Open access peer-reviewed chapter - ONLINE FIRST

Transplantation of MSCs for a Longstanding Engraftment and Maintenance of Bone Marrow Stroma

Written By

Aleksei E. Bigildeev

Submitted: 06 May 2025 Reviewed: 22 May 2025 Published: 09 July 2025

DOI: 10.5772/intechopen.1011167

Mesenchymal Stem Cell - Biology, Therapeutics, and Beyond IntechOpen
Mesenchymal Stem Cell - Biology, Therapeutics, and Beyond Edited by Rui Damásio Alvites

From the Edited Volume

Mesenchymal Stem Cell - Biology, Therapeutics, and Beyond [Working Title]

Dr. Rui Damásio Alvites and Prof. Ana Colette Maurício

Chapter metrics overview

1 Chapter Downloads

View Full Metrics

Abstract

The modern concept of allogeneic hematopoietic stem cell transplantation (alloHSCT) implies the transfer of the donor’s hematopoietic system exclusively, the essence of which is the infusion of hematopoietic stem cells (HSCs) in the form of a single-cell suspension into the body of the recipient, who is previously subjected to intensive high-dose chemotherapy in order to eradicate tumor cells and destroy his own hematopoiesis, including immune cells (to exclude immunological conflict). This concept does not include transplantation of BM stroma, because the results of early works dating back to the 70–80s of the twentieth century spoke in favor of the fact that mesenchymal stem cells (MSCs) and their progeny are not capable of transplantation. Today, there are strong reasons to state that such an assumption is wrong. Despite the low engraftment efficiency of MSCs demonstrated in the early studies, alternative targeted strategies could lead to successful MSC transplantation. In this chapter, the question of the possibility of BM stroma transplantation is considered, arguments in favor of the expediency of co-transplantation of MSCs and HSCs in the therapy of blood system diseases are given, and a new concept of co-transplantation of BM stroma and the hematopoietic system is proposed.

Keywords

  • bone marrow transplantation
  • MSCs
  • MSC transplantation
  • MSCs and HSCs co-transplantation
  • alternative concept of bone marrow transplantation
  • bone marrow stroma damage
  • MSCs stable engraftment

Viam supervadet vadens

1. Introduction

Allogeneic BM transplantation (alloBMT) is widely used for the therapy of a whole range of malignant hematologic diseases, primarily acute leukemia and aggressive lymphomas, as well as other diseases of the blood system, such as myelodysplastic syndrome (MDS) and aplastic anemia (AA). It is often the only way to achieve long-term remission in a patient or even cure the disease. There are several graft sources available for alloBMT, namely primary whole BM (BM), BM primed with granulocyte colony growth factor (G-CSF), mobilized CD34+ hematopoietic stem cells (HSCs), and umbilical cord blood HSCs. The current trend in hematology is that mobilized CD34+ cells are used progressively more often than other sources. These cells are mobilized from the donor’s BM into the peripheral blood (PB) using several stimulants and collected by apheresis. Compared to the whole BM as a source of graft, this approach has a number of advantages, including low invasiveness for a donor, no traumatization of the donor, the ability to control the cellularity of the transplant by selecting the optimal protocols of mobilization and apheresis, and the ability to prepare for collection of the cells on an outpatient basis. At the same time, this approach excludes transplantation of the stromal component of BM. After infusion of donor CD34+ cells, the hematopoietic tissue of the BM and immune system is replaced by donor hematopoietic cells, but the stromal tissue of the BM remains the recipient’s own. There are a number of fundamental problems associated with this approach, since the recipient’s own stroma is allogeneic to the hematopoietic graft, partially damaged by previous chemotherapy, and its properties are altered by tumor cells, which may lead to ineffective reconstitution of hematopoiesis or poor graft function and even graft failure. Moreover, the preserved host stroma can support the survival of leukemia stem cells (LSCs), which are a source of relapses and refractory course of the disease. Thus, there are significant arguments in favor of transplanting not only hematopoietic but also stromal tissue of the BM from the same donor. This will reduce the incidence of allogeneic graft failures, relapses, and refractory course of the disease and, ultimately, will increase the long-term survival of patients or will lead to the cure of those diseases that are currently considered to be incurable. Successful BM stroma transplantation will also increase the treatment efficiency of diseases associated with bone, fat, and cartilage dysfunction, as early attempts to use mesenchymal stromal cells showed promising but short-term clinical responses [1, 2, 3].

Despite the high relevance of BM stroma transplantation, to date, among hematologists, there is an entrenched view that stroma transplantation is impossible. This opinion is based on the results of early studies dating back to the 70–80s of the twentieth century stating that mesenchymal stem cells (MSCs) and their progeny are not capable of transplantation, at least via intravenous injection in the form of a single-cell suspension. Today, there are strong reasons to state that such a view is wrong. In this chapter, the question of the possibility of BM stroma transplantation is considered, arguments in favor of the expediency of co-transplantation of MSCs and HSCs in the therapy of blood system diseases are given, and a new concept of co-transplantation of BM stroma and the hematopoietic system is proposed.

2. Early studies stating that BM stroma is not transplantable

There are several key seminal studies in which the question of BM stroma transplantability was considered, and the authors concluded that the answer is “no.”

Several studies on this subject are common in their design. They investigated donor chimerism in plastic-adherent stromal cell layers of long-term bone marrow cultures (LTBMCs) developed from the BM of patients with different hematologic malignancies after HLA-identical or compatible alloBMT from donors of the opposite sex [4, 5, 6]. The authors stated that stromal cells within LTBMCs were exclusively of host origin. An analogous approach was implemented in a mouse model [7]. The authors did not detect donor cells in the stromal sublayer of LTBMCs derived from the BM of mice that had been previously irradiated and then transplanted with BM from donor mice whose genome contained the marker chromosome T6.

Attempts to clarify the transplantability of BM stroma were also made later, using other methods, in particular, in situ hybridization of a probe specific to the Y chromosome. It was used in trepan biopsies of the BM from patients who underwent alloBMT from a related HLA-identical donor of the opposite sex [8]. BM trepan biopsies were obtained from patients on days 3–203 after infusion of donor BM, fixed in formalin, decalcified, and hybridized with a Y-chromosome-specific DNA probe, and the probe was further visualized using two independent methods, and its presence in stromal and hematopoietic cells was assessed by optical microscopy. The advantage of this study is that the assessment of stromal cells’ identity does not require in vitro cell culture under conditions far from physiologic, and it is possible to study cells in their natural anatomical environment, taking into account their morphology. The authors concluded that the cells of the stromal system (adipocytes, fibroblasts, osteoblasts, osteocytes, and endothelial cells) in BM grafts were of host origin. The authors hypothesized that engraftment of BM stromal cell precursors did not occur as a result of alloBMT, in which the source of cells is BM.

Thus, we state that in most of the studies published in the 70s and 80s of the twentieth century, both on animal models and in studies involving humans, it was shown that in BM transplantations, in which a single-cell suspension of total BM cells without separation into fractions of hematopoietic and non-hematopoietic cells was used as a source of graft, donor stromal cells were not detected either in the primary BM of recipients after alloBMT or in the stromal sublayers of LTBMCs obtained from such BM. Although these studies were conducted on a methodological level that corresponded to the level of technological development of that time, they had several methodological flaws, such as insufficient sensitivity and utilizing cultured cells instead of primary BM.

The given analysis of studies concerning MSC and BM stroma transplantation allows us to formulate several key theses, which are most often given as reasons why stroma transplantation is impossible:

  • Nonspecific retention of MSCs in the lungs immediately after injection;

  • Absence of MSCs in the allogeneic recipient early after injection;

  • Absence of the mechanism of MSC homing to the BM;

  • Loss of MSC functions due to cell dissociation;

  • Absence of immune privileges of stromal cells in MSCs;

Further, we will consider experimental data on which these theses are based, and we will give data from modern research that show that the given statements are erroneous or, at least, can be questioned, and obstacles on the way to of MSC transplantation can be overcome.

