ. Genetically-manipulated adult stem cells as therapeutic agents and gene delivery vehicle for wound repair and regeneration. J Control Release. 2012;157(3):321–330.

204. Schäffler A, Büchler C. Concise review: adipose tissue-derived stromal cells—basic and clinical implications for novel cell-based therapies. Stem Cells. 2007;25(4):818–827.

205. Choi MR, Stanton-Maxey KJ, Stanley JK, et al. A cellular Trojan horse for delivery of therapeutic nanoparticles into tumors. Nano Lett. 2007;7(12):3759–3765.

206. Kelly PM, Davison RS, Bliss E, McGee JO. Macrophages in human breast disease: a quantitative immunohistochemical study. Br J Cancer. 1988;57(2):174–177.

207. Goswami S, Sahai E, Wyckoff JB, et al. Macrophages promote the invasion of breast carcinoma cells via a colony-stimulating factor-1/epidermal growth factor paracrine loop. Cancer Res. 2005;65(12):5278–5283.

208. Lewis C, Murdoch C. Macrophage responses to hypoxia: implications for tumor progression and anti-cancer therapies. Am J Pathol. 2005;167(3):627–635.

209. O’Sullivan C, Lewis CE, Harris AL, McGee JO. Secretion of epidermal growth factor by macrophages associated with breast carcinoma. Lancet. 1993;342(8864):148–149.

210. Tsutsui S, Yasuda K, Suzuki K, Tahara K, Higashi H, Era S. Macrophage infiltration and its prognostic implications in breast cancer: the relationship with VEGF expression and microvessel density. Oncol Rep. 2005;14(2):425–431.

211. al-Shukri S, Korneev IA. The prognostic factors in bladder cancer patients. Urol Nefrol (Mosk). 1996;6:49–53.

212. Hagemann T, Robinson SC, Schulz M, Trumper L, Balkwill FR, Binder C. Enhanced invasiveness of breast cancer cell lines upon co-cultivation with macrophages is due to TNF-alpha dependent up-regulation of matrix metalloproteases. Carcinogenesis. 2004;25(8):1543–1549.

213. Hanada T, Nakagawa M, Emoto A, Nomura T, Nasu N, Nomura Y. Prognostic value of tumor-associated macrophage count in human bladder cancer. Int J Urol. 2000;7(7):263–269.

214. Leek RD, Lewis CE, Whitehouse R, Greenall M, Clarke J, Harris AL. Association of macrophage infiltration with angiogenesis and prognosis in invasive breast carcinoma. Cancer Res. 1996;56(20):4625–4629.

215. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002;23(11):549–555.

216. Ohno S, Ohno Y, Suzuki N, et al. Correlation of histological localization of tumor-associated macrophages with clinicopathological features in endometrial cancer. Anticancer Res. 2004;24(5C):3335–3342.

217. Oosterling SJ, van der Bij GJ, Meijer GA, et al. Macrophages direct tumour histology and clinical outcome in a colon cancer model. J Pathol. 2005;207(2):147–155.

218. Wyckoff J, Wang W, Lin EY, et al. A paracrine loop between tumor cells and macrophages is required for tumor cell migration in mammary tumors. Cancer Res. 2004;64(19):7022–7029.

219. Kah JC, Wong KY, Neoh KG, et al. Critical parameters in the pegylation of gold nanoshells for biomedical applications: an in vitro macrophage study. J Drug Target. 2009;17(3):181–193.

220. Metz S, Bonaterra G, Rudelius M, Settles M, Rummeny EJ, Daldrup-Link HE. Capacity of human monocytes to phagocytose approved iron oxide MR contrast agents in vitro. Eur Radiol. 2004;14(10):1851–1858.

221. Oude Engberink RD, Blezer EL, Hoff EI, et al. MRI of monocyte infiltration in an animal model of neuroinflammation using SPIO-labeled monocytes or free USPIO. J Cereb Blood Flow Metab. 2008;28(4):841–851.

222. Dou H, Destache CJ, Morehead JR, et al. Development of a macrophage-based nanoparticle platform for antiretroviral drug delivery. Blood. 2006;108(8):2827–2835.

223. Dou H, Grotepas CB, McMillan JM, et al. Macrophage delivery of nanoformulated antiretroviral drug to the brain in a murine model of neuroAIDS. J Immunol. 2009;183(1):661–669.

224. Janeway CA Jr. Frontiers of the immune system. Nature. 1988;333(6176):804–806.

225. June CH. Principles of adoptive T cell cancer therapy. J Clin Invest. 2007;117(5):1204–1212.

226. Harlin H, Meng Y, Peterson AC, et al. Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Res. 2009;69(7):3077–3085.

227. Randolph GJ, Angeli V, Swartz MA. Dendritic-cell trafficking to lymph nodes through lymphatic vessels. Nat Rev Immunol. 2005;5(8):617–628.

228. Huppa JB, Davis MM. T-cell-antigen recognition and the immunological synapse. Nat Rev Immunol. 2003;3(12):973–983.

229. Ilett EJ, Barcena M, Errington-Mais F, et al. Internalization of oncolytic reovirus by human dendritic cell carriers protects the virus from neutralization. Clin Cancer Res. 2011;17(9):2767–2776.

230. Uto T, Akagi T, Yoshinaga K, Toyama M, Akashi M, Baba M. The induction of innate and adaptive immunity by biodegradable poly(gamma-glutamic acid) nanoparticles via a TLR4 and MyD88 signaling pathway. Biomaterials. 2011;32(22):5206–5212.

231. Vajkoczy P, Menger MD. Vascular microenvironment in gliomas. J Neurooncol. 2000;50(1–2):99–108.

232. Ischenko I, Seeliger H, Schaffer M, Jauch KW, Bruns CJ. Cancer stem cells: how can we target them? Curr Med Chem. 2008;15(30):3171–3184.

233. Pardridge WM. Blood-brain barrier drug targeting: the future of brain drug development. Mol Interv. 2003;3(2):90–105 151.

234. Anderson SA, Glod J, Arbab AS, et al. Noninvasive MR imaging of magnetically labeled stem cells to directly identify neovasculature in a glioma model. Blood. 2005;105(1):420–425.

235. Brekke C, Williams SC, Price J, Thorsen F, Modo M. Cellular multiparametric MRI of neural stem cell therapy in a rat glioma model. Neuroimage. 2007;37(3):769–782.

236. Bulte JW, Wu C, Brechbiel MW, et al. Dysprosium-DOTA-PAMAM dendrimers as macromolecular T2 contrast agents Preparation and relaxometry. Invest Radiol. 1998;33(11):841–845.

237. Bulte JW, Kraitchman DL. Monitoring cell therapy using iron oxide MR contrast agents. Curr Pharm Biotechnol. 2004;5(6):567–584.

238. Wu X, Hu J, Zhou L, et al. In vivo tracking of superparamagnetic iron oxide nanoparticle-labeled mesenchymal stem cell tropism to malignant gliomas using magnetic resonance imaging Laboratory investigation. J Neurosurg. 2008;108(2):320–329.

239. Barresi V, Belluardo N, Sipione S, Mudo G, Cattaneo E, Condorelli DF. Transplantation of prodrug-converting neural progenitor cells for brain tumor therapy. Cancer Gene Ther. 2003;10(5):396–402.

240. Ito S, Natsume A, Shimato S, et al. Human neural stem cells transduced with IFN-beta and cytosine deaminase genes intensify bystander effect in experimental glioma. Cancer Gene Ther. 2010;17(5):299–306.

241. Li S, Gao Y, Tokuyama T, et al. Genetically engineered neural stem cells migrate and suppress glioma cell growth at distant intracranial sites. Cancer Lett. 2007;251(2):220–227.

