1 INTRODUCTION
Based on World Health Organization (WHO), cancer is the second leading cause of death globally which was responsible for 8.8 million mortality in 2015. Nearly 1 in 6 deaths is due to cancer and the number of new cases is expected to rise by about 70% over the next 2 decades worldwide (Siegel, Miller, & Jemal, 2015; Torre et al., 2015). There are many types of cancer treatment including surgery, chemotherapy and radiotherapy (Amer, 2014). Cancerous patients had no survival benefits from current insufficient treatments and in most cases relapse and metastasis occurred (Nowakowski, Drela, Rozycka, Janowski, & Lukomska, 2016; Zhang et al., 2014). Such poor prognosis seems to be linked to detrimental effects on vital noncancerous bodily tissues, deficient drug concentrations in tumors also problems of accessing tumor sites principally in metastatic cancers, and systemic toxicity demonstrates the urgency to explore more effective anti-tumor therapy (Kim & Tannock, 2005; Liu et al., 2015; Pessino & Sobrero, 2006). Targeted therapy is emerging as a supplement or alternative to chemotherapy and/or radiation for various malignant diseases (Mirzaei, Naseri, Rezaee, et al., 2016; Mirzaei, Sahebkar, Salehi, et al., 2016; Mirzaei, Yazdi, Salehi, & Mirzaei, 2016). In the field of targeted therapy, gene therapy appears as a good substitute method for cancer treatment (Wu, Chang, & Huang, 2006). There are different approaches for cancer gene therapy including immunotherapy, oncolytic viruses and gene transfer (McCormick, 2001; Lin & Nemunaitis, 2004; Cross & Burmester, 2006; Mirzaei, Mirzaei, Lee, Hadjati, & Till, 2016; Mirzaei, Naseri, Rezaee, et al., 2016). Immunotherapy employ for immune system stimulation to destroy cancer cells (Blattman & Greenberg, 2004). Oncolytic viruses, that replicates within the cancer cell and cause cell destruction (Chiocca & Rabkin, 2014; Dwyer, Khan, Barry, O'Brien, & Kerin, 2010; Singh, Doley, Kumar, Sahoo, & Tiwari, 2012). Gene transfer serves as a new treatment approach that introduces foreign genes into cancerous cells to promote cell death or slow the progression of the cancer. Gene transfer represents the best way for cancer gene therapy. In this path, we could introduce multiple genes with completely different function to malignant cell such as pro-apoptotic genes, anti-angiogenesis genes, and suicide genes (Cao et al., 1998; Cross & Burmester, 2006; Persano, Crescenzi, & Indraccolo, 2007). Failure to distinguish between normal and tumor cells will probably remain a limiting factor for chemotherapy drugs but Suicide genes form the basis of a strategy for making cancer cells more vulnerable and susceptible to chemotherapy (Karjoo, Chen, & Hatefi, 2016b). Suicide gene therapy system is based on gene transfer into tumor cells, which leads to the exclusive expression of an enzyme able to convert a non-toxic prodrug into a lethal drug (Freeman, 2002; Izmirli, Sonmez, & Gogebakan, 2016; Touati et al., 2014). Achieving the specified purpose requires vehicles that encapsulate the gene and deliver it particularly to cancer cell and cancer local environment of the tumor. In gene therapy and especially cancer gene therapy, at first we need to utilize vehicle so called vector that includes a set of criteria; having tumor tropism (specificity for tumor microenvironments), do not able to elicit an immune response and safety (Hacein-Bey-Abina et al., 2002; Rajab, Nelson, Keung, & Conrad, 2013). From the first reports of gene therapy in 1960–1970 to present, there are three types of gene delivery systems into the target cell (Collins & Thrasher, 2015). Through to their high transduction efficiency, viral vectors are the most frequently used gene delivery strategies. Retrovirus, lentivirus, and adenovirus are common viral vectors. In the field of gene therapy with viral vectors there are two major obstacles in terms of the safety and toxicity of these vectors that limit the clinical potential of this approach (i.e., insertional mutagenesis of retroviral vectors and intensive immune reaction of adenoviral vectors) (Collins et al., 2008; Nayerossadat, Maedeh, & Ali, 2012; Rajab et al., 2013; Waehler, Russell, & Curiel, 2007). Molecular vectors including, naked DNA and application of cationic polymers such as polyethylenimine or poly-l-lysine, cationic peptides, and cationic liposomes. The major drawbacks of this approach are non-specific uptake of the DNA by cancer cells and toxicity of some cationic polymers such as lipoplexes (Dani, 1999; Miller & Vile, 1995; Nayerossadat et al., 2012; Simões et al., 2005; Zhang et al., 2014). Cellular vectors are promising vehicles to deliver various anticancer agents, including small molecule drugs, proteins, suicide genes, nanoparticles, and viruses. Cellular vectors have several advantages such as low immunogenicity, practically unlimited genetic material packaging capacity, simple and low-cost construction which makes them appropriate for large-scale production and most likely safe for clinical gene therapy (Link, Seregina, Traynor, & Burt, 2000; Ohlfest, Freese, & Largaespada, 2005).
Cell therapy is an effective therapeutic approach for treatment of various diseases such as cancer (Goradel et al., 2017; Mirzaei, Mirzaei, Lee, et al., 2016). Among of various cells which could be used for cell therapy, MSC are known as powerful tools for treatment of various diseases such as cancer. MSC can be utilized for targeted-gene delivery (Goradel et al., 2017; Mirzaei, Sahebkar, Jaafari, Goodarzi, & Mirzaei, 2016). MSCs have a low immunogenic potential besides the capacity to home and integrate in to the injured and inflamed sites. MSCs have indicated preferential tropism for tumor, so could transferred suicide genes and their products just in tumor local and reduce side effects about systemic toxicity and could achieve more efficient gene delivery to the target (Dvorak, 1986; Nauta & Fibbe, 2007; Uchibori, Tsukahara, Ohmine, & Ozawa, 2014). The aim of the present review is to discuss about (1) Suicide genes, known as good killers in cancer therapy; (2) Characteristics and application of MSCs in cancer therapy; (3) investigating previous research studies about MSCs in role of a vehicle for suicide genes.
2 SUICIDE GENES AS A GOOD KILLER
The term of gene directed enzyme prodrug therapy (GDEPT) is one of the promising alternatives to conventional chemotherapy because it minimizes the systemic toxicities of conventional chemotherapy drugs and refer to expression of a suicide gene in tumor cells for the in situ conversion of a pro-drug into cytotoxic metabolites (Greco & Dachs, 2001; Springer & Niculescu-Duvaz, 2000). The first attempt to use of suicide genes against cancer was in 1986 by Moolten et al. (Rajab et al., 2013). These points must consider in selection of suicide genes; Intended gene must encodes an enzyme that did not exist in normal cells or there is no enzyme with similar function in cells; Minimal toxicity of prodrug before activation in body and maximum toxicity after converting to active drug in tumor location; high kinetic enzyme activity or high affinity to target prodrug and finally active drug must diffuse in whole tumor mass by Bystander effects (Both, 2009; Rajab et al., 2013; Zarogoulidis et al., 2013). This mentions to the demolition of tumor cells that are not directly expressing the suicide gene. In this effect, gap junctions play substantial role via local diffusion of the active drug. Previous studies revealed that this approach could abolish tumors even if 10% of tumor cells expressed suicide genes, this efficiency is about bystander effects (Freeman et al., 1993; Mesnil & Yamasaki, 2000). There are several mechanisms for Bystander affect; intercellular communication via gap junctions, diffuse through the cell membrane, endocytosis of apoptotic vesicles and stimulation of the immune system against tumor. The gap junctional intercellular communication (GJIC) capacity varied among cancer cell lines and was dependent on the connexin 43 (cx43) expressions (Duarte, Carle, Faneca, De Lima, & Pierrefite-Carle, 2012; Huber, Austin, Richards, Davis, & Good, 1994; Pierrefite-Carle et al., 1999). The overexpression of Cx43 in glioma cells leading to the increase in the number of gap junctions and enhanced the bystander effect of HSV-TK as a suicide gene, reverse situation is in esophageal cancer (Huang et al., 2010; Matono et al., 2003). So evaluation of degree of GJIC has predictive value for determination of response to suicide genes that dependent on GJIC such as HSV-TK. Also studies reported that radiation could induce bystander effect, so suicide cancer therapy juxtaposed with the irradiation could increase tumor eradiation (Azzam & Little, 2004). In the following, we discussed two most applicable suicide genes in cancer gene therapy.
2.1 Cytosine deaminase (CD)/5-fluorocytosine (5-FC)
Although 5-flourouracil (5-FU) has been widely used in the past decades for the treatment of multiple cancers especially colorectal and breast cancers, but this drug failed to treat efficiently due to some limits: insufficient delivery of drug to cancer mass and adverse effect on body. High systemic toxicity made it unpleasant choice for patients (Cheung, Fralick, & Cheng, 2008; Longley, Harkin, & Johnston, 2003). Serious side effects could be reduced by gene-directed enzyme prodrug therapy depending on the bacterial and/or yeast cytosine deaminase (CD) enzyme activity with no equivalent in mammalian cells. CD gene codes cytosine deaminase, an enzyme express in a variety of bacteria, fungi and yeast that convert nontoxic prodrug 5-FC to its toxic metabolite 5-fluorouracil (5-FU) (Kucerova, Altanerova, Matuskova, Tyciakova, & Altaner, 2007; Kuriyama et al., 1999). 5-FU metabolites direct the formation of fraudulent 5FU-RNA and 5FU-DNA and inhibit RNA and DNA synthesis by thymidylate synthase blockage and finally apoptosis. Although there is no DNA synthesis in non-dividing cancer cells but high concentration of 5-FU could decrease mRNA level and subsequently protein starvation and cell death. 5-FU could exert toxic effects on neighboring cells through freely diffusion across the cellular membrane without GJIC mediation, owing to its small size and suitable charge (Gopinath & Ghosh, 2008; Karjoo, Chen, & Hatefi, 2016a). 5-FC, but not 5-FU crosses the blood brain barrier (Karjoo et al., 2016a; Ostertag et al., 2012). This capability seems to be another advantage of this gene, since it makes 5-FC appropriate for treatment of tumors in brain with difficult accessibility. Several reports revealed that CD/5FU system accompanied with radiotherapy will be more effective in tumor suppression (Kaliberov & Buchsbaum, 2012). Comparative studies showed that yeast CD produces 15-fold higher amount of 5-FU than bacterial CD (Kievit et al., 2000). Also results demonstrated that bifunctional yeast fusion gene CD::UPRT (cytosine deaminase::uracil phosphoribosyltransferase) could produce 100–10,000-higher amount of 5-FU compared with CD enzyme alone, in vitro and in vivo (Kanai et al., 1998; Tiraby et al., 1998). The ABC transporters are trans-membrane proteins that can facilitate the export of substrates through cellular membranes. ABC transporters participate in chemo-resistance of cultured cells. Actually multidrug resistance protein 5 (MRP5) and MRP8 confer resistance to 5-FU by increase the permeability of cellular membrane to monophosphate metabolites (Nambaru et al., 2011; Oguri et al., 2007). Matuskova et al. (2012) showed that silencing of the ABCC11 by RNA interference significantly sensitized breast carcinoma cell line (MDA-MB-231) to the CD::UPRT system.
2.2 Herpes simplex thymidine kinase (HSV-TK)/ganciclovir (GCV)
HSV-TK gene codes for a thymidine kinase that able to convert GCV to GCV monophosphate which is then turned into GCV-di and triphosphate by endogenous kinases. Insertion of GCV triphosphate to double stranded DNA leads to termination of DNA synthesis and finally continued with apoptosis as a result of maintenance of cell in S phase (Abate-Daga, Garcia-Rodríguez, Sumoy, & Fillat, 2010; Tomicic, Thust, & Kaina, 2002). Since HSV-TK has greater affinity for GCV in compare to endogenous kinases enzymes, the primary steps of GCV phosphorylation occurs mainly by viral TK. But GCV required very high dose rate to fully occupy the active site (Gallois-Montbrun, Veron, & Deville-Bonne, 2004; Karjoo et al., 2016a). This creates nonspecific toxicity such as immune suppression, acute depression of the bone-marrow, and finally insufficient tumor killing ability. Manipulation of enzyme gene to create a mutant HSV-TK that provide the same effect of wild type enzyme at lower levels of prodrug, can greatly reduce this obstacle (Balzarini et al., 2006; Black, Kokoris, & Sabo, 2001). HSV-TK/GCV system similar to CD/5-FC system has been shown bystander effect. The difference is that GCV trafficking is depending on the presence of gap junctions, which enable the exchange of toxic products between transduced and untransduced cells (Li Bi, Parysek, Warnick, & Stambrook, 1993; Mesnil, Piccoli, Tiraby, Willecke, & Yamasaki, 1996). As it seems CD/5-FC system has better performance because it is freely diffusible across the cellular membrane and independent of GJIC and connexins expression. Rainov et al. (2001) reported that Temozolomide enhances the efficiency of TK/GCV against malignant glioma. On the other hand, some ABC transporters such as ABCC4 and ABCG2 enhanced resistancy of transgene-expressing and bystander cells against TK/GCV system (Adachi et al., 2002). Also, based on previous studies, the origins of mutations are distinct in different tumor types so they show differences in sensitivity to various enzyme/prodrug systems (Huang et al., 2009; Jiang et al., 2014). This situation is seen more in single suicide gene therapy. Some results suggested that double suicide gene therapy with this genes had severer deadly effects than either one used alone (Aghi, Cho, Chiocca, Kramm, & Breakefield, 1998; Freytag, Rogulski, Paielli, Gilbert, & Kim, 1998; Uckert et al., 1998). On the other hand some reports shown that anticancer effect and survival rate were not significantly different between one and double suicide gene delivery, even single suicide gene systems may be preferable than combinations of the two systems (Chang et al., 2000; Moriuchi et al., 2002).