3. Re-evaluation of basic statements about the non-transplantability of BM stroma

3.1 Nonspecific retention of MSCs in the lungs immediately after injection?

The statement that MSCs are nonspecifically retained in lung tissue is based on the studies in which BM mesenchymal cells were labeled with a radioisotope marker and then injected intravenously into the recipient, and their fate was monitored [9]. The authors observed that after intravenous injection of labeled cells, which in this study were cultured mesenchymal stromal cells (which we will abbreviate hereinafter as “MCs,” and not primary mesenchymal stem cells of BM existing in vivo, which we denote here as “MSCs”), they, through the small circle of blood circulation, got nonspecifically stuck in the lungs, where they were detected 1 hour after intravenous injection. During the first 24 hours after infusion, MCs remained in the lungs, and after 72 hours, they could not be detected in the recipient’s body. The same authors subsequently published a review [10], in which they noted that most of the studies that investigated the ability of stromal cells to transplant used in vitro cultured MCs, which at that time were called “MSCs,” as an object of observation, and that the low ability of such cells to transplant may be associated with changes in their size, morphology, and surface phenotype, including activation of adhesion molecule expression as a result of in vitro cultivation. Indeed, it is known that cultivation of BM stromal cells on plastic leads to the fact that over time, small round-shaped cells turn into large spindle-shaped cells [11]. In another study, cultured MCs have been shown to become as large as 19 μm in diameter [12]. Since this diameter is larger than the size of pulmonary microcapillaries (5–10 μm), it is not surprising that such cells are retained in the lungs after intravenous injection. Besides, in one of the early studies, it was experimentally proved that pre-culturing reduces the ability of “MSCs” to migrate through the basal membrane [13], which creates difficulties for the migration of these cells. Considerations about the ratio of the linear size of cells and the diameter of microcapillaries in the lungs are important when considering the ability of MSCs to overcome this barrier, in light of the fact that “very small embryonic-like stem cells” (VSELs) are known. These cells have been isolated from mouse BM as a population of Sca-1+linCD45 cells expressing SSEA-1, Oct-4, Nanog, and Rex-1 markers characteristic of pluripotent stem cells. VSELs constitute ̴ 0.02% of BM mononuclear cells and are characterized by a small size (2–4 μm), smaller than the diameter of pulmonary capillaries [14]. In addition, VSELs express CXCR4 and thus are potentially capable of homing to the BM. A wide range of differentiation of VSELs has been demonstrated: they are able to differentiate into osteoblasts, cardiomyocytes, neurons, astrocytes, oligodendrocytes, Schwann cells, and insulin-producing cells [14, 15]. Thus, mouse BM contains a relatively rare cell population with osteogenic differentiation capacity that may escape the “lung trap.” It is possible that the BM contains other cell subpopulations with similar properties. Although no direct analog of VSELs has been found in adult humans, VSELs in humans are hypothetical, but we cannot exclude the theoretical possibility of discovering cells with similar properties in the future. For example, a population of small stem cells with diameters of up to 5 μm resembling VSELs was sorted recently from human embryonic stem cell (hESC) cultures [16]. These VSEL-like cells expressed stem-cell-related markers (CD133, SSEA-4) and markers of germinal lineage (DDX4/VASA, PRDM14). These cells were comparable to similar populations of small stem cells sorted from cell cultures of normal ovaries and were the predominant cells in the ascites of recurrent ovarian cancer.

3.2 Absence of MSCs in the organism of an allogeneic recipient in the early terms after injection?

The idea that MSCs may not survive for a long time after administration is based on the data indicating that most MSCs become apoptotic after administration [17], as well as on the results of early studies on the biodistribution of MCs [9]. However, several groups of investigators have shown in different kinds of model animals that MCs can be detected for long periods of time (up to several months) in a wide range of tissues, including BM, after systemic administration, even despite prior in vitro cultivation [18, 19, 20, 21, 22, 23, 24]. In one of these studies, PCR analysis showed that few donor murine cells were present in recipient mice after 1 week, but after 1–5 months, donor cells accounted for 1.5–12% of cells in bone, cartilage, and lung, as well as in BM and spleen [18]. The results demonstrated that at least some of the mesenchymal progenitor cells of BM expanded in culture can not only avoid arrest in lung microvessels and repopulate other organs and tissues but also survive long term in bone, cartilage, and lung and, more importantly, retain their functionality, since the authors were able to show that donor cells represented 2.5% of the chondrocytes isolated from the xiphoid and articular cartilage of the recipient. Interestingly, in these studies on the biodistribution of MCs, their frequency of detection and concentration in BM were often higher than in other tissues. This indicates the propensity of these cells to repopulate BM, which is not surprising if one assumes that the cells have a tropism for the microenvironment from which they were extracted. Recently, a systematic review has been published on the issue of MCs biodistribution [25], which shows that the biodistribution of MCs depends on the route of administration, that after intravenous administration MCs are detected in a wide range of organs and tissues, and that MCs are able to accumulate in damaged organs and tissues. In the majority of studies discussed, it is shown that the BM and spleen are the preferred sites of MCs homing. At the same time, the authors of a number of papers note that the efficiency of MCs homing into the BM is low. It is shown that the ratio of donor stromal cells is estimated as 0.1% of BM nuclear cells [18, 21, 22]. This may be due to the fact that the cited studies performed infusion of pre-cultured cells, and cultivation may result in a reduced ability of cells to invade through the vascular endothelial layer, as shown [13]. In those studies, where it is stated that mesenchymal cells are absent in the recipient’s organism after a short time, the sensitivity of the applied methods could be insufficient to observe rare cell subpopulations, which include MSCs (~0.01%–0.001% of nuclear BM cells).

It can be concluded that at intravenous injection of pre-expanded in vitro MCs, the majority of such cells are indeed retained in the lungs of the recipient for some time. However, this does not mean that rare cell subpopulations present in MC cultures, including cells resembling real MSCs by their properties, are also nonspecifically and irreversibly retained in the lung tissue. Low efficiency of homing, together with detection of cells in other organs and tissues, raised a question about the presence or absence of the mechanism of homing to the BM.

3.3 Lack of a mechanism for homing MSCs to BM?

An example of studies on which this statement is based is the article in which the arrest of MSCs in the lungs was shown [9]. In this study, the biodistribution of MSCs was monitored for 72 hours after intravenous injection of MSCs. At none of the investigated time points were injected MSCs detected in BM [9].

At the same time, by now there is a plenty of researches in which it is shown that intravenously injected stromal cells can, similarly to leukocytes, perform rolling and adhesion to vascular endothelial cells [26] and extravasate into various tissues, including BM, from the vascular channel through the basal membrane [27], although the number of cells migrating through the membrane is usually low [28]. In one of the already mentioned studies, the authors managed to detect eGFP-labeled cells in bones and BM of two recipients using PCR DNA ELISA analysis [19], and subsequently, the presence of MCs was detected in other organs and tissues, but using a more sensitive method [20]. The authors conclude that after intravenous infusion, MCs are predominantly localized in the BM, indicating the possibility of the existence of a mechanism of directed migration (homing) of these cells to the BM. Also, in another study, it was shown [29] that after intravenous injection of cells of stromal line GB1/6 to mice irradiated at doses of 3–8.5 Gy with additional irradiation of the right hind limb at a dose of 10–12.5 Gy, the injected cells were detected in situ in the hematopoietic sinuses of the recipient BM. Donor cells were undetectable 2 months after infusion in other organs—in the spleen, liver, and lungs. This indicates that intravenously infused stromal cells can infiltrate the vascular system of the BM and selectively accumulate in it [29].

Thus, both in animal models and in humans, it has been shown that after intravenous administration, stromal cells isolated from BM are capable of homing into their “native” BM niches, although with relatively low efficiency [28]. What limits the tropism and engraftment of MSCs in the BM remains not fully understood. There have been published papers showing the ability of cultured BM mesenchymal cells to migrate through the basal membrane and endothelial cell layer in model in vitro experiments and demonstrating the role of the metalloprotease MMP2, which is able to cleave type IV collagen (a major component of the basal membrane) in this process [27]. Interestingly, it is sufficient to culture primary BM-derived mesenchymal stromal cells for 24–48 hours for their ability to home to BM to be significantly reduced [13]. This information explains why, in many experiments on intravenous injection of multipotent mesenchymal stromal cells (MMSCs), it was not possible to observe significant homing of injected cells into the BM. At the time of publication of the described study, the mechanism of this phenomenon remained unknown, but much later it was found that MMSCs cultivation leads to a decrease in the expression of CXCR4 on their surface—a key molecule in the migration of various types of stem cells. The studies on the mechanisms of directed migration of MSCs and BM-MCs can be conditionally divided into those that show mobilization of stromal cells from BM into peripheral blood and those that study the mechanisms and pathways of migration of BM-MCs after their intravenous administration. It was noted that intravenously injected BM-MCs are able to accumulate in the sites of damage, hypoxia, and ischemia [30]. This ability of MMSCs was confirmed in myocardial infarction and ischemic brain injury [31, 32]. It has been hypothesized that proinflammatory cytokines, chemokines, including SDF-1α, and hypoxia factors, such as HIF1α, are involved in the mechanisms of MSCs migration to various tissues, and, in particular, to the BM, in which hypoxic conditions are maintained physiologically [30]. The following studies are devoted to this issue.

Among chemokines and their corresponding receptors, the SDF-1α/CXCR4 axis is a thoroughly studied system [30, 33, 34, 35, 36, 37]. This axis is known to be central to the homing of HSCs to the BM when administered intravenously [38], regulating the migration and homing of CXCR4+ hematopoietic progenitor cells, pre-B lymphocytes, and T-lymphocytes [33, 34, 35, 39]. More recent evidence suggests that in addition to HSCs, functional CXCR4 is expressed on the surface of various types of tissue-specific stem cells, such as neuronal stem cells [40], skeletal muscle satellite cells [41], liver oval cells [42], primordial germ cells [43], and other stem cell types including Muse cells and VSELs [44, 45].

There has been considerable evidence to suggest that the SDF-1α/CXCR4 axis is important for the migration of mesenchymal stromal/stem cells isolated from various sources [36, 37, 46, 47] and, more importantly, determines their ability to home to BM after intravenous administration [48]. Interestingly, data from previous studies suggested that a small subpopulation of cultured MSCs/MMSCs/BM-MCs initially maintains CXCR4 expression on the cell surface [48, 49], but upon ex vivo expansion, expression gradually declines and becomes barely detectable after prolonged cultivation [50]. This phenomenon may partly explain the fact that many researchers have failed to observe efficient repopulation of intravenously injected MSCs/MMSCs/BM-MCs after their preliminary expansion in vitro.

