Skip to main content

Mesenchymal stem cell-released oncolytic virus: an innovative strategy for cancer treatment

Abstract

Oncolytic viruses (OVs) infect, multiply, and finally remove tumor cells selectively, causing no damage to normal cells in the process. Because of their specific features, such as, the ability to induce immunogenic cell death and to contain curative transgenes in their genomes, OVs have attracted attention as candidates to be utilized in cooperation with immunotherapies for cancer treatment. This treatment takes advantage of most tumor cells' inherent tendency to be infected by certain OVs and both innate and adaptive immune responses are elicited by OV infection and oncolysis. OVs can also modulate tumor microenvironment and boost anti-tumor immune responses. Mesenchymal stem cells (MSC) are gathering interest as promising anti-cancer treatments with the ability to address a wide range of cancers. MSCs exhibit tumor-trophic migration characteristics, allowing them to be used as delivery vehicles for successful, targeted treatment of isolated tumors and metastatic malignancies. Preclinical and clinical research were reviewed in this study to discuss using MSC-released OVs as a novel method for the treatment of cancer.

Video Abstract

Introduction

In recent years, early diagnosis of some cancer types, along with the development of cancer-specific treatments, has led to an increase in cancer patients' survival rates [1]. However, the short half-life of several cancer-specific medications, restricted distribution to particular tumor types, and negative impacts on healthy tissues are important barriers to treatment. Actually, the primary goal of cancer treatment is to develop anticancer medications that effectively target malignant cells while preserving healthy tissue [2, 3]. A few instances of metastatic cancer have been effectively treated using traditional methods [4]. As a result, developing a novel therapeutic approach to inhibit metastasis is crucial, especially given the issues with current cancer treatment strategies, such as drug resistance and systemic side effects [5, 6].

Beyond the capacity of some viruses to mediate oncogenesis and their use in the development of immunotherapies, such as the cytomegalovirus (CMV) gliomagenesis and its implications in the development of CMV-specific adoptive T cell immunotherapies, viruses themselves can be used as therapeutic agents to target tumor cells [7]. In this way, oncolytic viruses (OVs) are defined as naturally occurring or genetically manipulated viruses that exclusively replicate and grow in tumor cells and kill them while sparing normal cells [8, 9]. Oncolytic viral therapy is a new strategy of cancer therapy that has shown promise in preclinical and clinical trials [10, 11]. Altered mutants of human viruses, wild-type animal viruses that are cytotoxic to human cancer cells, and live virus vaccines are among the viruses used in this therapy. Adenovirus, measles virus, reovirus, herpes simplex virus, vesicular stomatitis, Newcastle disease virus, vaccinia virus, and poliovirus are some of these viruses [12, 13]. OVs have the ability to directly lyse cancer cells but this is not the sole advantage of them; it is now well acknowledged that one of the most essential aspects of virotherapy is the cytotoxic immune response they can trigger or reactivate in patients, which results in therapeutic responses [14, 15] and was shown in glioblastoma, B cell malignancy, metastatic melanoma, and liver cancer [16,17,18,19,20]. Indeed, multiple investigators have reported on the possible use of OVs for cancer treatment, with a demonstration of long-term prognosis [21]. A variety of factors, including viral elimination by the immune system and viral uptake by tissues and organs, can influence viral effectiveness in reaching cancerous tissues [22, 23, 24]. To boost treatment efficacy, effective carrier vehicles are essential for delivering OVs to tumor sites. Adult stem or progenitor cells have been extracted from a variety of tissues, including the brain, heart, and kidney, and have shown promise in treating a variety of diseases [25,26,27]. In both in vitro and many murine cancer models, unmodified MSC has been demonstrated to have anti-tumor activities. This is due to antitumor substances generated by MSCs, which limit the growth of cancer cells including glioma, melanoma, lung cancer, hepatoma, and breast cancer [28,29,30,31,32,33]. Furthermore, MSCs have been utilized as carriers because of their known tumor-specific homing ability, which allows for the virus's safe transportation and releases on the tumor site [34,35,36,37]. Using MSCs might be a method to enhance the quantity of oncolytic virus given to patients while reducing side effects and avoiding direct tumor injections [38]. Altogether, in this paper we will review the features of MSCs and OVs as well as their activities against tumors, and also discuss challenges in using this strategy as an innovative cancer treatment.

The features of the MSC-based delivery of OV

As a novel cancer treatment method, virotherapy offers various benefits, including the likely absence of cross-resistance with traditional treatments and the ability to promote tumor elimination through a variety of pathways. In addition, MSCs are suitable carriers for anticancer viruses since they can home to tumor sites [39,40,41, 42], are easy to isolate and develop in vitro, and have strong metabolic activity, which is necessary for viral replication [34, 43].

MSCs as OV carriers

The majority of preclinical research has discovered efficient features for MSCs as OV carriers [44,45,46] and this strategy might be an effective way to promote oncolytic virotherapy efficiency [47,48,49,50,51,52,53,54]. Indeed, MSCs have been shown to be one of the best alternatives for OV chaperoning in the treatment of cancer due to their tumor-homing, inherent anticancer capacities, OVs preservation from neutralizing antibodies, and viral distribution to the tumor site via the Trojan horse strategy [53, 55, 56].

In a series of studies, MSC has been discovered to migrate to the area of injury, ischemia, and tumor sites via chemotaxis [57]. Although the mechanisms by which MSC migrate across the endothelium or to targeting sites remain unclear, extensive research has demonstrated that MSC migration is regulated by the cytokine/receptor pairs, such as SDF-1/CXCR4, SCF/c-Kit, HGF/c-Met, VEGF/VEGFR, PDGF/PDGFR, MCP-1/CCR2, and HMGB1/RAGE [58]. In the TME, many immune cells, as well as cancer cells, release soluble molecules that can directly influence MSC chemotaxis to injured tissues. The oxidative state, vascularization, and inflammatory condition of the tumor can all influence MSC migration efficiency at these locations [59]. It has been shown that interleukin-6 (IL-6) can promotes MSC tropism to cancer sites [60] and also this movement is IL-8-dependent in glioma [61]. Tumors can also recruit MSCs from other tissues, such as bone marrow (BM-MSCs) and adipose tissue (AD-MSCs), and promote their engraftment into the TME through inflammatory signals [62,63,64]. Local variables including hypoxia, cytokines, and Toll-like receptor (TLR) ligands stimulate recruited MSCs to multiply and express growth factors that enhance tissue regeneration at the site of injury [65]. Hepatic carcinoma [66], breast cancer [67], and glioma have all been demonstrated to attract MSCs [68].

This therapeutic strategy, in addition to offering substantial site-specificity, avoids potential issues associated with biological drug half-life limitations, as drug release might be tailored to be constant [43]. Moreover, it's hard to manage effective concentrations of anti-tumor drugs near the tumor for long periods of time [69]. For instance, in the case of brain tumors, the failure of substances to cross the blood–brain barrier is a concern. The use of MSCs as cellular delivery vehicles has been proposed as a novel approach to addressing these obstacles, allowing for a more precise and long-lasting therapeutic response than standard delivery methods would ordinarily allow [2]. Another unique aspect of employing MSCs as OV carriers is that they may function as biological manufacturers for viral genome replication, enhancing virus titer. This implies that a low initial dose of OVs for loading into MSCs is sufficient to deliver a high viral dose to tumor microenvironment. However, the specifics of OVs replication within MSCs remain unknown [70, 71]. As a result, when OVs are carried by MSCs, they leverage MSCs' natural affinity to reach tumor sites, improving OVs homing and promoting oncolysis.

Immunosuppressive activities of MSCs

Multiple studies revealed that MSCs have anti-inflammatory and immunosuppressive properties, so that, these cells generate and release a number of soluble cytokines, such as IL-6, IL-10, TGF-β1, heme oxygenase-1(HO-1), inducible nitric oxide synthase (iNOS), and indoleamine-2-dioxygenase-3(IDO) [72]. These cytokines are essential in immunosuppression and inhibiting B lymphocyte maturation and restricting their capacity to produce immunoglobulin [73,74,75], inhibiting the secretion of cytokines by helper T cells, reducing the cytotoxic actions of effector T lymphocytes [76], decreasing NK cell proliferation, cytotoxicity, and cytokine generation [77]. Furthermore, in a study on diabetic nephropathy in rats, it was shown that MSCs suppressed CD103+ DCs and CD68+ CD11c+ macrophages in the kidneys and alleviate renal injury [78,79,80,81]. They are also able to inhibit the differentiation of CD14+ monocytes and CD34+ progenitor cells into mature DCs [82], restrict DC differentiation and function [83], and thereby increase CD4 + CD25 + FOXP3+  T lymphocytes (Treg) development and generation of other regulatory immune subtypes, including CD8+ CD28−T lymphocytes [84, 85], IL-10-producing B lymphocytes [86], and IL-10-producing DCs [87]. These actions are essential MSC characteristics for inhibiting local inflammation during virotherapy and permitting the oncolytic virus to replicate and destroy cancer cells without immune constraint. [88].

Anti-tumor effects of MSCs

Several investigations have indicated the specific homing of oncolytic virus-loaded MSCs to tumor xenografts and consequent infection of tumor cells, results in diminished tumor sizes and a considerable improvement in the survival rates of treated animals [89,90,91,92,93,94,95]. MSCs enhance the proliferation of some tumor cell lines in vivo but not others. Variations in tumor types, MSC preparations, duration, and quantity of MSC delivery may cause differences in the functional role of MSCs in tumor growth [96]. Indeed, MSCs are thought to limit tumor development by interrupting the cell cycle, reducing proliferation, inhibiting the PI3K/AKT pathway, and expressing suppressor genes [97, 98]. In a breast cancer metastasis mouse model, the umbilical cord derived MSCs (UC-MSC) and AD-MSCs were administered, and it was shown that they could prevent lung metastasis and slow tumor development by cleavage of the poly (ADP-ribose) polymerase (PARP) and caspase-3, which could then trigger apoptosis [99]. Another study showed that murine bone marrow MSCs had a cytotoxic impact on the tumor in a melanoma murine model through the production of reactive oxygen species when in interact with endothelial cells located at the capillaries [100]. As a result, the tumor development was delayed and the endothelial cells undergo apoptosis. Nevertheless, the MSCs' cytotoxic effects were only apparent when they were implanted in large quantities [101]. In addition, in a mouse model of Kaposi sarcoma, human MSCs (hMSCs) administered intravenously (iv.) were found to home to carcinogenesis sites and potently decrease tumorigenesis. Cell contact via E-cadherin and Akt inhibition were essential for the suppression of the sarcoma cells proliferation [102]. MSCs have been also discovered to display anti-angiogenic properties in vitro and in melanoma murine models [101]. Moreover, AD-MSCs have both (pro- and anti-cancer) capabilities in breast [103] and prostate cancer [104].

Mechanisms of oncolytic virotherapy

Various mechanisms of action are used by oncolytic viruses. Selective replication inside tumor cells leads to a direct lytic impact on tumor cells and the development of a systemic anti-tumor immune response. Depending on the origin and kind of cancer cell, the viral vector's properties, and the interactions between the virus, TME, and host immune response, the proportional involvement of different mechanisms could differ [105]. Indeed, Oncolytic virotherapy depends on a balance of antiviral mechanisms that kill the virus and pro-immune mechanisms that detect cellular epitopes, TAAs, and neoantigens from virus-infected tumor cells [106]. In addition to their anti-tumor properties, OVs stimulate antiviral immunity against viral antigens from the resulting infection, which is a critical player during OV-based treatments due to its capability to create an advantageous microenvironment for the immune system's activity against specific cancer cell indicators [107, 108].

The two major methods by which OVs destroy tumors are direct cell death and the activation of anti-tumor immunity [109]. The first strategy employs the virus's biological life cycle; oncolytic viruses may destroy cancer cells infected with them by direct virus-induced cytotoxicity, which is regulated by a variety of cytotoxic immune activation pathways. Following cell proliferation and lysis, virions infect surrounding cells and repeat the lytic cycle, enabling the treatment to self-amplifying at the site of need [110]. This cycle repeats until the virus's replication is reduced or the number of vulnerable host cells is decreased [111]. On the other hand, OVs are one of the most well-known immunogenic cell death inducers, and they're more probably to be type II stimulators than type I stimulators [112]. Although the precise mechanisms by which oncological viruses operate are elusive, it is suspected that they function by regulating the infected cancer cell's molecular cell death machinery. The majority of cancer cells may be resistant to apoptosis [113] and according to researchers, they have several apoptosis evasion mechanisms but they may be driven to die by non-apoptotic processes. Recent studies demonstrated that OVs can induce immunogenic cell death (ICD) in cancer cells through immunogenic apoptosis, necroptosis, necrosis, autophagic cell death, and pyroptosis, which exposes calreticulin and HSPs to the cell surface and/or releases ATP, HMGB1, uric acid, and other DAMPs as well as PAMPs as danger signals, along with tumor-associated antigens (TAAs), to activate dendritic cells and elicit effective antitumor immunity [114, 115]. However, it was recently shown that dendritic cell-mediated anti-tumor immunity is compromised by cancer cells undergoing ferroptosis [116]. To respond to the tumor antigens, antigen-presenting cells (APCs) catch TAAs and neoantigens released by tumor cells, and then activate tumor-specific T lymphocytes [117, 118]. On the surface of tumor cells infected with OVs, there are virus-specific antigens, which help in their elimination by antiviral T lymphocytes [119]. As a result, even if the virus does not reproduce well, OVs can trigger an antitumor immune response [106]. Additionally, the breakdown of the tumor's immunological tolerance is acknowledged as a major feature of OV's method of action and can destroy the tumor [120, 121].

Additionally, the OV will infect tumor cells and control protein production, increasing viral macromolecule creation while also stimulating the expression and detection of danger signals. Indeed, these are the results of some signaling pathways that end in the release of DAMPs such as heat shock proteins (HSPs), calreticulin, uric acid, and ATP and cytokines including, interferons (IFNs), tumor necrosis factor-α (TNF-α) and IL-12, all of which help to improve immune responses [105, 106, 122, 123]. Pathogen-associated molecular patterns (PAMPs), such as nucleic acids, proteins, and viral capsid elements, are also released as a result of virus-induced tumor cell death [124, 125, 126, 127]. These compounds help counteract the immunosuppressive condition of the TME by promoting the migration and activation of macrophages, NK, DC, and tumor-specific cytotoxic T cells [128,129,130]. The TME is made up of tumor cells, local or penetrated non-transformed cells (e.g., cancer-associated fibroblasts, vascular endothelial cells, immune recruitment cells), secretory substances, and the extracellular matrix. In fact, this microenvironment is generally immunosuppressive and tumors produce soluble immunosuppressive agents such as nitric oxide and cytokines including IL-10 and TGF-β, resulting in active suppression of efficient anti-tumoral immune response [106, 131, 132]. Furthermore, Tregs and myeloid-derived suppressor cells (MDSCs) are directed to the TME, where they use the acquired immune response pathway's potential to detect and eliminate tumor cells [106, 132, 133].