2.3 Other important suicide genes
The other gene/prodrugs that commonly used as suicide genes including CYP2B6/cyclophosphamide (CPA), nitroreductase/CB1954 (NTR/CB1954), carboxylesterase (CE)/CPT-11 (irinotecan), and Inducible caspase-9 (iC9)/chemical inducer of dimerization (CID). Primary studies demonstrate that efficiency of CYP2B6 for cancer therapy is low; the poor results may be explained by low affinity of CYP2B6 to CPA. This can be solved by insertion mutations in CYP2B6 (CYP2B6TM; CYP2B6 triple mutant) and fusion with NADPH cytochrome P450 reductase (RED) (CYP2B6TM-RED) (Argos, 1990; Braybrooke et al., 2002; Nguyen et al., 2008; Touati et al., 2014). NfsB nitroreductase (NTR) isolated from Escherichia coli can convert CB 1954 (5-(aziridin-1-yl)-2,4-dinitrobenzamide) to a potent cytotoxic DNA chelating agent which can freely diffuse across the cell membrane and trigger extensive DNA damage and apoptosis in cancer cells. However, low activation rate of this non-natural substrate, CB1954, appears to restrict the impact of this enzyme. In addition CB1954 induced dose-dependent hepatotoxicity (Chung-Faye et al., 2001; Greco & Dachs, 2001; Green et al., 2013). Gene-directed enzyme prodrug therapy (GEPT) is another approach for chemotherapeutics selectivity improvement. One of such gene/prodrug to drug systems is carboxylesterase (CE)/campothecin (CPT-11) to SN-38 (7-ethyl-10-hydroxycamptothecin). CPT-11 could inhibit mammalian DNA topoisomerase I, while SN-38 Inhibits this enzyme activity approximately 1,000 times more effectively than CPT-11 (Pommier, 2006; Satoh et al., 1994; Wierdl et al., 2001). iC9 is another suicide context refer to an inducible caspase-9 actually a pro-apoptotic protein caspase9 that bind to a modified human FK-binding protein. AP1903, as chemical inducer of dimerization (CID) can activate iC9 and dimerized caspase-9 resulting in apoptosis (Ando et al., 2014; Boatright et al., 2003).
3 TALENT OF MSCS IN CANCER GENE THERAPY
Adult stem (AS) cells are a potentially valuable source in regenerative medicine. They are dividing to neural stem cells (NSCs) (ectoderm), hematopoietic stem cells (HSCs) (mesoderm), and MSCs (mesoderm) (Robinton & Daley, 2012; Weissman, 2000). In vitro expansion of NSCs is limited to the nervous system; they could not survive outside the nervous system. HSCs have been utilized in allogeneic cell therapy. Although bone marrow (BM) has been the principal source for the isolation of these cells, the harvesting of bone marrow entailed a highly invasive procedure with low cell yields (Prockop, 1997; Sylvester & Longaker, 2004). It is easier to work with MSCs, due to their less invasive and free of ethical issues harvest in compare with other sources (Amara, Touati, Beaune, & de Waziers, 2014; Cavarretta et al., 2010; Compte et al., 2009). MSCs were first obtained from bone marrow and defined as fibroblast-like multipotent stem cells by Friedenstein and coworkers in 1974 (Friedenstein, Chailakhjan, & Lalykina, 1970).
International Society for Cell Therapy (ISCT) proposed minimal criteria to describe human MSCs; adhere to plastic, ability to differentiate into adipocytes, osteoblasts, and chondrocytes, fibroblastoid phenotype and expression of cell surface markers CD105, CD73, CD271, CD90, and ganglioside GD2 in more than 95% of cells and absence of CD45, CD79a, or CD19, CD34, CD14, or CD11b, CD40, CD80, CD86, and the MHC II class cellular receptor HLA-DR (Baird, 2015; Dominici et al., 2006; Goradel et al., 2017; Horwitz et al., 2005; Mirzaei, Sahebkar, Avan, et al., 2016; Mirzaei, Sahebkar, et al., 2017; Mohammadi, Jaafari, Mirzaei, & Mirzaei, 2016). But it should be noted that there is no unique key determinant factor to distinguish MSCs, also the relative expression levels for these markers could be varied according to the species diversity, tissue sample and culture conditions. At first MSCs isolated from bone marrow (BM) but subsequently found in other source such as adipose tissue (AT), umbilical cord blood (UCB), peripheral blood, surrounding blood vessels, synovium, the circulatory system, dental pulp, and amniotic fluid (Morizono et al., 2003; Phinney & Prockop, 2007; Uchibori et al., 2014). In spite of the fact that MSCs only represent a small percentage of the total number of BM populating cells 0.01–0.001%), they extensively used by many researchers (Li, Fan, & Xiong, 2015). Since MSC isolation from BM recognized as an unpleasant experience, adipose tissue another valuable source of MSCs, has drown great attention in recent years (Kucerova et al., 2008).
Adipose tissue mostly isolated from subcutaneous tissue, we can get ∼40-fold higher yield of MSC from adipose tissue compared with the bone marrow. The rate of isolation of MSC from human AT is 100%. Great interest has developed in AT-MSC, which is free of ethical concerns and invasiveness (Kern, Eichler, Stoeve, Klüter, & Bieback, 2006). Moreover, it was reported that AT-MSC can be expanded long term without the loss of their phenotype. In contrary, BM-MSCs should be used during early cell passages (below P8) to prevent potential differentiation (Lee et al., 2006). UCB-derived MSC has significantly higher rate of expansion in compared with BM-MSC. Since these cells are obtained from placenta that discarded after birth, so the collection procedure is totally noninvasive and free from ethical considerations (Li et al., 2015). MSCs also have been exploited as tumor specific delivery vehicles for cancer gene therapy by multiple reasons; immunoprivileged status (due to lack of major histocompatibility complex MHC-II and only minimal MHC-I expression) and natural tropism for tumors and their metastases. MSCs do not motivate the host immune response and escape immunological rejection in allogeneic injection (Dvorak, 1986; Kidd et al., 2008; Zhang et al., 2014).
In addition MSCs can be efficiently transduced with viral or non-viral vectors containing a target gene. For example, in separate studies, Bak, Yang, and Wang (2010) and Jiang et al. (2014) demonstrated that hUCB-MSCs and hBM-MSCs can be simply infected by the lentivector and baculovirus respectively at a high efficiency of over 80%. MSCs mostly transduced through viral vectors including lentiviruses, retroviruses, baculovirus, and adenoviruses. All of these viral vehicles produce stable expression, while the later create transient expression of therapeutic genes. Non-viral vectors have been exploited in limited number of studies. The examples are nucleofection, spermine-pullulan (SP), PEIcyclodextrin, and cationic liposomes and cationic polymers (Gao et al., 2010; Uchibori et al., 2009; Zhang et al., 2014). Results revealed that in healthy animal models MSCs mostly migrate to lung, liver and bone but also MSCs have high migration potential to the tissue injury and inflammation (Hu, Fu, Tabata, & Gao, 2010; Kidd et al., 2008). For example, inflammation has long been associated with the development of cancer, MSCs exhibit strong tropism toward tumors and their metastases by expression of receptors such as CCR1, CCR4, CCR7, CCR9, CCR10, CXCR4, CXCR5, CXCR6, RAGE, CX3CR1, VEGFR, c-Kit, and c-Met for Infammatory chemokines and growth factors including SDF-1(CXCL12), MCP-1, HGF, IL-8, NT3, TGF-b, GCSF, VEGF, IL-8, IL-6, CCL2, HMGB1, and SCF (Honczarenko et al., 2006; Reagan & Kaplan, 2011; Ringe et al., 2007). CXCR4 is the most important chemokine receptor implicated in targeted homing of MSCs. MSCs expression profiling demonstrates that overexpression of CXCR4 and down regulation of MMP-2 have greater role in migration capability in to tumor sites, whereas knockdown of SDF-1 plays a negative role in stem cell recruitment to tumors (Menon et al., 2007; Song & Li, 2011). Sato et al demonstrated that epidermal growth factor receptor gene transfection conferred enhanced migratory activity toward GL261 gliomas or B16 melanoma in vivo (Sato et al., 2005). Moreover adhesion molecules, such as b1- and b2-integrins and L-selectin, are involved in MSC migration and homing to tumor site (Hu et al., 2010). A recent report on MSC behavior have highlighted the great impact of irradiation on MSC tropism and engraftment to tumor location, mediated at least in part by apoptosis and subsequent enhanced local release of inflammatory signals such as CCL2 and CCR8. For example, UCB-MSCs-based TRAIL gene delivery to irradiated glioma tumors enhanced apoptosis in glioma cells. Besides radiotherapy could increase MSC localization in LoVo, HT-29 (colon), and MDA-231 breast cancer cells (Kim et al., 2010; Klopp et al., 2007; Zielske, Livant, & Lawrence, 2009). Every safe cell-based therapy will require the capability to harness the unwanted growth of cells. Local injection of MSC developed some side effects, that is, ectopic ossification and calcification foci in mouse and rat models of myocardial infarction. Moreover bilateral diffuse pulmonary arises after bone marrow transplant in a dog. In a distinct study, spontaneous osteosarcoma formation in culture has been reported in murine-derived MSCs (Breitbach et al., 2007; Sale & Storb, 1983; Yoon, Park, Tkebuchava, Luedeman, & Losordo, 2004). Albeit MSCs have been utilized for treatment of many patients without major undesirable effects but some results, both in vitro and in vivo models suggest that must be cautions in this regard. MSCs exhibit pro-tumorigenic or tumor-supporting roles through inhibition of immune system and apoptosis, stimulation of EMT (epithelial–mesenchymal transition) and enhancement of angiogenesis, proliferation, migration, and metastasis (Djouad et al., 2003; Martin et al., 2010; Ramasamy et al., 2007). Unmodified MSC have antitumor properties in leukemias, glioma, and melanoma, and breast, hepatoma, and lung cancer cells (Ramasamy et al., 2007; Sun et al., 2009). In this regard, Ma et al. (2012) observed the growth inhibition of breast CSCs by hUCMSC both in vitro and in vivo. Ohlsson, Varas, Kjellman, Edvardsen, and Lindvall (2003) showed that mesenchymal progenitor (immortalized MSCs) are effective at inhibiting the in vivo proliferation of rat colon carcinoma cells. The underlying mechanism about antitumor effect of MSCs is likely related to down-regulation of Wnt, Akt, PI3K, and NF-kB signaling pathways by factors released from MSC in tumor position (Dai et al., 2011; Ma, Hao, Zhang, & Zhang, 2012). Some studies in order to increase the innate antitumor effects of MSCs carried out gene modification with oncogenes or other genes promoting cell proliferation but these modifications are associated with higher risk of tumorigenicity (Miletic et al., 2007). Engineered MSCs was first use for direct delivery of interferon ß (IFN-ß) gene to melanoma xenografts mice (Studeny et al., 2002). Considerable reduced in tumor growth rate and significant prolongation of survival of tumor-bearing mice motivated more researches to study therapeutic efficacy of genetically modified MSCs as cellular vehicles for several cancer treatments. MSCs explored for local secretion of therapeutic cytokine proteins such as; IFN-ß, IL-12, IL-24, and IL-2, delivery of prodrug activator genes (i.e., actually suicide genes) that will be explained in the following, amplifying and deliver of oncolytic viruses such as conditionally replicating oncolytic adenoviruses (CRAd), ICOVIR-5 and measles virus (MV), secretion of pro-apoptotic proteins such as TRAIL, TNF-related apoptosis ligand, anti-angiogenic agents such as TSP1 and endostatin and growth factor antagonists, that is, NK4 (antagonist of hepatocyte growth factor [HGF]) (Dwyer et al., 2010; Li, Ou, Wang, & Tang, 2016; Shah, 2012). MSCs loaded with nanoparticles containing chemotherapy drug, paclitaxel have been shown high efficient tumoricidal effect (Sadhukha, O'Brien, & Prabha, 2014).