Kyriakou et al. showed that despite low surface expression of CXCR4, human MSCs/MMSCs/BM-MCs migrated in response to SDF-1α in vitro, and this was enhanced when CXCR4 was overexpressed in cells by lentiviral transduction [51]. It was also shown that artificial overexpression of CXCR4 in human adipose tissue stromal cells (hADSCs) helps to direct these cells to damaged tissues in response to SDF-1α signaling [36]. Chen et al. observed the same correlation both in vitro and in vivo for CXCR4-transduced murine MSCs transplanted into irradiated mice [52]. Moreover, Bobis-Wozowikz described that transplantation of CXCR4-transduced human MMSCs isolated from adipose tissue into NOD/SCID mice resulted in increased engraftment to BM [53]. Interestingly, CXCR4 overexpression caused a significant increase in homing to the BM and spleen of animals that had previously received irradiation [51, 52]. This indicates that prior damage to the BM stroma may be a prerequisite or at least a factor contributing to the engraftment of intravenously injected mesenchymal progenitor cells in the recipient. It has been shown that irradiation of an animal with ionizing radiation or exposure to other DNA-toxic agents leads to increased expression of SDF-1α in BM [54], and this may be one of the mechanisms behind the increased ability of MSCs to engraft in the BM of irradiated mice [55].

Enhanced survival and ability of MSCs to migrate in vitro in response to hypoxia and cultivation in the presence of SDF-1α have been demonstrated [46, 56]. The ability of BM-MCs to migrate along the SDF1a gradient was experimentally demonstrated in a mouse model of hypoxic-ischemic brain injury (HIB), and this effect was abrogated by the CXCR4 inhibitor (AMD3100) [30].

These prerequisites, namely the expression of CXCR4 in BM-MCs at basal level and its activation in response to hypoxia, the local increase in SDF1α levels under hypoxic conditions, which can occur as a consequence of ischemization or injury, are also physiological in BM [57], together with the ability of BM-MCs to accumulate in vivo in ischemic and injured tissues, were the grounds to suggest that the SDF-1α/CXCR4 axis is the key and determines the ability of homing to BM not only of HSCs but also MSCs or their progeny.

Thus, summarizing the presented data, we can conclude that the ability of MSCs to chemotaxis along gradients of SDF1α is well documented. This is primarily shown for the migration of MSCs to injury sites. Among other things, it has been shown that CXCR4 expression is increased on the surface of MSCs in response to several chemokines, including SDF-1α. Stromal cells have been shown to be able to migrate along the SDF-1α gradient and penetrate the endothelial barrier in the BM from peripheral blood. At the same time, the efficiency of MSCs homing into BM is usually low, which is associated by researchers with a low level of expression of receptors to chemokines on the surface of primary, uncultured MSCs; an even greater decrease of expression of these receptors during in vitro cultivation of MSCs/BM-MCs isolated from the organism; and heterogeneity of the BM-MCs population.

3.4 Loss of MSC functions due to cell dissociation?

One of the significant arguments that can be heard in favor of the view that transplantation of BM stroma is impossible is the notion that MSCs lose their functional abilities to regenerate stromal tissue after dissociation of intercellular contacts and transformation of BM into a single-cell suspension before intravenous administration to the recipient. The conclusion that stromal cells are not capable of transplantation in the form of a single-cell suspension was made by several independent, authoritative researchers in the 70–80s of the twentieth century who conducted experiments on animals. In similar studies by Ivanov-Smolensky and Friedenstein, BM explants from (CBA x C57BL/6)F1 mice were used to investigate this issue. F1 mice were lethally irradiated at a dose of 11 Gy and reconstituted with BM from CBA mice, and BM explants were obtained from the recipients. Colonies of fibroblasts grew from the explants, which, judging by isoantigens in the indirect immunofluorescence reaction with anti-C57BL/6 serum, were of recipient origin [58, 59]. The same conclusion about the impossibility of stroma transplantation was made by Chertkov [60, 61]. In 1983 the authors demonstrated that if BM is explanted in culture as a whole fragment without cell separation and then the stromal sublayer is collected and transplanted under the kidney capsule, an ectopic focus of hematopoiesis is formed in the implantation site, i.e., MSCs fulfill their function and build a full-fledged hematopoietic microenvironment [60]. However, if BM is transformed into a single-cell suspension before explanting into the culture, the cellular composition of the stromal sublayer of the culture is quite different, and, most importantly, when it is implanted under the kidney capsule of the recipient mouse kidney, a focus of ectopic hematopoiesis is not formed, i.e., the function of MSCs is lost due to the preliminary dissociation of cells. Then, 2 years later, following a largely similar scheme to the experiments of Ivanov-Smolensky et al. [59], Chertkov et al. studied the cells of the stromal sublayer of LTBMCs from the BM of radiation chimeras, which were obtained by irradiating mice with a lethal dose of radiation and reconstituting hematopoiesis by intravenous injection of donor syngeneic or allogeneic BM in the form of a single-cell suspension [61]. The origin of stromal cells in the LTBMCs of chimeric BM was studied using double fluorochrome staining with mouse lineage-specific antiserums. It was shown that the plastic-adherent cells of the LTBMCs of allogeneic chimeras contained only recipient stromal progenitor cells. The authors concluded that stromal progenitor cells cannot be transplanted intravenously in mice.

It is interesting to note that J. Chertkov and A. Friedenstein published the results of their other experiments, in which the strength of the basis of the hypothesis put forward is questionable. Thus, for example, in the same issue of Experimental Hematology, I.L. Chertkov publishes another article [62], in which a similar experiment of forming the foci of ectopic hematopoiesis from a whole fragment of BM and from a single-cell suspension of BM, which was pelleted by centrifugation on a small fragment of a filter, was conducted. The difference between this study and the study described above by the same authors was that in the latter study, the stage of seeding the BM cells in culture was omitted. In this arrangement of the experiment, foci of ectopic hematopoiesis were formed in both cases. To the surprise of the authors, the dissociation of BM cells not only failed to prevent the formation of foci, but other way round the proportion of donor cells in such foci was increased. Also, Friedenstein published a study in which it was shown that transplantation of fibroblasts in the form of a suspension under the kidney capsule led to the formation of elements of BM stroma, including bone, and to the presence of hematopoietic cells in the implants, which indicated that dissociation of cells in these experiments still did not lead to a complete loss of MSC function [63].

The statement that MSCs lose their functions after dissociation of intercellular contacts directly contradicts later studies, where it was shown that MSCs are able to migrate in the organism to the microenvironment of solid tumors and to the site of inflammation [64, 65, 66]. For example, MSCs have been shown to be selectively recruited to injured or inflamed tissues due to the release of multiple inflammatory factors by injured tissues [67]. This process, in particular, is essential for the resolution of infection and bone regeneration. The ability of MSCs to migrate is increased during acute inflammation, a process associated with the release of tumor necrosis factor (TNF) and activation of the NF-κB pathway [68, 69]. It is known that MSCs migrate from the BM to the gastric cancer microenvironment [70]. Thus, MSCs or some of their descendants are able to move around the organism as separate cells, accumulate in inflamed or damaged tissues, and actively participate in regeneration processes. Consequently, dissociation of BM cells before their administration to the recipient does not necessarily mean the cessation or decrease of MSC functionality.

3.5 Lack of immune privileges for MSCs?

The question of immune privileges (IPs) of MSCs and other stromal progenitor cells has a long history of study. In vitro studies of immunomodulatory properties of MMSCs have shown that these cells are able to inhibit proliferation and activation of T-lymphocytes during cocultivation, express MHC class I at a low level, and do not express MHC class II on their surface [71, 72, 73]. It has also been shown that these cells and the extracellular vesicles and individual proteins and other molecules secreted by them are able to reduce the activation of immunologic responses in a number of autoimmune pathologies and in graft-versus-host disease after alloHSCT [74, 75, 76].

However, it was subsequently found that pre-cultured BM-MCs do not possess immune privileges, and their main part causes immune responses in the organism and is eliminated by the immune system in MHC-incompatible transplantation [77, 78]. However, even in the case of cultured cells, the possibility that among the total cell population, there is a rare population of cells still possessing IPs cannot be excluded. Indeed, compelling experimental evidence for IPs of BM-MCs in vivo has subsequently emerged and includes studies performed in mice, large primates, and humans. We will describe here only a few of them.

Long-term presence of donor BM stromal cells in the body of recipients after alloHSCT has been demonstrated: in the already mentioned study of the ability of BM-MCs to home to BM in primates, in addition to autoBMT, MHC-incompatible allogeneic combined infusion of HSCs and genetically labeled BM-MCs was performed [19]. It was shown that markers of donor MCs were detectable in the BM up to +76 days. The same authors published another study on primates, in which they injected autologous GFP-labeled MSCs into a baboon previously lethally irradiated and recovered by its own hematopoietic cells mobilized into PB before irradiation and a baboon that had not been irradiated or exposed to any cytotoxic therapy [20]. MSCs expressing immunogenic protein GFP persisted in the recipient’s organism for several months (up to 21 months) without being eradicated by the immune system of the organism [20]. For human allogeneic MSCs, their long-term (for several months) presence in the BM of a recipient allogeneic to the donor after intraosseous injection of a suspension of donor MSCs has been demonstrated [79, 80].

The IPs of MSCs have proven to be so strong that they allow these cells to survive in a xenogeneic organism. One study demonstrated the presence of xenogeneic human MSCs in the BM of immunodeficient (nude) mice for several months after intravenous infusion [23]. In another study, the authors transplanted a well-characterized population of human MSCs to sheep embryos at the early stages of pregnancy, before and after the expected development of immunological competence [81]. In this xenogeneic system, human MCs engrafted and persisted in various tissues for 13 months after transplantation. The transplanted human cells underwent site-specific differentiation into chondrocytes, adipocytes, myocytes, cardiomyocytes, BM stromal cells, and thymus stroma. Long-term engraftment was unexpectedly recorded even when cells were transplanted after the development of immunocompetence.