The immunostimulatory versus immunosuppressive behavior of the TME is controlled by cytokines and immune cells [107]. OVs enable proinflammatory cytokines to enter the TME, establishing a suitable condition for DC activation. By activating antigen presentation pathways in tumors, OVs stimulate DCs to detect tumor antigens [134, 135]. In this environment, OVs can outperform various evasion methods. For example, by treating with oncolytic reovirus, an ovarian cancer cell line expressed more MHC class I and other molecules related to antigen processing, such as the transporter associated with antigen processing (TAP) and β2-microglobulin (β2M). This action enhanced DC maturation, which resulted in adaptive immunological responses driven by CD8 T cells [136]. Moreover, in a murine model treated with an engineered adenovirus, many splenic CD11c CD8 DCs were found an tumor-infiltrating plasmacytoid DCs revealed a mature phenotype capable of priming tumor-specific cytotoxic T cell activities [137]. Furthermore, multiple studies have shown that other OVs, including vaccinia virus [138], measles virus [139], and HSV [140], can improve DC antigen presentation, which is commonly associated with enhanced expression of costimulatory/activation components such CD80, CD86, and MHC II.

The tumor-associated macrophage population is an important modulator of the immunostimulatory versus immunosuppressive behavior of the TME. Anti-viral and anti-cancer responses are related to M1 pro-inflammatory macrophages, whereas metastasis, angiogenesis, and inhibition of anti-cancer and anti-viral responses are related to M2 immunosuppressive macrophages [141,142,143]. As shown by oncolytic paramyxovirus infection of macrophages, OVs act as potent immunological triggers and are useful to modify the phenotypic activities of macrophages [144]. OVs are able to provide an inflammatory environment that encourages macrophage infiltration and activation. For instance, in a xenograft colorectal cancer model, it was shown that treatment with the oncolytic vaccinia virus GLV-1h68 caused a considerable increase in proinflammatory cytokines like IL-3, IL-6, IFN-, and CXCL10. It causes boosting the recruitment of proinflammatory macrophages to the tumor site [145]. Similar to this, a triple combined treatment including oncolytic HSV boosted macrophage recruitment and M1-like polarization, which helped to eliminate glioblastoma [146].

The typical antiviral response from healthy cells, which can limit OV reproduction and disseminate directly, is one of the earliest defenses to OVs. Type I IFN is one of the key drivers of this activity (IFN-a and IFN-b) [141, 142, 147, 148]. Type I IFN plays a significant part in anti-cancer responses by triggering immune cells inside the TME, including NK cells and CD8+ T cells, and pro-inflammatory cytokines, in addition to regulating the anti-viral condition [148, 149]. Because of its regulatory impact on NK cells and CD8+ T cells, Type I IFN enhances anti-tumor immune responses. Activated NK cells release type II interferon (IFN-γ), which suppresses angiogenesis, promotes apoptosis, and stimulates the immune system (by activating MHC class II in DCs, macrophage phagocytic activity, and CD8+ T cell responses) [107]. Type I IFN can also increase MHC class I expression in DCs, as well as co-stimulatory molecules (CD40, CD86) and the Th1 polarized response [141, 142] (Fig. 1). Furthermore, the capability of OV-infected tumor cells to produce type I IFN in each tumor site, as well as the potency by which certain OVs stimulate type I IFN signaling, varies significantly among OVs, and these facts all influence the OV's potential to activate the acquired immune system against viral and cancer antigens [142]. To achieve the appropriate balance between anti-viral and anti-cancer immunity, more research into the impact of each OV is required.

Fig. 1
figure 1

Mesenchymal stem cell-based delivery of oncolytic virus challenges. MSCs might stimulate angiogenesis, tumor cell proliferation, and metastasis by release large numbers of cytokines and growth factors, including VEGF, FGF-2, βFGF, PDGF, IL-8, IL-6, CXCL1, CCL5, and SDF-1. MET during metastasis tighten epithelial connections and make therapy challenging. Hypoxic circumstances have been observed to decrease viral proliferation and lytic capacity. Type I IFN hinder intra-tumoral spread of the OVs, moreover, infection with double-stranded RNA leads to PKR activation in the cell. Excessive viral replication may result in premature MSC lysis and reduce the efficiency. VEGF vascular endothelial growth factor, FGF-2 fibroblast growth factor 2, PDGF platelet-derived growth factor, IL interleukin, CXCL c-x-c motif chemokine ligand 1, CCL5 c–c motif chemokine ligand, SDF-1 stromal cell-derived factor 1, MET mesenchymal-to-epithelial transitions, IFN I type I interferon, PKR RNA-dependent protein kinase, eIF2α eukaryotic initiation factor 2 α

Angiogenesis is a cancer characteristic that involves the supply of nutrients and oxygen to tumor cells to enhance tumorigenesis [150, 151]. Several OVs have been demonstrated to have anti-angiogenic properties by inducing an acute disturbance of the tumor vasculature [152,153,154]. For instance, it has been found that the oncolytic vaccinia virus inhibits tumor angiogenesis, limits blood supply to tumor cells, and ultimately leads to hypoxia through impacting vascular cells [155, 156,157,158]. In addition, OVs can destroy uninfected cancer cells by damaging tumor blood vessels and enhancing particular antitumor immune responses. The transgene-encoded proteins produced by modified viruses can assist oncolytic viruses in killing uninfected cancer cells [159].

MSC-based delivery of oncolytic adenovirus (oAds)

Since the oAds replication could be limited to malignant cells, these viruses are being evaluated in clinical trials for the treatment of several malignancies [160,161,162]. (Table 1) In fact, after the oAds replicates in tumor cells, the cell is lysed and more infectious virions are produced, infecting and lysing adjacent tumor cells and consequently inducing endogenous tumor immune responses which has therapeutic benefits [163]. It has been revealed that MSCs are finally lysed by oAd replication which avoids any negative side effects related to stem cell viability in vivo [59, 164]. In addition, multiple investigations of individuals with ovarian cancer, melanoma, soft tissue or primary bone sarcoma, and other neoplasms have revealed that oAds have a high safety profile [10, 160, 161, 162, 165,166,167,168,169]. For instance, Garcia-Castro and colleagues demonstrated the safety and effectiveness of delivering numerous doses of autologous MSCs infected with ICOVIR-5, which is an optimized oncolytic adenovirus, to four children with metastatic neuroblastoma who had failed to respond to standard therapy [170]. An in vivo study also revealed that infected MSCs transport the combination of ICOVIR15 and Ad.iC9 to lung tumor sites effectively and specifically increase overall survival and tumor control [50]. Furthermore, in Stoff Khalili et al. research, hMSCs were utilized as intermediate vehicles for conditional replication oncolytic adenoviruses (CRAds) to target breast cancer and reduced the development of pulmonary metastasis, most likely due to viral replication in the hMSCs [90]. Pulmonary metastasis in patients with breast cancer is common characteristic [171, 172, 173] and is associated with lethal complications in these patients [174, 175]. Furthermore, when CRAd-loaded MSC was given intravenously to mice with solid ovarian cancer, it had a considerably stronger anticancer impact and a prolonged survival time than CRAd delivered directly [89]. Moreover, it has been shown that hepatocellular carcinoma (HCC)-targeted oAd can be loaded into MSCs successfully by viral capsid modification, and oAds induce significant cancer-specific death pathways through active viral reproduction in the MSC driver. Indeed, oAd-loaded MSCs improve both oAd and MSC safety features by reducing oAd hepatic sequestration and hepatotoxicity while improving MSC clearance through viral proliferation [59]. Also, Rincon et al. revealed that MSCs carrying the oncolytic adenovirus ICOVIR5 were therapeutically effective in treating lung cancer in mice by inhibiting tumor development and encouraging T cell migration to the tumors [176]. Similar to this, MSCs infected with the oncolytic adenovirus CRAd5/F11 prevented tumor growth in a colorectal cancer subcutaneous mouse xenograft model [177]. In the other study Kaczorowski et al. [178] removed the antiapoptotic gene E1B19K from oAd and replaced it with the cell death ligand TRAIL gene. After intravenous injection of infected MSCs, adenoviral capsid protein was found in tumor xenograft tissue but not in healthy tissue, indicating tumor-specific migration [178]. Similarly, direct in vivo therapy was associated with a significantly decreased tumor size, lower Ki67 and CD24 expression, and increased caspase activation [160]. Chastkofsky et al. evaluated the potential of MSC for OV delivery for the treatment of diffuse intrinsic pontine glioma (DIPG. They utilized the survivin promoter for conditional replication of OV. Their results revealed that cells and tumors have increased expression of survivin and cell surface proteins which provide effective OV entry and replication in DIPG cells and result in more prolonged survival [179]. To elaborate, survivin is an anti-apoptotic factor that facilitates the viability and survival of cells and has been identified as a target in the treatment of several autoimmune diseases and cancers [180, 181].

Table 1 Mesenchymal stem cell-released oncolytic virus for cancer treatment

In a mouse model of breast cancer with pulmonary metastases, Zhang et al. investigated the therapeutic effects of MSCs infused with OAd expressing decorin, a naturally occurring inhibitor of TGF-β signaling. They demonstrated that MSCs boosted the therapeutic benefits of oncolytic adenoviral administration and dissemination in tumor tissues [182]. Since the oncolytic adenovirus replicates in MSCs, it is essential to balance MSC viability with viral load in order to get the best therapeutic outcome. Recently, Zhang et al. developed an all-in-one Tet-on system that capable of controlling the reproduction of OAd. The new OAd expressing Endostatin and/or IL-24 was then introduced into hUCB-MSCs for the treatment of glioma. Their findings showed that this new OAd was capable of killing glioma cells with high efficiency while sparing healthy cells [183].

Interestingly, Barlabé et al. equipped the OAdv with a therapeutic transgene to provide the strongest anticancer effects. They demonstrated that, when menstrual blood-derived mesenchymal stem cells (MenSCs) are combined with ICOVIR15-cBiTE, an OAdv producing an EGFR-targeting bispecific T-cell engager (cBiTE), the antitumor effectiveness is increased in comparison to MenSCs loaded with the unarmed virus ICOVIR15 [194]. Combining MSC-delivered OV with prodrug activation is another strategy to achieve optimum effectiveness. Under these circumstances, MSCs might convert the simultaneously delivered prodrug into cytotoxic metabolites, causing oncolysis and inhibiting tumor development without being hazardous to the host's essential organs [195].

Adoptive cell treatments for solid tumors have a significant challenge due to the immunosuppressive TME [196]. Oncolytic immunotherapy using modified OAd by infecting tumor cells, may disrupt the TME [197]. It was recently revealed that lung cancers could be successfully treated in animal models using a combination of cell carrier-delivered OAd and chimeric antigen receptor (CAR-T) cells. A binary vector including an OAd and a helper-dependent Ad (HDAd; combinatorial Ad vector (Cad)) that expresses checkpoint PD-L1 blocker and IL-12 has been demonstrated by this work to be systemically delivered by MSCs [198]. The immune checkpoint ligand PD-L1 has been found to be overexpressed in a variety of solid tumors [199]. PD-L1 binds to its receptor PD-1, and blocking it has shown therapeutic potential in cancer therapy [200]. However, when PD-L1 blockade is combined with optimal dose IL-12 delivery, it induces a synergistic effect of enhancing anti-tumor immunity in cancer patients [201].

MSC-based delivery of oncolytic herpes simplex virus (oHSV)

The oHSV has been evaluated extensively in combination with MSCs and has demonstrated promising outcomes in the treatment of gliomas, metastatic melanomas, breast, and ovarian cancers, whether administered systemic [49, 184] or locally [185]. For example, in order to investigate efficiency of oHSV in immune-deficient and immune-competent murine models of melanoma brain metastasis, Du et al. utilized MSCs as cellular vehicles for oHSV and revealed that MSC-oHSV effectively detects metastatic tumor deposits in the brain, inhibits brain tumor development, and extends survival [184]. Furthermore, in a clinically relevant glioma model, Duebgen M, et al. infused hMSCs with oHSV and revealed that they successfully generated oHSV progeny, cause tumor resection and resistance and extended average lifespan in mice [185]. Indeed, one of the most important alternatives for GBM treatment is the oHSV, which is a naturally neurotrophic factor [8, 202, 203]. In addition, it has been shown that MSCs from various origins can be infected and loaded with a HER2-retargeted oncolytic HSV and the metastatic burden in the brain was shown to be reduced by more than half in NSG mice, which showed significant suppression of breast cancer brain metastases [49] (Table 1).

MSC-based delivery of oncolytic measles virus (oMV)

The oMV exhibits significant anticancer potential and is being studied as an innovative tumor therapy in a number of Phase I clinical studies [204, 205]. Indeed, oMV reproduction, protein expression, syncytia formation, and oncolysis have been reported in vivo, in a variety of human tumor xenografts, such as hematologic malignancies like lymphoma [206, 207] and myeloma [208], as well as solid tumors including ovarian cancer [209], glioblastoma [210], hepatocellular carcinoma [211], prostate cancer [212, 213], breast cancer [214], cervical cancer [215], and gastric cancer [216, 217]. On the other hand, MSCs have been demonstrated to be efficient carriers of attenuated oMV to ovarian tumors [94] and HCC [95]. For instance, in the hepatocellular carcinoma and murine model, two groups investigated intravenous single delivery of oMV-loaded BM-MSCs. Both measles antibody-naive and passively-immunized SCID mice showed a considerable decrease in tumor development when treated with oMV-infected BM-hMSCs [95]. Moreover, in an orthotopic ovarian cancer therapy model, treated mice with AD-MSCs as drivers of oMV survived longer in comparison to mice treated with a naked virus or uninfected MSC [53]. In addition, in a xenograft model, BM-hMSCs were shown to successfully delivering OMVs to precursor B-lineage acute lymphoblastic leukaemia (ALL) cells. Ex vivo loading of oMV into BM-MSCs was effective and oMV was replicated intracellularly without toxicity [186].

Early research [208, 210, 212, 218, 219, 220] reported an increase in apoptotic markers in infected cells, whereas Lampe et al. demonstrated viral cells die even when apoptosis inhibitors are used, indicating that alternative cell death pathways are involved [221]. Donnelly, OG et al. revealed that Measles virus causes immunogenic cell death (ICD) in human melanoma[222].