It has been showed that MSCs could have dual effects on cancerous condition. Anti-cancer and tumorgensis effects of MSCs led to some concerns about utilization of them as therapeutic agents. It has been showed that express of different types of MSCs surface markers play critical roles in the effect of MSCs in various conditions.
Finally, three main points regarding employing of MSCs as vectors remain to be defined: (1) MSCs are cells with active physiological process which using of them are not simple delivery platform. They could release a wide array of different molecules (i.e., growth factors, cytokines, and chemokines). Hence, they may provide various signals which could lead to enhancing of tumor burden and metastases. (2) Cancerous microenvironments may lead to inducing of malignant transformation in the injected MSCs. (3) How many MSCs need to be given and when and where they should be administrated. Hence, better understanding of MSCs physiology within the tumor sites, and more robust studies characterizing their homing mechanisms could enhance suggested therapies.
4 MSCS-EXOSOMES AS NEW THERAPEUTIC APPROACH FOR CANCER THERAPY
Among of various signaling and cellular effects of MSCs, exosomes released from MSCs have critical roles for transferring specific signals to host cells. Exosomes are known as nano vesicles which could carry a variety of molecules and markers such as DNAs, mRNAs, microRNAs, and proteins (Banikazemi et al., 2017; Borujeni et al., 2017; Mirzaei, Sahebkar, Jaafari, et al., 2016). The cargos are able to change behavior of host cells via targeting various cellular and molecular targets (Mirzaei, Sahebkar, Jaafari, et al., 2016; Saadatpour et al., 2016). For example, microRNAs (miRNAs) are one of important cargos which could be carried by exosomes. MiRNAs are small non-coding RNAs which have critical roles in regulating of a variety of vital biological processes such as growth, angiogenesis, and differentiation (Golabchi et al., 2017; Hashemi Goradel et al., 2017; Keshavarzi, Sorayayi, et al., 2017; Mashreghi et al., 2017; Mirzaei, Masoudifar, et al., 2017; Mirzaei, Yazdi, Salehi, et al., 2016; Rashidi, Malekzadeh, Goodarzi, Masoudifar, & Mirzaei, 2017; Salarinia et al., 2016). It has been showed that deregulation of them could be associated with imitation and progression of a wide range of diseases such as cardiovascular diseases, stroke, diabetes, and cancer (Fathullahzadeh, Mirzaei, Honardoost, Sahebkar, & Salehi, 2016; Gholamin et al., 2017; Hoseini et al., 2017; Keshavarzi, Darijani, et al., 2017; Mirzaei, 2017; Rabieian et al., 2017; Rashidi, Hoseini, Sahebkar, & Mirzaei, 2016; Simonian, Mosallayi, & Mirzaei, 2017). These molecules exert their effects via targeting various cellular and molecular pathways involved in various physiological processes (Gholamin et al., 2016; Mirzaei, Fathullahzadeh, et al., 2017; Mirzaei, Khataminfar, Mohammadparast, et al., 2016; Mirzaei, Momeni, et al., 2017; Mirzaei, Sahebkar, Mohammadi, et al., 2016; Mohammadi, Goodarzi, Jaafari, Mirzaei, & Mirzaei, 2016; Moridikia, Mirzaei, Sahebkar, & Salimian, 2017). Hence, targeting of miRNAs by exosomes could lead to change behavior of host cells and could be associated with emerging of diseases condition or therapeutic effects (Mirzaei, Gholamin, Shahidsales, et al., 2016; Mirzaei, Naseri, Rezaee, et al., 2016). Exosomes could be released from various types of cells such as cancer cells, and MSCs. These nano-particles could exert their inflammatory or anti-inflammatory effects on host cells (Rani, Ryan, Griffin, & Ritter, 2015). It has been showed that MSCs employed exosomes for paracrine functions. Paracrine functions of MSCs are known as one of important mechanisms involved in anti-inflammatory effects of MSCs (Rani et al., 2015). Hence, it seems that utilization of exosomes released from MSCs could be used as effective candidate for treatment of various diseases.
5 MSCS AS A VEHICLE FOR TARGETING SUICIDE GENES
Identification of new drug delivery platform is one of the major landscapes in cancer gene therapy (Mohammadi, Jaafari, et al., 2016). Despite a numerous of novel therapeutic approaches have emerged, none has yet confirmed the ability to cure various cancers. In this regard, MSCs emerge to be promising new therapeutic option. The natural tropism of MSCs for tumor sites makes them excellent vehicles for tumor-targeted therapies (Mohammadi, Jaafari, et al., 2016). Also, it has been showed that use of genetically modified MSCs may overcome limitations related with systemic administration of some cytokines and anti-neoplastic agents with a short half-life and high toxicity (Mohammadi, Jaafari, et al., 2016). Viral vectors for directed enzyme prodrug therapy (GDEPT) or suicide gene therapy have been utilized by multiple studies. This approach has some limitations due to its low specificity for tumor cells and systemic toxicity. Utilization of viral vectors for targeting various genes such as suicide gene are associated with various limitations such as high immunogenicity (Mirzaei, Sahebkar, Jaafari, et al., 2016). Hence, several studies applied improved viral vectors for overcoming to various limitations. Moreover, it has been showed that other vectors such as Piggybac could be used for targeting various genes and be associated with same or better results than viral vectors (Mirzaei, Sahebkar, Jaafari, et al., 2016). Hence, utilization of Piggybac or improved viral vectors could be employed as new delivery system with high potential for targeting various genes such as suicide genes. Genetically engineered MSCs using a GDEPT strategy could be a good resolve for these obstructions. MSC-targeted gene therapy is a two-step procedure. In the first step, the gene for the foreign enzyme (bacterial, yeast, or viral) is targeted to the tumor by transduction of MSCs. In the second step, transcription of the gene encoding the prodrug-drug converting enzyme could generate a lethal substance at the tumor site. With looking to previous studies we found that the retroviral suicide gene construct is often used for MSC transduction and most frequent transduced suicide genes are CD and HSV-TK. A large number of approaches have been applied using AT-MSCs and BM-MSCs as a delivery system for anti-neoplastic agents (Amara et al., 2014). A key characteristic of manipulated MSCs is that they have no difference with naïve MSCs in terms of proliferation, differentiation, and tumor homing potentials as well as surface antigenicity (Park et al., 2013). Until now many in vitro and in vivo studies carried out using MSCs in cancer therapy. A brief report of studies within this research area is embedded in Table 1. Due to this fact that MSCs express very low levels of receptors, like most healthy tissues, are unaffected by TRAIL-induced apoptosis. As a result, recruitment of MSCs secreting TRAIL brings brilliant results in cancer treatment (Grisendi et al., 2010; Kim et al., 2008; Loebinger, Eddaoudi, Davies, & Janes, 2009; Mohr et al., 2008). Moreover co-transfection of the TRAIL gene with some cases of double suicide gene approaches (i.e., HSV-TK) that confront with some difficulties including antagonistic antitumor activity, could improve the efficiency of tumoricidal effect (Kim et al., 2013; Martinez-Quintanilla et al., 2013). In order to achieve efficient homing of MSCs to the tumor site, the median amount of administered therapeutic cells recommended to be less than 10% of the tumor mass (Hung et al., 2005). Therefore, it is logical to use multiple injections of the therapeutic cells at a low dose rather than single injection at high dose. For example, Kim et al. (2013) reported that mouse with metastatic renal cell carcinoma (RCC) after two and three injections of small-divided doses of MSC/TRAIL-TK represents 50% and 100% survival, respectively. The potency of consecutive suicide gene therapy was also evaluated in another study in which repeated intracerebral injection of CDy:UPRT-AT-MSC lead to 88% increase in the survival time of rat with glioblastoma (Altanerova et al., 2012). Some studies employed combined therapeutic modalities for malignant disease treatment. Combination of suicide gene- MSCs with chemotherapy drug shows satisfying results in some cases. Ando et al. (2014) revealed that NSCLC progression prevents following proteasome inhibitor bortezomib administration, together with MSC- Ad.iC9. While, this drug is ineffective for the treatment of NSCLC, alone. In another study MSCs-TK and Valproic acid (VPA) has been used for treatment of glioma-bearing mice (Ryu et al., 2012). The combined treatment had dramatic inhibitory effects on tumor growth and prolonged the survival rate in mice.
Table 1.
Characteristic of previous studies in MSC-targeted suicide gene therapy
Type of cancer (cell line) |
Source of MSCs |
Type of suicide gene |
Vector |
Administration route |
References |
Ovarian carcinoma (SKOV3) |
hUCBMSCs |
HSV-tk/CD Combined treatment |
Lentivirus |
Just in vitro |
Jiang et al. (2014) |
Colon cancer (HT-29) |
hAT-MSC |
CD::UPRT |
Retrovirus |
Systemically administered |
Kucerova et al. (2007) |
bone metastatic PC (Du145, PC3, and LNCaP) |
hAT-MSC |
CD::UPRT |
Retrovirus |
Systemically administered |
Cavarretta et al. (2010) |
Glioma (9L) |
hBM-MSC |
Hsv-tk |
pcDNA3.1/Hygro(−) plasmid |
Local injection |
Mori et al. (2010) |
Glioma (9L) |
RatBM-MSC |
HSV-tk |
Retrovirus |
Local injection |
Miletic et al. (2007) |
Lewis lung carcinoma (LLC cell line) |
miceAT-MSC |
CD::UPRT & CD::UPRT::VP22 separately |
Plasmid |
Systemically administered |
Krassikova, Karshieva, Cheglakov, and Belyavsky (2016) |
Melanoma (A375) |
hAT-MSC |
CD::UPRT |
Retrovirus |
Systemically administered |
Kucerova et al. (2008) |
Gastric cancer (MKN45) |
hBM-MSC |
CD |
pcDNA-CD plasmid |
Systemically administered |
You et al. (2009) |
Non-small cell lung cancer |
hBM-MSC |
iC9 |
Adenovirus |
Systemically administered |
Ando et al. (2014) |
Breast cancer MDA-MB-231 |
hAT-MSC |
CD::UPRT or HSV-tk separately |
Retrovirus |
Systemically administered |
Matuskova et al. (2015) |
Renal cell carcinoma (RENCA) |
RatBM-MSC |
TRAIL-tk |
Adenovirus |
Systemically administered |
Kim et al. (2013) |
Glioma (C6) |
hAT-MSC |
CD::UPRT |
Retrovirus |
Local injection |
Altanerova et al. (2012) |
Mice bearing TC1-Luc2 tumors |
miceBM-MSC |
CYP2B6TM-RED |
Lentivirus |
Local injection |
Amara et al. (2016) |
Pulmonary melanoma (B16F10) |
RatBM-MSC |
CMV-tk |
Spermine-pullulan (SP) |
Systemically administered |
Zhang et al. (2014) |
Ovarian cancer (SKOV3) |
hBM-MSC |
TK007 & TK SR39 & CD:UPRT & NTR Separately |
Mammalian expression plasmid vectors |
Local injection |
Nouri, Wang, and Hatefi (2015) |
Glioma (8-MGBA, 42-MG-BA, and U-118 MG) |
hAT-MSC |
HSV-tk |
Retrovirus |
Systemically administered |
Matuskova et al. (2010) |
Melanoma (A375) & Glioma (8-MG-BA) |
hAT-MSC |
HSV-tk & CD::UPRT Separately |
Retrovirus |
Just in vitro |
Matuskova et al. (2012) |
Prostate cancer (TRAMPC1 & TRAMPC2) |
hAT-MSC & miceBM-MSC |
CD::UPRT |
Retrovirus |
Systemically administered |
Abrate et al. (2014) |
Glioma (9L) |
RatBM-MSC |
HSV-tk |
Retrovirus |
Local injection |
Uchibori et al. (2009) |
Glioma (U87MG) |
hBM-MSC |
CD |
Retrovirus |
Local injection |
Park et al. (2013) |
Hepatocellular carcinoma (Huh7) |
miceBM-MSC |
HSV-tk |
Plasmid |
Systemically administered |
Niess et al. (2011) |
Lung adenocarcinoma (A549) |
miceBM-MSC |
HSV-tk |
Retrovirus |
Just in vitro |
Yang et al. (2014) |
Prostate cancer (DU145 & PC3) |
SV40-hfBMSCsa |
HSV-tk |
Lentivirus |
Systemically administered |
Lee et al. (2013) |
Lung metastatic (By B16F10) |
RatBM-MSC |
HSV-tk |
Spermine-pullulan (SP) |
Systemically administered |
Zhang et al. (2015) |
Osteosarcoma (Cal72) |
hBM-MSC |
CD::UPRT |
Plasmid |
Systemically administered |
NguyenThai et al. (2015) |
Pancreatic carcinoma (Panc02) |
miceBM-MSC |
HSV-tk |
Plasmid |
Systemically administered |
Zischek et al. (2009) |
Glioma (TJ899, TJ905, U251, U87, & C6) |
RatBM-MSC |
HSV-tk |
Adenovirus |
Local injection |
Huang et al. (2010) |
Glioma (C6) |
RatBM-MSC |
CD |
Lentivirus |
Local injection |
Fei et al. (2012) |
Glioma (U-87) |
hAT-MSC |
HSV-tk |
Lentivirus |
Local injection |
de Melo et al. (2015) |
Glioma (C6/LacZ7) |
hBM-MSC |
CD |
Retrovirus |
Systemically administered |
Chang et al. (2010) |
Glioma (C6) |
RatBM-MSC |
HSV-tk |
Retrovirus |
Local injection |
Gu, Li, Tokuyama, Yokota, and Namba (2010) |
Glioma (9L) |
RatBM-MSC |
CD |
Adenovirus |
Local injection |
Kosaka et al. (2012) |
Glioma (U-87) |
hBM-MSC |
CD |
Retrovirus |
Local injection |
Jung et al. (2015) |
prostate cancer (PC3,DU145) & breast cancer (MCF7) & mouse fibrosarcoma (RIF1) |
RatBM-MSC |
HSV-tk |
Lentivirus |
Systemically administered |
Song et al. (2010) |
Glioma (C6) |
RatBM-MSC |
HSV-tk |
Retrovirus |
Local injection |
Amano, Gu, Koizumi, Tokuyama, and Namba (2011) |
Glioma (C6) |
RatBM-MSC |
HSV-tk |
Retrovirus |
Local injection |
Amano, Gu, Koizumi, Tokuyama, and Namba (2011) |
Glioma (U-87) |
hBM-MSC |
HSV-tk |
Adenovirus |
Local injection |
Ryu et al. (2012) |
Glioma (U87MG) |
hBM-MSC |
HSV-tk |
Baculovirus |
Systemically administered |
Bak et al. (2010) |
6 FUTURE PERSPECTIVE
Current data indicate that cancer incidence is steadily increasing in the World. There is high mortality statistic available in current decades. All of these highlighted the urgent need for more safer and more effective therapies. One of the principal challenges along cancer treatment is how to destroy malignant tumors without damaging healthy cells. A new approach that shows great promise in this area is employment of a suicide gene. In this way we need an appropriate carrier for therapeutic gene delivery specific to cancer site. The application of anti-cancer gene-expressing MSCs for targeted cancer therapy is a novel and promising strategy. MSCs with important characteristic such as strong tumor tropism, unlimited packaging capacity and unique immunologic tolerance, could overcome current obstacles and successfully deliver these suicide genes. Although MSCs have anticancer capacity but based on some reports could have positive role in tumor progression. Eventually more researches required to find novel insights into MSCs biology, potential clinical application, and molecular mechanism for homing to tumor site.