Recently, it was shown that MSCs have pronounced IPs in vivo in adult mice: MSCs expressing immunogenic marker GFP survived and maintained their functionality for 6 weeks in immunologically complete adult mice [82, 83]. It is known that in vitro cultivation significantly changes the properties of cells extracted from the organism. For example, it has been shown that MMSCs cultivation leads to the induction of MHC expression, changes in the expression of genes encoding proinflammatory cytokines [73, 84], changes in the global DNA methylation and gene expression profile, and the occurrence of chromosomal rearrangements [85, 86]. As a consequence, it is legitimate to suggest that the immunological characteristics of cells in vivo and in vitro differ significantly.

Thus, it can be concluded that in mammalian BM, there are one or more distinct cell populations that have IPs and can be successfully transplanted to an MHC-compatible or incompatible recipient and survive in their body despite an active immune system.

If we think about stroma transplantation in patients as a component of alloBMT, we can assume that the probability of survival of these cells is much higher than in other settings, because at the moment of infusion of donor cells and for a certain period of time after it, the patient is in deep agranulocytosis, and the activity of the remaining circulating T-lymphocytes of the recipient is significantly reduced. Humoral immune response to MSCs and their differentiating progeny is also unlikely in such a situation. If combined transplantation of MSCs and HSCs is successful, the recipient becomes a “chimera:” his hematopoiesis and immune system become donor and, consequently, syngeneic with injected MCs, so there is no reason for a systemic immune response to injected donor MCs. However, local immune reactions presumably can be observed due to the preserved resident macrophages of the recipient residing in organs and tissues and residual circulating T-lymphocytes.

It can be concluded that for today there are reliable and numerous experimental confirmations that at intravenous injection of whole BM cell suspension containing true multipotent, not subjected to pre-cultivation MSCs, the latter are able to survive for a long time in the recipient’s organism, migrate into BM using specialized molecular mechanisms, and perform their direct function—to participate in regeneration and maintenance of BM stromal tissue.

4. New concept of bone marrow transplantation that includes a change of both stromal and hematopoietic components to donor origin

At the same time, there were studies that testified in favor of the fact that transplantation of elements of the stromal component of BM is still possible. This was demonstrated by independent teams of researchers for small mammals (mice), large mammals (primates, dogs), and humans [13, 28, 29, 55, 87, 88, 89, 90, 91].

MSCs are an integral component of BM stroma and can be considered its key element. Such perception of MSCs is generally accepted because these cells are capable of de novo formation of a full-fledged hematopoietic territory with appropriate spatial structure and biochemical composition, containing all the necessary cellular elements of the stroma, including adipocytes and their precursors adipoblasts, osteocytes and their precursors osteoblasts, reticular cells, pericytes, and other types of stromal cells [92, 93, 94, 95]. To date, a number of key properties of MSCs have been demonstrated, namely:

  • the ability of donor allogeneic MSCs to survive in the recipient’s body for a long period of time; and in the recipient’s body [18, 19, 20, 21, 22, 23, 24];

  • the ability of intravenously injected MSCs to repopulate the BM due to specialized molecular mechanisms [19, 27];

  • the ability of MSCs to maintain their functionality (to differentiate in the osteogenic direction and proliferation) after their dissociation and intravenous injection in the form of a single-cell suspension into the recipient’s body [55];

  • immune privileges of MSCs, i.e., their ability to survive despite the recipient’s immune system being activated against their antigens [82, 83];

This creates a fundamental basis for the development of protocols for effective MSC transplantation and BM stroma replacement in patients with hematologic diseases, including oncology, as well as in patients with skeletal diseases, including genetic diseases. Successful BM stroma transplantation also has the potential to improve the effectiveness of treatment of diseases associated with dysfunction of bone, fat, and cartilage tissue [1, 2, 92]. However, given the failure of numerous studies of BM stroma transplantation attempts described above, what might be the key to success?

Many researchers note the ability of MSCs to migrate to the areas of ischemic and injured tissues, but none of the reviewed studies, in particular in the field of hematology, proposes to increase the efficiency of homing and engraftment of MSCs into the recipient’s BM during alloBMT by targeted and intentional damage of BM stromal tissue before infusion of donor BM. Meanwhile, it can be one of the reasonable tactics, since the function of MSCs is to regenerate the damaged tissue, first of all, the stromal tissue.

I present here a new concept of BM transplantation that includes transplantation of both MSCs and HSCs from the same donor, where transplantation means not only injection of cells but also their stable engraftment and participation in regeneration and maintenance of the corresponding tissue. The central idea of this concept is the necessity of sufficient damage to the BM stroma in the recipient before BM graft infusion. There are preclinical studies that suggest MSCs may constitute a powerful means of treating tissue lesions caused by ionizing radiation, either after accidental exposure to radioactivity or as a side effect of clinical radiotherapy. Animal studies and early clinical experiences suggest a role for MSCs in the regeneration of these tissue lesions, both by differentiating into functional parenchymal cells and by creating a nurturing microenvironment for other cells [96]. We have our own experimental data demonstrating that prior severe damage to the recipient BM stroma is a necessary condition of effective and lasting engraftment of donor MSCs. However, more research is needed to fully reveal the regenerative potential of MSCs in the BM [97].

4.1 The strategy

This alternative concept consists of the following key steps/stages (Figure 1).

Figure 1.

The main steps of the alternative strategy for sequential co-transplantation of BM stroma and HSCs.

A hypothetical timeline for the four-step protocol is as follows. Eradication of BM stroma could be done after induction therapy and simultaneously with consolidation chemotherapy. Infusion of donor MSCs should be done in advance, for example, at day-14, in order to give the cells some time to home to the BM, engraft, and reconstitute BM stroma. Infusion of HSCs on day 0. Supportive and regenerative therapy for at least 3 months after HSCs infusion. This timeline is speculative and theoretical; a real-time course should be established in future experiments.