MSC-based delivery of oncolytic myxoma virus (MYXY)

In order to cure a malignant brain tumor in mice, human adipose-derived MSCs (AD-MSCs) have been coupled with MYXV, which expresses the reporter gene green fluorescent protein (GFP) [187]. This study established the potential of AD-MSCs to produce new viral particles as well as the ability of infected cells to adhere to tumors when administered intravenously. As a consequence, mice treated with the OV-loaded MSCs had a much higher survival rate than mice treated with MSCs alone.

BM-MSCs have also shown to be permissive to MYXV replication [188]. Furthermore, the scientists showed improved antitumor activity in an immunological competent lung melanoma model following intravenous treatment, in comparison to MYXV monotherapy, employing an IL-15-armed MYXV to infect BM-MSCs. Only animals treated with the virus without cell carriers showed an enhanced proportion of circulating NK cells, suggesting that MSCs may have inhibited the immune system from recognizing the infection. Finally, tumors from animals treated with MYXV-IL-15-loaded MSCs showed an increase in pro-inflammatory cytokines, programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1), and infiltration of effector T cells, suggesting a potential antitumor immune response as a result of the therapy.

In a separate investigation, recombinant MYXV, which encodes murine LIGHT, also known as tumor necrosis factor ligand superfamily member 14 (TNFSF14), was used to infect AD-MSCs ex vivo. Results from this study showed that mice with orthotopically induced pancreatic ductal adenocarcinoma (PDAC) had increased trafficking into the pancreas compared to tumor-free animals, which led to the extended survival of the treated PDAC-seeded animals and the increased expression of important adaptive immune response markers. Administered IP and pre-loaded ADSCs with transgene-armed MYXV really enable more efficient oncolytic virus ferrying to PDAC sites and promote better tumor regression [189]. Additionally, it has been shown that combining the pre-loaded LIGHT (TNFSF14)-Armed Myxoma virus with Gemcitabine (as an antimetabolite) may be a potential strategy to enhance the therapeutic benefits of vMyx-LIGHT/ADSCs against PDAC in vivo [190] (Table 1).

MSC-based delivery of oncolytic Reovirus

A wide range of human malignancies, including acute myeloid leukemia, have been treated with reovirus, a naturally occurring OV [223]. The potential use of an oncolytic reovirus created by Reolysin®, (pelareorep; wild-type reovirus; Serotype 3 Dearing; Oncolytics Biotech Inc.), for the treatment of various tumor cells was examined in several clinical trials [224]. Reolysin®, a novel systemically administered promising anti-cancer drug for pancreatic, ovarian, and malignant glioma tumors, received FDA approval in 2015 [225, 226]. The oncolytic reovirus's anti-cancer effect against the glioblastoma multiforme (GBM) cell line may be enhanced by AD-MSCs, which are a vulnerable host for the virus [227]. In vitro cancer treatment using MSCs that have been infected with reovirus type-3 Dearing (T3D) has also been examined. According to the study's findings, reovirus-infected AD-MSCs in TC-1 cells produce more NO and TNF-α than normal while producing less TGF-β1 and IL-10. Additionally, TC-1 cells were co-cultured with infected AD-MSCs, which greatly enhanced apoptosis when compared to the control [192].

In a different study, Babaei et al. examined the anti-tumor potential of adipose-derived mesenchymal stem cells (AD-MSCs) as a novel delivery system for the Dearing strain of reovirus (ReoT3D), which has the highest capacity to eradicate cancer cells among other strains in a murine model of colorectal cancer. They found that ReoT3D and MSCs together were more effective for therapy than ReoT3D and MSCs alone [191]. Wang et al. demonstrated that Human UC-MSCs harboring reovirus display antitumor effectiveness impacts for AML in the presence of Nabs via increasing CXCL10 production from hUC-MSCs [193] (Table 1).

Clinical studies using MSC-OV

Only a few clinical trials have used MSC-OV to treat cancer patients (Table 2). Autologous MSCs loaded with oAd (ICOVIR) or CELYVIR were employed in the first-in-human trial for the treatment of pediatric refractory metastatic neuroblastoma (NCT01844661) [38].

Table 2 Clinical trials on the MSC-OV utilization in cancer patients

Another phase I/II clinical trial examined the adverse effects and optimal dosage of MSCs infected with the oncolytic measles virus that encodes the thyroidal sodium iodide symporter (MV-NIS) and how effectively it treats patients with recurrent ovarian, primary peritoneal, or fallopian tube cancer (NCT02068794). Also, another phase I trial, evaluated the optimum dose and side effects of the oncolytic adenovirus DNX-2401-loaded hBM-MSCs in treating patients with recurrent high-grade glioma through intra-arterial administration (NCT03896568). Also, the safety and efficacy parameters of AloCELYVIR in Metastatic uveal melanoma patients with hepatic metastases were examined in phase I/II clinical trial (NCT05047276) (Table 2).

Challenges

Despite progress in our understanding of MSC-based OV delivery, there are still significant challenges ahead, which raises questions and concerns that are debatable and scientists have proposed a number of solutions to these problems, which are detailed below (Fig. 2).

Fig. 2
figure 2

MSCs feature as OVs carriers and mechanisms of MSC-released OVs in cancer treatment. OVs are maintained by MSCs from immune system responses. MSCs migrate to the tumor site via chemotaxis. There are two major methods by which OVs destroy tumors are direct cell death and the activation of anti-tumor immunity. Tumor cells secret and release DAMPs such as HSPs, calreticulin, uric acid, and ATP and cytokines including, IFNs, TNF-α and IL-12, and PAMPs, such as nucleic acids, proteins, and viral capsid elements as a result of OVs infection and oncolysis. These compounds help counteract the immunosuppressive condition of the TME by promoting the migration and activation of MQs, NK cells, DCs, and tumor-specific cytotoxic T cells. Normal cells antiviral response also includes type I IFN which can play a significant part in anti-cancer responses by triggering immune cells inside the TME. DAMPs damage-associated molecular patterns, HSPs heat shock proteins, ATP adenosine triphosphate, IFNs interferons, TNF-α tumor necrosis factor-α, PAMPs pathogen-associated molecular patterns, MQs macrophages, NK cells natural killer cells, DCs dendritic cells, TME tumor microenvironment

MSC tumorigenesis

The interaction of tumor cells with healthy cells and the stroma in the TME is getting important since these interactions have a role in critical stages of tumor progression including angiogenesis, immunomodulatory, metastasis, invasion, and apoptotic resistance [113, 228, 229]. Moreover, reports have argued that MSCs enhance or suppress tumor growth and metastasis through a variety of mechanisms, including the secretion of soluble molecules that trigger or repress innate and adaptive immune responses, activate or reduce angiogenesis, and sustain the cancer stem cell environment [230, 231, 232]. Indeed, MSCs can alter the rate of tumorigenesis depending on the circumstances [36, 233]. According to studies, MSCs release trophic factors that can boost tissue angiogenesis, cell proliferation, and cell survival [234,235,236]. For example, large numbers of cytokines and growth factors that stimulate angiogenesis, including VEGF, FGF-2, βFGF, PDGF, IL-8, IL-6, angiopoietin, and TGF, are released by MSCs and contribute to the development of tumor angiogenesis [237,238,239]. Additionally, cancer-associated fibroblasts (CAFs) actively encourage tumor angiogenesis by producing chemokines and cytokines such as IL-4, IL-8, IL-6, TNF, CXCL12, TGF, and VEGF which have anti-inflammatory and pro-angiogenic properties [240]. CAFs differentiation is enhanced by interactions between MSC and tumor cells [237, 241, 242]. Furthermore, MSCs stimulate epithelial-mesenchymal transition (EMT) by secreting growth factors and cytokines such as HGF, PDGF, EGF, and TGF, which cause the production of transcriptional regulators of EMT like Slug, Snail, Zeb1, and Twist [243, 244]. Evidence suggests that abnormal EMT enhances tumor metastasis, drug resistance, and tumor growth [245]. The epithelial cell-related proteins E-cadherin, ZO-1, and -catenin/plakoglobin are downregulated during the EMT while mesenchymal proteins including fibronectin, N-cadherin, smooth muscle actin, and vimentin are increased [71, 246]. In addition through the release of chemokines such as CXCL1, CCL5, CXCL5, CXCL8, and CXCL7 by MSCs, tumor cell migration to metastatic sites is accelerated [247, 248]. It has been also shown that MSCs release large amounts of CXCL12 (SDF-1), which control the invasion and migration of tumor cells that express CXCR4 [249, 250]. MSCs polarization in response to substances released by the tumor, is another explanation for the conflicting results, which either forces the cells into a tumor-promoting or suppressive action. Accordingly, an in vitro co-culture of MSC1 with several cancer cell lines reduced the growth of tumors, while the MSC2 co-culture had a contradictory outcome. Similarly, MSC1 treatment of tumors established in immune-competent mice reduced tumor growth and metastasis, whereas MSC2 treatment promoted tumor development and dissemination [159]. Indeed, inhibiting or activating certain MSC TLRs might be a promising approach for enhancing the anticancer effects of OV oncolysis by adding MSC immune-stimulatory features.

As a result, a greater understanding of the particular molecular processes behind these pro-tumorigenic actions is essential for further improving anti-cancer therapeutic approaches. Enhancing the therapeutic effects for cancer patients would be achievable through the reduction of MSC recruitment into tumor areas and the suppression of their tumor-supportive actions, particularly with the combination of other therapeutic methods, such as immunotherapy.

MSC lysis

However, while viral replication within MSCs is a desirable feature, excessive viral replication may result in premature MSC lysis and reduce the overall efficiency [59]. It has been demonstrated that a large initial viral dosage reduces total MSC survival with just a little increase in overall viral production [59].

Antiviral responses

Patients’ antiviral immune responses are a major concern, limiting the impact of oncolytic viruses [251, 252]. The patient's antiviral defense is triggered and recruited to limit virus reproduction and dissemination, resulting in viral elimination and the treatment impact being lost [106]. Antiviral cytokines, including various forms of IFN, are a barrier to an efficient anti-tumor response to OV because hinder intra-tumoral spread of the OVs [253]. Several studies have utilized histone deacetylase (HDAC) inhibitors to promote epigenetic changes and reduce antiviral cytokine responses in the TME to resolve this challenge [254,255,256]. Furthermore, infection with double-stranded RNA leads to PKR activation in the cell. PKR can phosphorylate the α-subunit of eukaryotic initiation factor 2 α (eIF2α) which keeps it inactive and prevents viral protein production [257, 258]. However, in vitro and in vivo studies have shown that Sunitinib inhibits the antiviral enzymes RNase L and PKR, which impedes antiviral innate immune responses [258].

OV optimizing

When determining the ideal OV treatment method, inherent qualities should be regarded. Each OV family will have its genomic complexity, replication methods, lytic qualities, transgene packaging capacity, and immune response-inducing ability to activate anti-tumor immunity. Since different OVs have distinct tumor tropisms, it has been difficult to identify precise molecular biomarkers that predict particular anti-tumor efficacy for each OV [133, 259]. Moreover, optimizing the initial OVs loading dosage is a critical factor that improves loading efficacy and affects treatment effectiveness [260]. Additionally, the oncolytic virus lifecycle's time is an essential factor in the carrier cells' tumor-homing potential. Indeed, before the viral progeny is released, the delivery cell must concentrate in tumor sites to provide effective delivery of the therapeutic virus [261].

Targeting methods

MSCs can be targeted using a variety of methods, such as physical, physiological, and biological ones that attempt to increase their density in a specific area [262]. Physical targeting is inserting cells directly into the area that requires therapy via surgical methods or guiding techniques like catheters or external magnets [263, 264, 265]. In addition, a different approach is to contain therapeutic cells in a matrix or devices that maintain cells in the transplant area [262]. For example, it has been demonstrated that MSC encapsulation in a biodegradable, synthetic extracellular matrix dramatically boosted their survival in the GBM excision cavity while permitting the production of anti-cancer proteins [185, 266, 267]. Another method employs physiological mechanisms like the systemic circulation to transfer the cells rather than active cell-mediated migration [262]. For instance, cells frequently become caught in the lungs' capillaries. To distribute MSC-mediated treatments to the lungs, this effect can be used [54, 90]. The MSCs trapping in the lung seems to be dependent on several factors, such as administration route and vessel size. Intravenous administration of MSCs results in an accumulation of cells in the lungs, which are then redistributed to the kidneys, spleen, and liver. Arterial injection bypasses the lungs, so MSCs are widely distributed throughout the rest of the body. There is insufficient systemic biodistribution during intramuscular, intraarticular and intradermal administration [268, 269]. In spite of the route of injection of MSCs [66], the size of the vessels are also play a key role in MSCs trapping in lung. Scherpfer et al. showed that the average size of MSCs is larger than the size of pulmonary capillaries. Therefore, large amounts of administered MSCs can become trapped in the capillaries of the lung and prevent access to other organs. In addition, vasodilator could be reduced lung localization [270].

Furthermore, biological targeting techniques have been developed to satisfy the demand for greater target stringency following systemic administration of MSCs, particularly when the disease to be treated is extensive, as in metastases [271, 272]. It includes evidence-based techniques aiming at enhancing MSC homing, binding selectivity to a target tissue, and persistence within the target environment [262]. Indeed, to regulate MSC homing potential, various approaches have been established, including altering the MSC culture conditions to promote the production of homing-related compounds, redesigning the cell membrane to increase homing, and adjusting the target tissue to better attract MSCs [273].

OV penetration

Epithelial junctions function as an obstacle to the intracellular infiltration of OVs, particularly adenoviruses, in carcinomas [274]. Indeed, phenotype changes during metastasis, including EMT and later mesenchymal-to-epithelial transitions (MET), which tighten epithelial connections and make therapy challenging [275, 276]. Yumul et al. created epithelial junction openers (JO) by modifying Ad5Δ24. They found that oncolytic Ads that express JO had a substantially higher anti-tumor activity than unmodified viruses [274]. Moreover, the extracellular matrix (ECM) and cellular connections are significant barriers that are related to the spread and penetration of OVs. In fact, OVs must cross the complex ECM to reach tumor cells and lysis them [277]. Pre-treatment of cancer with collagenase [278] or co-administration of hyaluronidase with oncolytic adenoviruses [279] resulted in increased viral dissemination. Additionally, altering OVs to express matrix metalloproteinases-1 and -8 causes cancer-associated sulfated glycosaminoglycans to be degraded, resulting in improved viral dispersion and treatment efficiency [280]. Tumor cell apoptosis also promotes viral dissemination. For instance, Nagano et al. found that cytotoxic substances induced apoptosis and activated caspase-8, resulting in greater intratumoral uptake and anti-cancer effect of oncolytic HSV. They hypothesized that reducing or eliminating apoptotic tumor cells resulted in channel-like structures and empty areas, enabling oncolytic HSV to disseminate more easily [281].