REFERENCES
-
Abate-Daga, D.,
Garcia-Rodríguez, L.,
Sumoy, L., &
Fillat, C.
(2010). Cell cycle control pathways act as conditioning factors for TK/GCV sensitivity in pancreatic cancer cells.
Biochimica et Biophysica Acta (BBA)-Molecular Cell Research,
1803(10), 1175–1185.
-
Abrate, A.,
Buono, R.,
Canu, T.,
Esposito, A.,
Del Maschio, A.,
Lucianò, R., …
Benigni, F.
(2014). Mesenchymal stem cells expressing therapeutic genes induce autochthonous prostate tumour regression.
European Journal of Cancer,
50(14), 2478–2488.
-
Adachi, M.,
Sampath, J.,
Lan, L.-B.,
Sun, D.,
Hargrove, P.,
Flatley, R., …
Matherly, L.
(2002). Expression of MRP4 confers resistance to ganciclovir and compromises bystander cell killing.
Journal of Biological Chemistry,
277(41), 38998–39004.
-
Aghi, M.,
Cho, T.-C.,
Chiocca, E. A.,
Kramm, C. M., &
Breakefield, X. O.
(1998). Synergistic anticancer effects of ganciclovir/thymidine kinase and 5-fluorocytosine/cytosine deaminase gene therapies.
Journal of the National Cancer Institute,
90(5), 370–380.
-
Altanerova, V.,
Cihova, M.,
Babic, M.,
Rychly, B.,
Ondicova, K.,
Mravec, B., &
Altaner, C.
(2012). Human adipose tissue-derived mesenchymal stem cells expressing yeast cytosinedeaminase::uracil phosphoribosyltransferase inhibit intracerebral rat glioblastoma.
International Journal of Cancer,
130(10), 2455–2463.
-
Amano, S.,
Gu, C.,
Koizumi, S.,
Tokuyama, T., &
Namba, H.
(2011). Timing of ganciclovir administration in glioma gene therapy using HSVtk gene-transduced mesenchymal stem cells.
Cancer Genomics-Proteomics,
8(5), 245–250.
-
Amano, S.,
Gu, C.,
Koizumi, S.,
Tokuyama, T., &
Namba, H.
(2011). Tumoricidal bystander effect in the suicide gene therapy using mesenchymal stem cells does not injure normal brain tissues.
Cancer Letters,
306(1), 99–105.
-
Amara, I.,
Pramil, E.,
Senamaud-Beaufort, C.,
Devillers, A.,
Macedo, R.,
Lescaille, G., …
Beaune, P.
(2016). Engineered mesenchymal stem cells as vectors in a suicide gene therapy against preclinical murine models for solid tumors.
Journal of Controlled Release,
239, 82–91.
-
Amara, I.,
Touati, W.,
Beaune, P., &
de Waziers, I.
(2014). Mesenchymal stem cells as cellular vehicles for prodrug gene therapy against tumors.
Biochimie,
105, 4–11.
-
Amer, M. H.
(2014). Gene therapy for cancer: Present status and future perspective.
Molecular and Cellular Therapies,
2(1), 27.
-
Ando, M.,
Hoyos, V.,
Yagyu, S.,
Tao, W.,
Ramos, C. A.,
Dotti, G., …
Bouchier-Hayes, L.
(2014). Bortezomib sensitizes non-small cell lung cancer to mesenchymal stromal cell-delivered inducible caspase-9-mediated cytotoxicity.
Cancer Gene Therapy,
21(11), 472–482.
-
Argos, P.
(1990). An investigation of oligopeptides linking domains in protein tertiary structures and possible candidates for general gene fusion.
Journal of Molecular Biology,
211(4), 943–958.
-
Azzam, E. I., &
Little, J. B.
(2004). The radiation-induced bystander effect: Evidence and significance.
Human & Experimental Toxicology,
23(2), 61–65.
-
Baird, S.
(2015). Mesenchymal stem cells: How can we realize their therapeutic potential in cancer therapy?
Journal of Clinical & Experimental Pathology,
1(3), 25. https://doi.org/10.1186/scrt25
-
Bak, X.,
Yang, J., &
Wang, S.
(2010). Baculovirus-transduced bone marrow mesenchymal stem cells for systemic cancer therapy.
Cancer Gene Therapy,
17(10), 721–729.
-
Balzarini, J.,
Liekens, S.,
Solaroli, N.,
El Omari, K.,
Stammers, D. K., &
Karlsson, A.
(2006). Engineering of a single conserved amino acid residue of herpes simplex virus type 1 thymidine kinase allows a predominant shift from pyrimidine to purine nucleoside phosphorylation.
Journal of Biological Chemistry,
281(28), 19273–19279.
-
Banikazemi, Z.,
Haji, H. A.,
Mohammadi, M.,
Taheripak, G.,
Iranifar, E.,
Poursadeghiyan, M., …
Mirzaei, H.
(2017). Diet and cancer prevention: Dietary compounds, dietary microRNAs and dietary exosomes.
Journal of Cellular Biochemistry,
28(10), 26244.
-
Black, M. E.,
Kokoris, M. S., &
Sabo, P.
(2001). Herpes simplex virus-1 thymidine kinase mutants created by semi-random sequence mutagenesis improve prodrug-mediated tumor cell killing.
Cancer Research,
61(7), 3022–3026.
-
Blattman, J. N., &
Greenberg, P. D.
(2004). Cancer immunotherapy: A treatment for the masses.
Science,
305(5681), 200–205.
-
Boatright, K. M.,
Renatus, M.,
Scott, F. L.,
Sperandio, S.,
Shin, H.,
Pedersen, I. M., …
Green, D. R.
(2003). A unified model for apical caspase activation.
Molecular Cell,
11(2), 529–541.
-
Borujeni, M. J. S.,
Esfandiary, E.,
Taheripak, G.,
Codoner-Franch, P.,
Alonso-Iglesias, E., &
Mirzaei, H.
(2017). Molecular aspects of diabetes mellitus: Resistin, microRNA and exosome.
Journal of Cellular Biochemistry,
8(10), 26271.
-
Both, G. W.
(2009). Gene-directed enzyme prodrug therapy for cancer: A glimpse into the future?
Discovery Medicine,
8(42), 97–103.
-
Braybrooke, J.,
Slade, A.,
Gibson, R.,
Deplanque, G.,
Madhusudan, S., &
Makris, A.
(2002). Phase one study of MetXia-P450 gene therapy for patients with advanced breast cancer or melanoma.
11(4): 1512–1520.
-
Breitbach, M.,
Bostani, T.,
Roell, W.,
Xia, Y.,
Dewald, O.,
Nygren, J. M., …
Hescheler, J.
(2007). Potential risks of bone marrow cell transplantation into infarcted hearts.
Blood,
110(4), 1362–1369.
-
Cao, L.,
Kulmburg, P.,
Veelken, H.,
Mackensen, A.,
Mézes, B.,
Lindemann, A., …
Rosenthal, F. M.
(1998). Cytokine gene transfer in cancer therapy.
Stem Cells,
16(S2), 251–260.
-
Cavarretta, I. T.,
Altanerova, V.,
Matuskova, M.,
Kucerova, L.,
Culig, Z., &
Altaner, C.
(2010). Adipose tissue-derived mesenchymal stem cells expressing prodrug-converting enzyme inhibit human prostate tumor growth.
Molecular Therapy,
18(1), 223–231.
-
Chang, D. Y.,
Yoo, S. W.,
Hong, Y.,
Kim, S.,
Kim, S. J.,
Yoon, S. H., …
Kim, S. S.
(2010). The growth of brain tumors can be suppressed by multiple transplantation of mesenchymal stem cells expressing cytosine deaminase.
International Journal of Cancer,
127(8), 1975–1983.
-
Chang, J. W.,
Lee, H.,
Kim, E.,
Lee, Y.,
Chung, S. S., &
Kim, J.-H.
(2000). Combined antitumor effects of an adenoviral cytosine deaminase/thymidine kinase fusion gene in rat C6 glioma.
Neurosurgery,
47(4), 931–939.
-
Cheung, W. Y.,
Fralick, R., &
Cheng, S.
(2008). The confused cancer patient: A case of 5-fluorouracil-induced encephalopathy.
Current Oncology,
15(5), 234–236.
-
Chiocca, E. A., &
Rabkin, S. D.
(2014). Oncolytic viruses and their application to cancer immunotherapy.
Cancer Immunology Research,
2(4), 295–300.
-
Chung-Faye, G.,
Palmer, D.,
Anderson, D.,
Clark, J.,
Downes, M.,
Baddeley, J., …
Seymour, L.
(2001). Virus-directed, enzyme prodrug therapy with nitroimidazole reductase.
Clinical Cancer Research,
7(9), 2662–2668.
-
Collins, M., &
Thrasher, A.
(2015). Gene therapy: Progress and predictions.
Proceedings Biological Sciences,
282(1821), 20143003. https://doi.org/10.1098/rspb.2014.3003
-
Collins, S. A.,
Guinn, B. A.,
Harrison, P. T.,
Scallan, M. F.,
O'Sullivan, G. C., &
Tangney, M.
(2008). Viral vectors in cancer immunotherapy: Which vector for which strategy?