The first step is “Eradication of the recipient’s BM stroma.” The goal of this step is to eradicate the host’s MSCs in the BM and/or significantly damage the patient’s BM stroma. This is necessary so that the donor MSCs, which will be injected into the recipient at the next step, would get into the organism in which the function of the BM stroma is lost/damaged and, reacting to this, carry out their direct duty—to regenerate the lost/damaged tissue. A direct analogy can be made here with alloHSCT. This procedure is aimed not only at the elimination of tumor cells but also at almost complete destruction of the patient’s own hematopoiesis, including his immune system, as well as emptying hematopoietic niches in the BM. Destruction of the immune system and emptying of hematopoietic niches create favorable conditions for the introduced donor’s HSCs to engraft by populating empty niches without experiencing pressure from the recipient’s immunity, activating their regenerative program, and fully restoring hematopoiesis. It is likely that successful engraftment of donor HSCs and reconstitution of hematopoiesis is not in the least due to the fact that HSCs enter the body with the lost/damaged hematopoietic tissue and activate their regenerative function to replenish it. At this step, various strategies and approaches can be used, the detailed development of which is still to come. One such approach is nonspecific stromal damage by radiation or polychemotherapy (PCT). Several studies that have demonstrated successful engraftment of the donor’s stroma have used high doses of radiation [29, 89], so in principle, this approach is feasible. However, it should be remembered that stromal cells are much more resistant to ionizing radiation [96, 98] than hematopoietic cells. This means that if ionizing radiation is considered as a factor capable of damaging MSCs and applied as TBI, it may require too high doses of radiation to effectively suppress the stromal component of BM. The side effects of using such doses may be acute radiation sickness, high cytotoxicity, and the occurrence of secondary tumors. It is doubtful that in the case of TBI it will be possible to achieve doses that would, on the one hand, lead to destruction of BM stroma or at least MSCs within the stroma and, on the other hand, be acceptable in terms of toxicity/complications profile. Radiation therapy is actively developing, and methods of precision irradiation of cells with a precision of up to 1 millimeter are available today, which is a promising alternative to TBI. The development of 3D-4D conformal radiation therapy approaches, such as CyberKnife radiosurgery technology, which is used for radiosurgical treatment of solid tumors, primarily brain tumors and vascular malformations, has become more accessible nowadays. This technology allows irradiation of areas with an accuracy of less than a millimeter and, importantly, affords continuous tracking of the target position and correction of the course of irradiation when the target shifts with respiration or when the patient moves intentionally or not. This makes it possible to selectively and precisely apply ionizing radiation to all areas where BM is concentrated—bones of the skull, ribs, spine, pelvis, and epiphyses of limb bones. Another method of stromal eradication may be the administration of chemotherapeutic drugs that have a cytotoxic effect on stromal cells. It is difficult to think of any significant selectivity with respect to them, although it can be assumed that those drugs used for the treatment of malignant bone tumors and, in particular, osteosarcomas—tumors arising from osteoblasts—may be more effective here than other substances [99]. These drugs include cisplatin, doxorubicin, ifosfamide, etoposide, epirubicin, and methotrexate [100]. This list is certainly not exhaustive; additional information can be drawn from the Russian national clinical guidelines for the treatment of malignant bone tumors or the US National Comprehensive Cancer Network (NCCN) national clinical cancer guidelines [100]. Other chemotherapeutic agents with distinct effects on stromal cells can be selected in functional tests on stromal cell lines (e.g., OP-9) [101, 102], fibroblasts [103], or on primary cultures of BM-derived MMSCs, adipose tissue, or other sources [104], although it is most appropriate to test drugs specifically on primary, previously uncultured BM stromal cells, since this tissue is the therapeutic target in this case, and cultured MSCs isolated from different sources other than BM vary in their ability to proliferate, differentiate, migrate, and secrete extracellular matrix, which may result in different sensitivity to the drugs being tested [105], and cell culturing itself leads to changes in cell characteristics, as mentioned above. It is also worth noting that the tumor microenvironment (TME) is currently the focus of attention of oncologists and oncology researchers, and there are actively developing therapeutic approaches that target TME. Tumor-associated fibroblasts (CAFs) are one of the significant components of TME, and attempts are being made to targetedly eradicate these cells in order to improve the effectiveness of the treatment of primary and metastasizing solid tumors [106]. The most effective drugs, which were discovered in these studies, can be considered as promising candidates for the eradication of BM stromal cells within the proposed new approach to alloBMT and treatment of stromal tissue diseases. Another therapeutic tactic for stromal eradication may be targeting certain cell subpopulations, in particular, MSCs in BM or their differentiated progeny involved in niches for HSCs, such as osteoblasts. Such tactics require knowledge about the surface markers specific to these cells. Unfortunately, to date, there is no unambiguous opinion on the surface immunophenotype of BM MSCs: different researchers mention different surface markers and believe that cells expressing them satisfy the definition of MSCs. Some authors argue that MSCs are cells expressing Lepr and Cxcl12 [107]. It has also been shown that the population of stem/progenitor mesenchymal cells in BM is significantly enriched in a subpopulation of CD271+ cells [108, 109]. Other researchers consider MSCs to be pericytes characterized by the expression of smooth muscle α-actin (Acta2) [107, 110]. Finally, CD45Nes+ cells are considered MSCs [93]. They are exclusively located along BM arterioles and are a component of hematopoietic niches that regulate the quiescent state of HSCs [111]. The mentioned subpopulations can partially overlap with each other [111], but the ability to transfer hematopoietic activity in vivo has been clearly demonstrated so far only for Nes+ MSCs [93]. In addition, previously described Muse cells are characterized as SSEA3+ cells [112], and VSEL cells are described as Sca1+CXCR4+SSEA1+linCD45 [14]. By definition, MSCs are capable of differentiating in at least three classical directions: osteogenic, adipogenic, and chondrogenic. This ability ensures the formation and maintenance of the basic cellular elements of the BM stroma. As a result of MSC differentiation in the osteogenic direction, osteoblast-osteocyte precursors are formed, the main function of which is the formation of new bone tissue due to active secretion of extracellular matrix substances. Some studies claim that osteoblasts are direct participants of niches for HSCs and their progeny in the BM [113, 114, 115]. Considering the important role of osteoblasts in HSC maintenance, a special term, “osteoblastic niche” of HSCs, was even introduced. However, it should be noted that in a number of papers, authors have disputed the importance of osteoblasts in HSC maintenance [116]. Osteoblasts are exclusively present in BM, lining the inner surface of bone. Osteoblasts are characterized by the expression of the surface membrane proteins CD10 [117], CD44 [118], CD46, CD55, and CD59 [119], as well as the recently discovered A7 antigen, which has strong specificity [120]. There is reason to suggest that osteoblasts are a favorable target for chemotherapy aimed at damaging the stroma as part of the preparatory step before its transplantation, because osteoblasts express a number of soluble extracellular matrix proteins, such as osteocalcin, osteopontin, procollagen, procollagen peptidase, and bone sialoprotein. The content of these proteins can be measured in peripheral blood, and their levels can serve to assess the efficiency of osteoblast destruction along with soluble forms of the osteoblast surface antigens described above. Moreover, since osteoblasts are present specifically in bones, while MSCs are distributed throughout the body, this makes the former a more specific target, and their destruction, presumably, may have fewer undesirable side effects on the body than the destruction of MSCs. Thus, targeted chemotherapy can be directed at osteoblasts either separately or in addition to targeted therapy directed at MSCs. The arsenal of methods for targeting specific cell types is currently quite wide and includes not only chemical agents but also various types of antibodies (monoclonal antibodies, bispecific antibodies, and mini-antibodies conjugated to a cytostatic agent) [121, 122], and T-lymphocytes or NK cells with a chimeric T cell receptor (CAR-T, CAR-NT cells, respectively) engineered to specifically recognize a particular target [123, 124, 125, 126]. Such CAR-T or CAR-NT cells can be generated against a single target in a cell population selected for exposure. CAR-T and CAR-NT cells can be used in cocktails for a more efficient cytotoxic effect against a selected cell subpopulation [127].

Various combinations of nonspecific and targeted drugs directed at different types of target cells and at different antigens on the surface of these cells are possible. This part requires detailed development in preclinical studies.

Theoretically, such a strategy can radically transform the schemes of induction and consolidation therapy before alloBMT: such schemes and regimens can be developed that will be primarily directed not so much at hematopoietic, tumor, and immune cells but at BM stroma. No niches for HSCs—no hematopoiesis and no leukemia. At the same time, it is likely that PCT regimens will retain drugs aimed at reducing the total mass of leukemia cells in the blood/BM in order to reduce the negative impact of these cells on the body. An additional option for promising regimens may be to affect T regulatory cell (Treg) populations in the recipient, as it is these cells, together with MSCs, that confer immune privileges to HSCs in the BM, keeping the latter dormant, and thus may contribute to the persistence of leukemia stem cells in the BM [128, 129]. On the other hand, Tregs may affect the manifestation and severity of GVHD [130], so a balance between benefit and risk must be maintained when targeting Tregs.

The second step of the proposed concept is “Preparing the foothold,” which is an infusion of donor whole BM cell suspension or selected subpopulations of MSCs. The aim of this stage is to repopulate the vacant BM territory with donor MSCs so that they begin to recreate the hematopoietic microenvironment in the BM and build niches for HSCs. It is important to emphasize that the aim of this stage is not to reconstitute hematopoiesis at the expense of donor hematopoietic cells. The efficiency of engraftment of donor HSCs and, as a consequence, donor chimerism of hematopoietic tissue will be low due to the fact that at this time point in the recipient’s body, there will be significantly damaged or destroyed niches for them. Consequently, at this stage, not the whole BM but only its stromal component or separate stromal cell subpopulations, in particular, MSCs, if it is possible to isolate them in any way, are feasible to be infused. On the other hand, infusion of whole BM implies infusion of various subpopulations of immune cells, which may support the recipient’s immunity for some time at the expense of granulocytes and memory lymphocytes.

The third stage is “Airborne.” This stage consists of the infusion of donor HSCs, as is currently done in alloHSCT. This (second) infusion should probably be performed some time (what time, it is to be determined) after the first one so that MSCs have time to build new niches for HSCs. This will allow the HSCs to inhabit the niches/territories recreated for them and engraft effectively. Probably due to the fact that the stroma recovery may not be complete or the efficiency of its transplantation will not be too high, it will be necessary to adjust the therapeutic doses of CD34+ cells administered to the recipient toward their increase.

The fourth stage is “Support/Supply Pathway Establishment.” Damaging the BM stroma can have serious negative consequences for the patient’s body. The injected donor MSCs may not be able to cope effectively with the task of building a new hematopoietic territory throughout the body. They may need support. What kind of support is the best, should be subject of further research. It could be supportive infusions of CAR-MSCs, which have pronounced homing ability to BM and significant immune modulatory effects [131, 132], or agents that stimulate regeneration of nerves in the BM [133], or chemotherapeutic agents used for prophylaxis and treatment of GVHD.

At each of these stages, it is advisable to utilize factors that can increase the efficiency of BM stromal transplantation; their combined application can result in a significantly increased likelihood of high-quality, stable, complete BM stromal transplantation. These factors may include effective eradication of the recipient BM stroma. It has been shown that irradiation of an animal with ionizing radiation or exposure to another DNA-toxic agent, such as 5-fluorouracil (5-FU) or cyclophosphamide (Cy), leads to increased expression of SDF-1 in the BM [54]. The ability of BM-MCs to migrate along the SDF1a gradient was experimentally demonstrated [30]. Together with overexpression of CXCR4 on BM-MCs, it could facilitate the engraftment of injected cells [52]. Another factor that can increase the efficiency of MSC transplantation is the introduction of an increased amount of these cells. In those studies, in which it was possible to demonstrate long-term presence of donor stromal cells in the recipient’s organism, the authors attributed the success to the increased dose of injected cells [28, 55, 89, 90]. Another factor that is likely to be significant in the engraftment of stromal cells is the use of vasodilators. It has been shown that intravenous injection of rat BM-MCs after vasodilator administration increased the efficiency of accumulation of radioactive label embedded in the cells by 50% compared to a group that was not injected with a vasodilator [21]. Finally, after infusion of mesenchymal cells into the recipient, it is advisable to consider supportive therapy that will promote engraftment of the injected cells, their proliferation, and differentiation for complete reconstruction of the stromal component of the bone marrow. Many cytokines have a stimulatory effect on mesenchymal progenitor cells. For example, it has been shown that the basic fibroblast growth factor FGF2 stimulates the proliferation of BM-MCs and their differentiation into tenocytes [134]. Transforming growth factor beta (TGFb) takes part in adipogenic differentiation of MSCs [135]. Also, immunity factors have an important influence on MSCs. For example, it has been shown that interleukin-1 beta is a growth factor for stromal progenitor cells [136, 137], and tumor necrosis factor (TNF) and interferon gamma can influence the immunomodulatory properties of BM-MCs [138, 139]. It is reasonable to consider the possibility of supportive therapy aimed at the regeneration of BM stroma and hematopoiesis. Sensory neuronal signaling is known to modulate the functionality of MSCs and osteoclasts in BM [140]. It has been shown that active proliferation of tumor cells in BM in acute myeloid leukemia (AML) has a deleterious and persistent effect on the sympathetic neuronal component of HSC niches in human BM, which can lead to long-term hematologic dysfunction [133].

4.2 Risks of the proposed strategy

Despite the considerations presented about the potential benefits of BM stroma transplantation, it is necessary to evaluate all possible risks to the patient and to ensure that the safety and clinical efficacy of this procedure are appropriate. The risks associated with the proposed procedure of MSC transplantation after prior eradication therapy are highlighted in this chapter. Given the need for high-dose radiation therapy and/or potential intensification of induction and consolidation protocols to achieve profound damage to the stromal component of the BM, the risks of secondary tumors, a higher incidence of infectious complications, and associated mortality as well as some other risks should be evaluated [141, 142, 143].

4.2.1 Risks associated with damage to the BM stroma and failure of donor MSC engraftment

Potential transplantation protocols will require approaches that would result in significant damage and possibly even the eradication of the recipient’s BM stroma. And if no engraftment of donor MSCs and regeneration of BM stroma follow, there will be no engraftment of hematopoietic tissue due to a lack of necessary niches for HSCs. This may result in extremely severe complications, long rehabilitation, and even a lethal outcome for the patient. There is also a risk of such undesirable phenomena as osteogenesis disorder with the formation of severe progressive osteoporosis (progressive bone loss). The risk of immune rejection of MSCs is another factor that should be addressed. To minimize these risks, it is important to establish and develop new criteria for donor selection for MSCs and HSCs co-transplantation and methods that facilitate BM stroma regeneration [131, 132, 133].

4.2.2 Risk associated with MSC-driven support of tumor cells

Tumor recurrence is one of the leading problems that arise after alloHSCT. Here, the risk of MSC application is connected with the fact that these donor cells can perform not only antitumor function and replace tumor-remodeled stroma with a healthy microenvironment but also get involved in the tumor microenvironment again, preserving and stimulating malignant cells [144, 145, 146]. Some effects exerted by MSCs may depend on the histological type of the tumor and its molecular characteristics, while others may be similar for different tumor types [147, 148]. For example, several studies have shown that MSCs exert antitumor effects on breast and lung cancer cell lines in vitro [149, 150] and on pancreatic tumors in vivo [151]. However, other studies demonstrate that, in contrast, MSCs promote proliferation of breast cancer and melanoma cells when co-cultured with tumor cell lines in vitro [152, 153]. They also enhance tumor growth when administered to mice with lung or prostate cancer [154, 155]. Most of the work in the field of investigating the effect of MSCs directly on tumor cells and on the progression and metastasis concerns solid tumors [148, 156, 157], but the risks and benefits of MSCs have also been investigated in the field of malignant hematologic diseases [143, 158, 159, 160]. In addition, MSCs isolated from the BM of a healthy donor can induce drug resistance of hematopoietic tumor cells, both cell lines and primary tumor cells, including CML, OML, and T- and B-ALL cells [161, 162, 163, 164, 165, 166, 167].

This determines an important principle of using MSCs in clinical practice during their transplantation as a part of the strategy for joint sequential transplantation of BM stroma and hematopoietic tissue: MSCs should be used for the purpose of BM stroma transplantation, if possible, during complete remission of the underlying disease.

4.2.3 Spontaneous transformation of stromal cells in culture

The possibility of spontaneous transformation of mesenchymal cells themselves cannot be discounted. Intense ex vivo expansion of any cell type can lead to malignant transformation through a selection of rapidly dividing cells, which increases the risk of accumulation of genetic and epigenetic alterations and may ultimately lead to spontaneous aberrant transformations. BM-MCs cultured in vitro are no exception, although data regarding the resistance of these cells to the accumulation of genetic abnormalities in culture and the probability of their spontaneous transformation are contradictory. Some studies claim that long-term cultivation does not reveal chromosomal abnormalities and telomerase activity up to 60 population doublings [168, 169], and does not lead to tumor formation when transplanted into immunodeficient mice [169, 170]. In other studies, the authors report that after prolonged ex vivo expansion, human mesenchymal stromal cells derived from adipose tissue or BM were prone to malignant transformation, had an increased proliferation rate, acquired an altered immune phenotype and cell morphology compared to typical BM-MCs, and carried cytogenetic abnormalities [171, 172].

This allows for the formulation of one more principle of MSC use: for BM stroma transplantation, it is advisable to use primary, not pre-cultured MSCs or BM-MCs, minimally expanded in culture.

4.2.4 Tumors from primary uncultured MSCs

It should also be kept in mind that tumors originating in the body from cells of mesenchymal nature are known, namely malignant tumors of bone, such as osteosarcoma, Ewing’s sarcoma, chondrosarcoma, fibrosarcoma of bone, and others [100]. These diseases are caused by different populations of BM stromal cells that are descendants of MSCs, such as osteoblasts in the case of osteosarcoma [173, 174]. The frequency of such tumors is relatively low, but still, before using MSCs and their derivatives for the purpose of their transplantation from a donor, it is necessary to have criteria that can confirm that normal, non-pathological cells are being injected. At least, it is required to exclude diagnoses with involvement of donor mesenchymal cells in tumorigenesis, and it is better to test MSCs for genetic abnormalities associated with the risk of tumor transformation of these cells.

4.2.5 Secondary tumors as a result of high-dose radiation therapy

It is known that radiation therapy carries the risk of developing not only acute radiation sickness but also secondary tumors [175, 176, 177]. In addition, mesenchymal cells and MSCs in particular are much more resistant to the effects of radiation than HSCs [98]. Therefore, the use of TBI to damage MSCs would be associated with high doses that would be necessary to achieve the desired effect, and this in turn is associated with high toxicity and the likelihood of secondary tumor development. Therefore, this approach appears to be ineffective. To overcome these difficulties and increase the efficacy and safety of MSC eradication in the recipient, it is necessary to develop targeting and low-toxic ways to target MSCs and other stromal cell subpopulations of BM, such as 3D and 4D conformal radiation therapy, CAR-T, CAR-NK cells targeting MSCs, monoclonal antibodies, and their conjugates with cytotoxic drugs.

4.2.6 MSCs as potential carriers of infections

Another aspect that should be considered when using MSCs in clinical practice is the possibility of their infection by various pathogens. It is known that part of MSCs in vivo are pericytes that are in close contact with blood vessels [178], making MSCs ideal guardians on the path of infection, and at the same time, targets for viruses. It has been shown that MSCs can be infected by various members of the Herpesviridae family, H5N1 avian influenza virus, Zika virus, and others [179]. These reservoirs are difficult to eliminate by the immune system, as BM MSCs have been shown to have immune privileges [83], which may be part of the immune-tolerant microenvironment of the BM [128, 157]. This allows for the formulation of one more principle of MSCs utilization: before MSCs introduction into the recipient’s organism, it is necessary to test the cells for pathogen infection.

Summarizing the data presented in this section, it can be stated that the use of BM-MCs and MSCs in clinical practice is associated with a number of significant risks. These risks can be minimized by using primary MSCs that have not undergone ex vivo expansion in culture, as well as by using MSCs in complete remission of malignant disease of the blood system. An alternative strategy of BM transplantation involving combined sequential transplantation of MSCs and HSCs does not involve prior ex vivo culturing of MC-MSCs and therefore avoids the risks associated with the accumulation of genetic abnormalities in culture. Nevertheless, regardless of the preliminary cultivation, before transplantation of donor MSCs, they should be carefully tested for the presence of genetic abnormalities and infectious agents, even in the case of a healthy donor, in whose blood no infection is detected.

5. Conclusion

Science, and medical science in particular, is not only the expansion of knowledge about the world and, in particular, about the principles of functioning of the human body and other living beings, but also the narrowing of the realm of the impossible. What was considered impossible a few decades ago is now becoming not only achievable but even routine. Those diseases that were considered incurable are gradually losing their status. The successes of surgery, transplantology, and other branches of medicine are vivid examples of this. In my opinion, now it is time to raise the question of whether the combined transplantation of BM stromal and hematopoietic tissue for the purpose of therapy of malignant and non-malignant diseases of the blood system and skeletal diseases is really impossible. A lot of studies performed on various animal species and on humans show that all major fundamental obstacles for this are absent. For example, it was shown that MSCs are able to retain their functionality after intravenous injection in the form of single-cell suspension into the recipient’s body, have a specialized molecular mechanism of homing into the BM, and are able not only to survive but also to divide and differentiate in the necessary stromal directions in the recipient’s BM. The main problem at present remains the extremely low efficiency of engraftment of donor mesenchymal cells, and its solution will open the way for fundamentally new schemes of therapy of the abovementioned diseases. In my opinion, one of the possible solutions here is preliminary destruction or significant damage of the recipient’s BM stroma, which is necessary to activate the regenerative potential of the injected stem cells, as well as the use of uncultured true MSCs or more universal stem cells for transplantation, the presence of which in the mammalian organisms has been recently shown. Obviously, this approach carries many potential risks. What is the risk of targeted destruction of stem cells that maintain stromal tissues? Or what are the consequences of the destruction of osteoblasts responsible for bone regeneration and maintenance of HSCs? To what extent will this aggravate the situation of a patient who has also had his own hematopoiesis and immune system destroyed as part of induction and consolidation therapy before alloBMT? Are there any immunological risks? These questions must be answered in many, many experimental animal studies. Some risks can be considered to be low. For example, since a patient is in deep agranulocytosis before infusion of donor cells of alloBMT and because MSCs have immune privileges, the probability that infused MSCs or their differentiated progeny will be rejected seems to be rather low. Some risks are indeed high, for example, the risk of low efficiency of MSCs engraftment to the BM. Although if one thinks about alloBMT, it could also be considered a wild idea: the deliberate destruction of the entire hematopoiesis along with the immune system. It would seem to be incompatible with life. Nevertheless, today tens of thousands of people all over the world undergo this procedure, and many take undoubted risks for the sake of a clear goal—to get rid of deadly cancer and no less dangerous other diseases. And it often succeeds! So, for all the potential risks and difficulties, the emerging benefits of the proposed approach to combined stromal and hematopoietic transplantation seem as attractive as the devastating consequences. It will reduce the incidence of BM graft failure, leukemia and lymphoma relapses, and refractory courses of these diseases, and thus help save many lives. There is a long road ahead, but the one who walks it will conquer it.

Acknowledgments

I would like to thank my teacher, Nina Drize, for the development of the ability to think critically, without which the birth of these ideas would not have been possible. I express my heartfelt gratitude to my colleagues who participated in writing this chapter, discussing the ideas presented here, searching for sources of information, and providing support while working on this text. My heartfelt thanks to my family and friends for their continuous support and understanding during the long work on the manuscript. The study was conducted under the state assignment of Lomonosov Moscow State University.

References

  1. 1. Horwitz EM, Prockop DJ, Gordon PL, Koo WWK, Fitzpatrick LA, Neel MD, et al. Clinical responses to bone marrow transplantation in children with severe osteogenesis imperfecta. Blood. 2001;97:1227-1231
  2. 2. Horwitz EM, Le Blanc K, Dominici M, Mueller I, Slaper-Cortenbach I, Marini FC, et al. Clarification of the nomenclature for MSC: The International Society for Cellular Therapy position statement. Cytotherapy. 2005;7:393-395
  3. 3. Horwitz EM, Prockop DJ, Fitzpatrick LA, Koo WWK, Gordon PL, Neel M, et al. Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta. Nature Medicine. 1999;5:309-313
  4. 4. Golde DW, Hocking WG, Quan SG, Sparkes RS, Gale RP. Origin of human bone marrow fibroblasts. British Journal of Haematology. 1980;44:183-187
  5. 5. Laver J, Jhanwar S, O’Reilly R, Castro-Malaspina H. Host origin of the human hematopoietic microenvironment following allogeneic bone marrow transplantation. Blood. 1987;70:1966-1968
  6. 6. Simmons PJ, Przepiorka D, Thomas ED, Torok-Storb B. Host origin of marrow stromal cells following allogeneic bone marrow transplantation. Nature. 1987;328:429-432
  7. 7. Bentley S, Knutsen T, Whang-Peng J. The origin of the hematopoietic microenvironment in continuous bone marrow culture. Experimental Hematology. 1982;10:367-372
  8. 8. Athanasou NA, Quinn J, Brenner MK, Prentice HG, Graham A, Taylor S, et al. Origin of marrow stromal cells and haemopoietic chimaerism following bone marrow transplantation determined by in situ hybridisation. British Journal of Cancer. 1990;61:385-389
  9. 9. Eggenhofer E, Benseler V, Kroemer A, Popp FC, Geissler EK, Schlitt HJ, et al. Mesenchymal stem cells are short-lived and do not migrate beyond the lungs after intravenous infusion. Frontiers in Immunology. 2012;3:1-8
  10. 10. Eggenhofer E, Luk F, Dahlke MH, Hoogduijn MJ. The life and fate of mesenchymal stem cells. Frontiers in Immunology. 2014;5:1-6
  11. 11. Colter DC, Class R, DiGirolamo CM, Prockop DJ. Rapid expansion of recycling stem cells in cultures of plastic-adherent cells from human bone marrow. Proceedings of the National Academy of Sciences of the United States of America. 2000;97:3213-3218
  12. 12. Schrepfer S, Deuse T, Reichenspurner H, Fischbein MP, Robbins RC, Pelletier MP. Stem cell transplantation: The lung barrier. Transplantation Proceedings. 2007;39:573-576
  13. 13. Rombouts WJC, Ploemacher RE. Primary murine MSC show highly efficient homing to the bone marrow but lose homing ability following culture. Leukemia. 2003;17:160-170
  14. 14. Kucia M, Reca R, Campbell FR, Zuba-Surma E, Majka M, Ratajczak J, et al. A population of very small embryonic-like (VSEL) CXCR4(+)SSEA-1(+)Oct-4+ stem cells identified in adult bone marrow. Leukemia. 2006;20:857-869
  15. 15. Abouzaripour M, Kashani IR, Pasbakhsh P, Atlasy N. Intravenous transplantation of very small embryonic like stem cells in treatment of diabetes mellitus. Avicenna Journal of Medical Biotechnology. 2015;7:22
  16. 16. Virant-klun I, Skerl P, Novakovic S, Vrtacnik-bokal E, Smrkolj S. Similar population of cd133+ and ddx4+ vsel-like stem cells sorted from human embryonic stem cell, ovarian, and ovarian cancer ascites cell cultures: The real embryonic stem cells? Cells. 11 Jul 2019;8(7):706. DOI: 10.3390/cells8070706
  17. 17. Liu XB, Chen H, Chen HQ, Zhu MF, Hu XY, Wang YP, et al. Angiopoietin-1 preconditioning enhances survival and functional recovery of mesenchymal stem cell transplantation. Journal of Zhejiang University Science B. 2012;13:616-623
  18. 18. Pereira RF, Halford KW, O’Hara MD, Leeper DB, Sokolov BP, Pollard MD, et al. Cultured adherent cells from marrow can serve as long-lasting precursor cells for bone, cartilage, and lung in irradiated mice. Proceedings of the National Academy of Sciences of the United States of America. 1995;92:4857
  19. 19. Devine SM, Bartholomew AM, Mahmud N, Nelson M, Patil S, Hardy W, et al. Mesenchymal stem cells are capable of homing to the bone marrow of non-human primates following systemic infusion. Experimental Hematology. 2001;29:244-255
  20. 20. Devine SM, Cobbs C, Jennings M, Bartholomew A, Hoffman R. Mesenchymal stem cells distribute to a wide range of tissues following systemic infusion into nonhuman primates. Blood. 2003;101:2999-3001
  21. 21. Gao J, Dennis JE, Muzic RF, Lundberg M, Caplan AI. The dynamic in vivo distribution of bone marrow-derived mesenchymal stem cells after infusion. Cells, Tissues, Organs. 2001;169:12-20
  22. 22. Erices AA, Allers CI, Conget PA, Rojas CV, Minguell JJ. Human cord blood-derived mesenchymal stem cells home and survive in the marrow of immunodeficient mice after systemic infusion. Cell Transplantation. 2003;12:555-561
  23. 23. Allers C, Sierralta WD, Neubauer S, Rivera F, Minguell JJ, Conget PA. Dynamic of distribution of human bone marrow-derived mesenchymal stem cells after transplantation into adult unconditioned mice. Transplantation. 2004;78:503-508
  24. 24. Bensidhoum M, Chapel A, Francois S, Demarquay C, Mazurier C, Fouillard L, et al. Homing of in vitro expanded Stro-1- or Stro-1+ human mesenchymal stem cells into the NOD/SCID mouse and their role in supporting human CD34 cell engraftment. Blood. 2004;103:3313-3319
  25. 25. Sanchez-Diaz M, Quiñones-Vico MI, de la Torre RS, Montero-Vílchez T, Sierra-Sánchez A, Molina-Leyva A, et al. Biodistribution of mesenchymal stromal cells after administration in animal models and humans: A systematic review. Journal of Clinical Medicine. 29 Jun 2021;10(13):2925
  26. 26. Rüster B, Göttig S, Ludwig RJ, Bistrian R, Müller S, Seifried E, et al. Mesenchymal stem cells display coordinated rolling and adhesion behavior on endothelial cells. Blood. 2006;108:3938-3944
  27. 27. De Becker A, Van Hummelen P, Bakkus M, Broek IV, De Wever J, De Waele M, et al. Migration of culture-expanded human mesenchymal stem cells through bone marrow endothelium is regulated by matrix metalloproteinase-2 and tissue inhibitor of metalloproteinase-3. Haematologica. 2007;92:440-449
  28. 28. Cilloni D, Carlo-Stella C, Falzetti F, Sammarelli G, Regazzi E, Colla S, et al. Limited engraftment capacity of bone marrow–derived mesenchymal cells following T-cell–depleted hematopoietic stem cell transplantation. Blood. 2000;96:3637-3643
  29. 29. Anklesaria P, Kase K, Glowacki J, Holland CA, Sakakeeny MA, Wright JA, et al. Engraftment of a clonal bone marrow stromal cell line in vivo stimulates hematopoietic recovery from total body irradiation. Proceedings of the National Academy of Sciences of the United States of America. 1987;84:7681-7685
  30. 30. Yu Q, Liu L, Lin J, Wang Y, Xuan X, Guo Y, et al. SDF-1α/CXCR4 axis mediates the migration of mesenchymal stem cells to the hypoxic-ischemic brain lesion in a rat model. Cell Journal. 2015;16:440-447
  31. 31. Barbash IM, Chouraqui P, Baron J, Feinberg MS, Etzion S, Tessone A, et al. Systemic delivery of bone marrow-derived mesenchymal stem cells to the infarcted myocardium: Feasibility, cell migration, and body distribution. Circulation. 2003;108:863-868
  32. 32. Ji JF, He BP, Dheen ST, Tay SSW. Interactions of chemokines and chemokine receptors mediate the migration of mesenchymal stem cells to the impaired site in the brain after hypoglossal nerve injury. Stem Cells. 2004;22:415-427
  33. 33. Aiuti A, Webb IJ, Bleul C, Springer T, Gutierrez-Ramos JC. The chemokine SDF-1 is a chemoattractant for human CD34+ hematopoietic progenitor cells and provides a new mechanism to explain the mobilization of CD34+ progenitors to peripheral blood. The Journal of Experimental Medicine. 1997;185:111-120
  34. 34. Ma Q, Jones D, Borghesani PR, Segal RA, Nagasawa T, Kismimoto T, et al. Impaired B-lymphopoiesis, myelopoiesis, and derailed cerebellar neuron migration in CXCR4- and SDF-1-deficient mice. Proceedings of the National Academy of Sciences of the United States of America. 1998;95:9448-9453
  35. 35. Kim CH, Broxmeyer HE. Chemokines: Signal lamps for trafficking of T and B cells for development and effector function. Journal of Leukocyte Biology. 1999;65:6-15
  36. 36. Cho HH, Kyoung KM, Seo MJ, Kim YJ, Bae YC, Jung JS. Overexpression of CXCR4 increases migration and proliferation of human adipose tissue stromal cells. Stem Cells and Development. 2006;15:853-864
  37. 37. Wu Y, Zhao RCH. The role of chemokines in mesenchymal stem cell homing to myocardium. Stem Cell Reviews and Reports. 2012;8:243-250
  38. 38. Peled A, Petit I, Kollet O, Magid M, Ponomaryov T, Byk T, et al. Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science. 1999;283:845-848
  39. 39. Nagasawa T, Hirota S, Tachibana K, Takakura N, Nishikawa SI, Kitamura Y, et al. Defects of B-cell lymphopoiesis and bone-marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature. 1996;382:635-638
  40. 40. Lazarini F, Tham TN, Casanova P, Arenzana-Seisdedos F, Dubois-Dalcq M. Role of the alpha-chemokine stromal cell-derived factor (SDF-1) in the developing and mature central nervous system. Glia. 2003;42:139-148
  41. 41. Ratajczak MZ, Majka M, Kucia M, Drukala J, Pietrzkowski Z, Peiper S, et al. Expression of functional CXCR4 by muscle satellite cells and secretion of SDF-1 by muscle-derived fibroblasts is associated with the presence of both muscle progenitors in bone marrow and hematopoietic stem/progenitor cells in muscles. Stem Cells. 2003;21:363-371
  42. 42. Hatch HM, Zheng D, Jorgensen ML, Petersen BE. SDF-1alpha/CXCR4: A mechanism for hepatic oval cell activation and bone marrow stem cell recruitment to the injured liver of rats. Cloning and Stem Cells. 2002;4:339-351
  43. 43. Ara T, Nakamura Y, Egawa T, Sugiyama T, Abe K, Kishimoto T, et al. Impaired colonization of the gonads by primordial germ cells in mice lacking a chemokine, stromal cell-derived factor-1 (SDF-1). Proceedings of the National Academy of Sciences of the United States of America. 2003;100:5319-5323
  44. 44. Wojakowski W, Tendera M, Michałowska A, Majka M, Kucia M, Maślankiewicz K, et al. Mobilization of CD34/CXCR4+, CD34/CD117+, c-met+ stem cells, and mononuclear cells expressing early cardiac, muscle, and endothelial markers into peripheral blood in patients with acute myocardial infarction. Circulation. 2004;110:3213-3220
  45. 45. Iseki M, Kushida Y, Wakao S, Akimoto T, Mizuma M, Motoi F, et al. Muse cells, nontumorigenic pluripotent-like stem cells, have liver regeneration capacity through specific homing and cell replacement in a mouse model of liver fibrosis. Cell Transplantation. 2017;26:821-840
  46. 46. Liu X, Duan B, Cheng Z, Jia X, Mao L, Fu H, et al. SDF-1/CXCR4 axis modulates bone marrow mesenchymal stem cell apoptosis, migration and cytokine secretion. Protein & Cell. 2011;2:845-854
  47. 47. Cornelissen AS, Maijenburg MW, Nolte MA, Voermans C. Organ-specific migration of mesenchymal stromal cells: Who, when, where and why? Immunology Letters. 2015;168:159-169
  48. 48. Wynn RF, Hart CA, Corradi-Perini C, O’Neill L, Evans CA, Wraith JE, et al. A small proportion of mesenchymal stem cells strongly expresses functionally active CXCR4 receptor capable of promoting migration to bone marrow. Blood. 2004;104:2643-2645
  49. 49. Sordi V, Malosio ML, Marchesi F, Mercalli A, Melzi R, Giordano T, et al. Bone marrow mesenchymal stem cells express a restricted set of functionally active chemokine receptors capable of promoting migration to pancreatic islets. Blood. 2005;106:419-427
  50. 50. Honczarenko M, Le Y, Swierkowski M, Ghiran I, Glodek AM, Silberstein LE. Human bone marrow stromal cells express a distinct set of biologically functional chemokine receptors. Stem Cells. 2006;24:1030-1041
  51. 51. Kyriakou C, Rabin N, Pizzey A, Nathwani A, Yong K. Factors that influence short-term homing of human bone marrow-derived mesenchymal stem cells in a xenogeneic animal model. Haematologica. 2008;93:1457-1465
  52. 52. Chen W, Li M, Cheng H, Yan Z, Cao J, Pan B, et al. Overexpression of the mesenchymal stem cell Cxcr4 gene in irradiated mice increases the homing capacity of these cells. Cell Biochemistry and Biophysics. 2013;67:1181-1191
  53. 53. Bobis-Wozowicz S, Miekus K, Wybieralska E, Jarocha D, Zawisz A, Madeja Z, et al. Genetically modified adipose tissue−derived mesenchymal stem cells overexpressing CXCR4 display increased motility, invasiveness, and homing to bone marrow of NOD/SCID mice. Experimental Hematology. 2011;39:686-696.e4
  54. 54. Ponomaryov T, Peled A, Petit I, Taichman RS, Habler L, Sandbank J, et al. Induction of the chemokine stromal-derived factor-1 following DNA damage improves human stem cell function. The Journal of Clinical Investigation. 2000;106:1331-1339
  55. 55. Piersma AH, Ploemacher RE, Brockbank KGM. Transplantation of bone marrow fibroblastoid stromal cells in mice via the intravenous route. British Journal of Haematology. 1983;54:285-290
  56. 56. Meng SS, Xu XP, Chang W, Lu ZH, Huang LL, Xu JY, et al. LincRNA-p21 promotes mesenchymal stem cell migration capacity and survival through hypoxic preconditioning. Stem Cell Research & Therapy. 25 Oct 2018;9(1):280
  57. 57. Ceradini DJ, Kulkarni AR, Callaghan MJ, Tepper OM, Bastidas N, Kleinman ME, et al. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nature Medicine. 2004;10:858-864
  58. 58. Friedenstein A, Ivanov-Smolenski A, Chajlakjan R, Gorskaya U, Kuralesova A, Latzinik N, et al. Origin of bone marrow stromal mechanocytes in radiochimeras and heterotopic transplants. Experimental Hematology. 1978;6:440-444
  59. 59. Ivanov-Smolenskiĭ AA, Chaĭlakhian RK, Gerasimov IV, Gorskaia IF, Kuralesova AI. Origin of stromal bone marrow mechanocytes according to the results of typing them by isoantigens and chromosomal markers. Ontogenez. 1978;9:245-252
  60. 60. Chertkov JL, Drize NJ, Gurevitch OA, Udalov GA. Hemopoietic stromal precursors in long-term culture of bone marrow: I. Precursor characteristics, kinetics in culture, and dependence on quality of donor hemopoietic cells in chimeras. Experimental Hematology. 1983;11:231-242
  61. 61. Chertkov J, Drize N, Gurevitch O, Samoylova R. Origin of hemopoietic stromal progenitor cells in chimeras. Experimental Hematology. 1985;13:1217-1222
  62. 62. Chertkov J, Drize N, Gurevitch O. Hemopoietic stromal precursors in long-term culture of bone marrow: II. Significance of initial packing for creating a hemopoietic microenvironment and maintaining stromal precursors in the culture. Experimental Hematology. 1983;11:243-248
  63. 63. Friedenstein AJ, Chailakhyan RK, Latsinik NV, Panasyvk AF, Keiliss-Borok IV. Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues. Cloning in vitro and retransplantation in vivo. Transplantation. 1974;17:331-340
  64. 64. Ahn SY. The role of MSCs in the tumor microenvironment and tumor progression. Anticancer Research. 2020;40:3039-3047
  65. 65. Gu F, