Summary:
Multiple stem cell types exhibit inherent tropism for cancer, and engineered stem cells have been utilized as therapeutic agents to specifically target cancer cells. Recently, stem cells have been engineered to target multiple surface-receptors on tumor cells, as well as endothelial and immune cells in the tumor microenvironment. In this review, we discuss the rationales and strategies of developing multiple receptor targeted stem cells, their mechanisms of action, and the promises and challenges they hold as cancer therapeutics.
Introduction:
Since the identification of aberrant signal transduction through cell surface receptors as a common feature of many cancer types, the targeting of these pathways has proven to be a promising route in the development of cancer therapies. By tailoring therapeutic agents to selectively target surface receptors indicative of malignant cells, the cytotoxicity to neighboring cells can be significantly limited (1,2). However, resistance, both inherent and acquired, is a significant limitation on the efficacy monospecific antibodies and ligands targeting cell surface receptors due to the activation of alternative signaling pathways and receptors in cancer cells (3). Heterogeneity within and between tumors also limits the functionality of therapies targeting a single tumor biomarker (4,5,6). Due to its role in the tumor progression and resistance, the tumor microenvironment has also become a promising target for immune-based therapy (7). Therefore, targeting of multiple cell surface receptors on cancer cells and associated cells has the potential to target heterogeneous tumors, as well as impact the tumor microenvironment, and, therefore, has become an exciting new direction for targeted therapy in cancer (8) (Table 1).
Table 1:
Cell surface receptors expressed on tumor cells and within the tumor microenvironment that have been or have the potential to be* utilized in stem cell-delivered cell surface receptor targeting therapies and their respective targeting agents. Cell surface receptors with differential or unique expression on the surface of tumor cells or cells of the tumor microenvironment are attractive targets for cell surface receptor targeting therapies. *Stem cell (SC) delivered anti-tumor receptor targeting agents have not yet been explored for this receptor type. Abbreviations: TNFR – tumor necrosis factor receptor, DR4/5 – death receptor 4/5, EGFR – epidermal growth factor receptor, IFNAR - Interferon-α/β cell surface receptor complex, GPCR – G protein coupled receptor, BnR – Bombesin receptor, SSTR – somatostatin receptor, ET – endothelin receptors, FR – folate receptor, TfR – transferrin receptor, FGFR – fibroblast growth receptors, TRAIL - Tumor necrosis factor-related apoptosis inducing ligand, oHSV – oncolytic herpes simplex virus, TSP – thrombospondin, scFV- single chain variable fragment, BC – breast cancer, UC – uterine cancer, NB – neuroblastoma, GBM – glioblastoma, NK – natural killer AML – acute myeloid leukemia, ALL – acute lymphoblastic leukemia, LC – lung cancer, OC – ovarian cancer.
Receptor type | SC delivered anti-tumor targeting agent | Expression in tumor cells and cells of the tumor microenvironment | References |
---|---|---|---|
TNFRs | |||
Death receptors 4 and 5 (DR4/5) | TRAIL | Expressed in non-malignant and malignant cells, suggested to be expressed at higher levels in malignant cells | 120,121 |
CD137/4–1BB | CD137/4–1BB ligand (CD137L/4–1BBL) | Expressed on T cells | 86 |
EGFRs | |||
EGFRvIII | EGFR targeting antibodies or nanobodies (ENb) | Expressed only in glioma, novel variant | 80 |
EGFR2/HER2 | anti-HER2 antibodies (trastuzaumab) | Overexpressed in BC, gastric, cervical, UC, gallbladder and testicular cancers, bladder carcinomas, and extrahepatic cholangiocarcinomas | 122 |
Nectin-1 | oHSV | Expressed in neurons, as well as a variety of cancers including NB, GBM, and squamous cell carcinoma. Often predicative of oHSV susceptibility. | 123,124 |
IFNAR | Type I interferons (IFNα, IFNβ) | Expressed on a variety of tumor cells (fibrosarcoma, cervical, colon, and lung carcinoma, etc.) and immune cells (T cells, NK cells, and dendritic cells) | 64–66,125 |
CD36 | TSP | Expressed on endothelial cells,present in highly vascularized tumors such as GBM | 62 |
CD20 | anti-CD20 antibodies (rituximab, scFv–CD20) | Exclusively expressed in B cell precursors, mature B cells, and B cell lymphoma (Hodgkin’s and non-Hodgkin lymphoma) | 84 |
CD33 | anti-CD33 antibodies (gemtuzumab ozogamicin (Mylotarg), anti- CD33–anti-CD3 bsAb) |
Overexpressed on of the majority of AML cells | 87 |
CD19 | anti-CD19 antibodies (blinatumomab, anti-CD3–antiCD19 TandAb) | Expressed in B-precursor ALL and B cell lymphoma | 87 |
CD3 | anti-CD3 antibodies (anti-CD33–anti-CD3 bsAB, TandAb, blinatunmomab, anti-CD3scFv) | Expressed on T cells | 86 |
GPCRs | |||
BnR | * | Expressed in GBM, NB, non-small cell and small cell LC (novel receptor), intestinal, thymic, gastrointestinal, lung, and bronchial carcinoid tumors, OC, pancreatic, prostate, BC, head and neck, colon, UC, and renal cancers, Ewing sarcoma, pituitary tumors | 126,127 |
SSTRs | * | Expressed in small cell LC, prostate, BC, gastric cancers, neuroendocrine tumors, colorectal and hepatocellular carcinoma | 128 |
ET | * | Expressed in melanoma | 129,130 |
Integrins | |||
ανβn (ex ανβ3) | * | Expressed in activated endothelial cells and tumor cells (U87MG GBM cells) | 131,132 |
α−3 | * | Expressed in OC and BC, and melanoma | 133 |
FR (α, β, γ) | * | Expressed in OCand BC, endometrial and renal cell carcinomas, lung andenocarcinomas and mesotheliomas | 134 |
TfRs | |||
TfR1 (CD71) | * | Overexpressed in glioma and GBM, expressed in BC and OC | 135–137 |
FGFRs | * | Alterations in expression in BC, bladder, prostate, LC, and haematological malignancies | 138 |
The multiple receptor targeting strategy is still limited by the unfavorable pharmacokinetics of potential therapeutics, due to rapid clearance from the tumor site and limited bio-transformation and availability. Additionally, the difficulty in maintaining sustained concentrations of biologically based therapeutics at the site of the tumor, particularly in the case of solid tumors where therapeutics are often unable reach the core of the tumor (9). The tumor-tropic nature of adult stem cells through solid tumors and to the sites of micro-metastases has made them attractive candidates for use as therapeutics in cancer. Stem cells can be engineered to stably express and then release biological therapeutics, thus becoming factories for the production of therapeutics, which can improve the half-lives of these agents and provide continuous exposure at the tumor-site (9,10). This circumvents the short half-lives of therapeutics such as antibodies and anti-cancer proteins, by permitting site-specific and long-term delivery of therapeutic agents. Stem cells delivered therapeutics are readily accessable at the site of tumors and can infiltrate solid tumors more readily than biological therapies delivered using traditional methods (9,10). The goal of this review is to discuss the recent advances the development of multiple cell surface receptor targeting therapies through the use engineered stem cells.