Current Gene Therapy,
8(2), 66–78.
-
Compte, M.,
Cuesta, A. M.,
Sánchez-Martín, D.,
Alonso-Camino, V.,
Vicario, J. L.,
Sanz, L., &
Álvarez-Vallina, L.
(2009). Tumor immunotherapy using gene-modified human mesenchymal stem cells loaded into synthetic extracellular matrix scaffolds.
Stem Cells,
27(3), 753–760.
-
Cross, D., &
Burmester, J. K.
(2006). Gene therapy for cancer treatment: Past, present and future.
Clinical Medicine & Research,
4(3), 218–227.
-
Dai, L.-J.,
Moniri, M. R.,
Zeng, Z.-R.,
Zhou, J. X.,
Rayat, J., &
Warnock, G. L.
(2011). Potential implications of mesenchymal stem cells in cancer therapy.
Cancer Letters,
305(1), 8–20.
-
Dani, S.
(1999). The challenge of vector development in gene therapy.
Brazilian Journal of Medical and Biological Research,
32(2), 133–145.
-
de Melo, S. M.,
Bittencourt, S.,
Ferrazoli, E. G.,
da Silva, C. S.,
da Cunha, F. F.,
da Silva, F. H., …
Martin, P. K. M.
(2015). The anti-tumor effects of adipose tissue mesenchymal stem cell transduced with HSV-Tk gene on U-87-driven brain tumor.
PLoS ONE,
10(6), e0128922.
-
Djouad, F.,
Plence, P.,
Bony, C.,
Tropel, P.,
Apparailly, F.,
Sany, J., …
Jorgensen, C.
(2003). Immunosuppressive effect of mesenchymal stem cells favors tumor growth in allogeneic animals.
Blood,
102(10), 3837–3844.
-
Dominici, M.,
Le Blanc, K.,
Mueller, I.,
Slaper-Cortenbach, I.,
Marini, F.,
Krause, D., …
Horwitz, E.
(2006). Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement.
Cytotherapy,
8(4), 315–317.
-
Duarte, S.,
Carle, G.,
Faneca, H.,
De Lima, M. C. P., &
Pierrefite-Carle, V.
(2012). Suicide gene therapy in cancer: Where do we stand now?
Cancer Letters,
324(2), 160–170.
-
Dvorak, H. F.
(1986). Tumors: Wounds that do not heal. Similarities between tumor stroma generation and wound healing.
The New England Journal of Medicine,
315(26), 1650–1659.
-
Dwyer, R. M.,
Khan, S.,
Barry, F. P.,
O'Brien, T., &
Kerin, M. J.
(2010). Advances in mesenchymal stem cell-mediated gene therapy for cancer.
Stem Cell Research & Therapy,
1(3), 25.
-
Fathullahzadeh, S.,
Mirzaei, H.,
Honardoost, M. A.,
Sahebkar, A., &
Salehi, M.
(2016). Circulating microRNA-192 as a diagnostic biomarker in human chronic lymphocytic leukemia.
Cancer Gene Therapy,
23(10), 327–332.
-
Fei, S.,
Qi, X.,
Kedong, S.,
Guangchun, J.,
Jian, L., &
Wei, Q.
(2012). The antitumor effect of mesenchymal stem cells transduced with a lentiviral vector expressing cytosine deaminase in a rat glioma model.
Journal of Cancer Research and Clinical Oncology,
138(2), 347–357.
-
Freeman, S. M.
(2002). Suicide gene therapy. Cancer gene therapy.
Advances in Experimental Medicine and Biology,
465: 411–422.
-
Freeman, S. M.,
Abboud, C. N.,
Whartenby, K. A.,
Packman, C. H.,
Koeplin, D. S.,
Moolten, F. L., &
Abraham, G. N.
(1993). The “bystander effect”: Tumor regression when a fraction of the tumor mass is genetically modified.
Cancer Research,
53(21), 5274–5283.
-
Freytag, S. O.,
Rogulski, K. R.,
Paielli, D. L.,
Gilbert, J. D., &
Kim, J. H.
(1998). A novel three-pronged approach to kill cancer cells selectively: Concomitant viral, double suicide gene, and radiotherapy.
Human Gene Therapy,
9(9), 1323–1333.
-
Friedenstein, A.,
Chailakhjan, R., &
Lalykina, K.
(1970). The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells.
Cell Proliferation,
3(4), 393–403.
-
Gallois-Montbrun, S.,
Veron, M., &
Deville-Bonne, D.
(2004). Antiviral nucleoside analogs phosphorylation by nucleoside diphosphate kinase.
Mini Reviews in Medicinal Chemistry,
4(4), 361–369.
-
Gao, J.-Q.,
Zhao, Q.-Q.,
Lv, T.-F.,
Shuai, W.-P.,
Zhou, J.,
Tang, G.-P., …
Hu, Y.-L.
(2010). Gene-carried chitosan-linked-PEI induced high gene transfection efficiency with low toxicity and significant tumor-suppressive activity.
International Journal of Pharmaceutics,
387(1), 286–294.
-
Gholamin, S.,
Mirzaei, H.,
Razavi, S. M.,
Hassanian, S. M.,
Saadatpour, L.,
Masoudifar, A., …
Avan, A.
(2017). GD2-targeted immunotherapy and potential value of circulating microRNAs in neuroblastoma.
Journal of Cellular Physiology,
1(10), 25793.
-
Gholamin, S.,
Pasdar, A.,
Khorrami, M. S.,
Mirzaei, H.,
Mirzaei, H. R.,
Salehi, R., …
Avan, A.
(2016). The potential for circulating microRNAs in the diagnosis of myocardial infarction: A novel approach to disease diagnosis and treatment.
Current Pharmaceutical Design,
22(3), 397–403.
-
Golabchi, K.,
Soleimani-Jelodar, R.,
Aghadoost, N.,
Momeni, F.,
Moridikia, A.,
Nahand, J. S., …
Mirzaei, H.
(2017). MicroRNAs in retinoblastoma: Potential diagnostic and therapeutic biomarkers.
Journal of Cellular Physiology,
28(10), 26070.
-
Gopinath, P., &
Ghosh, S. S.
(2008). Implication of functional activity for determining therapeutic efficacy of suicide genes in vitro.
Biotechnology Letters,
30(11), 1913–1921.
-
Goradel, N. H.,
Hoor, F. G.,
Negahdari, B.,
Malekshahi, Z. V.,
Hashemzehi, M.,
Masoudifar, A., &
Mirzaei, H.
(2017). Stem cell therapy: A new therapeutic option for cardiovascular diseases.
Journal of Cellular Biochemistry,
25(10), 26169.
-
Greco, O., &
Dachs, G. U.
(2001). Gene directed enzyme/prodrug therapy of cancer: Historical appraisal and future prospectives.
Journal of Cellular Physiology,
187(1), 22–36.
-
Green, L. K.,
Storey, M. A.,
Williams, E. M.,
Patterson, A. V.,
Smaill, J. B.,
Copp, J. N., &
Ackerley, D. F.
(2013). The flavin reductase MsuE is a novel nitroreductase that can efficiently activate two promising next-generation prodrugs for gene-directed enzyme prodrug therapy.
Cancers,
5(3), 985–997.
-
Grisendi, G.,
Bussolari, R.,
Cafarelli, L.,
Petak, I.,
Rasini, V.,
Veronesi, E., …
Barti-Juhasz, H.
(2010). Adipose-derived mesenchymal stem cells as stable source of tumor necrosis factor-related apoptosis-inducing ligand delivery for cancer therapy.
Cancer Research,
70(9), 3718–3729.
-
Gu, C.,
Li, S.,
Tokuyama, T.,
Yokota, N., &
Namba, H.
(2010). Therapeutic effect of genetically engineered mesenchymal stem cells in rat experimental leptomeningeal glioma model.
Cancer Letters,
291(2), 256–262.
-
Hacein-Bey-Abina, S.,
Le Deist, F.,
Carlier, F.,
Bouneaud, C.,
Hue, C.,
De Villartay, J.-P., …
Dupuis-Girod, S.
(2002). Sustained correction of X-linked severe combined immunodeficiency by ex vivo gene therapy.
New England Journal of Medicine,
346(16), 1185–1193.
-
Hashemi Goradel, N.,
Ghiyami-Hour, F.,
Jahangiri, S.,
Negahdari, B.,
Sahebkar, A.,
Masoudifar, A., &
Mirzaei, H.
(2017). Nanoparticles as new tools for inhibition of cancer angiogenesis.
Journal of Cellular Physiology,
25(10), 26029.
-
Honczarenko, M.,
Le, Y.,
Swierkowski, M.,
Ghiran, I.,
Glodek, A. M., &
Silberstein, L. E.
(2006). Human bone marrow stromal cells express a distinct set of biologically functional chemokine receptors.
Stem Cells,
24(4), 1030–1041.
-
Horwitz, E.,
Le Blanc, K.,
Dominici, M.,
Mueller, I.,
Slaper-Cortenbach, I.,
Marini, F. C., …
Keating, A.
(2005). Clarification of the nomenclature for MSC: The International Society for Cellular Therapy position statement.
Cytotherapy,
7(5), 393–395.
-
Hoseini, Z.,
Sepahvand, F.,
Rashidi, B.,
Sahebkar, A.,
Masoudifar, A., &
Mirzaei, H.
(2017). NLRP3 inflammasome: Its regulation and involvement in atherosclerosis.
Journal of Cellular Physiology,
27(10), 25930.
-
Hu, Y.-L.,
Fu, Y.-H.,
Tabata, Y., &
Gao, J.-Q.
(2010). Mesenchymal stem cells: A promising targeted-delivery vehicle in cancer gene therapy.
Journal of Controlled Release,
147(2), 154–162.
-
Huang, Q.,
Liu, X.,
Kang, C.,
Wang, G.,
Zhong, Y., &
Pu, P.
(2010). The anti-glioma effect of suicide gene therapy using BMSC expressing HSV/TK combined with overexpression of Cx43 in glioma cells.
Cancer Gene Therapy,
17(3), 192–202.
-
Huang, S.,
Zhang, D.,
Han, J.,
Zhang, N.,
Zhang, S.,
Mu, W., &
Wei, F.
(2009). Radiosensitization and anti-tumour effects of cytosine deaminase and thymidine kinase fusion suicide gene in human adenoid cystic carcinoma cells.
Journal of International Medical Research,
37(2), 479–490.
-
Huber, B. E.,
Austin, E. A.,
Richards, C. A.,
Davis, S. T., &
Good, S. S.
(1994). Metabolism of 5-fluorocytosine to 5-fluorouracil in human colorectal tumor cells transduced with the cytosine deaminase gene: Significant antitumor effects when only a small percentage of tumor cells express cytosine deaminase.
Proceedings of the National Academy of Sciences of the United States of America,
91(17), 8302–8306.
-
Hung, S.-C.,
Deng, W.-P.,
Yang, W. K.,
Liu, R.-S.,
Lee, C.-C.,
Su, T.-C., …
Chen, W.-H.
(2005). Mesenchymal stem cell targeting of microscopic tumors and tumor stroma development monitored by noninvasive in vivo positron emission tomography imaging.
Clinical Cancer Research,
11(21), 7749–7756.
-
Izmirli, M.,
Sonmez, D., &
Gogebakan, B.
(2016). The war against cancer: Suicide gene therapy.
Advances in Modern Oncology Research,
8(8), e70594.
-
Jiang, J.,
Wei, D.,
Sun, L.,
Wang, Y.,
Wu, X.,
Li, Y., …
Wei, Z.
(2014). A preliminary study on the construction of double suicide gene delivery vectors by mesenchymal stem cells and the in vitro inhibitory effects on SKOV3 cells.
Oncology Reports,
31(2), 781–787.
-
Jung, J. H.,
Kim, A. A.,
Chang, D.-Y.,
Park, Y. R.,
Suh-Kim, H., &
Kim, S.-S.
(2015). Three-dimensional assessment of bystander effects of mesenchymal stem cells carrying a cytosine deaminase gene on glioma cells.
American Journal of Cancer Research,
5(9), 2686.
-
Kaliberov, S. A., &
Buchsbaum, D. J.
(2012). Cancer treatment with gene therapy and radiation therapy.
Advances in Cancer Research,
115, 221.
-
Kanai, F.,
Kawakami, T.,
Hamada, H.,
Sadata, A.,
Yoshida, Y.,
Tanaka, T., …
Omata, M.
(1998). Adenovirus-mediated transduction of Escherichia coli uracil phosphoribosyltransferase gene sensitizes cancer cells to low concentrations of 5-fluorouracil.
Cancer Research,
58(9), 1946–1951.