242. Li S, Tokuyama T, Yamamoto J, Koide M, Yokota N, Namba H. Potent bystander effect in suicide gene therapy using neural stem cells transduced with herpes simplex virus thymidine kinase gene. Oncology. 2005;69(6):503–508.

243. Li S, Tokuyama T, Yamamoto J, Koide M, Yokota N, Namba H. Bystander effect-mediated gene therapy of gliomas using genetically engineered neural stem cells. Cancer Gene Ther. 2005;12(7):600–607.

244. Zhao M, Liang C, Li A, et al. Magnetic paclitaxel nanoparticles inhibit glioma growth and improve the survival of rats bearing glioma xenografts. Anticancer Res. 2010;30(6):2217–2223.

245. Kim SK, Cargioli TG, Machluf M, et al. PEX-producing human neural stem cells inhibit tumor growth in a mouse glioma model. Clin Cancer Res. 2005;11(16):5965–5970.

246. Lorico A, Mercapide J, Solodushko V, Alexeyev M, Fodstad O, Rappa G. Primary neural stem/progenitor cells expressing endostatin or cytochrome P450 for gene therapy of glioblastoma. Cancer Gene Ther. 2008;15(9):605–615.

247. van Eekelen M, Sasportas LS, Kasmieh R, et al. Human stem cells expressing novel TSP-1 variant have anti-angiogenic effect on brain tumors. Oncogene. 2010;29(22):3185–3195.

248. Ehtesham M, Kabos P, Kabosova A, Neuman T, Black KL, Yu JS. The use of interleukin 12-secreting neural stem cells for the treatment of intracranial glioma. Cancer Res. 2002;62(20):5657–5663.

249. Stagg J, Lejeune L, Paquin A, Galipeau J. Marrow stromal cells for interleukin-2 delivery in cancer immunotherapy. Hum Gene Ther. 2004;15(6):597–608.

250. Xu G, Jiang XD, Xu Y, et al. Adenoviral-mediated interleukin-18 expression in mesenchymal stem cells effectively suppresses the growth of glioma in rats. Cell Biol Int. 2009;33(4):466–474.

251. Gunnarsson S, Bexell D, Svensson A, Siesjo P, Darabi A, Bengzon J. Intratumoral IL-7 delivery by mesenchymal stromal cells potentiates IFNγ-transduced tumor cell immunotherapy of experimental glioma. J Neuroimmunol. 2010;218(1–2):140–144.

252. Corsten MF, Miranda R, Kasmieh R, Krichevsky AM, Weissleder R, Shah K. MicroRNA-21 knockdown disrupts glioma growth in vivo and displays synergistic cytotoxicity with neural precursor cell delivered S-TRAIL in human gliomas. Cancer Res. 2007;67(19):8994–9000.

253. Ehtesham M, Kabos P, Gutierrez MA, et al. Induction of glioblastoma apoptosis using neural stem cell-mediated delivery of tumor necrosis factor-related apoptosis-inducing ligand. Cancer Res. 2002;62(24):7170–7174.

254. Shah K, Tung CH, Yang K, Weissleder R, Breakefield XO. Inducible release of TRAIL fusion proteins from a proapoptotic form for tumor therapy. Cancer Res. 2004;64(9):3236–3242.

255. Zhu D, Lam DH, Purwanti YI, et al. Systemic delivery of fusogenic membrane glycoprotein-expressing neural stem cells to selectively kill tumor cells. Mol Ther. 2013;21(8):1621–1630.

256. O’Neal DP, Hirsch LR, Halas NJ, Payne JD, West JL. Photo-thermal tumor ablation in mice using near infrared-absorbing nanoparticles. Cancer Lett. 2004;209(2):171–176.

257. Alizadeh D, Zhang L, Hwang J, Schluep T, Badie B. Tumor-associated macrophages are predominant carriers of cyclodextrin-based nanoparticles into gliomas. Nanomedicine. 2010;6(2):382–390.