Stem cell types:
Stem cells (SCs) are the source of all embryogenic tissue generation and any continued regeneration of tissue throughout adulthood. A variety of SC types have been explored as potential biologically based therapeutics, and from these studies adult stem cells seem to have the most promise for use in clinical studies (11,12,13). Embryonic SCs (ESCs) were the first SC type to be explored for possible sources of stem cell therapy but were abandoned primarily due to ethical concerns in the sourcing of these cells (14,15). Therefore, adult SCs, which can be successfully isolated from various sources within the body, have become the focus of stem cell-based therapies. The three primary types of adult SCs that have been explored for these purposes are mesenchymal SCs (MSC), neural SCs (NSCs), and induced pluripotent SCs (iPSCs). MSCs can be derived from bone marrow, adipose tissue, umbilical cord blood (UCB), and various other tissues (16). NSCs, also called neural precursor cells, do not refer to a single cell type but rather a heterogeneous population of self-renewing and multipotent cells present in both the developing and adult central nervous system (17). NSCs have shown particular promise in developing cell-based therapies for neurodegenerative disorders, but also hold potential for other conditions including certain cancers (12,17,18,19,20). iPSCs, which are generated from the reprogramming of somatic cells by expression of defined transcription factors, have emerged as a popular and effective SC type for therapy. These cells can be derived from a wide variety of starting cells, providing incredible accessibility compared to other SC types for the development of iPSC-based therapies and disease models (11,21). Engineered hematopoietic stem cells (HSCs) have also been explored for their potential to generate sustained anti-tumor effect by engraftment of engineered HSCs expressing either T-cell receptors (TCRs) or chimeric antigen receptors (CARs) (22). However, HSCs have not been explored for bifunctional tumor cell surface receptor targeting.
Stem cell homing and immunogenicity
SCs offer the opportunity to improve the efficacy of biological therapeutics by improving pharmacokinetics and biotransformation and producing continuous potent concentrations of therapeutics at the tumor site. Two properties, the abilities to evade the host immune system and to home to tumor loci, are necessary to establish an effective cellular therapy; however, if and to what extent these characteristics are present in SCs is still subject to debate. SCs have been documented to have immunosuppressive properties via the expression of cytokines and growth factors that can impact immune pathways within a host organism (23,24,25). The implantation of foreign SCs, both adult SCs and induced allogeneic donor SCs, has still been shown to elicit an immune response and lead to rejection of the cells in vivo (26,27,28,29,30). SCs have also been shown to have inherent immuno-modulatory effects. NSC implantation in the brain has been shown to induce an immunological response, indicated by infiltration of lymphocytes, and to induce of pro-inflammatory cytokines IL-1β and TNFα in the brain (25). Allogeneic MSCs have been shown to inhibit the activation of CD4+ T cells and to alter the humoural immune response both in vitro and in vivo (29). Furthermore in contrast to ESC derived cells, iPSCs have been shown to induce a T-cell dependent immune response in syngeneic recipients. This immunogenicity was attributed to differential cell surface marker expression and may in fact limit the efficacy of iPSC-based therapy (26). While allogeneic MSCs are less immunogenic than other allogeneic non-SC cell types, such as fibroblasts, they are not entirely immune privileged but rather they are able to escape host rejection transiently (29,31).
The second necessary characteristic for cellular delivery, migratory potential, was first demonstrated for neural SCs (NSC) and neural progenitor cells in xenograft mouse models (10). NSCs have been shown not only to integrate into primary tumors, but also to track to micro-metastases that are typical of brain tumors like glioblastoma (32). Tumor tropic characteristics have also been demonstrated in numerous SC types (33,34,35). Although the molecular mechanisms of tumor tropism are not yet completely understood, several chemokine-chemokine receptor pathways have been implicated in this characteristic. The most well studied of these is stromal cell-derived factor 1 (SDF1) and its receptor CXC-chemokine receptor 4 (CXCR4), which have been shown to have a significant role in the migration of NSCs, MSCs, ESCs, HSCs, and iPSCs (33,34,36–40). Other chemokine-chemokine receptor pairs such as SCF-c-Kit, HGF-c-Met, VEGF-VEGFR, PDGF-PDGFR, MCP-1-CCR2, and HMGB1-RAGE as well as cytokines that co-operate with G-protein coupled receptors and growth factor receptors have also been shown to be involved in stem cell recruitment and migration (35). The nature of the SC population such as heterogeneity, culture conditions, and expression of migratory factors can influence the migration of SCs to tumor sites (10). Additionally, the tumor microenvironment, particularly degree of hypoxia, extent of vascularization, and inflammation, can also play a role in the SC migration toward tumor loci (10). Tropism to tumor loci is a key feature of a variety of SC types that make them attractive for cell therapy in cancer, and the ability to use or enhance this feature may improve therapeutic potential of SCs. Better understanding the factors that influence SC tumor tropism and immune evasion can aid in developing SCs as tools for therapeutic delivery by capitalizing on the extent to which these characteristics are intrinsic and by potentially modifying the SCs or the tumor environment to promote these characteristics.
Creating cell surface receptor targeted stem cells
Unmodified SCs have been shown to have anti-tumor effects primarily attributed to factors secreted by the SCs as well as physical interactions between the SCs and tumor cells (41–48). Particularly, the antitumor effects of MSCs have been attributed in part to enhancing the host antitumor immune responses, demonstrating the benefits of the inherent immune-modulatory effects of SCs (43). However, quite promisingly SCs have been modified to target cancer cell surface receptors, greatly improving their anti-cancer efficacy. Commonly, this is achieved by viral transduction of SCs using lentivirus (LV) or safer alternatives such as retrovirus or adeno-associated virus (AAV) (10). Among the viral vectors, LVs have the unique ability to infect non-dividing cells, and therefore have been widely utilized in the creation of engineered stem cells. The anti-tumor agents that have been used to target cell surface receptors through engineered SCs include traditional biologics, such as antibodies and ligands, as well as oncolytic viruses (Figure 1).
Figure 1. Engineered receptor targeted stem cells promote tumor cell death:
SCs can be modified in a variety of ways to generate an anti-tumor response by targeting cell surface receptors. SC engineered to express therapeutic proteins or antibodies that function directly on tumor cells (TRAIL, EGF agonists, anti-CD20, CD33, CD19) or that act on the tumor microenvironment to elicit an immune response (IL-10, IL-18, IFNα/β) or on surround cells (stromal effectors or blood vessel effects (aaTSP-1). SCs loaded with oncolytic virus amplify within the SC eventually leading to its rupture and release of viral progeny. These viral particles will go on to infect tumor cells, killing them and inducing an immune response at the tumor site. Abbreviations: SC- stem cell, TRAIL- tumor necrosis factor-related apoptosis inducing ligand, IL- interleukin, IFN- interferon, EGF- epidermal growth factor, aaTSP-1- anti-angiogenic thrombospondin,
Targeting of Multiple Cell Surface Receptors
Single ligand targeting multiple receptors on tumor cells or receptors on tumor associated cell types:
While the use of SCs has been shown to improve the delivery of therapies targeting cell surface receptors, the targeting of multiple receptors with a single therapy has to potential to combat resistance and tumor heterogeneity (Figure 2). Pro-apoptotic protein tumor necrosis factor-related apoptosis inducing ligand (TRAIL), binds to death receptors 4 and 5 (DR4/5), which are expressed in a tumor specific manner and induce caspase mediated apoptosis upon TRAIL binding (49,50,51). In mouse models of highly malignant brain tumors, glioblastomas (GBM), both bone marrow-derived MSC and NSCs engineered to express a secretable (S) form of TRAIL (S-TRAIL) have shown anti-tumor effects (51,52,53,54). Additionally, MSC-TRAIL has recently been shown to be effective in preclinical models of pancreatic ductal adenocarcinoma, Ewing sarcoma, lung cancer, and melanoma (55,56,57,58). Photochemical internalization and nanoparticle-mediated transfections have also been used to engineer TRAIL secreting MSCs which also demonstrated an anti-tumor effect in preclinical models, indicating the potential for safer engineering of MSCs which may improve translation to the clinic (59,60). In addition to molecules that directly target multiple receptors on tumor cells, anti-angiogenic agents, such as three type 1 repeats (3TSR) of thrombospondin (TSP)-1, act as indirect effectors on tumor associated endothelial cells and blocking the ability of tumors to stimulate new blood vessel formation (61). Studies have shown that MSCs engineered to express anti-angiogenic (aa) 3TSR have anti-tumor efficacy by binding to CD36 receptors on tumor associated endothelial cells and preventing angiogenesis in a mouse model of GBM (61,62). aa3TSR also has the ability to bind to CD36 receptors on tumor cells and has been shown to upregulate expression of DR4/5 in tumor cells, priming these cells for treatment by S-TRAIL (62). Interferon (IFN)β belongs to type I interferons that bind to the interferon-α/β cell surface receptor complex (IFNAR) (63) and has direct anti-tumor activity (64,65,66). IFNβ also acts as an immunostimulatory molecule by indirectly provoking an antitumor response via modulation of the immune system (67,68,69). However, despite these bi-functional modes of action, the clinical translation of IFNβ treatments for cancer so far has been restricted by its short half-life and systemic toxicity (70,71,72). MSCs engineered to express mouse IFNβ have been shown to target multiple cell types in a resection model of mouse GBM. Encapsulated MSC-IFNβ placed in the tumor resection cavity, were able to simultaneously recruit CD4/CD8 T cells to the site of tumor resection and induce apoptosis in tumor cells, leading to increased survival in mice bearing GBMs (73).