Hypoxic effect

Hypoxia is a characteristic of solid tumors that emerges throughout the formation and development of the tumor and has been demonstrated to have paradoxical impacts on OVs [282]. Hypoxic circumstances have been observed to decrease viral proliferation and lytic capacity without changing the expression of surface receptors [283, 284]. Because hypoxia may cause cell cycle arrest, this feature might influence the capability of oADs and other viruses that rely on cell cycle advancement to reproduce [284]. Clarke et al. created an oncolytic adenovirus in which the expression of the E1A gene is regulated by the hypoxia-response factor-containing promoter to counteract hypoxic suppression of viral reproduction and to get the benefits of hypoxic conditions for homing [285]. However, in 2009, two groups revealed that a hypoxic condition increases oncolytic HSV viral proliferation [286, 287]. This might be due to HSV's intrinsic affinity to low-oxygen cells or DNA damage caused by oxygen-derived free radicals, which promotes HSV reproduction [286].

Treatment durability

Tumors frequently recur after great initial treatment results. Stem cell treatment using a single substance, like other chemotherapies, isn't always successful in removing tumors [288, 289]. As a result, a reasonable medication combination should be determined [290]. Many different combination therapies have been tried to see whether they can help with treatment persistence. For instance, irradiating cancer cells leads them to release molecules that promote MSC penetration across integral basement membranes, resulting in an increase in the amount of MSCs in cancers [291].

Modification

In addition to their inherent potential to lyse cancer cells, OVs can be modified to improve their lytic activity. For example, adenoviruses expressing the herpes simplex virus-1 thymidine kinase (HSV-1 TK) under the osteocalcin promoter have been designed to target bone cancers. HSV-1 TK could convert thymidine analogs, such as ganciclovir into monophosphates, which stop DNA synthesis and trigger cell death By incorporating into the DNA of reproducing cells [292, 293, 294]. OVs have been modified to improve immune responses even further. Most transgenes are designed to induce an adaptive immune response against cancer antigens or to contribute to the treatment of immune cell-depleted malignancies. Including, cytokines, chemokines, inhibitory receptors, co-stimulatory receptors, bispecific cell engagers, immunological ligands, and combinations of any of these [133, 295, 296]. For instance, researchers have engineered oncolytic viruses which can express IL-2, IL-12, IL-15, IL-6, IL-21, IL-18, IL-24, and granulocyte–macrophage colony-stimulating factor (GM-CSF) and activate various aspects of the immune system [295, 296]. Moreover, the immunosuppressive TME might be altered by inserting an immune stimulatory chemical into OV genomes. The most often utilized example is GM-CSF, which has been inserted into OV genomes as an immune stimulatory molecule to promote the maturation and recruitment of APCs, particularly DCs, as well as the recruitment of tumor antigen-specific T cells and NK cells [109]. On the other hand, one aspect of transgenic-armed OVs is that immune activation can be delayed depending on the viral promoter that regulates the transgene or by controlling protein translation. To avoid an overly-rapid immune response, the expression of transgenes should be postponed until the viral oncolysis is at its maximum [297]. Additionally, the kind of transgenic and the number of transgenes that may be included in a single viral construct are both influenced by the type of virus. Unlike DNA viruses, which can handle more transgenes without harming replication, RNA viruses generally have a shorter genome and can only encode a restricted number of them [133]. Furthermore, a modified oncolytic adenovirus expressing the TRAIL gene was recently utilized to treat a mouse model of pancreatic ductal adenocarcinoma (PDAC), a malignant and lethal malignancy with a poor prognosis and few treatment options. The study revealed that in a PDAC animal model, AD-MSCs carrying TRAIL specifically homed to the cancer site and significantly slowed tumor growth, with no toxicity or adverse effects [178].

MSCs can be also genetically manipulated or preconditioned to increase their intrinsic features, such as improved migration, adhesion, and survival, as well as reduced premature aging. For example, to improve MSC migration, CXCR1, 4, and 7 were overexpressed which CXCR1 binds to IL-8 and CXCR4 and CXCR7 bind to SDF-1. Also for increasing MSC adhesion ability, MSCs were genetically engineered to express higher levels of integrin-linked kinase (ILK) [298].

Conclusion and prospect

MSCs improve the anticancer efficacy of virotherapy in a variety of ways. Indeed, MSCs act as a reproduction site for OVs, allowing for the generation of more virions, which is advantageous for virotherapy. In addition, MSCs' tumor tropism and immunosuppressive activity enable the virus to specifically target the cancer site, increasing viral spread, and survival. MSCs, on the other hand, generate cytokines that attract immune cells to the TME, increasing the anticancer immune response. Moreover, oncolysis triggers the production of danger signal including TAAs and DAMPs/PAMPs, which stimulate local anticancer immune responses and alter the TME from immunosuppressive to immunostimulatory [45, 51].

Integrating MSCs with more effective OVs is a reasonable move towards enhancing therapeutic outcomes. Currently, there are four ongoing clinical trials using the OV-loaded MSCs for cancer therapy, which offer up a wide range of combinations with MSCs [299].

Altogether, further development of MSCs-OVs therapies may rely on a multifaceted strategy to select design parameters to improve the safety profile and efficacy of carrier cells, improve viral replication in MSCs, and establish patient eligibility criteria. For overcoming these obstacles some efforts have performed. For example, by manipulating the MSCs, it is feasible to enhance the clinical result. Polymers or other viral capsids might potentially be used to improve infectivity and viral replication [300, 301]. Also to control the adenovirus’s replication within MSCs, an all-in-one Tet-on system has been developed, which could help future studies to reach the optimum therapeutic effect of the oncolytic virus [183].

To summarize, although existing clinical trials will help to clarify the therapeutic efficacy of MSCs as OV cell carriers, further efforts should be undertaken to translate current viral and cellular preclinical achievements to the clinic, either as monotherapy or in combination with radiation, chemotherapy, or even immunotherapies.

Availability of data and materials

Not applicable.

Abbreviations

OVs:

Oncolytic viruses

TME:

Tumor microenvironment

MSCs:

Mesenchymal stem cells

UC-MSC:

Umbilical cord derived MSCs

hMSCs:

Human MSCs

BM-MSCs:

Bone marrow MSCs

DAMPs:

Damage-associated molecular patterns

PAMPs:

Pathogen-associated molecular patterns

oAds:

Oncolytic adenovirus

CRAds:

Conditional replication oncolytic adenoviruses

oHSV:

Herpes simplex virus

oMV:

Oncolytic measles virus

EMT:

Epithelial-mesenchymal transition

MET:

Mesenchymal-epithelial transitions

References

  1. Amri A, Soltanian AR, Borzouei S. Survival rates and prognostic factors of thyroid cancer: a retrospective cohort study. J Parathyr Dis. 2022;10: e11162.

    Article  Google Scholar 

  2. Stuckey DW, Shah K. Stem cell-based therapies for cancer treatment: Separating hope from hype. Nat Rev Cancer. 2014;14(10):683–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Engel J, Lategahn J, Rauh D. Hope and disappointment: covalent inhibitors to overcome drug resistance in non-small cell lung cancer. ACS Med Chem Lett. 2016;7:2–5.

    Article  CAS  PubMed  Google Scholar 

  4. Jamali S, et al. Strategy for treating the gastric cancer: a systematic review and meta-analysis. Int J Sci Res Dent Med Sci. 2020;2(1):6–11.

    CAS  Google Scholar 

  5. Ardalan M. Parathyroid carcinoma; an updated mini-review on current trends. J Parathyr Dis. 2016;4(2):57–9.

    Google Scholar 

  6. Roayaei M, Andalib Z, Akhavan A. The effect of oral zinc sulfate on prevention of chemotherapy induced oral mucositis in breast cancer patients treated with adriamycin and cyclophosphamide; a double-blind randomized clinical trial. J Nephropharmacol. 2023;12(1):e10533.

    Article  Google Scholar 

  7. Daei Sorkhabi A, et al. The basis and advances in clinical application of cytomegalovirus-specific cytotoxic T cell immunotherapy for glioblastoma multiforme. Front Oncol. 2022;12:818447.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Aghi M, Martuza RL. Oncolytic viral therapies–the clinical experience. Oncogene. 2005;24(52):7802–16.

    Article  CAS  PubMed  Google Scholar 

  9. Parato KA, et al. Recent progress in the battle between oncolytic viruses and tumours. Nat Rev Cancer. 2005;5(12):965–76.

    Article  CAS  PubMed  Google Scholar 

  10. Liu T-C, Galanis E, Kirn D. Clinical trial results with oncolytic virotherapy: a century of promise, a decade of progress. Nat Clin Pract Oncol. 2007;4(2):101–17.

    Article  CAS  PubMed  Google Scholar 

  11. Lawler SE, Chiocca EA. Oncolytic virus-mediated immunotherapy: a combinatorial approach for cancer treatment. J Clin Oncol. 2015;33(25):2812–4.

    Article  CAS  PubMed  Google Scholar 

  12. Romero D. Oncolytic viruses prime antitumour immunity. Nat Rev Clin Oncol. 2018;15(3):135–135.

    Article  PubMed  Google Scholar 

  13. Engeland CE, Bell JC. Introduction to oncolytic virotherapy. Oncolytic Viruses. 2020. https://doi.org/10.1007/978-1-4939-9794-7_1.

    Article  PubMed  Google Scholar 

  14. Lichty BD, et al. Going viral with cancer immunotherapy. Nat Rev Cancer. 2014;14(8):559–67.

    Article  CAS  PubMed  Google Scholar 

  15. Scott EM, et al. Solid tumor immunotherapy with T cell engager-armed oncolytic viruses. Macromol Biosci. 2018;18(1):1700187.

    Article  Google Scholar 

  16. Andtbacka RH, et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol. 2015;33(25):2780–8.

    Article  CAS  PubMed  Google Scholar 

  17. Markert JM, et al. Phase Ib trial of mutant herpes simplex virus G207 inoculated pre-and post-tumor resection for recurrent GBM. Mol Ther. 2009;17(1):199–207.

    Article  CAS  PubMed  Google Scholar 

  18. Cripe TP, et al. Phase 1 study of intratumoral Pexa-Vec (JX-594), an oncolytic and immunotherapeutic vaccinia virus, in pediatric cancer patients. Mol Ther. 2015;23(3):602–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Heo J, et al. Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer. Nat Med. 2013;19(3):329–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Russell SJ, et al. Remission of disseminated cancer after systemic oncolytic virotherapy. Mayo Clin Proc. 2014;89(7):926–33.

    Article  PubMed  Google Scholar 

  21. Elankumaran S, et al. Type I interferon-sensitive recombinant newcastle disease virus for oncolytic virotherapy. J Virol. 2010;84(8):3835–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Roy DG, Bell JC, Bourgeois-Daigneault M-C. Magnetic targeting of oncolytic VSV-based therapies improves infection of tumor cells in the presence of virus-specific neutralizing antibodies in vitro. Biochem Biophys Res Commun. 2020;526(3):641–6.

    Article  CAS  PubMed  Google Scholar 

  23. Schirrmacher V, van Gool S, Stuecker W. Breaking therapy resistance: An update on oncolytic Newcastle disease virus for improvements of cancer therapy. Biomedicines. 2019;7(3):66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Howells A, et al. Oncolytic viruses—interaction of virus and tumor cells in the battle to eliminate cancer. Front Oncol. 2017;7:195.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Corsten MF, Shah K. Therapeutic stem-cells for cancer treatment: hopes and hurdles in tactical warfare. Lancet Oncol. 2008;9(4):376–84.

    Article  PubMed  Google Scholar 

  26. Teo AK, Vallier L. Emerging use of stem cells in regenerative medicine. Biochem J. 2010;428(1):11–23.

    Article  CAS  PubMed  Google Scholar 

  27. Seif F, Kheirollah A, Babaahmadi-Rezaei H. Efficient isolation and identification of primary endothelial cells from bovine aorta by collagenase P. Immunopathol Persa. 2020;6(2):e15.

    Article  Google Scholar 

  28. Maestroni G, Hertens E, Galli P. Factor (s) from nonmacrophage bone marrow stromal cells inhibit Lewis lung carcinoma and B16 melanoma growth in mice. Cell Mol Life Sci CMLS. 1999;55(4):663–7.

    Article  CAS  PubMed  Google Scholar 

  29. Nakamura K, et al. Antitumor effect of genetically engineered mesenchymal stem cells in a rat glioma model. Gene Ther. 2004;11(14):1155–64.

    Article  CAS  PubMed  Google Scholar 

  30. Qiao C, et al. Human mesenchymal stem cells isolated from the umbilical cord. Cell Biol Int. 2008;32(1):8–15.

    Article  CAS  PubMed  Google Scholar 

  31. Qiao L, et al. Suppression of tumorigenesis by human mesenchymal stem cells in a hepatoma model. Cell Res. 2008;18(4):500–7.

    Article  CAS  PubMed  Google Scholar 

  32. Ahn J-O, et al. Human adipose tissue-derived mesenchymal stem cells inhibit melanoma growth in vitro and in vivo. Anticancer Res. 2015;35(1):159–68.

    CAS  PubMed  Google Scholar 

  33. He N, et al. MSCs inhibit tumor progression and enhance radiosensitivity of breast cancer cells by down-regulating Stat3 signaling pathway. Cell Death Dis. 2018;9(10):1026.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Willmon C, et al. Cell carriers for oncolytic viruses: Fed Ex for cancer therapy. Mol Ther. 2009;17(10):1667–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Xia X, et al. Mesenchymal stem cells as carriers and amplifiers in CRAd delivery to tumors. Mol Cancer. 2011;10(1):134.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Beckermann B, et al. VEGF expression by mesenchymal stem cells contributes to angiogenesis in pancreatic carcinoma. Br J Cancer. 2008;99(4):622–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kallifatidis G, et al. Improved lentiviral transduction of human mesenchymal stem cells for therapeutic intervention in pancreatic cancer. Cancer Gene Ther. 2008;15(4):231–40.

    Article  CAS  PubMed  Google Scholar 

  38. Ruano D, et al. First-in-human, first-in-child trial of autologous MSCs carrying the oncolytic virus Icovir-5 in patients with advanced tumors. Mol Ther. 2020;28(4):1033–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Tang Y, et al. In vivo tracking of neural progenitor cell migration to glioblastomas. Hum Gene Ther. 2003;14(13):1247–54.

    Article  CAS  PubMed  Google Scholar 

  40. Aboody KS, et al. Neural stem cells display extensive tropism for pathology in adult brain: evidence from intracranial gliomas. Proc Natl Acad Sci. 2000;97(23):12846–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Shah K, et al. Glioma therapy and real-time imaging of neural precursor cell migration and tumor regression. Ann Neurol Off J Am Neurol Assoc Child Neurol Soc. 2005;57(1):34–41.

    CAS  Google Scholar 

  42. Sasportas LS, et al. Assessment of therapeutic efficacy and fate of engineered human mesenchymal stem cells for cancer therapy. Proc Natl Acad Sci. 2009;106(12):4822–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Shah K. Mesenchymal stem cells engineered for cancer therapy. Adv Drug Deliv Rev. 2012;64(8):739–48.

    Article  CAS  PubMed  Google Scholar 

  44. Banijamali RS, et al. Kinetics of oncolytic reovirus T3D replication and growth pattern in mesenchymal stem cells. Cell J (Yakhteh). 2020;22(3):283.

    Google Scholar 

  45. Keshavarz M, et al. Oncolytic Newcastle disease virus delivered by Mesenchymal stem cells-engineered system enhances the therapeutic effects altering tumor microenvironment. Virol J. 2020;17(1):64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hai C, et al. Application of mesenchymal stem cells as a vehicle to deliver replication-competent adenovirus for treating malignant glioma. Chin J Cancer. 2012;31(5):233.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Kazimirsky G, et al. Mesenchymal stem cells enhance the oncolytic effect of Newcastle disease virus in glioma cells and glioma stem cells via the secretion of TRAIL. Stem Cell Res Ther. 2016;7(1):149.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Melen GJ, et al. Influence of carrier cells on the clinical outcome of children with neuroblastoma treated with high dose of oncolytic adenovirus delivered in mesenchymal stem cells. Cancer Lett. 2016;371(2):161–70.

    Article  CAS  PubMed  Google Scholar 

  49. Leoni V, et al. Systemic delivery of HER2-retargeted oncolytic-HSV by mesenchymal stromal cells protects from lung and brain metastases. Oncotarget. 2015;6(33):34774.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Hoyos V, et al. Mesenchymal stromal cells for linked delivery of oncolytic and apoptotic adenoviruses to non-small-cell lung cancers. Mol Ther. 2015;23(9):1497–506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Franco-Luzón L, et al. Systemic oncolytic adenovirus delivered in mesenchymal carrier cells modulate tumor infiltrating immune cells and tumor microenvironment in mice with neuroblastoma. Oncotarget. 2020;11(4):347–61.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Gimenez-Sanchez A, et al. Cellular Virotherapy increases tumor-infiltrating lymphocytes (TIL) and decreases their PD-1+ subsets in mouse immunocompetent models. Cancers. 2020;12(7):1920.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Mader EK, et al. Mesenchymal stem cell carriers protect oncolytic measles viruses from antibody neutralization in an orthotopic ovarian cancer therapy model. Clin Cancer Res. 2009;15(23):7246–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hakkarainen T, et al. Human mesenchymal stem cells lack tumor tropism but enhance the antitumor activity of oncolytic adenoviruses in orthotopic lung and breast tumors. Hum Gene Ther. 2007;18(7):627–41.

    Article  CAS  PubMed  Google Scholar 

  55. Collet G, et al. Trojan horse at cellular level for tumor gene therapies. Gene. 2013;525(2):208–16.

    Article  CAS  PubMed  Google Scholar 

  56. Ferguson SD, et al. Crossing the boundaries: stem cells and gene therapy. Discov Med. 2010;9(46):192.

    PubMed  PubMed Central  Google Scholar 

  57. Salmasi Z, et al. Mesenchymal stem cells engineered by modified polyethylenimine polymer for targeted cancer gene therapy, in vitro and in vivo. Biotechnol Prog. 2020;36(6): e3025.

    Article  CAS  PubMed  Google Scholar 

  58. Momin EN, et al. The oncogenic potential of mesenchymal stem cells in the treatment of cancer: directions for future research. Curr Immunol Rev. 2010;6(2):137–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Yoon A-R, et al. Mesenchymal stem cell–mediated delivery of an oncolytic adenovirus enhances antitumor efficacy in hepatocellular carcinoma. Cancer Res. 2019;79(17):4503–14.

    Article  CAS  PubMed  Google Scholar 

  60. Rattigan Y, et al. Interleukin 6 mediated recruitment of mesenchymal stem cells to the hypoxic tumor milieu. Exp Cell Res. 2010;316(20):3417–24.

    Article  CAS  PubMed  Google Scholar 

  61. Ringe J, et al. 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. J Cell Biochem. 2007;101(1):135–46.

    Article  CAS  PubMed  Google Scholar 

  62. Kidd S, et al. Origins of the tumor microenvironment: Quantitative assessment of adipose-derived and bone marrow–derived stroma. PLoS ONE. 2012;7(2): e30563.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Briko A, Kapravchuk V, Kobelev A, Hammoud A, Leonhardt S, Ngo C, Gulyaev Y, Shchukin, S. A way of bionic control based on EI, EMG, and FMG signals. Sensors (Basel, Switzerland) 2021;22(1):152. https://doi.org/10.3390/s22010152.

    Article  PubMed  Google Scholar 

  64. Briko A, Kapravchuk V, Kobelev A, Tikhomirov A, Hammoud A, Al-Harosh M, Leonhardt S, Ngo C, Gulyaev Y, Shchukin S. Determination of the geometric parameters of electrode systems for electrical impedance myography: a preliminary study. Sensors (Basel, Switzerland) 2021;22(1):97. https://doi.org/10.3390/s22010097.

    Article  PubMed  Google Scholar 

  65. Rustad KC, Gurtner GC. Mesenchymal stem cells home to sites of injury and inflammation. Adv Wound Care. 2012;1(4):147–52.

    Article  Google Scholar 

  66. Xie C, et al. Systemically infused mesenchymal stem cells show different homing profiles in healthy and tumor mouse models. Stem Cells Transl Med. 2017;6(4):1120–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Ma F, et al. Human umbilical cord mesenchymal stem cells promote breast cancer metastasis by interleukin-8-and interleukin-6-dependent induction of CD44+/CD24-cells. Cell Transplant. 2015;24(12):2585–99.

    Article  PubMed  Google Scholar 

  68. Smith CL, et al. Pre-exposure of human adipose mesenchymal stem cells to soluble factors enhances their homing to brain cancer. Stem Cells Transl Med. 2015;4(3):239–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Khalili M, et al. Evaluation of the survival rate and clinical outcome of nanodrug administration for the treatment of lung cancer: a systematic review and meta-analysis. Int J Sci Res Dent Med Sci. 2022;4(3):140–7.

    Google Scholar 

  70. Yoon A, et al. Mesenchymal stem cell–mediated delivery of an oncolytic adenovirus enhances antitumor efficacy in hepatocellular carcinomasystemic delivery of oncolytic adenovirus with MSC. Cancer Res. 2019;79(17):4503–14.

    Article  CAS  PubMed  Google Scholar 

  71. Dadras F, Sheikh V, Khoshjou F. Epithelial and endothelial mesenchymal transition and their role in diabetic kidney disease. J Renal Inj Prev. 2017;7(1):1–6.

    Article  Google Scholar 

  72. Gao F, et al. Mesenchymal stem cells and immunomodulation: current status and future prospects. Cell Death Dis. 2016;7(1):e2062–e2062.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Khare D, et al. Mesenchymal stromal cell-derived exosomes affect mRNA expression and function of B-lymphocytes. Front Immunol. 2018;9:3053.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Corcione A, et al. Human mesenchymal stem cells modulate B-cell functions. Blood. 2006;107(1):367–72.

    Article  CAS  PubMed  Google Scholar 

  75. Palaniappan S, et al. Effect of Lung Specific Yoga Mudras on Pulmonary Function Tests in subjects with FEV1% predicted values less than 80%. Int J Sci Res Dent Med Sci. 2021;3(3):117–21.

    Google Scholar 

  76. Rozenberg A, et al. Human mesenchymal stem cells impact Th17 and Th1 responses through a prostaglandin E2 and myeloid-dependent mechanism. Stem Cells Transl Med. 2016;5(11):1506–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Spaggiari GM, et al. Mesenchymal stem cells inhibit natural killer–cell proliferation, cytotoxicity, and cytokine production: role of indoleamine 2, 3-dioxygenase and prostaglandin E2. Blood J Am Soc Hematol. 2008;111(3):1327–33.

    CAS  Google Scholar 

  78. Carreras-Planella L, et al. Immunomodulatory effect of MSC on B cells is independent of secreted extracellular vesicles. Front Immunol. 2019;10:1288.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Wilson A, et al. Isolation and characterisation of human adipose-derived stem cells. In: Immunological tolerance. Springer; 2019. p. 3–13.

    Chapter  Google Scholar 

  80. Zhang F, et al. Mesenchymal stem cells alleviate rat diabetic nephropathy by suppressing CD103+ DCs-mediated CD8+ T cell responses. J Cell Mol Med. 2020;24(10):5817–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Feng Y, Li F, Yan J, Guo X, Wang F, Shi H, Du J, Zhang H, Gao Y, Li D, Yao Y, Hu W, Han J, Zhang M, Ding R, Wang X, Huang C, Zhang J. Pan-cancer analysis and experiments with cell lines reveal that the slightly elevated expression of DLGAP5 is involved in clear cell renal cell carcinoma progression. Life Sciences. 2021;287:120056

    Article  CAS  PubMed  Google Scholar 

  82. Xu L-L, et al. Impaired function of bone marrow mesenchymal stem cells from immune thrombocytopenia patients in inducing regulatory dendritic cell differentiation through the Notch-1/Jagged-1 signaling pathway. Stem cells Dev. 2017;26(22):1648–61.

    Article  CAS  PubMed  Google Scholar 

  83. Jiang X-X, et al. Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells. Blood. 2005;105(10):4120–6.

    Article  CAS  PubMed  Google Scholar 

  84. Liu Q, et al. Human mesenchymal stromal cells enhance the immunomodulatory function of CD8+ CD28− regulatory T cells. Cell Mol Immunol. 2015;12(6):708–18.

    Article  CAS  PubMed  Google Scholar 

  85. El Omar R, et al. Immunomodulation of endothelial differentiated mesenchymal stromal cells: impact on T and NK cells. Immunol Cell Biol. 2016;94(4):342–56.

    Article  PubMed  Google Scholar 

  86. Cho K-A, et al. Mesenchymal stem cells ameliorate B-cell-mediated immune responses and increase IL-10-expressing regulatory B cells in an EBI3-dependent manner. Cell Mol Immunol. 2017;14(11):895–908.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Liu X, et al. Mesenchymal stem/stromal cells induce the generation of novel IL-10–dependent regulatory dendritic cells by SOCS3 activation. J Immunol. 2012;189(3):1182–92.

    Article  CAS  PubMed  Google Scholar 

  88. Ahmed AU, et al. Bone marrow mesenchymal stem cells loaded with an oncolytic adenovirus suppress the anti-adenoviral immune response in the cotton rat model. Mol Ther. 2010;18(10):1846–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Komarova S, et al. Mesenchymal progenitor cells as cellular vehicles for delivery of oncolytic adenoviruses. Mol Cancer Ther. 2006;5(3):755–66.

    Article  CAS  PubMed  Google Scholar 

  90. Stoff-Khalili MA, et al. Mesenchymal stem cells as a vehicle for targeted delivery of CRAds to lung metastases of breast carcinoma. Breast Cancer Res Treat. 2007;105(2):157–67.

    Article  PubMed  Google Scholar 

  91. Sonabend AM, et al. Mesenchymal stem cells effectively deliver an oncolytic adenovirus to intracranial glioma. Stem cells. 2008;26(3):831–41.

    Article  CAS  PubMed  Google Scholar 

  92. Yong RL, et al. Human bone marrow–derived mesenchymal stem cells for intravascular delivery of oncolytic adenovirus Δ24-RGD to human gliomas. Cancer Res. 2009;69(23):8932–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Ahmed AU, 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):1559–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Mader EK, et al. Optimizing patient derived mesenchymal stem cells as virus carriers for a phase I clinical trial in ovarian cancer. J Transl Med. 2013;11(1):20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Ong H-T, et al. Systemically delivered measles virus-infected mesenchymal stem cells can evade host immunity to inhibit liver cancer growth. J Hepatol. 2013;59(5):999–1006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Klopp AH, et al. Concise review: dissecting a discrepancy in the literature: do mesenchymal stem cells support or suppress tumor growth? Stem cells. 2011;29(1):11–9.

    Article  CAS  PubMed  Google Scholar 

  97. Chulpanova DS, et al. Application of mesenchymal stem cells for therapeutic agent delivery in anti-tumor treatment. Front Pharmacol. 2018;9:259.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Aliyari-Serej Z, et al. Relation between Immune cell response and stemness genes expression in breast cancer; a new approach in NANOG gene and Let7-a expression in breast cancer cell lines. Immunopathol Persa. 2020;6(2):e21.

    Article  Google Scholar 

  99. Sun B, et al. Therapeutic potential of mesenchymal stromal cells in a mouse breast cancer metastasis model. Cytotherapy. 2009;11(3):289–98.

    Article  CAS  PubMed  Google Scholar 

  100. Sui X, Zhang R, Liu S, Duan T, Zhai L, Zhang M, Han X, Xiang Y, Huang X, Lin H, Xie T. RSL3 drives ferroptosis through GPX4 inactivation and ROS production in colorectal cancer. Front Pharmacol. 2018. https://doi.org/10.3389/fphar.2018.01371

  101. Otsu K, et al. Concentration-dependent inhibition of angiogenesis by mesenchymal stem cells. Blood J Am Soc Hematol. 2009;113(18):4197–205.

    CAS  Google Scholar 

  102. Khakoo AY, et al. Human mesenchymal stem cells exert potent antitumorigenic effects in a model of Kaposi’s sarcoma. J Exp Med. 2006;203(5):1235–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Kucerova L, et al. Tumor-driven molecular changes in human mesenchymal stromal cells. Cancer Microenviron. 2015;8(1):1–14. https://doi.org/10.1007/s12307-014-0151-9.

    Article  CAS  PubMed  Google Scholar 

  104. Cavarretta IT, et al. Adipose tissue–derived mesenchymal stem cells expressing prodrug-converting enzyme inhibit human prostate tumor growth. Mol Ther. 2010;18(1):223–31.

    Article  CAS  PubMed  Google Scholar 

  105. Kaufman HL, Kohlhapp FJ, Zloza A. Oncolytic viruses: a new class of immunotherapy drugs. Nat Rev Drug Discov. 2015;14(9):642–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Gujar S, et al. Antitumor benefits of antiviral immunity: an underappreciated aspect of oncolytic virotherapies. Trends Immunol. 2018;39(3):209–21.

    Article  CAS  PubMed  Google Scholar 

  107. De Matos AL, Franco LS, McFadden G. Oncolytic viruses and the immune system: the dynamic duo. Mol Ther Methods Clin Dev. 2020;17:349–58.

    Article  Google Scholar 

  108. Liu S, Li Q, Chen K, Zhang Q, Li G, Zhuo L, Zhai B, Sui X, Hu X, Xie T. The emerging molecular mechanism of m6A modulators in tumorigenesis and cancer progression. Biomed Pharmacother. 2020;127:110098. https://doi.org/10.1016/j.biopha.2020.110098.

    Article  CAS  PubMed  Google Scholar 

  109. Jhawar SR, et al. Oncolytic viruses—natural and genetically engineered cancer immunotherapies. Front Oncol. 2017;7:202.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Mullen JT, Tanabe KK. Viral oncolysis. Oncologist. 2002;7(2):106–19.

    Article  CAS  PubMed  Google Scholar 

  111. Hamid O, et al. Oncolytic immunotherapy: unlocking the potential of viruses to help target cancer. Cancer Immunol Immunother. 2017;66(10):1249–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Serrano-del Valle A, et al. Immunogenic cell death and immunotherapy of multiple myeloma. Front Cell Dev Biol. 2019;7:50.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022;12(1):31–46.

    Article  CAS  PubMed  Google Scholar 

  114. Guo ZS, Liu Z, Bartlett DL. Oncolytic immunotherapy: dying the right way is a key to eliciting potent antitumor immunity. Front Oncol. 2014;4:74.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Aaes TL, et al. Vaccination with necroptotic cancer cells induces efficient anti-tumor immunity. Cell Rep. 2016;15(2):274–87.

    Article  CAS  PubMed  Google Scholar 

  116. Wiernicki B, et al. Cancer cells dying from ferroptosis impede dendritic cell-mediated anti-tumor immunity. Nat Commun. 2022;13(1):3676.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Pol JG, Bridle BW, Lichty BD. Detection of tumor antigen-specific T-cell responses after oncolytic vaccination. In: Oncolytic Viruses. Springer; 2020. p. 191–211.

    Chapter  Google Scholar 

  118. Keshavarz M, et al. Oncolytic paramyxoviruses-induced autophagy; a prudent weapon for cancer therapy. J Biomed Sci. 2019;26(1):48.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Sobol PT, et al. Adaptive antiviral immunity is a determinant of the therapeutic success of oncolytic virotherapy. Mol Ther. 2011;19(2):335–44.

    Article  CAS  PubMed  Google Scholar 

  120. Workenhe ST, Verschoor ML, Mossman KL. The role of oncolytic virus immunotherapies to subvert cancer immune evasion. Future Oncol. 2015;11(4):675–89.

    Article  CAS  PubMed  Google Scholar 

  121. van Vloten JP, et al. Critical interactions between immunogenic cancer cell death, oncolytic viruses, and the immune system define the rational design of combination immunotherapies. J Immunol. 2018;200(2):450–8.

    Article  PubMed  Google Scholar 

  122. Marelli G, et al. Oncolytic viral therapy and the immune system: a double-edged sword against cancer. Front Immunol. 2018;9:866.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Bai Y, et al. Updates to the antitumor mechanism of oncolytic virus. Thorac Cancer. 2019;10(5):1031–5.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Garg AD, Agostinis P. Cell death and immunity in cancer: from danger signals to mimicry of pathogen defense responses. Immunol Rev. 2017;280(1):126–48.

    Article  CAS  PubMed  Google Scholar 

  125. Jiang H, Fueyo J. Healing after death: antitumor immunity induced by oncolytic adenoviral therapy. Oncoimmunology. 2014;3(7): e947872.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Jamali S, et al. Prevalence of malignancy and chronic obstructive pulmonary disease among patients with COVID-19: a systematic review and meta-analysis. Int J Sci Res Dent Med Sci. 2020;2(2):52–8.

    CAS  Google Scholar 

  127. Zhang Z, Cui F, Cao C, Wang Q, Zou Q. Single-cell RNA analysis reveals the potential risk of organ-specific cell types vulnerable to SARS-CoV-2 infections. Comput Biol Med 2022;140:105092. https://doi.org/10.1016/j.compbiomed.2021.105092.

    Article  CAS  Google Scholar 

  128. Xu Q, et al. Evaluation of Newcastle disease virus mediated dendritic cell activation and cross-priming tumor-specific immune responses ex vivo. Int J Cancer. 2020;146(2):531–41.

    Article  CAS  PubMed  Google Scholar 

  129. Kepp O, et al. Consensus guidelines for the detection of immunogenic cell death. Oncoimmunology. 2014;3(9): e955691.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Reale A, et al. Perspectives on immunotherapy via oncolytic viruses. Infect Agents Cancer. 2019;14(1):5.

    Article  Google Scholar 

  131. Filley AC, Dey M. Immune system, friend or foe of oncolytic virotherapy? Front Oncol. 2017;7:106.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Prestwich RJ, et al. Oncolytic viruses: a novel form of immunotherapy. Expert Rev Anticancer Ther. 2008;8(10):1581–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Harrington K, et al. Optimizing oncolytic virotherapy in cancer treatment. Nat Rev Drug Discovery. 2019;18(9):689–706.

    Article  CAS  PubMed  Google Scholar 

  134. Rotte A, Jin J, Lemaire V. Mechanistic overview of immune checkpoints to support the rational design of their combinations in cancer immunotherapy. Ann Oncol. 2018;29(1):71–83.

    Article  CAS  PubMed  Google Scholar 

  135. Cai L, et al. Defective HLA class I antigen processing machinery in cancer. Cancer Immunol Immunother. 2018;67(6):999–1009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Gujar S, et al. Multifaceted therapeutic targeting of ovarian peritoneal carcinomatosis through virus-induced immunomodulation. Mol Ther. 2013;21(2):338–47.

    Article  CAS  PubMed  Google Scholar 

  137. Lapteva N, et al. Attraction and activation of dendritic cells at the site of tumor elicits potent antitumor immunity. Mol Ther. 2009;17(9):1626–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Greiner S, et al. The highly attenuated vaccinia virus strain modified virus Ankara induces apoptosis in melanoma cells and allows bystander dendritic cells to generate a potent anti-tumoral immunity. Clin Exp Immunol. 2006;146(2):344–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Guillerme J-B, et al. Measles virus vaccine-infected tumor cells induce tumor antigen cross-presentation by human plasmacytoid dendritic cellscross-presentation of human tumor antigen by pDC. Clin Cancer Res. 2013;19(5):1147–58.

    Article  CAS  PubMed  Google Scholar 

  140. Benencia F, et al. Herpes virus oncolytic therapy reverses tumor immune dysfunction and facilitates tumor antigen presentation. Cancer Biol Ther. 2008;7(8):1194–205.

    Article  CAS  PubMed  Google Scholar 

  141. Denton NL, et al. Tumor-associated macrophages in oncolytic virotherapy: friend or foe? Biomedicines. 2016;4(3):13.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Marchini A. et al., Immune conversion of tumor microenvironment by oncolytic viruses: the protoparvovirus H-1PV case study. Front Immunol 2019: p. 1848.

  143. Denton NL, et al. Myelolytic treatments enhance oncolytic herpes virotherapy in models of Ewing sarcoma by modulating the immune microenvironment. Mol Ther Oncolytics. 2018;11:62–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Tan DQ, et al. Macrophage response to oncolytic paramyxoviruses potentiates virus-mediated tumor cell killing. Eur J Immunol. 2016;46(4):919–28.

    Article  CAS  PubMed  Google Scholar 

  145. Ehrig K, et al. Growth inhibition of different human colorectal cancer xenografts after a single intravenous injection of oncolytic vaccinia virus GLV-1h68. J Transl Med. 2013;11(1):79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Saha D, Martuza RL, Rabkin SD. Macrophage polarization contributes to glioblastoma eradication by combination immunovirotherapy and immune checkpoint blockade. Cancer Cell. 2017;32(2):253-267. e5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Russell L, et al. Oncolytic viruses: priming time for cancer immunotherapy. BioDrugs. 2019;33(5):485–501.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Jin HY, Wang ZA. Global stabilization of the full attraction-repulsion Keller-Segel system. Discrete Contin Dyn Syst Ser A. 2020;40(6):3509–27. https://doi.org/10.3934/dcds.2020027.

    Article  Google Scholar 

  149. Dharwadkar A, et al. Cytological study of salivary gland lesions along with histopathological correlation in a Tertiary Care Centre. Int J Sci Res Den Med Scis. 2022;4(3):101–9.

    Google Scholar 

  150. Hashemi Goradel N, et al. Nanoparticles as new tools for inhibition of cancer angiogenesis. J Cell Physiol. 2018;233(4):2902–10.

    Article  CAS  PubMed  Google Scholar 

  151. Goradel NH, et al. Melatonin as an angiogenesis inhibitor to combat cancer: mechanistic evidence. Toxicol Appl Pharmacol. 2017;335:56–63.

    Article  CAS  PubMed  Google Scholar 

  152. Angarita FA, et al. Mounting a strategic offense: fighting tumor vasculature with oncolytic viruses. Trends Mol Med. 2013;19(6):378–92.

    Article  CAS  PubMed  Google Scholar 

  153. Breitbach CJ, et al. Oncolytic vaccinia virus disrupts tumor-associated vasculature in humans. Cancer Res. 2013;73(4):1265–75.

    Article  CAS  PubMed  Google Scholar 

  154. Bejarano MT, Merchan JR. Targeting tumor vasculature through oncolytic virotherapy: recent advances. Oncolytic Virother. 2015;4:169.

    CAS  Google Scholar 

  155. Hammoud A, Tikhomirov AN, Shaheen Z. Automatic bio-impedance signal analysis: smoothing processes efficacy evaluation in determining the vascular tone type. In: 2021 Ural symposium on biomedical engineering, radioelectronics and information technology (USBEREIT). IEEE. 2021. pp. 0113–0116.

  156. Haka AS, et al. In vivo margin assessment during partial mastectomy breast surgery using Raman spectroscopy. Cancer Res. 2006;66(6):3317–22.

    Article  CAS  PubMed  Google Scholar 

  157. Breitbach CJ, et al. Targeted inflammation during oncolytic virus therapy severely compromises tumor blood flow. Mol Ther. 2007;15(9):1686–93.

    Article  CAS  PubMed  Google Scholar 

  158. Hammoud A, Tikhomirov A, Myasishcheva G, Shaheen Z, Volkov A, Briko A, Shchukin S. Multi-channel bioimpedance system for detecting vascular tone in human limbs: an approach. Sensors (Basel, Switzerland) 2021;22(1):138. https://doi.org/10.3390/s22010138.

    Article  Google Scholar 

  159. Russell SJ, Peng K-W, Bell JC. Oncolytic virotherapy. Nat Biotechnol. 2012;30(7):658–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Alemany R, Balagué C, Curiel DT. Replicative adenoviruses for cancer therapy. Nat Biotechnol. 2000;18(7):723–7.

    Article  CAS  PubMed  Google Scholar 

  161. Nettelbeck DM. Virotherapeutics: conditionally replicative adenoviruses for viral oncolysis. Anticancer Drugs. 2003;14(8):577–84.

    Article  CAS  PubMed  Google Scholar 

  162. Yamamoto M, Curiel DT. Current issues and future directions of oncolytic adenoviruses. Mol Ther. 2010;18(2):243–50.

    Article  CAS  PubMed  Google Scholar 

  163. Pol J, et al. Trial Watch: oncolytic viruses for cancer therapy. Oncoimmunology. 2014;3: e28694.

    Article  PubMed  PubMed Central  Google Scholar 

  164. He X, Zhu Y, Yang L, Wang Z, Wang Z, Feng J, Wen X, Cheng L, Zhu R. MgFe-LDH nanoparticles: a promising leukemia inhibitory factor replacement for self-renewal and pluripotency maintenance in cultured mouse embryonic stem cells. Adv Sci (Weinheim, Baden-Wurttemberg, Germany). 2021;8(9):2003535. https://doi.org/10.1002/advs.202003535.

    Article  CAS  Google Scholar 

  165. Bramante S, et al. Serotype chimeric oncolytic adenovirus coding for GM-CSF for treatment of sarcoma in rodents and humans. Int J Cancer. 2014;135(3):720–30.

    Article  CAS  PubMed  Google Scholar 

  166. Liikanen I, et al. Oncolytic adenovirus with temozolomide induces autophagy and antitumor immune responses in cancer patients. Mol Ther. 2013;21(6):1212–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Kanerva A, et al. Antiviral and antitumor T-cell immunity in patients treated with GM-CSF–coding oncolytic adenovirus. Clin Cancer Res. 2013;19(10):2734–44.

    Article  CAS  PubMed  Google Scholar 

  168. Kim KH, et al. A phase I clinical trial of Ad5/3-Δ24, a novel serotype-chimeric, infectivity-enhanced, conditionally-replicative adenovirus (CRAd), in patients with recurrent ovarian cancer. Gynecol Oncol. 2013;130(3):518–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Bramante S, et al. Treatment of melanoma with a serotype 5/3 chimeric oncolytic adenovirus coding for GM-CSF: Results in vitro, in rodents and in humans. Int J Cancer. 2015;137(7):1775–83.

    Article  CAS  PubMed  Google Scholar 

  170. Garcia-Castro J, 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):476–83.

    Article  CAS  PubMed  Google Scholar 

  171. Barahman M, Bahadoram M, Mahmoudian-Sani M. Frequency of triple negative breast cancer in referrals patients to an oncology radiotherapy section. J Prev Epidemiol. 2021;6(1): e09.

    Article  Google Scholar 

  172. Khalili SM, et al. Comparing the quality of life of women suffering from breast cancer receiving palliative care and ordinary care. Immunopathol Persa. 2020;6(2):e22.

    Article  Google Scholar 

  173. Sadr Z, et al. Study of family medicine role in breast cancer prevention: a systematic review. J Prev Epidemiol. 2021;6(2):e32–e32.

    Article  Google Scholar 

  174. Fouladi N, et al. Factors associated with delayed diagnosis of breast cancer; a study in North-West of Iran. Immunopathol Persa. 2019;6(1):e02.

    Article  Google Scholar 

  175. Darakhshandeh A, Momenzadeh M. Cachexia and anorexia in cancer; a systematic review. Immunopathol Persa. 2020;7(2):e20.

    Article  Google Scholar 

  176. Rincón E, et al. Mesenchymal stem cell carriers enhance antitumor efficacy of oncolytic adenoviruses in an immunocompetent mouse model. Oncotarget. 2017;8(28):45415.

    Article  PubMed  PubMed Central  Google Scholar 

  177. Guo Y, et al. Menstrual blood-derived stem cells as delivery vehicles for oncolytic adenovirus virotherapy for colorectal cancer. Stem cells Dev. 2019;28(13):882–96.

    Article  CAS  PubMed  Google Scholar 

  178. Kaczorowski A, et al. Delivery of improved oncolytic adenoviruses by mesenchymal stromal cells for elimination of tumorigenic pancreatic cancer cells. Oncotarget. 2016;7(8):9046.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Chastkofsky MI, et al. Mesenchymal stem cells successfully deliver oncolytic virotherapy to diffuse intrinsic pontine GliomaStem cells deliver virotherapy to DIPG. Clin Cancer Res. 2021;27(6):1766–77.

    Article  CAS  PubMed  Google Scholar 

  180. Shomali N, et al. Survivin; a novel therapeutic target that correlates with survival of autoreactive T lymphocytes obtained from patients with ankylosing spondylitis. Gene. 2022;844: 146829.

    Article  CAS  PubMed  Google Scholar 

  181. Shomali N, et al. Dysregulation of survivin-targeting microRNAs in autoimmune diseases: new perspectives for novel therapies. Front Immunol. 2022;13: 839945.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Zhang Y, et al. Therapeutic effects of mesenchymal stem cells loaded with oncolytic adenovirus carrying decorin on a breast cancer lung metastatic mouse model. Mol Ther Oncolytics. 2022;24:486–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Zhang J, et al. Systemic administration of mesenchymal stem cells loaded with a novel oncolytic adenovirus carrying IL-24/endostatin enhances glioma therapy. Cancer Lett. 2021;509:26–38.

    Article  CAS  PubMed  Google Scholar 

  184. Du W, et al. Stem cell-released oncolytic herpes simplex virus has therapeutic efficacy in brain metastatic melanomas. Proc Natl Acad Sci. 2017;114(30):E6157–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Duebgen M, et al. Stem cells loaded with multimechanistic oncolytic herpes simplex virus variants for brain tumor therapy. JNCI J Natl Cancer Inst. 2014;106(6):dju090.

    Article  PubMed  Google Scholar 

  186. Castleton A, et al. Human mesenchymal stromal cells deliver systemic oncolytic measles virus to treat acute lymphoblastic leukemia in the presence of humoral immunity. Blood J Am Soc Hematol. 2014;123(9):1327–35.

    CAS  Google Scholar 

  187. Josiah DT, et al. Adipose-derived stem cells as therapeutic delivery vehicles of an oncolytic virus for glioblastoma. Mol Ther. 2010;18(2):377–85.

    Article  CAS  PubMed  Google Scholar 

  188. Jazowiecka-Rakus J, et al. Myxoma virus-loaded mesenchymal stem cells in experimental oncolytic therapy of murine pulmonary melanoma. Mol Ther Oncol. 2020;18:335–50.

    Article  CAS  Google Scholar 

  189. Jazowiecka-Rakus J, et al. Myxoma virus expressing LIGHT (TNFSF14) pre-loaded into adipose-derived mesenchymal stem cells is effective treatment for murine pancreatic adenocarcinoma. Cancers. 2021;13(6):1394.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Jazowiecka-Rakus J, et al. Combination of LIGHT (TNFSF14)-armed myxoma virus pre-loaded into ADSCs and gemcitabine in the treatment of experimental orthotopic murine pancreatic adenocarcinoma. Cancers. 2022;14(8):2022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Babaei A, et al. Mesenchymal stem cells loaded with oncolytic reovirus enhances antitumor activity in mice models of colorectal cancer. Biochem Pharmacol. 2021;190: 114644.

    Article  CAS  PubMed  Google Scholar 

  192. Banijamali RS, et al. Mesenchymal stem cells support delivery and boost the efficacy of oncolytic reoviruses in TC-1 tumor cells. J Cell Biochem. 2021;122(10):1360–75.

    Article  CAS  PubMed  Google Scholar 

  193. Wang X, et al. Mesenchymal stem cell carriers enhance antitumor efficacy induced by oncolytic reovirus in acute myeloid leukemia. Int Immunopharmacol. 2021;94: 107437.

    Article  CAS  PubMed  Google Scholar 

  194. Barlabé P, et al. Enhanced antitumor efficacy of an oncolytic adenovirus armed with an EGFR-targeted BiTE using menstrual blood-derived mesenchymal stem cells as carriers. Cancer Gene Ther. 2020;27(5):383–8.

    Article  PubMed  Google Scholar 

  195. Ho C-T, et al. Combination of mesenchymal stem cell-delivered oncolytic virus with prodrug activation increases efficacy and safety of colorectal cancer therapy. Biomedicines. 2021;9(5):548.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Keshavarz A, et al. Recent findings on chimeric antigen receptor (CAR)-engineered immune cell therapy in solid tumors and hematological malignancies. Stem Cell Res Ther. 2022;13(1):482.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Mardi A, et al. Biological causes of immunogenic cancer cell death (ICD) and anti-tumor therapy; combination of Oncolytic virus-based immunotherapy and CAR T-cell therapy for ICD induction. Cancer Cell Int. 2022;22(1):169.

    Article  Google Scholar 

  198. McKenna MK, et al. Mesenchymal stromal cell delivery of oncolytic immunotherapy improves CAR-T cell antitumor activity. Mol Ther. 2021;29(5):1808–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Darwish MM, et al. Prognostic implication of PD-L1 expression and associated tumor infiltrating lymphocytes in metastatic breast cancer. Immunopathol Persa. 2021;8(1):e18.

    Article  Google Scholar 

  200. Sorkhabi AD, et al. Cancer combination therapies by silencing of CTLA-4, PD-L1, and TIM3 in osteosarcoma. IUBMB Life. 2022;74(9):908–17.

    Article  CAS  PubMed  Google Scholar 

  201. Rahimi Kalateh Shah Mohammad G, et al. Cytokines as potential combination agents with PD-1/PD-L1 blockade for cancer treatment. J Cell Physiol. 2020;235(7–8):5449–60.

    Article  CAS  PubMed  Google Scholar 

  202. Varghese S, Rabkin SD. Oncolytic herpes simplex virus vectors for cancer virotherapy. Cancer Gene Ther. 2002;9(12):967–78.

    Article  CAS  PubMed  Google Scholar 

  203. Hoffmann D, Wildner O. Comparison of herpes simplex virus-and conditionally replicative adenovirus-based vectors for glioblastoma treatment. Cancer Gene Ther. 2007;14(7):627–39.

    Article  CAS  PubMed  Google Scholar 

  204. Msaouel P, Dispenzieri A, Galanis E. Clinical testing of engineered oncolytic measles virus strains in the treatment of cancer: an overview. Curr Opin Mol Ther. 2009;11(1):43.

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Lech PJ, Russell SJ. Use of attenuated paramyxoviruses for cancer therapy. Expert Rev Vaccines. 2010;9(11):1275–302.

    Article  CAS  PubMed  Google Scholar 

  206. Dai X-M, et al. Targeted disruption of the mouse colony-stimulating factor 1 receptor gene results in osteopetrosis, mononuclear phagocyte deficiency, increased primitive progenitor cell frequencies, and reproductive defects. Blood J Am Soc Hematol. 2002;99(1):111–20.

    CAS  Google Scholar 

  207. Künzi V, et al. Recombinant measles virus induces cytolysis of cutaneous T-cell lymphoma in vitro and in vivo. J Investig Dermatol. 2006;126(11):2525–32.

    Article  PubMed  Google Scholar 

  208. Peng K-W, et al. Systemic therapy of myeloma xenografts by an attenuated measles virus. Blood J Am Soc Hematol. 2001;98(7):2002–7.

    CAS  Google Scholar 

  209. Peng K-W, et al. Intraperitoneal therapy of ovarian cancer using an engineered measles virus. Cancer Res. 2002;62(16):4656–62.

    CAS  PubMed  Google Scholar 

  210. Phuong LK, et al. Use of a vaccine strain of measles virus genetically engineered to produce carcinoembryonic antigen as a novel therapeutic agent against glioblastoma multiforme. Cancer Res. 2003;63(10):2462–9.

    CAS  PubMed  Google Scholar 

  211. Blechacz B, et al. Engineered measles virus as a novel oncolytic viral therapy system for hepatocellular carcinoma. Hepatology. 2006;44(6):1465–77.

    Article  CAS  PubMed  Google Scholar 

  212. Liu C, et al. Prostate-specific membrane antigen retargeted measles virotherapy for the treatment of prostate cancer. Prostate. 2009;69(10):1128–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Yazdani A, et al. Evaluation of the urine mRNA-PCA3 expression level in prostate patients; comparison between benign prostatic hyperplasia and cancer. Immunopathol Persa. 2022;8(2):e15207–e15207.

    Article  Google Scholar 

  214. Iankov ID, et al. Demonstration of anti-tumor activity of oncolytic measles virus strains in a malignant pleural effusion breast cancer model. Breast Cancer Res Treat. 2010;122(3):745–54.

    Article  PubMed  Google Scholar 

  215. Wang B, et al. Deficiency of caspase 3 in tumor xenograft impairs therapeutic effect of measles virus Edmoston strain. Oncotarget. 2015;6(18):16019.

    Article  PubMed  PubMed Central  Google Scholar 

  216. Lv Y, et al. A recombinant measles virus vaccine strain rMV-Hu191 has oncolytic effect against human gastric cancer by inducing apoptotic cell death requiring integrity of lipid raft microdomains. Cancer Lett. 2019;460:108–18.

    Article  CAS  PubMed  Google Scholar 

  217. Hasanpour Dehkordi A, Keikhaei B, Bahadoram M, Razmjoo S, Barahman M. Keep the corners; impact of chemotherapy on renal function. J Nephropathol. 2020;9(1):e02.

    Article  Google Scholar 

  218. Gauvrit A, et al. Measles virus induces oncolysis of mesothelioma cells and allows dendritic cells to cross-prime tumor-specific CD8 response. Cancer res. 2008;68(12):4882–92.

    Article  CAS  PubMed  Google Scholar 

  219. Zhao D, et al. Live attenuated measles virus vaccine induces apoptosis and promotes tumor regression in lung cancer. Oncol Rep. 2013;29(1):199–204.

    Article  CAS  PubMed  Google Scholar 

  220. McDonald CJ, et al. A measles virus vaccine strain derivative as a novel oncolytic agent against breast cancer. Breast Cancer Res Treat. 2006;99(2):177–84.

    Article  CAS  PubMed  Google Scholar 

  221. Lampe J, et al. An armed oncolytic measles vaccine virus eliminates human hepatoma cells independently of apoptosis. Gene Ther. 2013;20(11):1033–41.

    Article  CAS  PubMed  Google Scholar 

  222. Donnelly OG, et al. Measles virus causes immunogenic cell death in human melanoma. Gene Ther. 2013;20(1):7–15.

    Article  CAS  PubMed  Google Scholar 

  223. Hall K, et al. Reovirus-mediated cytotoxicity and enhancement of innate immune responses against acute myeloid leukemia. BioResearch Open Access. 2012;1(1):3–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Chakrabarty R, et al. The oncolytic virus, pelareorep, as a novel anticancer agent: a review. Investig New Drugs. 2015;33(3):761–74.

    Article  CAS  Google Scholar 

  225. Ferhat M. Oncolytic viruses: the next major breakthrough in cancer treatment. J Hum Virol Retrovirol. 2017;5(1):00141.

    Google Scholar 

  226. Li J, Zhao J, Tan T, Liu M, Zeng Z, Zeng Y, Zhang L, Fu C, Chen D, Xie T. Nanoparticle drug delivery system for glioma and its efficacy improvement strategies: a comprehensive review. Int J Nanomed. 2020;15:2563–82. https://doi.org/10.2147/IJN.S243223.

    Article  Google Scholar 

  227. Babaei A, et al. In vitro anti-cancer activity of adipose-derived mesenchymal stem cells increased after infection with oncolytic reovirus. Adv Pharm Bull. 2021;11(2):361.

    CAS  PubMed  Google Scholar 

  228. Kaneko Y, et al. Bone marrow-derived stem cell therapy for metastatic brain cancers. Cell Transplant. 2015;24(4):625–30.

    Article  PubMed  Google Scholar 

  229. Khosravian M, Momenzadeh M, Koosha F, Alimohammadi N, Kianpour N. Lung cancer risk and the inhibitors of angiotensin converting enzyme; an updated review on recent evidence. Immunopathol Persa. 2022;8(1):e19.

    Article  Google Scholar 

  230. Yulyana Y, et al. Paracrine factors of human fetal MSCs inhibit liver cancer growth through reduced activation of IGF-1R/PI3K/Akt signaling. Mol Ther. 2015;23(4):746–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Lin HD, et al. Human umbilical cord wharton’s jelly stem cell conditioned medium induces tumoricidal effects on lymphoma cells through hydrogen peroxide mediation. J Cell Biochem. 2016;117(9):2045–55.

    Article  CAS  PubMed  Google Scholar 

  232. Lin W, et al. Mesenchymal stem cells and cancer: clinical challenges and opportunities. BioMed Res Int. 2019;2019:2820853.

    Article  PubMed  PubMed Central  Google Scholar 

  233. Block GJ, et al. Multipotent stromal cells are activated to reduce apoptosis in part by upregulation and secretion of stanniocalcin-1. Stem cells. 2009;27(3):670–81.

    Article  CAS  PubMed  Google Scholar 

  234. Sakaida I, et al. Transplantation of bone marrow cells reduces CCl4-induced liver fibrosis in mice. Hepatology. 2004;40(6):1304–11.

    Article  PubMed  Google Scholar 

  235. Wang L, et al. Hepatic vascular endothelial growth factor regulates recruitment of rat liver sinusoidal endothelial cell progenitor cells. Gastroenterology. 2012;143(6):1555-1563. E2.

    Article  CAS  PubMed  Google Scholar 

  236. Kim SU, et al. The Laennec staging system for histological sub-classification of cirrhosis is useful for stratification of prognosis in patients with liver cirrhosis. J Hepatol. 2012;57(3):556–63.

    Article  PubMed  Google Scholar 

  237. Spaeth EL, et al. Mesenchymal stem cell transition to tumor-associated fibroblasts contributes to fibrovascular network expansion and tumor progression. PLoS ONE. 2009;4(4): e4992.

    Article  PubMed  PubMed Central  Google Scholar 

  238. Feng B, Chen L. Review of mesenchymal stem cells and tumors: executioner or coconspirator? Cancer Biother Radiopharm. 2009;24(6):717–21.