258. Kennedy LC, Bear AS, Young JK, et al. T cells enhance gold nanoparticle delivery to tumors in vivo. Nanoscale Res Lett. 2011;6(1):283.

259. Cole C, Qiao J, Kottke T, et al. Tumor-targeted, systemic delivery of therapeutic viral vectors using hitchhiking on antigen-specific T cells. Nat Med. 2005;11(10):1073–1081.

260. Ilett EJ, Prestwich RJ, Kottke T, et al. Dendritic cells and T cells deliver oncolytic reovirus for tumour killing despite pre-existing anti-viral immunity. Gene Ther. 2009;16(5):689–699.

261. Power AT, Bell JC. Taming the Trojan horse: optimizing dynamic carrier cell/oncolytic virus systems for cancer biotherapy. Gene Ther. 2008;15(10):772–779.

262. Russell SJ, Peng KW. The utility of cells as vehicles for oncolytic virus therapies. Curr Opin Mol Ther. 2008;10(4):380–386.

263. Alley SC, Okeley NM, Senter PD. Antibody–drug conjugates: targeted drug delivery for cancer. Curr Opin Chem Biol. 2010;14(4):529–537.

264. Banerjee R, Katsenovich Y, Lagos L, McIintosh M, Zhang X, Li CZ. Nanomedicine: magnetic nanoparticles and their biomedical applications. Curr Med Chem. 2010;17(27):3120–3141.

265. Stephan MT, Moon JJ, Um SH, Bershteyn A, Irvine DJ. Therapeutic cell engineering with surface-conjugated synthetic nanoparticles. Nat Med. 2010;16(9):1035–1041.

266. Cheng H, Kastrup CJ, Ramanathan R, et al. Nanoparticulate cellular patches for cell-mediated tumoritropic delivery. ACS Nano. 2010;4(2):625–631.

267. Nakamura K, Ito Y, Kawano Y, et al. Antitumor effect of genetically engineered mesenchymal stem cells in a rat glioma model. Gene Ther. 2004;11(14):1155–1164.

268. Hong X, Miller C, Savant-Bhonsale S, Kalkanis SN. Antitumor treatment using interleukin-12-secreting marrow stromal cells in an invasive glioma model. Neurosurgery. 2009;64(6):1139–1146 [discussion 1146–1137].

269. Nakamizo A, Marini F, Amano T, et al. Human bone marrow-derived mesenchymal stem cells in the treatment of gliomas. Cancer Res. 2005;65(8):3307–3318.

270. Kim SM, Lim JY, Park SI, et al. Gene therapy using TRAIL-secreting human umbilical cord blood-derived mesenchymal stem cells against intracranial glioma. Cancer Res. 2008;68(23):9614–9623.

271. Menon LG, Kelly K, Yang HW, Kim SK, Black PM, Carroll RS. Human bone marrow-derived mesenchymal stromal cells expressing S-TRAIL as a cellular delivery vehicle for human glioma therapy. Stem Cells. 2009;27(9):2320–2330.

272. Sasportas LS, Kasmieh R, Wakimoto H, et al. Assessment of therapeutic efficacy and fate of engineered human mesenchymal stem cells for cancer therapy. Proc Natl Acad Sci USA. 2009;106(12):4822–4827.

273. Miletic H, Fischer Y, Litwak S, et al. Bystander killing of malignant glioma by bone marrow-derived tumor-infiltrating progenitor cells expressing a suicide gene. Mol Ther. 2007;15(7):1373–1381.

274. Kinoshita Y, Kamitani H, Mamun MH, et al. A gene delivery system with a human artificial chromosome vector based on migration of mesenchymal stem cells towards human glioblastoma HTB14 cells. Neurol Res. 2010;32(4):429–437.

275. Uchibori R, Okada T, Ito T, et al. Retroviral vector-producing mesenchymal stem cells for targeted suicide cancer gene therapy. J Gene Med. 2009;11(5):373–381.