Figure 2. Potentiating stem cell efficacy through bifunctional SCs:
The efficacy of engineered SCs can be improved by targeting multiple cell surface receptors with a single bifunctional SC, increasing specificity and overcoming resistance. Bifunctional SCs 1) release a single molecule, such as TRAIL, that simultaneously targets two cell surface receptors (DR4 and DR5); 2) release two different molecules that target distinct cell surface receptors, such as TRAIL and aaTSP-1, or release a bifunctional fusion protein to target two different cell surface receptors, such as ENb-TRAIL or scFv-CD20-TRAIL 3) release bifunctional fusion proteins to target and recruit immune cells from the tumor microenvironment such as T cells (anti-CD33-antiCD3 and anti-CD19-anti-CD3) bsAbs fusion proteins. Abbreviations: SC- stem cell, TRAIL-tumor necrosis factor-related apoptosis inducing ligand, 3TSP-1- anti-angiogenic thrombospondin, ENb- epidermal growth factor receptor nanobody, bsAb-bispecific antibody
SC engineered to express ligands targeting multiple receptors on cells in the tumor microenvironment:
Because tumor cells and the associated cells within the tumor microenvironment express a wide variety of cell surface receptors, one therapeutic strategy is to engineer SCs to simultaneously release multiple therapeutic proteins targeting multiple receptors on tumor and associated cells in the tumor microenvironment (74) (Figure 2). A growing number of studies are adopting this approach, utilizing bimodal SCs and SCs secreting bifunctional molecules and often taking advantage of synergistic biological relationships between two therapies. MSCs expressing both aa3TSR and S-TRAIL have been shown to target both DR4/5 on tumor cells and CD36 on tumor cells and tumor associated endothelial cells in TRAIL resistant GBMs, leading to extended survival times in mouse models (62). The dual expression of cytokines by SCs to target tumor cells and immune cells has also been shown to be effective. In a rat tumor model, SCs expressing both IL-18 and IFNβ significantly prolonged survival and inhibited tumor growth both by promoting tumor cell apoptosis and stimulating antitumor cytokine production and CD4/CD8 T cell infiltration of the intracranial gliomas. The robust antitumor response of MSC-IL18 and IFNβ indicated a synergistic effect of the two immunostimulatory cytokines when used in combination (75). Similarly, MSCs engineered to co-express the cytokines IL-10 and IFNγ reduced tumor growth in a mouse model of hepatocellular carcinoma by cell cycle arrest, demonstrated by increased expression of cell cycle inhibitors p21 and p27, as well as modulation of the MAPK pathway (76).
Stem cells expressing antibodies and ligands targeting multiple receptors:
Traditional antibodies have a highly conserved tetrameric structure consisting of a pair of heavy chains and a pair of light chains linked by disulphide bonds. Alternative antibody structures have been developed, including heavy chain-only antibodies (77): which do not have a CH1 domain, antigen-binding fragment or single-chain variable fragments (scFv): which contain only two variable domains, one from each of the heavy and light chains, and nanobodies: which consist of the only the heavy-chain variable fragment (VHH) and are on the scale of nanometres (8). Antibody fragments have increased ability to penetrate tumor tissues, due to their small size, but their efficacy is limited because of their short half-life and rapid renal clearance in vivo (78).
SCs can be modified by viral or non-viral methods to express either full antibodies or antibody fragments (79). The epidermal growth factor receptor (EGFR), has also been a target of SC delivered antibody therapy. SCs engineered to express EGFR and its glioma associated variant (EGFRvIII) targeting nanobodies (ENb), which consist solely of the antigen-specific domain (VHH), have been shown to reduce EGFR signaling in vitro and inhibit tumor growth in a mouse model of gliomas (80). In an effort to take advantage of the efficient cell-mediated delivery of antibodies for ligand-based therapies, researchers recently engineered NSCs containing phosphatase and tensin homolog (PTEN) with the leader sequence from human light-chain immunoglobulin G (IgG) to target inactivated PTEN signaling in GBM. This antibody-ligand fusion molecule showed increased secretion from NSCs and potential to transfer to neighboring cells, demonstrating an increased potential for NSC delivered PTEN therapy (81).
Although the area of SC delivered bifunctional proteins is still in its infancy, several studies have explored the delivery of single bi-functional molecules targeting two different cell surface receptors (9) (Figure 2). Due to the low expression levels of DR 4/5 and EGFR in adult SCs, SCs can be engineered to express a nanobody against EGFR (ENb) fused to TRAIL (ENb-TRAIL) without significant auto-toxicity. SC-ENb-TRAIL was shown to outperform SC-ENb alone in mouse tumor models by inhibiting EGFR and inducing caspase-mediate cell death (80). Recent studies have shown that ENb-TRAIL blocks EGFR signaling via the binding of ENb to EGFR which in turn induces DR5 clustering at the plasma membrane and thereby primes tumor cells to caspase-mediated apoptosis (82). MSCs expressing a dimeric EGFR-specific diabody single-chain TRAIL (Db-scTRAIL) were also shown to have antitumor activity against colorectal cancer in vitro and in vivo, particularly in combination with bortezomib, a proteasome inhibitor (83). MSCs loaded with a bifunctional protein consisting of scFv-CD20 fused with S-TRAIL was shown to inhibit cell proliferation and induce apoptosis in CD20 positive lymphoma cells. Additionally, in a xenograft model of B-cell lymphoma, i.v. injection of MSC-scFvCD20-TRAIL significantly inhibited tumor growth when compared to MSC-TRAIL without any noticeable toxicity (84).
T-cell engaging bispecific antibodies (bsAbs) are a promising tool for cancer treatment as they establish a transient synapse between T cells and a tumor cell by surface antigen binding on tumor cell with one arm and simultaneously recruiting T cells via the signal transmitting CD3 domain of the T-cell receptor complex (85) (Figure 2). Previous studies have explored MSCs for delivering a fully humanized anti-CD33-anti-CD3 bsAb, which is capable of redirecting human T cells against CD33-expressing leukemic cells. MSC expressing anti-CD33-anti-CD3 bsAb were shown to redirect T cells efficiently against CD33 presenting target cells and eliminated autologous acute myeloid leukemia (AML) cells over time. The immune response against AML cells was further enhanced further by providing T cells an additional co-stimulus via the CD137-CD137 ligand axis through MSCs expressing CD137L (86). More recent studies have shown that MSCs secreting TandAb, a tetravalent bispecific tandem diabody for CD3 and CD19, were also effective in a mouse model of B-cell lymphoma by inhibiting tumor growth (87).