    CAS  PubMed  Google Scholar 

  239. Du WJ, et al. Heterogeneity of proangiogenic features in mesenchymal stem cells derived from bone marrow, adipose tissue, umbilical cord, and placenta. Stem Cell Res Ther. 2016;7(1):163.

    Article  PubMed  PubMed Central  Google Scholar 

  240. Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016;16(9):582–98.

    Article  CAS  PubMed  Google Scholar 

  241. Ishihara S, et al. Mechano-signal transduction in mesenchymal stem cells induces prosaposin secretion to drive the proliferation of breast cancer cells. Cancer Res. 2017;77(22):6179–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Barcellos-de-Souza P, et al. Mesenchymal stem cells are recruited and activated into carcinoma-associated fibroblasts by prostate cancer microenvironment-derived TGF-β1. Stem cells. 2016;34(10):2536–47.

    Article  CAS  PubMed  Google Scholar 

  243. Martin F, et al. Potential role of mesenchymal stem cells (MSCs) in the breast tumour microenvironment: stimulation of epithelial to mesenchymal transition (EMT). Breast Cancer Res Treat. 2010;124(2):317–26.

    Article  CAS  PubMed  Google Scholar 

  244. Xue Z, et al. Mesenchymal stem cells promote epithelial to mesenchymal transition and metastasis in gastric cancer though paracrine cues and close physical contact. J Cell Biochem. 2015;116(4):618–27.

    Article  CAS  PubMed  Google Scholar 

  245. Shahverdi M, et al. The regulatory role of autophagy-related miRNAs in lung cancer drug resistance. Biomed Pharmacother. 2022;148: 112735.

    Article  CAS  PubMed  Google Scholar 

  246. Jafari M, et al. Tempol effect on epithelial-mesenchymal transition induced by hyperglycemia. J Nephropathol. 2017;6(1):1–4.

    Article  PubMed  Google Scholar 

  247. Karnoub AE, et al. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 2007;449(7162):557–63.

    Article  CAS  PubMed  Google Scholar 

  248. McAndrews KM, et al. Mesenchymal stem cells induce directional migration of invasive breast cancer cells through TGF-β. Sci Rep. 2015;5(1):16941.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Eckert F, et al. Potential role of CXCR4 targeting in the context of radiotherapy and immunotherapy of cancer. Front Immunol. 2018;9:3018.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Ma M, et al. Mesenchymal stromal cells may enhance metastasis of neuroblastoma via SDF-1/CXCR4 and SDF-1/CXCR7 signaling. Cancer Lett. 2011;312(1):1–10. https://doi.org/10.1016/j.canlet.2011.06.028.

    Article  CAS  PubMed  Google Scholar 

  251. Zemp FJ, et al. Cellular factors promoting resistance to effective treatment of glioma with oncolytic myxoma virus. Cancer Res. 2014;74(24):7260–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Peng K, et al. Using clinically approved cyclophosphamide regimens to control the humoral immune response to oncolytic viruses. Gene Ther. 2013;20(3):255–61.

    Article  CAS  PubMed  Google Scholar 

  253. Ferguson MS, Lemoine NR, Wang Y, Systemic delivery of oncolytic viruses: hopes and hurdles. Adv Virol. 2012. 2012.

  254. Gao Y, Zhang H, Lirussi F, Garrido C, Ye X, Xie T. Dual inhibitors of histone deacetylases and other cancerrelated targets: a pharmacological perspective. Biochem Pharmacol. 2020;182:114224. https://doi.org/10.1016/j.bcp.2020.114224.

    Article  CAS  PubMed  Google Scholar 

  255. Otsuki A, et al. Histone deacetylase inhibitors augment antitumor efficacy of herpes-based oncolytic viruses. Mol Ther. 2008;16(9):1546–55.

    Article  CAS  PubMed  Google Scholar 

  256. Cody JJ, Markert JM, Hurst DR. Histone deacetylase inhibitors improve the replication of oncolytic herpes simplex virus in breast cancer cells. PLoS ONE. 2014;9(3): e92919.

    Article  PubMed  PubMed Central  Google Scholar 

  257. Sadler AJ, Williams BR. Interferon-inducible antiviral effectors. Nat Rev Immunol. 2008;8(7):559–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  258. Jha BK, et al. Inhibition of RNase L and RNA-dependent protein kinase (PKR) by sunitinib impairs antiviral innate immunity. J Biol Chem. 2011;286(30):26319–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  259. Bell J, McFadden G. Viruses for tumor therapy. Cell Host Microbe. 2014;15(3):260–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  260. Moreno R, et al. Enhanced antitumor efficacy of Oncolytic adenovirus–loaded menstrual blood–derived Mesenchymal stem cells in combination with peripheral blood mononuclear cells. Mol Cancer Ther. 2019;18(1):127–38.

    Article  CAS  PubMed  Google Scholar 

  261. Thaci B, et al. Pharmacokinetic study of neural stem cell-based cell carrier for oncolytic virotherapy: targeted delivery of the therapeutic payload in an orthotopic brain tumor model. Cancer Gene Ther. 2012;19(6):431–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. Roth J, Curiel D, Pereboeva L. Cell vehicle targeting strategies. Gene Ther. 2008;15(10):716–29.

    Article  CAS  PubMed  Google Scholar 

  263. Arbab AS, et al. In vivo trafficking and targeted delivery of magnetically labeled stem cells. Hum Gene Ther. 2004;15(4):351–60.

    Article  CAS  PubMed  Google Scholar 

  264. Fiarresga A, et al. Intracoronary delivery of human mesenchymal/stromal stem cells: insights from coronary microcirculation invasive assessment in a swine model. PLoS ONE. 2015;10(10): e0139870.

    Article  PubMed  PubMed Central  Google Scholar 

  265. Silva LH, et al. Magnetic targeting as a strategy to enhance therapeutic effects of mesenchymal stromal cells. Stem Cell Res Ther. 2017;8(1):58.

    Article  PubMed  PubMed Central  Google Scholar 

  266. Kauer TM, et al. Encapsulated therapeutic stem cells implanted in the tumor resection cavity induce cell death in gliomas. Nat Neurosci. 2012;15(2):197–204.

    Article  CAS  Google Scholar 

  267. Shah K. Encapsulated stem cells for cancer therapy. Biomatter. 2013;3(1): e24278.

    Article  PubMed  PubMed Central  Google Scholar 

  268. Sanchez-Diaz M, et al. Biodistribution of mesenchymal stromal cells after administration in animal models and humans: a systematic review. J Clin Med. 2021;10(13):2925.

    Article  PubMed  PubMed Central  Google Scholar 

  269. Hammoud A, Sharay EY, Tikhomirov AN. Newtonian and non-Newtonian pulsatile flows through carotid artery bifurcation based on CT image geometry. In: AIP Conference Proceedings, Vol. 2171. AIP Publishing LLC. 2019. P. 110022.

  270. Schrepfer S, et al. Stem cell transplantation: the lung barrier. In: Transplantation proceedings. Elsevier. 2007.

  271. Rosenblum D, et al. Progress and challenges towards targeted delivery of cancer therapeutics. Nat Commun. 2018;9(1):1410.

    Article  PubMed  PubMed Central  Google Scholar 

  272. Hooshyar N, Hassanzadeh S, Nasri H. Acute and chronic kidney disease in lung cancer; an update. J Ren Endocrinol. 2018;4:e22.

    Google Scholar 

  273. De Becker A, Van Riet I. Homing and migration of mesenchymal stromal cells: how to improve the efficacy of cell therapy? World J Stem Cells. 2016;8(3):73.

    Article  PubMed  PubMed Central  Google Scholar 

  274. Yumul R, et al. Epithelial junction opener improves oncolytic adenovirus therapy in mouse tumor models. Hum Gene Ther. 2016;27(4):325–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  275. Turley EA, et al. Mechanisms of disease: epithelial–mesenchymal transition—does cellular plasticity fuel neoplastic progression? Nat Clin Pract Oncol. 2008;5(5):280–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  276. Christiansen JJ, Rajasekaran AK. Reassessing epithelial to mesenchymal transition as a prerequisite for carcinoma invasion and metastasis. Cancer res. 2006;66(17):8319–26.

    Article  CAS  PubMed  Google Scholar 

  277. Wojton J, Kaur B. Impact of tumor microenvironment on oncolytic viral therapy. Cytokine Growth Factor Rev. 2010;21(2–3):127–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  278. Kuriyama N, et al. Pretreatment with protease is a useful experimental strategy for enhancing adenovirus-mediated cancer gene therapy. Hum Gene Ther. 2000;11(16):2219–30.

    Article  CAS  PubMed  Google Scholar 

  279. de Heer P, et al. Cyclooxygenase 2 expression in rectal cancer is of prognostic significance in patients receiving preoperative radiotherapy. Clin Cancer Res. 2007;13(10):2955–60.

    Article  PubMed  Google Scholar 

  280. Mok W, Boucher Y, Jain RK. Matrix metalloproteinases-1 and-8 improve the distribution and efficacy of an oncolytic virus. Cancer res. 2007;67(22):10664–8.

    Article  CAS  PubMed  Google Scholar 

  281. Nagano S, et al. Cancer cell death enhances the penetration and efficacy of oncolytic herpes simplex virus in tumors. Cancer res. 2008;68(10):3795–802.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  282. Wong HH, Lemoine NR, Wang Y. Oncolytic viruses for cancer therapy: overcoming the obstacles. Viruses. 2010;2(1):78–106.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  283. Shen B, Hermiston T. Effect of hypoxia on Ad5 infection, transgene expression and replication. Gene Ther. 2005;12(11):902–10.

    Article  CAS  PubMed  Google Scholar 

  284. Shen B, Bauzon M, Hermiston T. The effect of hypoxia on the uptake, replication and lytic potential of group B adenovirus type 3 (Ad3) and type 11p (Ad11p). Gene Ther. 2006;13(12):986–90.

    Article  CAS  PubMed  Google Scholar 

  285. Hernandez-Alcoceba R, et al. New oncolytic adenoviruses with hypoxia-and estrogen receptor-regulated replication. Hum Gene Ther. 2002;13(14):1737–50.

    Article  CAS  PubMed  Google Scholar 

  286. Aghi MK, et al. Hypoxia enhances the replication of oncolytic herpes simplex virus. Mol Ther. 2009;17(1):51–6.

    Article  CAS  PubMed  Google Scholar 

  287. Fasullo M, Burch A, Britton A. Hypoxia enhances the replication of oncolytic herpes simplex virus in p53-breast cancer cells. Cell Cycle. 2009;8(14):2194–7.

    Article  CAS  PubMed  Google Scholar 

  288. Dinita Devi N, et al. Askin tumor: a case report of a rare tumor. Int J Sci Res Dent Med Sci. 2021;3(3):153–5.

    Google Scholar 

  289. Shariati Y, et al. Evaluate the clinical outcome of nanoparticle albumin-bound paclitaxel on breast cancer treatment: a systematic review and meta-analysis. Int J Sci Res Dent Med Sci. 2022;4(3):127–33.

    Google Scholar 

  290. Bagó JR, Sheets KT, Hingtgen SD. Neural stem cell therapy for cancer. Methods. 2016;99:37–43.

    Article  PubMed  Google Scholar 

  291. Zielske SP, Livant DL, Lawrence TS. Radiation increases invasion of gene-modified mesenchymal stem cells into tumors. Int J Radiat Oncol Biol Phys. 2009;75(3):843–53.

    Article  CAS  PubMed  Google Scholar 

  292. Kubo H, et al. Phase I dose escalation clinical trial of adenovirus vector carrying osteocalcin promoter-driven herpes simplex virus thymidine kinase in localized and metastatic hormone-refractory prostate cancer. Hum Gene Ther. 2003;14(3):227–41.

    Article  CAS  PubMed  Google Scholar 

  293. Alvarez RD, Curiel DT. A phase I study of recombinant adenovirus vector-mediated intraperitoneal delivery of herpes simplex virus thymidine kinase (HSV-TK) gene and intravenous ganciclovir for previously treated ovarian and extraovarian cancer patients. University of Alabama Comprehensive Cancer Center, Birmingham, AL. Hum Gene Ther. 1997;8(5):597–613.

    Article  CAS  PubMed  Google Scholar 

  294. Hu S, Hui Z, Lirussi F, Garrido C, Ye X, Xie T. Small molecule DNA-PK inhibitors as potential cancer therapy: a patent review (2010–present). Expert Opin Ther Pat. 2021;31(5):435–52. https://doi.org/10.1080/13543776.2021.1866540.

    Article  CAS  Google Scholar 

  295. De Graaf J, et al. Armed oncolytic viruses: A kick-start for anti-tumor immunity. Cytokine Growth Factor Rev. 2018;41:28–39.

    Article  PubMed  PubMed Central  Google Scholar 

  296. Pearl TM, et al. Oncolytic virus-based cytokine expression to improve immune activity in brain and solid tumors. Mol Ther Oncolytics. 2019;13:14–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  297. Twumasi-Boateng K, et al. Oncolytic viruses as engineering platforms for combination immunotherapy. Nat Rev Cancer. 2018;18(7):419–32.

    Article  CAS  PubMed  Google Scholar 

  298. Ocansey DKW, et al. Improved therapeutics of modified mesenchymal stem cells: an update. J Transl Med. 2020;18(1):42.

    Article  PubMed  PubMed Central  Google Scholar 

  299. Zheng M, et al. Oncolytic viruses for cancer therapy: barriers and recent advances. Mol Ther Oncol. 2019;15:234–47.

    Article  Google Scholar 

  300. Na Y, et al. Systemic administration of human mesenchymal stromal cells infected with polymer-coated oncolytic adenovirus induces efficient pancreatic tumor homing and infiltration. J Control Release. 2019;305:75–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  301. Moreno R, et al., Human menstrual blood-derived mesenchymal stem cells as potential cell carriers for oncolytic adenovirus. Stem Cells Int 2017. 3615729.

Download references

Acknowledgements

Not applicable

Funding

No Funders.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the conception and the main idea of the work. NGD, AIG, MEA, AOZ, MJA, RSZ, MAJ, SAJA, and JAR drafted the main text, figures, and tables. MD supervised the work and provided the comments and additional scientific information. YSA, NMM, YFM, and RAS also reviewed and revised the text. All authors read and approved the final version of the work to be published.

Corresponding author

Correspondence to Mohammad Darvishi.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

There is no conflict of interests.

Additional information

Publisher's Note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ghasemi Darestani, N., Gilmanova, A.I., Al-Gazally, M.E. et al. Mesenchymal stem cell-released oncolytic virus: an innovative strategy for cancer treatment. Cell Commun Signal 21, 43 (2023). https://doi.org/10.1186/s12964-022-01012-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-022-01012-0

Keywords