-
Karjoo, Z.,
Chen, X., &
Hatefi, A.
(2016a). Progress and problems with the use of suicide genes for targeted cancer therapy.
Advanced Drug Delivery Reviews,
99, 113–128.
-
Karjoo, Z.,
Chen, X., &
Hatefi, A.
(2016b). Progress and problems with the use of suicide genes for targeted cancer therapy.
Advanced Drug Delivery Reviews,
99(Pt A), 113–128.
-
Kern, S.,
Eichler, H.,
Stoeve, J.,
Klüter, H., &
Bieback, K.
(2006). Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue.
Stem Cells,
24(5), 1294–1301.
-
Keshavarzi, M.,
Darijani, M.,
Momeni, F.,
Moradi, P.,
Ebrahimnejad, H.,
Masoudifar, A., &
Mirzaei, H.
(2017). Molecular imaging and oral cancer diagnosis and therapy.
Journal of Cellular Biochemistry,
8(10), 26042.
-
Keshavarzi, M.,
Sorayayi, S.,
Jafar Rezaei, M.,
Mohammadi, M.,
Ghaderi, A.,
Rostamzadeh, A., …
Mirzaei, H.
(2017). MicroRNAs-based imaging techniques in cancer diagnosis and therapy.
Journal of Cellular Biochemistry,
29(10), 26012.
-
Kidd, S.,
Spaeth, E.,
Klopp, A.,
Andreeff, M.,
Hall, B., &
Marini, F.
(2008). The (in) auspicious role of mesenchymal stromal cells in cancer: Be it friend or foe.
Cytotherapy,
10(7), 657–667.
-
Kievit, E.,
Nyati, M. K.,
Ng, E.,
Stegman, L. D.,
Parsels, J.,
Ross, B. D., …
Lawrence, T. S.
(2000). Yeast cytosine deaminase improves radiosensitization and bystander effect by 5-fluorocytosine of human colorectal cancer xenografts.
Cancer Research,
60(23), 6649–6655.
-
Kim, J. J., &
Tannock, I. F.
(2005). Repopulation of cancer cells during therapy: An important cause of treatment failure.
Nature Reviews Cancer,
5(7), 516–525.
-
Kim, S. M.,
Lim, J. Y.,
Park, S. I.,
Jeong, C. H.,
Oh, J. H.,
Jeong, M., …
Jeun, S.-S.
(2008). Gene therapy using TRAIL-secreting human umbilical cord blood-derived mesenchymal stem cells against intracranial glioma.
Cancer Research,
68(23), 9614–9623.
-
Kim, S. M.,
Oh, J. H.,
Park, S. A.,
Ryu, C. H.,
Lim, J. Y.,
Kim, D. S., …
Jeun, S. S.
(2010). Irradiation enhances the tumor tropism and therapeutic potential of tumor necrosis factor-related apoptosis-inducing ligand-secreting human umbilical cord blood-derived mesenchymal stem cells in glioma therapy.
Stem Cells,
28(12), 2217–2228.
-
Kim, S. W.,
Kim, S. J.,
Park, S. H.,
Yang, H. G.,
Kang, M. C.,
Choi, Y. W., …
Sung, Y. C.
(2013). Complete regression of metastatic renal cell carcinoma by multiple injections of engineered mesenchymal stem cells expressing dodecameric TRAIL and HSV-TK.
Clinical Cancer Research,
19(2), 415–427.
-
Klopp, A. H.,
Spaeth, E. L.,
Dembinski, J. L.,
Woodward, W. A.,
Munshi, A.,
Meyn, R. E., …
Marini, F. C.
(2007). Tumor irradiation increases the recruitment of circulating mesenchymal stem cells into the tumor microenvironment.
Cancer Research,
67(24), 11687–11695.
-
Kosaka, H.,
Ichikawa, T.,
Kurozumi, K.,
Kambara, H.,
Inoue, S.,
Maruo, T., …
Date, I.
(2012). Therapeutic effect of suicide gene-transferred mesenchymal stem cells in a rat model of glioma.
Cancer Gene Therapy,
19(8), 572–578.
-
Krassikova, L. S.,
Karshieva, S. S.,
Cheglakov, I. B., &
Belyavsky, A. V.
(2016). Combined treatment, based on lysomustine administration with mesenchymal stem cells expressing cytosine deaminase therapy, leads to pronounced murine Lewis lung carcinoma growth inhibition.
The Journal of Gene Medicine,
18(9), 220–233.
-
Kucerova, L.,
Altanerova, V.,
Matuskova, M.,
Tyciakova, S., &
Altaner, C.
(2007). Adipose tissue-derived human mesenchymal stem cells mediated prodrug cancer gene therapy.
Cancer Research,
67(13), 6304–6313.
-
Kucerova, L.,
Matuskova, M.,
Pastorakova, A.,
Tyciakova, S.,
Jakubikova, J.,
Bohovic, R., …
Altaner, C.
(2008). Cytosine deaminase expressing human mesenchymal stem cells mediated tumour regression in melanoma bearing mice.
The Journal of Gene Medicine,
10(10), 1071–1082.
-
Kuriyama, S.,
Mitoro, A.,
Yamazaki, M.,
Tsujinoue, H.,
Nakatani, T.,
Akahane, T., …
Fukui, H.
(1999). Comparison of gene therapy with the herpes simplex virus thymidine kinase gene and the bacterial cytosine deaminase gene for the treatment of hepatocellular carcinoma.
Scandinavian Journal of Gastroenterology,
34(10), 1033–1041.
-
Lee, R. H.,
Hsu, S. C.,
Munoz, J.,
Jung, J. S.,
Lee, N. R.,
Pochampally, R., &
Prockop, D. J.
(2006). A subset of human rapidly self-renewing marrow stromal cells preferentially engraft in mice.
Blood,
107(5), 2153–2161.
-
Lee, W. Y.,
Zhang, T.,
Lau, C. P.,
Wang, C.,
Chan, K.-M., &
Li, G.
(2013). Immortalized human fetal bone marrow-derived mesenchymal stromal cell expressing suicide gene for anti-tumor therapy in vitro and in vivo.
Cytotherapy,
15(12), 1484–1497.
-
Li Bi, W.,
Parysek, L. M.,
Warnick, R., &
Stambrook, P. J.
(1993). In vitro evidence that metabolic cooperation is responsible for the bystander effect observed with HSV tk retroviral gene therapy.
Human Gene Therapy,
4(6), 725–731.
-
Li, S.,
Ou, M.,
Wang, G., &
Tang, L.
(2016). Application of conditionally replicating adenoviruses in tumor early diagnosis technology, gene-radiation therapy and chemotherapy.
Applied Microbiology and Biotechnology,
100(19), 8325–8335.
-
Li, Z.,
Fan, D., &
Xiong, D.
(2015). Mesenchymal stem cells as delivery vectors for anti-tumor therapy.
Stem Cell Investigation,
2, 6. https://doi.org/10.3978/j.issn.2306-9759.2015.03.01
-
Lin, E., &
Nemunaitis, J.
(2004). Oncolytic viral therapies.
Cancer Gene Therapy,
11(10), 643–664.
-
Link, C. J.,
Seregina, T.,
Traynor, A., &
Burt, R. K.
(2000). Cellular suicide therapy of malignant disease.
Stem Cells,
18(3), 220–226.
-
Liu, X.,
Hu, J.,
Sun, S.,
Li, F.,
Cao, W.,
Wang, Y., …
Yu, Z.
(2015). Mesenchymal stem cells expressing interleukin-18 suppress breast cancer cells in vitro.
Experimental and Therapeutic Medicine,
9(4), 1192–1200.
-
Loebinger, M. R.,
Eddaoudi, A.,
Davies, D., &
Janes, S. M.
(2009). Mesenchymal stem cell delivery of TRAIL can eliminate metastatic cancer.
Cancer Research,
69(10), 4134–4142.
-
Longley, D. B.,
Harkin, D. P., &
Johnston, P. G.
(2003). 5-fluorouracil: Mechanisms of action and clinical strategies.
Nature Reviews Cancer,
3(5), 330–338.
-
Ma, Y.,
Hao, X.,
Zhang, S., &
Zhang, J.
(2012). The in vitro and in vivo effects of human umbilical cord mesenchymal stem cells on the growth of breast cancer cells.
Breast Cancer Research and Treatment,
133(2), 473–485.
-
Martin, F.,
Dwyer, R.,
Kelly, J.,
Khan, S.,
Murphy, J.,
Curran, C., …
Barry, F.
(2010). Potential role of mesenchymal stem cells (MSCs) in the breast tumour microenvironment: Stimulation of epithelial to mesenchymal transition (EMT).
Breast Cancer Research and Treatment,
124(2), 317–326.
-
Martinez-Quintanilla, J.,
Bhere, D.,
Heidari, P.,
He, D.,
Mahmood, U., &
Shah, K.
(2013). Therapeutic efficacy and fate of bimodal engineered stem cells in malignant brain tumors.
Stem Cells,
31(8), 1706–1714.
-
Mashreghi, M.,
Azarpara, H.,
Bazaz, M. R.,
Jafari, A.,
Masoudifar, A.,
Mirzaei, H., &
Jaafari, M. R.
(2017). Angiogenesis biomarkers and their targeting ligands as potential targets for tumor angiogenesis.
Journal of Cellular Physiology,
13(10), 26049.
-
Matono, S.,
Tanaka, T.,
Sueyoshi, S.,
Yamana, H.,
Fujita, H., &
Shirouzu, K.
(2003). Bystander effect in suicide gene therapy is directly proportional to the degree of gap junctional intercellular communication in esophageal cancer.
International Journal of Oncology,
23(5), 1309–1316.
-
Matuskova, M.,
Baranovicova, L.,
Kozovska, Z.,
Durinikova, E.,
Pastorakova, A.,
Hunakova, L., …
Kucerova, L.
(2012). Intrinsic properties of tumour cells have a key impact on the bystander effect mediated by genetically engineered mesenchymal stromal cells.
The Journal of Gene Medicine,
14(12), 776–787.
-
Matuskova, M.,
Hlubinova, K.,
Pastorakova, A.,
Hunakova, L.,
Altanerova, V.,
Altaner, C., &
Kucerova, L.
(2010). HSV-tk expressing mesenchymal stem cells exert bystander effect on human glioblastoma cells.
Cancer Letters,
290(1), 58–67.
-
Matuskova, M.,
Kozovska, Z.,
Toro, L.,
Durinikova, E.,
Tyciakova, S.,
Cierna, Z., …
Kucerova, L.
(2015). Combined enzyme/prodrug treatment by genetically engineered AT-MSC exerts synergy and inhibits growth of MDA-MB-231 induced lung metastases.
Journal of Experimental & Clinical Cancer Research,
34(1), 33.
-
McCormick, F.
(2001). Cancer gene therapy: Fringe or cutting edge?
Nature Reviews Cancer,
1(2), 130–141.
-
Menon, L. G.,
Picinich, S.,
Koneru, R.,
Gao, H.,
Lin, S. Y.,
Koneru, M., …
Banerjee, D.
(2007). Differential gene expression associated with migration of mesenchymal stem cells to conditioned medium from tumor cells or bone marrow cells.
Stem Cells,
25(2), 520–528.
-
Mesnil, M.,
Piccoli, C.,
Tiraby, G.,
Willecke, K., &
Yamasaki, H.
(1996). Bystander killing of cancer cells by herpes simplex virus thymidine kinase gene is mediated by connexins.
Proceedings of the National Academy of Sciences United States of America,
93(5), 1831–1835.
-
Mesnil, M., &
Yamasaki, H.
(2000). Bystander effect in herpes simplex virus-thymidine kinase/ganciclovir cancer gene therapy: Role of gap-junctional intercellular communication1.
Cancer Research,
60(15), 3989–3999.
-
Miletic, H.,
Fischer, Y.,
Litwak, S.,
Giroglou, T.,
Waerzeggers, Y.,
Winkeler, A., …
Stenzel, W.
(2007). Bystander killing of malignant glioma by bone marrow-derived tumor-infiltrating progenitor cells expressing a suicide gene.
Molecular Therapy,
15(7), 1373–1381.
-
Miller, N., &
Vile, R.
(1995). Targeted vectors for gene therapy.
FASEB Journal,
9(2), 190–199.
-
Mirzaei, H.
(2017). Stroke in women: Risk factors and clinical biomarkers.
Journal of Cellular Biochemistry,
12(10), 26130.
-
Mirzaei, H.,
Fathullahzadeh, S.,
Khanmohammadi, R.,
Darijani, M.,
Momeni, F.,
Masoudifar, A., …
Mirzaei, H. R.
(2017). State of the art in microRNA as diagnostic and therapeutic biomarkers in chronic lymphocytic leukemia.
Journal of Cellular Physiology,
13(10), 25799.
-
Mirzaei, H.,
Gholamin, S.,
Shahidsales, S.,
Sahebkar, A.,
Jaafari, M. R.,
Mirzaei, H. R., …
Avan, A.
(2016). MicroRNAs as potential diagnostic and prognostic biomarkers in melanoma.
European Journal of Cancer,
53, 25–32.
-
Mirzaei, H.,
Khataminfar, S.,
Mohammadparast, S.,
Sales, S. S.,
Maftouh, M.,
Mohammadi, M., …
Avan, A.
(2016). Circulating microRNAs as potential diagnostic biomarkers and therapeutic targets in gastric cancer: Current status and future perspectives.
Current Medicinal Chemistry,
23(36), 4135–4150.
-
Mirzaei, H.,
Masoudifar, A.,
Sahebkar, A.,
Zare, N.,
Nahand, J. S.,
Rashidi, B., …
Jaafari, M. R.
(2017). MicroRNA: A novel target of curcumin in cancer therapy.
Journal of Cellular Physiology,
15(10), 26055.
-
Mirzaei, H.,
Momeni, F.,
Saadatpour, L.,
Sahebkar, A.,
Goodarzi, M.,
Masoudifar, A., …
Jaafari, M. R.
(2017). MicroRNA: Relevance to stroke diagnosis, prognosis, and therapy.
Journal of Cellular Physiology,
9(10), 25787.
-
Mirzaei, H.,
Naseri, G.,
Rezaee, R.,
Mohammadi, M.,
Banikazemi, Z.,
Mirzaei, H. R., …
Sahebkar, A.
(2016). Curcumin: A new candidate for melanoma therapy?
International Journal of Cancer,
139(8), 1683–1695.
-
Mirzaei, H.,
Sahebkar, A.,
Avan, A.,
Jaafari, M. R.,
Salehi, R.,
Salehi, H., …
Mirzaei, H. R.
(2016). Application of mesenchymal stem cells in melanoma: A potential therapeutic strategy for delivery of targeted agents.
Current Medicinal Chemistry,
23(5), 455–463.
-
Mirzaei, H.,
Sahebkar, A.,
Jaafari, M. R.,
Goodarzi, M., &
Mirzaei, H. R.
(2016). Diagnostic and therapeutic potential of exosomes in cancer: The beginning of a new tale?
Journal of Cellular Physiology,
14(10), 25739.
-
Mirzaei, H.,
Sahebkar, A.,
Jaafari, M. R.,
Hadjati, J.,
Javanmard, S. H.,
Mirzaei, H. R., &
Salehi, R.
(2016). PiggyBac as a novel vector in cancer gene therapy: Current perspective.
Cancer Gene Therapy,
23(2–3), 45–47.
-
Mirzaei, H.,
Sahebkar, A.,
Shiri, L.,
Moridikia, A.,
Nazari, S.,
Nahand, J. S., …
Jaafari, M. R.
(2017). Therapeutic application of multipotent stem cells.
Journal of Cellular Physiology,
5(10), 25990.
-
Mirzaei, H.,
Yazdi, F.,
Salehi, R., &
Mirzaei, H. R.
(2016). SiRNA and epigenetic aberrations in ovarian cancer.
Journal of Cancer Research and Therapeutics,
12(2), 498–508.
-
Mirzaei, H. R.,
Mirzaei, H.,
Lee, S. Y.,
Hadjati, J., &
Till, B. G.
(2016). Prospects for chimeric antigen receptor (CAR) gammadelta T cells: A potential game changer for adoptive T cell cancer immunotherapy.
Cancer Letters,
380(2), 413–423.
-
Mirzaei, H. R.,
Sahebkar, A.,
Mohammadi, M.,
Yari, R.,
Salehi, H.,
Jafari, M. H., …
Mirzaei, H.
(2016). Circulating microRNAs in hepatocellular carcinoma: Potential diagnostic and prognostic biomarkers.
Current Pharmaceutical Design,
22(34), 5257–5269.
-
Mirzaei, H. R.,
Sahebkar, A.,
Salehi, R.,
Nahand, J. S.,
Karimi, E.,
Jaafari, M. R., &
Mirzaei, H.
(2016). Boron neutron capture therapy: Moving toward targeted cancer therapy.
Journal of Cancer Research and Therapeutics,
12(2), 520–525.
-
Mohammadi, M.,
Goodarzi, M.,
Jaafari, M. R.,
Mirzaei, H. R., &
Mirzaei, H.
(2016). Circulating microRNA: A new candidate for diagnostic biomarker in neuroblastoma.
Cancer Gene Ther,
23(11), 371–372.
-
Mohammadi, M.,
Jaafari, M. R.,
Mirzaei, H. R., &
Mirzaei, H.
(2016). Mesenchymal stem cell: A new horizon in cancer gene therapy.
Cancer Gene Therapy,
23(9), 285–286.
-
Mohr, A.,
Lyons, M.,
Deedigan, L.,
Harte, T.,
Shaw, G.,
Howard, L., …
Zwacka, R.
(2008). Mesenchymal stem cells expressing TRAIL lead to tumour growth inhibition in an experimental lung cancer model.
Journal of cellular and molecular medicine,
12(6b), 2628–2643.
-
Mori, K.,
Iwata, J.,
Miyazaki, M.,
Osada, H.,
Tange, Y.,
Yamamoto, T., …
Shiroishi, T.
(2010). Bystander killing effect of tymidine kinase gene-transduced adult bone marrow stromal cells with ganciclovir on malignant glioma cells.
Neurologia Medico-Chirurgica,
50(7), 545–553.
-
Moridikia, A.,
Mirzaei, H.,
Sahebkar, A., &
Salimian, J.
(2017). MicroRNAs: Potential candidates for diagnosis and treatment of colorectal cancer.
Journal of Cellular Physiology,
16(10), 25801.
-
Moriuchi, S.,
Wolfe, D.,
Tamura, M.,
Yoshimine, T.,
Miura, F.,
Cohen, J., &
Glorioso, J.
(2002). Double suicide gene therapy using a replication defective herpes simplex virus vector reveals reciprocal interference in a malignant glioma model.
Gene Therapy,
9(9), 584.
-
Morizono, K.,
De Ugarte, D. A.,
Zhu, M.,
Zuk, P.,
Elbarbary, A.,
Ashjian, P., …
Hedrick, M. H.
(2003). Multilineage cells from adipose tissue as gene delivery vehicles.
Human Gene Therapy,
14(1), 59–66.
-
Nambaru, P. K.,
Hübner, T.,
Köck, K.,
Mews, S.,
Grube, M.,
Payen, L., …
Rimmbach, C.
(2011). Drug efflux transporter multidrug resistance-associated protein 5 affects sensitivity of pancreatic cancer cell lines to the nucleoside anticancer drug 5-fluorouracil.
Drug Metabolism and Disposition,
39(1), 132–139.
-
Nauta, A. J., &
Fibbe, W. E.
(2007). Immunomodulatory properties of mesenchymal stromal cells.
Blood,
110(10), 3499–3506.
-
Nayerossadat, N.,
Maedeh, T., &
Ali, P. A.
(2012). Viral and nonviral delivery systems for gene delivery.
Advanced Biomedical Research,
1(1), 27.
-
Nguyen, T.-A.,
Tychopoulos, M.,
Bichat, F.,
Zimmermann, C.,
Flinois, J.-P.,
Diry, M., …
Beaune, P.
(2008). Improvement of cyclophosphamide activation by CYP2B6 mutants: From in silico to ex vivo.
Molecular Pharmacology,
73(4), 1122–1133.
-
NguyenThai, Q. A.,
Sharma, N.,
Luong, D. H.,
Sodhi, S. S.,
Kim, J. H.,
Kim, N., …
Jeong, D. K.
(2015). Targeted inhibition of osteosarcoma tumor growth by bone marrow-derived mesenchymal stem cells expressing cytosine deaminase/5-fluorocytosine in tumor-bearing mice.
The Journal of Gene Medicine,
17(3–5), 87–99.
-
Niess, H.,
Bao, Q.,
Conrad, C.,
Zischek, C.,
Notohamiprodjo, M.,
Schwab, F., …
Nelson, P. J.
(2011). Selective targeting of genetically engineered mesenchymal stem cells to tumor stroma microenvironments using tissue-specific suicide gene expression suppresses growth of hepatocellular carcinoma.
Annals of Surgery,
254(5), 767–775.
-
Nouri, F. S.,
Wang, X., &
Hatefi, A.
(2015). Genetically engineered theranostic mesenchymal stem cells for the evaluation of the anticancer efficacy of enzyme/prodrug systems.
Journal of Controlled Release,
200, 179–187.
-
Nowakowski, A.,
Drela, K.,
Rozycka, J.,
Janowski, M., &
Lukomska, B.
(2016). Engineered mesenchymal stem cells as an anti-cancer trojan horse.
Stem Cells and Development,
25(20), 1513–1531.
-
Oguri, T.,
Bessho, Y.,
Achiwa, H.,
Ozasa, H.,
Maeno, K.,
Maeda, H., …
Ueda, R.
(2007). MRP8/ABCC11 directly confers resistance to 5-fluorouracil.
Molecular Cancer Therapeutics,
6(1), 122–127.
-
Ohlfest, J. R.,
Freese, A. B., &
Largaespada, D. A.
(2005). Nonviral vectors for cancer gene therapy: Prospects for integrating vectors and combination therapies.
Current Gene Therapy,
5(6), 629–641.
-
Ohlsson, L. B.,
Varas, L.,
Kjellman, C.,
Edvardsen, K., &
Lindvall, M.
(2003). Mesenchymal progenitor cell-mediated inhibition of tumor growth in vivo and in vitro in gelatin matrix.
Experimental and Molecular Pathology,
75(3), 248–255.
-
Ostertag, D.,
Amundson, K. K.,
Lopez Espinoza, F.,
Martin, B.,
Buckley, T.,
Da Silva, A. P. G., …
Ibañez, C. E.
(2012). Brain tumor eradication and prolonged survival from intratumoral conversion of 5-fluorocytosine to 5-fluorouracil using a nonlytic retroviral replicating vector.
Neuro-Oncology,
14(2), 145–159.
-
Park, J. S.,
Chang, D.-Y.,
Kim, J.-H.,
Jung, J. H.,
Park, J.,
Kim, S.-H., …
Suh-Kim, H.
(2013). Retrovirus-mediated transduction of a cytosine deaminase gene preserves the stemness of mesenchymal stem cells.
Experimental & Molecular Medicine,
45(2), e10.
-
Persano, L.,
Crescenzi, M., &
Indraccolo, S.
(2007). Anti-angiogenic gene therapy of cancer: Current status and future prospects.
Molecular Aspects of Medicine,
28(1), 87–114.
-
Pessino, A., &
Sobrero, A.
(2006). Optimal treatment of metastatic colorectal cancer.
Expert Review of Anticancer Therapy,
6(5), 801–812.
-
Phinney, D. G., &
Prockop, D. J.
(2007). Concise review: Mesenchymal stem/multipotent stromal cells: The state of transdifferentiation and modes of tissue repair—Current views.
Stem Cells,
25(11), 2896–2902.
-
Pierrefite-Carle, V.,
Baqué, P.,
Gavelli, A.,
Mala, M.,
Chazal, M.,
Gugenheim, J., …
Rossi, B.
(1999). Cytosine deaminase/5-fluorocytosine-based vaccination against liver tumors: Evidence of distant bystander effect.
Journal of the National Cancer Institute,
91(23), 2014–2019.
-
Pommier, Y.
(2006). Topoisomerase I inhibitors: Camptothecins and beyond.
Nature Reviews Cancer,
6(10), 789–802.
-
Prockop, D. J.
(1997). Marrow stromal cells as stem cells for nonhematopoietic tissues.
Science,
276(5309), 71–74.
-
Rabieian, R.,
Boshtam, M.,
Zareei, M.,
Kouhpayeh, S.,
Masoudifar, A., &
Mirzaei, H.
(2017). Plasminogen activator inhibitor type-1 as a regulator of fibrosis.
Journal of Cellular Biochemistry,
18(10), 26146.
-
Rainov, N. G.,
Fels, C.,
Droege, J. W.,
Schäfer, C.,
Kramm, C. M., &
Chou, T.-C.
(2001). Temozolomide enhances herpes simplex virus thymidine kinase/ganciclovir therapy of malignant glioma.
Cancer Gene Therapy,
8(9), 662.
-
Rajab, K.,
Nelson, P.,
Keung, E., &
Conrad, C.
(2013). Suicide gene therapy against cancer.