Stem cells loaded with bi-functional Oncolytic Viruses targeting multiple receptors
Oncolytic viruses (OVs) are viruses that either naturally or by genetic modification have the ability to selectively infect and replicate in tumor cells, while sparing surrounding healthy tissue (88,89). Oncolytic viruses make use of cell surface receptors to gain entry into cells (89). However, clearance of the virus by the host immune system and off-target infection of noncancerous cells makes systemic administration of OVs ineffective, while insufficient viral spread in intratumoral injection also limits this route of delivery (90). SCs have been explored as a possible means of overcoming issues in OV delivery by shielding the virus from anti-viral immunity and directing viral particles more directly toward the tumor. A variety of SC types, including MSCs, have been explored for the delivery of oncolytic adenovirus either systemically or intratumorally (91,92,93,94,95). However, only limited number of OVs have been engineered that offer the utilization of at least two different cell surface receptors, one for the viral entry and other for the ligand/antibody expressed by the SC loaded with OVs. Recently, MSCs loaded with adenovirus were used to modify hepatocellular carcinoma (HCC) cells in vivo to present anti-CD3scfv on their surface. Once presented, this surface marker activated lymphocytes leading to cancer cell specific lysis and inhibited tumor growth in mouse models (96). Furthermore, OVs have been genetically engineered to express various agents targeting tumor cell receptors and/or immune cell receptors with the intention of improving their specificity and efficacy. oHSV engineered with TRAIL has been shown to inhibit tumor progression and prolong survival in mice bearing resistant intracerebral tumors (97). While MSC delivered oncolytic adenovirus expression TRAIL has been shown to be effective in preclinical models of pancreatic cancer, demonstrating the potential for SC delivery of OVs with additional anti-tumor agents (98).
Stem cell released Extracellular Vesicles
A variety of cells types release extracellular vesicles (EVs), which can contained a variety of biological molecules including mRNA, miRNA, proteins and lipids (99). EVs may be taken up by target cells by fusion with the plasma membrane, binding to receptors on the cell surface, or by endocytosis by phagocytosis (100). Two of the most well described ligand-receptor complexes involved in exosome endocytosis are low-density lipoprotein (LDL) and its receptor (LDLR), and transferrin (Tf) and the transferrin receptor (TfR). Other protein-protein interactions linked to EV intake include lectins, adhesion molecules, such as cadherins and integrins, heparin sulfate proteoglycans, and T-cell immunoglobulins and mucins, including EGF/EGFR (101). EVs, which include both exosomes and microvesicles, may be responsible for the paracrine influences of MSCs on adjacent cells by transfer of biological material to enhance communication between cell types (102,103). MSC delivered EV-miRNA mimics (miR-124 and miR-125) to GBM cells, which are deficient in expression of these miRNAs has been shown to have antitumor effects in a mouse model of GBM (104). In addition to miRs, MSCs have also been used to delivery anti-miRs, which block miR function. MSC derived exosomes were used to deliver anti-miR-9 to GBM cells sensitizing these cells to temozolomide, and thus reverse chemoresistance (100,105) Exosomes from MSCs engineered to express an siRNA against GRP78, which is overexpressed in Sorafenib resistant cancer cells, were also shown to effective in combination with Sorafenib against hepatocellular carcinoma cells in vitro and in vivo, further demonstrating the ability SC derived exosomes to reverse chemoresistance (106). SC derived exosomes have also been used to target the tumor microenvironment. Exosomes derived form mESCs engineered to express granulocyte-macrophage colony-stimulating factor (GM-CSF) were shown to inhibit the growth mouse models of lung cancer by activating CD8+ T effector responses and acting as a prophylactic cancer vaccine (107). Menstrual MSC derived exosomes have been identified as inhibitors of angiogenesis in prostate, breast cancer, and oral squamous cell carcinomas by a reduction of VEGF secretion by endothelial cells leading to anti-tumor effects (108,109).
Perspectives
The use of engineered SCs to target multiple cell surface receptors has the potential to overcome some of the most difficult challenges in developing effective cancer treatments: resistance, tumor heterogeneity, and poor pharmacokinetics. The success of anti-cancer SCs in preclinical studies has been exciting for researchers in the field; however, as these therapies begin to be translated to the clinic, significant caution must be taken to ensure safety and efficacy of these therapies. Improvements in the homing of SCs, developing effective methods of non-viral engineering, and a strong understanding of the mechanisms underlying the efficacy of anti-tumor SCs are all essential for the continued pursuit of these therapies.
Recent studies have aimed to address clinical limitations on the use of engineered SCs, such as the use of viral transduction in the generation of therapeutic SC lines, which has safety concerns due to immunogenicity and oncogenic effects of viruses (59). The use of non-viral polylysine-modified polyethylenimine (PEI-PLL) copolymer-based transfection to generate engineered MSCs (110) indicate that such methods may hold to key for safe and expedient translation of modified SCs to clinical settings. A further limitation in the clinical application of SC delivered cancer therapies is the potential for immune rejection after implantation. Therefore, the generation of immune evasive SCs may hold the potential for advancing these therapies. Recently the studies have demonstrated methods of generating hypoimmunogenic or immunosuppressive SCs either by inactivation of the major histocompatibility complex (MHC) class I and II genes and activation of CD47 or by ex vivo induction of apoptosis (111,112). Both of these strategies could improve the safety of SC based therapies by reducing the risk of adverse immune response to transplantation.
The continued development of more clinically relevant tumor models will improve the potential for therapeutic success of treatments developed in the lab. Profiling of patient-derived tumors can be used to establish new targets for therapies, aid in understanding the mechanisms of existing therapies, and determine what tumors will respond to what therapies based on their specific cancer profile (113). Engineered SCs offer the opportunity to develop more personalized medicine, where the therapeutic SCs can be developed to have optimal efficacy against a specific tumor and its profile. Furthermore, establishing more true-to-life animal models would aid in the screening of SC based therapies. As of now, most animal models rely on the surgical introduction of cancer cells from cancer cell lines into immunocompromised host animals. This model poorly reflects the true development and subsequent environment of tumors and have limited utility in screening therapies, especially when there is an immunomodulatory component. Genetically engineered mouse model that develop tumors without introduction of outside cells or incorporating patient-derived tumor samples into animal models may hold more utility in testing therapies that depend significantly on the tumor microenvironment (10).
Clinical trials testing gene engineered SC targeting single cell surface receptors have been completed or are currently active. Genexine, Inc. sponsored trial, to establish the safety and efficacy of MSCs expressing IL-12 (GX-051) in patients with head and neck cancers in 2014. trial was conducted at MD Anderson Cancer Center in 2015 to establish tolerable doses of MSCs expressing IFNβ in patients with ovarian cancer. Furthermore, trial is currently recruiting patients for MSCs expressing TRAIL (TACTICAL) for non-small cell lung cancer. MSCs loaded with oncolytic adenovirus (ICOVIR-5) were also explored for their safety and efficacy in metastatic neuroblastoma and melanoma (114,115). Finally, clinical trials for NSCs expressing cytosine deaminase in combination with 5-Fluorocytosine () or 5-Fluorocytosine and Leucovorin () have either been completed or are active respectively. While these single receptor targeting SC delivered therapies are being explored in the clinic, multiple cell surface receptor targeting SC based therapies have not yet reached clinical trial stage.