276. Gu C, Li S, Tokuyama T, Yokota N, Namba H. Therapeutic effect of genetically engineered mesenchymal stem cells in rat experimental leptomeningeal glioma model. Cancer Lett. 2010;291(2):256–262.

277. Sonabend AM, Ulasov IV, Tyler MA, Rivera AA, Mathis JM, Lesniak MS. Mesenchymal stem cells effectively deliver an oncolytic adenovirus to intracranial glioma. Stem Cells. 2008;26(3):831–841.

278. Josiah DT, Zhu D, Dreher F, Olson J, McFadden G, Caldas H. Adipose-derived stem cells as therapeutic delivery vehicles of an oncolytic virus for glioblastoma. Mol Ther. 2010;18(2):377–385.

279. Negroni L, Samson M, Guigonis JM, Rossi B, Pierrefite-Carle V, Baudoin C. Treatment of colon cancer cells using the cytosine deaminase/5-fluorocytosine suicide system induces apoptosis, modulation of the proteome, and Hsp90beta phosphorylation. Mol Cancer Ther. 2007;6(10):2747–2756.

280. Altanerova V, Cihova M, Babic M, et al. Human adipose tissue-derived mesenchymal stem cells expressing yeast cytosinedeaminase:uracil phosphoribosyltransferase inhibit intracerebral rat glioblastoma. Int J Cancer. 2012;130(10):2455–2463.

281. Pulkkanen KJ, Yla-Herttuala S. Gene therapy for malignant glioma: current clinical status. Mol Ther. 2005;12(4):585–598.

282. Amano S, Li S, Gu C, et al. Use of genetically engineered bone marrow-derived mesenchymal stem cells for glioma gene therapy. Int J Oncol. 2009;35(6):1265–1270.

283. Song C, Xiang J, Tang J, et al. Thymidine kinase gene modified bone marrow mesenchymal stem cells as vehicles for antitumor therapy. Hum Gene Ther. 2011;22(4):439–449.

284. Mori K, Iwata J, Miyazaki M, et al. Bystander killing effect of tymidine kinase gene-transduced adult bone marrow stromal cells with ganciclovir on malignant glioma cells. Neurol Med Chir (Tokyo). 2010;50(7):545–553.

285. Choi SA, Lee JY, Wang KC, et al. Human adipose tissue-derived mesenchymal stem cells: characteristics and therapeutic potential as cellular vehicles for prodrug gene therapy against brainstem gliomas. Eur J Cancer. 2012;48(1):129–137.

286. Djouad F, Plence P, Bony C, et al. Immunosuppressive effect of mesenchymal stem cells favors tumor growth in allogeneic animals. Blood. 2003;102(10):3837–3844.

287. von Bahr L, Batsis I, Moll G, et al. Analysis of tissues following mesenchymal stromal cell therapy in humans indicates limited long-term engraftment and no ectopic tissue formation. Stem Cells. 2012;30(7):1575–1578.

288. DeFrancesco L. Landmark approval for Dendreon’s cancer vaccine. Nat Biotechnol. 2010;28(6):531–532.

289. Noh YW, Jang YS, Ahn KJ, Lim YT, Chung BH. Simultaneous in vivo tracking of dendritic cells and priming of an antigen-specific immune response. Biomaterials. 2011;32(26):6254–6263.

290. Azzi J, Tang L, Moore R, et al. Polylactide-cyclosporin a nanoparticles for targeted immunosuppression. FASEB J. 2010;24(10):3927–3938.

291. Kravtzoff R, Colombat PH, Desbois I, et al. Tolerance evaluation of L-asparaginase loaded in red blood cells. Eur J Clin Pharmacol. 1996;51(3–4):221–225.

292. Brandenberger R, Burger S, Campbell A, Fong T, Lapinskas E, Rowley JA. Cell therapy bioprocessing. BioProcess Int. 2011;9(s1):30–37.

..................Content has been hidden....................

You can't read the all page of ebook, please click here login for view all page.
Reset
3.129.21.47