Journal of Genetic Syndromes and Gene Therapy,
4(9), 1–8.
-
Ramasamy, R.,
Lam, E. W.,
Soeiro, I.,
Tisato, V.,
Bonnet, D., &
Dazzi, F.
(2007). Mesenchymal stem cells inhibit proliferation and apoptosis of tumor cells: Impact on in vivo tumor growth.
Leukemia,
21(2), 304–310.
-
Rani, S.,
Ryan, A. E.,
Griffin, M. D., &
Ritter, T.
(2015). Mesenchymal stem cell-derived extracellular vesicles: Toward cell-free therapeutic applications.
Molecular Therapy,
23(5), 812–823.
-
Rashidi, B.,
Hoseini, Z.,
Sahebkar, A., &
Mirzaei, H.
(2016). Anti-atherosclerotic effects of vitamins D and E in suppression of atherogenesis.
Journal of Cellular Physiology,
14(10), 25738.
-
Rashidi, B.,
Malekzadeh, M.,
Goodarzi, M.,
Masoudifar, A., &
Mirzaei, H.
(2017). Green tea and its anti-angiogenesis effects.
Biomedicine & Pharmacotherapy,
89, 949–956.
-
Reagan, M. R., &
Kaplan, D. L.
(2011). Concise review: Mesenchymal stem cell tumor-homing: Detection methods in disease model systems.
Stem Cells,
29(6), 920–927.
-
Ringe, J.,
Strassburg, S.,
Neumann, K.,
Endres, M.,
Notter, M.,
Burmester, G. R., …
Sittinger, M.
(2007). Towards in situ tissue repair: Human mesenchymal stem cells express chemokine receptors CXCR1, CXCR2 and CCR2, and migrate upon stimulation with CXCL8 but not CCL2.
Journal of Cellular Biochemistry,
101(1), 135–146.
-
Robinton, D. A., &
Daley, G. Q.
(2012). The promise of induced pluripotent stem cells in research and therapy.
Nature,
481(7381), 295–305.
-
Ryu, C. H.,
Park, K. Y.,
Kim, S. M.,
Jeong, C. H.,
Woo, J. S.,
Hou, Y., &
Jeun, S.-S.
(2012). Valproic acid enhances anti-tumor effect of mesenchymal stem cell mediated HSV-TK gene therapy in intracranial glioma.
Biochemical and Biophysical Research Communications,
421(3), 585–590.
-
Saadatpour, L.,
Fadaee, E.,
Fadaei, S.,
Nassiri Mansour, R.,
Mohammadi, M.,
Mousavi, S. M., …
Mirzaei, H.
(2016). Glioblastoma: Exosome and microRNA as novel diagnosis biomarkers.
Cancer Gene Therapy,
23(12), 415–418.
-
Sadhukha, T.,
O'Brien, T. D., &
Prabha, S.
(2014). Nano-engineered mesenchymal stem cells as targeted therapeutic carriers.
Journal of Controlled Release,
196, 243–251.
-
Salarinia, R.,
Sahebkar, A.,
Peyvandi, M.,
Mirzaei, H. R.,
Jaafari, M. R.,
Riahi, M. M., …
Mirzaei, H.
(2016). Epi-drugs and Epi-miRs: Moving beyond current cancer therapies.
Current Cancer Drug Targets,
16(9), 773–788.
-
Sale, G., &
Storb, R.
(1983). Bilateral diffuse pulmonary ectopic ossification after marrow allograft in a dog. Evidence for allotransplantation of hemopoietic and mesenchymal stem cells.
Experimental Hematology,
11(10), 961–966.
-
Sato, H.,
Kuwashima, N.,
Sakaida, T.,
Hatano, M.,
Dusak, J. E.,
Fellows-Mayle, W. K., …
Pollack, I. F.
(2005). Epidermal growth factor receptor-transfected bone marrow stromal cells exhibit enhanced migratory response and therapeutic potential against murine brain tumors.
Cancer Gene Therapy,
12(9), 757–768.
-
Satoh, T.,
Hosokawa, M.,
Atsumi, R.,
Suzuki, W.,
Hakusui, H., &
Nagai, E.
(1994). Metabolic activation of CPT-11, 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxycamptothecin, a novel antitumor agent, by carboxylesterase.
Biological and Pharmaceutical Bulletin,
17(5), 662–664.
-
Shah, K.
(2012). Mesenchymal stem cells engineered for cancer therapy.
Advanced Drug Delivery Reviews,
64(8), 739–748.
-
Siegel, R. L.,
Miller, K. D., &
Jemal, A.
(2015). Cancer statistics, 2015.
CA: A Cancer Journal for Clinicians,
65(1), 5–29.
-
Simões, S.,
Filipe, A.,
Faneca, H.,
Mano, M.,
Penacho, N.,
Düzgünes, N., &
Pedroso de Lima, M.
(2005). Cationic liposomes for gene delivery.
Expert Opinion on Drug Delivery,
2(2), 237–254.
-
Simonian, M.,
Mosallayi, M., &
Mirzaei, H.
(2017). Circulating miR-21 as novel biomarker in gastric cancer: Diagnostic and prognostic biomarker.
Journal of Cancer Research and Therapeutics, [Epub ahead of print].
-
Singh, P. K.,
Doley, J.,
Kumar, G. R.,
Sahoo, A., &
Tiwari, A. K.
(2012). Oncolytic viruses & their specific targeting to tumour cells.
Indian Journal of Medical Research,
136(4), 571.
-
Song, C., &
Li, G.
(2011). CXCR4 and matrix metalloproteinase-2 are involved in mesenchymal stromal cell homing and engraftment to tumors.
Cytotherapy,
13(5), 549–561.
-
Song, C.,
Xiang, J.,
Tang, J.,
Hirst, D. G.,
Zhou, J.,
Chan, K.-M., &
Li, G.
(2010). Thymidine kinase gene modified bone marrow mesenchymal stem cells as vehicles for antitumor therapy.
Human Gene Therapy,
22(4), 439–449.
-
Springer, C. J., &
Niculescu-Duvaz, I.
(2000). Prodrug-activating systems in suicide gene therapy.
The Journal of Clinical Investigation,
105(9), 1161–1167.
-
Studeny, M.,
Marini, F. C.,
Champlin, R. E.,
Zompetta, C.,
Fidler, I. J., &
Andreeff, M.
(2002). Bone marrow-derived mesenchymal stem cells as vehicles for interferon-β delivery into tumors.
Cancer Research,
62(13), 3603–3608.
-
Sun, B.,
Roh, K.-H.,
Park, J.-R.,
Lee, S.-R.,
Park, S.-B.,
Jung, J.-W., …
Kang, K.-S.
(2009). Therapeutic potential of mesenchymal stromal cells in a mouse breast cancer metastasis model.
Cytotherapy,
11(3), 289–298.
-
Sylvester, K. G., &
Longaker, M. T.
(2004). Stem cells: Review and update.
Archives of Surgery,
139(1), 93–99.
-
Tiraby, M.,
Cazaux, C.,
Baron, M.,
Drocourt, D.,
Reynes, J.-P., &
Tiraby, G.
(1998). Concomitant expression of E. coli cytosine deaminase and uracil phosphoribosyltransferase improves the cytotoxicity of 5-fluorocytosine.
FEMS Microbiology Letters,
167(1), 41–49.
-
Tomicic, M. T.,
Thust, R., &
Kaina, B.
(2002). Ganciclovir-induced apoptosis in HSV-1 thymidine kinase expressing cells: Critical role of DNA breaks, Bcl-2 decline and caspase-9 activation.
Oncogene,
21(14), 2141.
-
Torre, L. A.,
Bray, F.,
Siegel, R. L.,
Ferlay, J.,
Lortet-Tieulent, J., &
Jemal, A.
(2015). Global cancer statistics, 2012.
CA: A Cancer Journal for Clinicians,
65(2), 87–108.
-
Touati, W.,
Tran, T.,
Seguin, J.,
Diry, M.,
Flinois, J.-P.,
Baillou, C.,
Lescaille, G., …
Lemoine, F.
(2014). A suicide gene therapy combining the improvement of cyclophosphamide tumor cytotoxicity and the development of an anti-tumor immune response.
Current Gene Therapy,
14(3), 236–246.
-
Uchibori, R.,
Okada, T.,
Ito, T.,
Urabe, M.,
Mizukami, H.,
Kume, A., &
Ozawa, K.
(2009). Retroviral vector-producing mesenchymal stem cells for targeted suicide cancer gene therapy.
The Journal of Gene Medicine,
11(5), 373–381.
-
Uchibori, R.,
Tsukahara, T.,
Ohmine, K., &
Ozawa, K.
(2014). Cancer gene therapy using mesenchymal stem cells.
International Journal of Hematology,
99(4), 377–382.
-
Uckert, W.,
Kammertöns, T.,
Haack, K.,
Qin, Z.,
Gebert, J.,
Schendel, D., &
Blankenstein, T.
(1998). Double suicide gene (cytosine deaminase and herpes simplex virus thymidine kinase) but not single gene transfer allows reliable elimination of tumor cells in vivo.
Human Gene Therapy,
9(6), 855–865.
-
Waehler, R.,
Russell, S. J., &
Curiel, D. T.
(2007). Engineering targeted viral vectors for gene therapy.
Nature Reviews Genetics,
8(8), 573–587.
-
Weissman, I. L.
(2000). Translating stem and progenitor cell biology to the clinic: Barriers and opportunities.
Science,
287(5457), 1442–1446.
-
Wierdl, M.,
Morton, C. L.,
Weeks, J. K.,
Danks, M. K.,
Harris, L. C., &
Potter, P. M.
(2001). Sensitization of human tumor cells to CPT-11 via adenoviral-mediated delivery of a rabbit liver carboxylesterase.
Cancer Research,
61(13), 5078–5082.
-
Wu, H.-C.,
Chang, D.-K., &
Huang, C.-T.
(2006). Targeted therapy for cancer.
Journal of Cancer Molecules,
2(2), 57–66.
-
Yang, K.,
Xu, W.-G.,
Liu, Y.-Z.,
Meng, X.-R.,
Chen, P., &
Wu, L.-C.
(2014). Study on the effect of BMSCs-EGFP-tk as mediator of HSV1-tk/GCV suicide gene therapy directed against A549 in vitro.
International Journal of Clinical and Experimental Medicine,
7(9), 3080.
-
Yoon, Y.-S.,
Park, J.-S.,
Tkebuchava, T.,
Luedeman, C., &
Losordo, D. W.
(2004). Unexpected severe calcification after transplantation of bone marrow cells in acute myocardial infarction.
Circulation,
109(25), 3154–3157.
-
You, M. H.,
Kim, W. J.,
Shim, W.,
Lee, S. R.,
Lee, G.,
Choi, S., …
Han, S. U.
(2009). Cytosine deaminase-producing human mesenchymal stem cells mediate an antitumor effect in a mouse xenograft model.
Journal of Gastroenterology and Hepatology,
24(8), 1393–1400.
-
Zarogoulidis, P.,
Darwiche, K.,
Sakkas, A.,
Yarmus, L.,
Huang, H.,
Li, Q., …
Malecki, M.
(2013). Suicide gene therapy for cancer-current strategies.
Journal of Genetic Syndrome & Gene Therapy,
4. pii: 16849.
-
Zhang, T.-Y.,
Huang, B.,
Wu, H.-B.,
Wu, J.-H.,
Li, L.-M.,
Li, Y.-X., …
Tabata, Y.
(2015). Synergistic effects of co-administration of suicide gene expressing mesenchymal stem cells and prodrug-encapsulated liposome on aggressive lung melanoma metastases in mice.
Journal of Controlled Release,
209, 260–271.
-
Zhang, T.-Y.,
Huang, B.,
Yuan, Z.-Y.,
Hu, Y.-L.,
Tabata, Y., &
Gao, J.-Q.
(2014). Gene recombinant bone marrow mesenchymal stem cells as a tumor-targeted suicide gene delivery vehicle in pulmonary metastasis therapy using non-viral transfection.
Nanomedicine,
10(1), 257–267.
-
Zielske, S. P.,
Livant, D. L., &
Lawrence, T. S.
(2009). Radiation increases invasion of gene-modified mesenchymal stem cells into tumors.
International Journal of Radiation Oncology* Biology* Physics,
75(3), 843–853.
-
Zischek, C.,
Niess, H.,
Ischenko, I.,
Conrad, C.,
Huss, R.,
Jauch, K.-W., …
Bruns, C.
(2009). Targeting tumor stroma using engineered mesenchymal stem cells reduces the growth of pancreatic carcinoma.
Annals of Surgery,
250(5), 747–753.