SC based therapies, particularly those targeting multiple aberrant signaling pathways, offer the potential to significantly improve the efficacy of biologically based therapies that have previously shown limited efficacy due to unfavorable pharmacokinetics, resistance, or tumor heterogeneity. Engineered SC-based therapies can also be combined with other non-cell delivered immune-based therapies or traditional chemotherapies (116,117) to potentially further enhance efficacy by targeting multiple pathways simultaneously. Furthermore, engineered SC-based therapies have an enormous potential to be used in combination with other cell based cancer therapies, such as CAR-T cells. However, as with all newly developing therapies, caution must be taken to prioritize safety and preserve the integrity of field moving forward. As clinical trials have begun, the safety of these therapies will continue to be tested rigorously while preclinical trials will continue to explore the potential of these therapies. SCs engineered to express receptor-targeted ligands have also been frequently combined with suicide therapy like HSV-TK to provide synergistic therapeutic effect and improve the safety of therapeutic SCs by ensuring their eradication post-treatment (110,118). Recently, the novel suicide gene iCasp9, which binds to the bioinert small molecule (AP20187) leading to cell death, has been explored in combination with MSC-TRAIL with promising results demonstrating coexistence between the suicide and anti-cancer mechanisms (119).
References
- 1.Richter M, Zhang H. Receptor-Targeted Cancer Therapy. DNA Cell Biol 2005;24(5), 271–282. [DOI] [PubMed] [Google Scholar]
- 2.Mendelsohn J, Baselga J The EGF receptor family as targets for cancer therapy. Oncogene 2000;19(56):6550–6565. [DOI] [PubMed] [Google Scholar]
- 3.Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer 2013;13:714–726. [DOI] [PubMed] [Google Scholar]
- 4.Eirew P, Steif A, Khattra J et al. Dynamics of genomic clones in breast cancer patient xenografts at single-cell resolution. Nature 2015;518(7539):422–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Patel AP, Tirosh I, Trombetta JJ et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 2014;344(6190):1396–1401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Verhaak RG, Hoadley KA, Purdom E et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010;17(1):98–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med 2013;19(11):1423–1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Zhu Y, Choi SH, Shah K. Multifunctional receptor-targeting antibodies for cancer therapy. Lancet Oncol 2015;16(15):e543–e554. [DOI] [PubMed] [Google Scholar]
- 9.Choi SH, Shah K. Engineering bifunctional proteins and stem cells: next generation of targeted cancer therapies. Discov Med 2016;22(120):157–166. [PubMed] [Google Scholar]
- 10.Stuckey DW, Shah K. Stem cell-based therapies for cancer treatment: separating hope from hype. Nat Rev Cancer 2014;14(10):683–691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.de Lazaro I, Yilmazer A, Kostarelos, K. Induced pluripotent stem (iPS) cells: A new source for cell-based therapeutics? J Control Release 2014;185:37–44. [DOI] [PubMed] [Google Scholar]
- 12.Nam H, Lee KH, Nam DH, Joo KM. Adult human neural stem cell therapeutics: Current developmental status and prospect. World J Stem Cell 2015;7(1):126–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Savla JJ, Nelson BC, Perry CN, Adler ED. Induced pluripotent stem cells for the study of cardiovascular disease. J Am Coll Cardiol 2014;64(5),512–519. [DOI] [PubMed] [Google Scholar]
- 14.Reubinoff BE, Pera MF, Fong CY et al. Embryonic stem cell lines from human blastocysts: Somatic differentiation in vitro. Nat Biotechnol 2000;18(4):399–404. [DOI] [PubMed] [Google Scholar]
- 15.Winkler J, Hescheler J, Sachinidis A. Embryonic stem cells for basic research and potential clinical applications in cardiology. Biochim Biophys Acta 2005;1740(2):240–248. [DOI] [PubMed] [Google Scholar]
- 16.Sun Y, Williams A, Waisbourd M et al. Stem cell therapy for glaucoma: Science or snake oil? Surv Ophthalmol 2015;60(2):93–105. [DOI] [PubMed] [Google Scholar]
- 17.Martino G, Pluchino S. The therapeutic potential of neural stem cells. Nat Rev Neurosci 2006;7(5):395–406. [DOI] [PubMed] [Google Scholar]
- 18.Kim SU. Human neural stem cells genetically modified for brain repair in neurological disorders. Neuropathology 2004;24(3):159–171. [DOI] [PubMed] [Google Scholar]
- 19.Kim SU, Lee HJ, Kim YB. Neural stem cell-based treatment for neurodegenerative diseases. Neuropathology 2013;33(5):491–504. [DOI] [PubMed] [Google Scholar]
- 20.Shah K Imaging neural stem cell fate in mouse model of glioma. Curr Protoc Stem Cell Biol 2009;Chapter 5:Unit 5A.1. [DOI] [PubMed] [Google Scholar]
- 21.Abbott A Cell rewind wins medicine Nobel. Nature 2012;490(7419):151–152. [DOI] [PubMed] [Google Scholar]
- 22.Adair JE, Kubek SP, Kiem HP. Hematopoietic Stem Cell Approaches to Cancer. Hematol Oncol Clin North Am 2017;31(5):897–912. [DOI] [PubMed] [Google Scholar]
- 23.Jones BJ, McTaggart SJ. Immunosuppression by mesenchymal stromal cells: from culture to clinic. Exp Hematol 2008;36(6):733–741. [DOI] [PubMed] [Google Scholar]
- 24.Bernardo ME Fibbe WE. Mesenchymal stromal cells: sensors and switchers of inflammation. Cell Stem Cell 2013;13(4):392–402. [DOI] [PubMed] [Google Scholar]
- 25.Milwid JM, Elman JS, Li M et al. Enriched protein screening of human bone marrow mesenchymal stromal cell secretions reveals MFAP5 and PENK as novel IL-10 modulators. Mol Ther 2014;22(5):999–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Muraoka K, Shingo T, Yasuhara T et al. The high integration and differentiation potential of autologous neural stem cell transplantation compared with allogeneic transplantation in adult rat hippocampus. Exp Neurol 2006;199(2):311–327. [DOI] [PubMed] [Google Scholar]
- 27.Zhao T, Zhang ZN, Rong Z, Xu Y. Immunogenicity of induced pluripotent stem cells. Nature 2011;474(7350):212–215. [DOI] [PubMed] [Google Scholar]
- 28.Eliopoulos N, Stagg J, Lejeune L et al. Allogeneic marrow stromal cells are immune rejected by MHC class I- and class II-mismatched recipient mice. Blood 2005;106(13), 4057–4065. [DOI] [PubMed] [Google Scholar]
- 29.Zangi L, Margalit R, Reich-Zeliger S et al. Direct imaging of immune rejection and memory induction by allogeneic mesenchymal stromal cells. Stem Cells 2009;27(11):2865–2874. [DOI] [PubMed] [Google Scholar]
- 30.Schu S, Nosov M, O’Flynn L et al. Immunogenicity of allogeneic mesenchymal stem cells. J Cell Mol Med 2012;16(9):2094–2103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Ankrum JA, Ong JF, Karp JM. Mesenchymal stem cells: immune evasive, not immune privileged. Nat Biotechnol 2014;32(3):252–260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Park SA, Ryu CH, Kim SM et al. CXCR4-transfected human umbilical cord blood-derived mesenchymal stem cells exhibit enhanced migratory capacity toward gliomas. Int J Oncol 2011;38(1):97–103. [PubMed] [Google Scholar]
- 34.Koizumi S, Gu C, Amano S et al. Migration of mouse-induced pluripotent stem cells to glioma-conditioned medium is mediated by tumor-associated specific growth factors. Oncol Lett 2011;2(2):283–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Momin EN, Vela G, Zaidi HA, Quinones-Hinojosa A. The oncogenic potential of mesenchymal stem cells in the treatment of cancer: Directions for future research. Curr Immunol Rev 2010;6(2):137–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Xu Q, Yuan X, Xu M et al. Chemokine CXC receptor 4--mediated glioma tumor tracking by bone marrow--derived neural progenitor/stem cells. Mol Cancer Ther 2009;8(9):2746–2753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Suarez-Alvarez B, Lopez-Vazquez A, Lopez-Larrea C. Mobilization and homing of hematopoietic stem cells. Adv Exp Med Biol 2012;741:152–170. [DOI] [PubMed] [Google Scholar]
- 38.Shi M, Li J, Lio L et al. Regulation of CXCR4 expression in human mesenchymal stem cells by cytokine treatment: role in homing efficiency in NOD/SCID mice. Haematologica 2007;92:897–904. [DOI] [PubMed] [Google Scholar]
- 39.Wynn RF, Hart CA, Corradi-Perini C et al. A small proportion of mesenchymal stem cells strongly expresses functionally active CXCR4 receptor capable of promoting migration to bone marrow. Blood 2004;104(9):2643–2645. [DOI] [PubMed] [Google Scholar]
- 40.Guo Y, Hangoc G, Bian H et al. SDF-1/CXCL12 enhances survival and chemotaxis of murine embryonic stem cells and production of primitive and definitive hematopoietic progenitor cells. Stem Cells 2005;23(9):1324–1332. [DOI] [PubMed] [Google Scholar]
- 41.Ding DC, Chang YH, Shyu WC, Lin SZ. Human umbilical cord mesenchymal stem cells: A new era for stem cell therapy. Cell Transplant 2015;24(3):339–347. [DOI] [PubMed] [Google Scholar]
- 42.Hendijani F, Javanmard SH, Sadeghi-Aliabadi H. Human Wharton’s jelly mesenchymal stem cell secretome display antiproliferative effect on leukemia cell line and produce additive cytotoxic effect in combination with doxorubicin. Tissue Cell 2015;47(3):229–234. [DOI] [PubMed] [Google Scholar]
- 43.Kawabata A, Ohta N, Seiler G et al. Naive rat umbilical cord matrix stem cells significantly attenuate mammary tumor growth through modulation of endogenous immune responses. Cytotherapy 2014;15(5):586–597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Wu S, Ju GQ, Du T et al. Microvesicles derived from human umbilical cord Wharton’s jelly mesenchymal stem cells attenuate bladder tumor cell growth in vitro and in vivo. PLoS One 2013;8(4):e61366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Yang C, Lei D, Ouyang W et al. Conditioned media from human adipose tissue-derived mesenchymal stem cells and umbilical cord-derived mesenchymal stem cells efficiently induced the apoptosis and differentiation in human glioma cell lines in vitro. Biomed Res Int 2014; 2014:109389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Motaln H, Gurden K, Hren M et al. Human mesenchymal stem cells exploit the immune response mediating chemokines to impact the phenotype of glioblastoma. Cell Transplant 2012;21(7):1529–1545. [DOI] [PubMed] [Google Scholar]
- 47.Schichor C, Albrecht V, Korte B et al. Mesenchymal stem cells and glioma cells form a structural as well as a functional syncytium in vitro. Exp Neurol 2012;234(1):208–219. [DOI] [PubMed] [Google Scholar]
- 48.Qiao L, Xu Z, Zhao T et al. Suppression of tumorigenesis by human mesenchymal stem cells in a hepatoma model. Cell Res 2008;18(4):500–507. [DOI] [PubMed] [Google Scholar]
- 49.Jo M, Kim TH, Seol DW et al. Apoptosis induced in normal human hepatocytes by tumor necrosis factor-related apoptosis-inducing ligand. Nat Med 2000;6(5):564–567. [DOI] [PubMed] [Google Scholar]
- 50.Thorburn A Death receptor-induced cell killing. Cell Signal 2004;16(2):139–144. [DOI] [PubMed] [Google Scholar]
- 51.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Shah K, Bureau E, Kim DE et al. Glioma therapy and real-time imaging of neural precursor cell migration and tumor regression. Ann Neurol 2005;57(1):34–41. [DOI] [PubMed] [Google Scholar]
- 53.Tang XJ, Lu JT, Tu HJ et al. TRAIL-engineered bone marrow-derived mesenchymal stem cells: TRAIL expression and cytotoxic effects on C6 glioma cells. Anticancer Res 2014;34(2):729–734. [PubMed] [Google Scholar]
- 54.Wang XJ, Xiang BY, Ding YH et al. Human menstrual blood-derived mesenchymal stem cells as a cellular vehicle for malignant glioma gene therapy. Oncotarget 2017;8(35):58309–58321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Spano C, Grisendi G, Golinelli G et al. Soluble TRAIL armed human MSC as gene therapy for pancreatic cancer. Sci Rep 2019;9(1):1788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Guiho R, Biteau K, Grisenid G et al. TRAIL delivered by mesenchymal stromal/stem cells counteracts tumor development in orthotropic Ewing sarcoma models. Int J Cancer 2016;139(12):2802–2811. [DOI] [PubMed] [Google Scholar]
- 57.Yan C, Song X, Yu W. Human umbilical cord mesenchymal stem cells delivering sTRAIL home to lung cancer mediated by MCP-1/CCR2 axis and exhibit antitumor effects. Tumour Biol 2016;37(6):8425–8435. [DOI] [PubMed] [Google Scholar]
- 58.Jing HX, Duan de J, Zhou H et al. Adipose-derived mesenchymal stem cell-facilitated TRAIL expression in melanoma treatment in vitro. Mol Med Rep 2016;14(1):195–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Han J, Hwang HS, Na K. TRAIL-secreting human mesenchymal stem cells engineering by non-viral vector and photochemical internalization for pancreatic cancer gene therapy. Biomaterials 2018;182:258–268. [DOI] [PubMed] [Google Scholar]
- 60.Jiang X, Fitch S, Wang C et al. Nanoparticle engineered TRAIL-overexpressing adipose-derived stem cell target and eradicate glioblastoma via intracranial delivery. Proc Natl Acad Sci USA 2016;113(48):13857–13862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Choi SH, Tamura K, Khajuria RK et al. Anti-angiogenic variant of TSP-1 targets TRAIL-resistant brain tumor cells. Mol Ther 2015; 23(2):235–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Plantanias LC. Mechanism of type-I- and type-II-interferon-mediated signaling. Nat Rev immunol 2005;5(5):375–386. [DOI] [PubMed] [Google Scholar]
- 64.Dong Z, Juang SH, Kumar R et al. Suppression of tumorigenicity and metastasis in murine UV-2237 fibrosarcoma cells by infection with a retroviral vector harboring the interferon-beta gene. Cancer Immunol Immunother 1998;46(3):137–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Qin XQ, Tao N, Dergay A et al. Interferon beta gene therapy inhibits tumor formation and causes regression of establish tumors in immune-deficient mice. Proc Natl Acad Sci USA 1998;95(24):14411–14416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lykhova AA, Kudryavets YI, Strokovska LI et al. Suppression of proliferation, tumorigenicity and metastasis of lung cancer cells after their transduction by interferon-beta gene in baculovirus vector. Cytokine 2015;71(2):318–326. [DOI] [PubMed] [Google Scholar]
- 67.Fuertes MB, Kacha AK, Kline J et al. Host type I IFN signal are required for antitumor CD8+ T cell response through CD8α+ dendritic cells. J Exp Med 2011;208(10):2005–2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Jablonska J, Leschner S, Westphal K, et al. Neutrophils responsive to endogenous IFN-beta regulate tumor angiogenesis and growth in a mouse tumor model. J Clin Invest 2010;120(4):1151–1164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Tough DF, Borrow P, Sprent J. Induction of bystander T cell proliferation by viruses and type I interferon in vivo. Science. 1996;272(5270):1947–1950. [DOI] [PubMed] [Google Scholar]
- 70.Fierlbeck G, Ulmer A, Schreiner T et al. Pharmacodynamics of recombinant IFN-beta during long-term treatment of malignant melanoma. J Interferon Cytokine Res 1996;16(10):777–781. [DOI] [PubMed] [Google Scholar]
- 71.Salmon P, Le Contonnec JY, Galazka A, et al. Pharmacokinetic and pharmacodynamics of recombinant human interferon-beta in healthy male volunteers. J Interferon Cytokine Res 1996;16(10):759–764. [DOI] [PubMed] [Google Scholar]
- 72.Trinchieri G Type I interferon: friend or foe? J Exp Med.2010; 207(10):2053–2063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Choi SH. Suckey DW, Pignatta S et al. Tumor resection recruit effector T ell and boosts therapeutic effect of encapsulates stem cells expressing IFNβ in glioblastomas. Clin Cancer Res 2017;23(22):7047–7058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Shah K Stem cell-based therapies for tumors in the brain: are we there yet? Neuro oncol 2016;18(8):1066–1078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Xu G, Guo Y, Seng Z et al. Bone marrow-derived mesenchymal stem cells co-expressing interleukin-18 and interferon-beta exhibit potent antitumor effect against intracranial glioma in rats. Oncol Rep 2015;34(4):1915–1922. [DOI] [PubMed] [Google Scholar]
- 76.Wang H, Wang J, Shi X, Ding Y. Genetically engineered bone marrow-derived mesenchymal stem cells co-expressing IFN-γ and IL-10 inhibit hepatocellular carcinoma by modulating MAPK pathway. J BUON 2017b;22(6):1515–1524. [PubMed] [Google Scholar]
- 77.Hamers-Casterman C, Atarhouch T, Muyldermans S et al. Naturally occurring antibodies devoid of light chains. Nature 1993;363:446–448. [DOI] [PubMed] [Google Scholar]
- 78.Chapman AP, Antoniw P, Spitali M et al. Therapeutic antibody fragments with prolonged in vivo half-lives. Nat Biotechnol 1999;17:780–783. [DOI] [PubMed] [Google Scholar]
- 79.Frank RT, Najbauer J, Aboody KS. Concise Review: Stem Cells As an Emerging Platform for Antibody Therapy of Cancer. Stem Cells 2010;28(11)2084–2087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.van de Water JA, Bagci-Onder T, Agarwal AS et al. Therapeutic stem cells expressing variants of EGFR-specific nanobodies have antitumor effects. Proc Natl Acad Sci USA 2012;109(41):16642–16647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Lavictoire SJ, Gont A, Julian LM et al. Engineering PTEN-L for cell-mediated delivery. Mol Ther Methods Clin Dev 2017;9:12–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Zhu Y, Bassoff N, Reinshagen C et al. Bi-specific molecule against EGFR and death receptors simultaneously targets proliferation and death pathways in tumors. Sci Rep 2017;7(1):2602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Marini I, Siegnemun M, Hutt M et al. Antitumor Activity of a Mesenchymal Stem Cell Line Stably Secreting a Tumor-Targeted TNF-Related Apoptosis-Inducing Ligand Fusion Protein. Front Immunol 2017;8:536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Yan C, Li S, Li Z et al. Human umbilical cord mesenchymal stem cells as vehicles of CD20-specific TRAIL fusion protein delivery: a double-target therapy against non-Hodgkin’s lymphoma. Mol Pharm 2013;10(1):142–151. [DOI] [PubMed] [Google Scholar]
- 85.Stamova S, Felmann A, Cartellieri M et al. Generation of single-chain bispecific green fluorescent protein fusion antibodies for imaging of antibody-induced T cell synapses. Anal Biochem 2012;423(2):261–268. [DOI] [PubMed] [Google Scholar]
- 86.Aliperta R, Cartellieri M, Feldmann A et al. Bispecific antibody releasing-mesenchymal stromal cell machinery for retargeting T cells towards acute myeloid leukemia blasts. Blood Cancer J 2015;5(9):e348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Zhang X, Yang Y, Zhang L et al. Mesenchymal stromal cells as vehicles of tetravalent bispecific Tandab (CD3/CD19) for the treatment of B cell lymphoma combined with IDO pathway inhibitor d-1-methyl-tryptophan. J Hematol Oncol. 2017;10(1):56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Aghi M, Martuza RL. Oncolytic viral therapies-The clinical experience. Oncogene 2005; 24(52):7802–7816. [DOI] [PubMed] [Google Scholar]
- 89.Parato KA, Senger D, Forsyth PA, Bell JC. Recent progress in the battle between oncolytic viruses and tumors. Nat Rev Cancer 2005;5(12):965–976. [DOI] [PubMed] [Google Scholar]
- 90.Nakashima H, Kaur B, Chiocca EA. Directing systemic oncolytic viral delivery to tumors via carrier cells. Cytokine Growth Factor Rev 2010;21(2–3):119–126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Ahmed AU, Tyler MA, Thaci B et al. A comparative study of neural and mesenchymal stem cell-based carriers for oncolytic adenovirus in a model of malignant glioma. Mol Pharm 2011;8(5):1559–1572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Sonabend AM, Ulasov IV, Tyler MA et al. Mesenchymal stem cells effectively deliver an oncolytic adenovirus to intracranial glioma. Stem Cells 2008;26(3):831–841. [DOI] [PubMed] [Google Scholar]
- 93.Yong RL, Shinojima N, Fueyo J et al. Human bone marrow-derived mesenchymal stem cells for intravascular delivery of oncolytic adenovirus Delta24-RGD to human gliomas. Cancer Res 2009;69(23):8932–8940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Moreno R, Reojas LA, Villellas FV et al. Human menstrual blood-derived mesenchymal stem cells as potential cell carriers for oncolytic adenovirus. Stem Cells Int 2017;3615729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Ricon E, Cejalva T, Kanojia D et al. Mesenchymal stem cell carriers enhance antitumor efficacy of oncolytic adenoviruses in an immunocompetent mouse model. Oncotarget 2017;8(28):45415–45431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Zhang Q, Yuan XF, Lu Y et al. Surface expression of anti-CD3scfv stimulates locoregional immunotherapy against hepatocellular carcinoma depending on the E1A-engineered human umbilical cord mesenchymal stem cells. Int J Cancer 2017b;141(7):1445–1457. [DOI] [PubMed] [Google Scholar]
- 97.Tamura K, Wakimoto H, Agarwal AS et al. Multimechanistic tumor targeted oncolytic virus overcomes resistance in brain tumors. Mol Ther 2013;21(1)68–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Kaczorowski A, Hammer K, Liu L et al. Delivery of improved oncolytic adenoviruses by mesenchymal stromal cells for elimination of tumorigenic pancreatic cancer cells. Oncotarget 2016;7(8):9046–9059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Nawaz M, Fatima F, Vallabhaneni KC et al. Extracellular vesicles: evolving factors in stem cell biology. Stem Cells Int 2016;2016:1073140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Wu J, Qu Z, Fei ZW et al. Role of stem cell-derived exosomes in cancer. Oncol Lett 2017;13(5):2855–2866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Gonda A, Kabagwira J, Senthil GN et al. Internalization of exosomes through receptor-mediated endocytosis. Mol Cancer Res 2019;17(2)337–347. [DOI] [PubMed] [Google Scholar]
- 102.Bruno S, Collino F, Iavello A, Camussi G. Effects of mesenchymal stromal cell-derived extracellular vesicles on tumor growth. Front Immunol 2014;5:382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Favaro E, Carpanetto A, Lamorte S et al. Human mesenchymal stem cell-derived microvesicles modulate T cell response to islet antigen glutamic acid decarboxylase in patients with type 1 diabetes. Diabetologia 2014;57(8):1664–1673. [DOI] [PubMed] [Google Scholar]
- 104.Lee HK, Finniss S, Cazacu S et al. Mesenchymal stem cells deliver synthetic microRNA mimics to glioma cells and glioma stem cells and inhibit their cell migration and self-renewal. Oncotarget 2013;4(2):346–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Munoz JL, Bliss SA, Greco SJ et al. Delivery of functional anti-miR-9 by mesenchymal stem cell-derived exosomes to glioblastoma multiforme cells conferred chemosensitivity. Mol Ther Nucleic Acids 2013;2:e126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Li H, Yang C, Shi Y, Zhao L. Exosomes derived from siRNA against GRP78 modified bone-marrow-derived mesenchymal stem cells suppress Sorafenib resistance in hepatocellular carcinoma. J Nanobiotechnology 2018;16(1):103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Yaddanapudi K, Meng S, Whitt AG et al. Exosomes from GM-CSF expressing embryonic stem cells are an effective prophylactic vaccine for cancer prevention. Oncoimmunology 2019;8(3):1561119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Rosenberger L, Ezquer M, Lillo-Vera F et al. Stem cell exosomes inhibit angiogenesis and tumor growth of oral squamous cell carcinoma. Sci Rep 2019;9(1):663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Alcayaga-Miranda F, Gonzàlez PL, Lopez-Verrilli A et al. Prostate tumor-induced angiogenesis is blocked by exosomes derived from menstrual stem cells through the inhibition of reactive oxygen species. Oncotarget 2016;7(28):44462–44477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Malik YS, Sheikh MA, Xing Z et al. Polylysine-modified polyethylenimine polymer can generate genetically engineered mesenchymal stem cells for combinational suicidal gene therapy in glioblastoma. Acta Biomater 2018;80:144–153. [DOI] [PubMed] [Google Scholar]
- 111.Deuse T, Hu X, Gravina A et al. Hypoimmunogenic derivatives of induced pluripotent stem cells evade immune rejection in fully immunocompetent allogeneic recipients. Nat Biotechnol 2019;37(3):252–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Galleu A, Riffo-Vasquez Y, Trento C et al. Apoptosis in mesenchymal stromal cells induces in vivo recipient-mediated immunomodulation. Sci Transl Med 2017;9(416). [DOI] [PubMed] [Google Scholar]
- 113.Garraway LA, Lander ES. Lessons from the cancer genome. Cell 2013;153(1):17–37. [DOI] [PubMed] [Google Scholar]
- 114.Garcia-Casto J, Alemany R, Cascallo M et al. Treatment of metastatic neuroblastoma with systemic oncolytic virotherapy delivered by autologous mesenchymal stem cells: an exploratory study. Cancer Gene Ther 2010;17(7):476–483. [DOI] [PubMed] [Google Scholar]
- 115.Garcia M, Moreno R, Gil M et al. A phase I trial of oncolytic adenovirus ICOVIR-5 administered intravenously to melanoma patients. Hum Gene Ther Clin Dev 2018;10.1089/humc.2018.107. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
- 116.Du W, Seah I, Bougazzoul O et al. Stem cell-released oncolytic herpes simplex virus has therapeutic efficacy in brain metastatic melanomas. PNAS USA 2017;114(30):E6157–E6165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Redjal N, Zhu Y, Shah K. Combination of systemic chemotherapy with local stem cell delivered S-TRAIL in resected brain tumors. Stem Cells 2015;33(1):101–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Martinez-Quintanilla J, Bhere D, Heidari P et al. Therapeutic efficacy and fate of bimodal engineered stem cells in malignant brain tumors. Stem Cells 2013;31(8):1706–1714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Rossignoli F, Grisendi G, Spano C et al. Inducible Caspase9-mediated suicide gene for MSC-based cancer gene therapy. Cancer Gene Ther 2019;26(1–2):11–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Ashkenazi A Directing cancer cells to self-destruct with pro-apoptotic receptor agonists. Nat Rev Drug Discov 2008;7(12):1011–1012. [DOI] [PubMed] [Google Scholar]
- 121.Rowinsky EK. Curtailing the high rate of late-stage attrition of investigational therapeutics against unprecedented targets in patients with lung and other malignancies. Clin Cancer Res 2004;10(12 Pt 2):4220s–4226s. [DOI] [PubMed] [Google Scholar]
- 122.Yan M, Schwaederle M, Arguello D et al. HER2 expression status in diverse cancers: review of results from 37,992 patients. Cancer Metastasis Rev 2015;34:157–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Wang PY, Swain HM, Kunkler AL et al. Neuroblastomas vary widely in their sensitivities to herpes simplex virotherapy unrelated to virus receptors and susceptibility. Gene Ther 2016;23(2):135–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Spear PG. Herpes simplex virus: receptors and ligands for cell entry. Cell Microbiol. 2004;6(5):401–410. [DOI] [PubMed] [Google Scholar]
- 125.Parker BS, Rautela J, Hertzog PJ Antitumour actions of interferons: implications for cancer therapy. Nat Rev Cancer 2016;16(3):131–144. [DOI] [PubMed] [Google Scholar]
- 126.Jensen RT, Bettey JF, Spindel ER, Benya RV. International Union of Pharmacology. LXVIII. Mammalian Bombesin Receptors: Nomenclature, Distribution, Pharmacology, Signaling, and Functions in Normal and Disease States. Pharmacol Rev 2008;60(1):1–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Fathi Z, Corjay MH, Shapira H et al. BRS-3: novel bombesin receptor subtype selectively expressed in testis and lung carcinoma cells. J Biol Chem 1993;268(8):5979–5984. [PubMed] [Google Scholar]
- 128.Akhtar MJ, Ahamed M, Alhadlaq HA et al. Targeted anticancer therapy: Overexpressed receptors and nanotechnology. Clin Chim Acta 2014;436:78–92. [DOI] [PubMed] [Google Scholar]
- 129.Bittner M, Meltzer P, Chen Y et al. Molecular classification of cutaneous malignant melanoma by gene expression profiling. Nature 2000;406 (6795):536–540. [DOI] [PubMed] [Google Scholar]
- 130.Ross DT, Scherf U, Eisen MB et al. Systematic variation in gene expression patterns in human cancer cell lines. Nat Genet 2000;24(3):227–235. [DOI] [PubMed] [Google Scholar]
- 131.Chen X, Plasencia C, Hou Y, Neamanti N. Synthesis and biological evaluation of dimeric RGD peptide-paclitaxel conjugate as a model for integrin-targeted drug delivery. J Med Chem 2005;48(18):1098–1106. [DOI] [PubMed] [Google Scholar]
- 132.Cai W, Niu G, Chen X. Imaging of integrins as biomarkers for tumour angiogenesis. Curr Pharm Des 2008;14(28):2943–2973. [DOI] [PubMed] [Google Scholar]
- 133.Mizejewski GJ. Role of integrins in cancer: survey of expression patterns. Proc Soc Exp Biol Med 1999;222(2):124–138. [DOI] [PubMed] [Google Scholar]
- 134.Parker N, Turk MJ, Westrick E et al. Folate receptor expression in carcinomas and normal tissues determined by a quantitative radioligand binding assay. Anal Biochem. 2005;338(2):284–293. [DOI] [PubMed] [Google Scholar]
- 135.Recht L, Torres CO, Smith TW et al. Transferrin receptor in normal and neoplastic brain tissue: implications for brain-tumour immunotherapy. J Neurosurg 1990;72(6):941–945. [DOI] [PubMed] [Google Scholar]
- 136.Rychtarcikova Z, Lettlova S, Tomkova V et al. Tumor initiating cells of breast and prostate origin show alterations in the expression of genes related to iron metabolism. Oncotarget 2017;8(4):6376–6398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Basuli D, Tesfay L, Deng Z et al. Iron addiction: a novel therapeutic target in ovarian cancer. Oncogene 2017;36(29):4089–4099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Wesche J, Haglung K, Haugsten EM. Fibroblast growth factors and their receptors in cancer. Biochem J 2011;437(2):1999–213. [DOI] [PubMed] [Google Scholar]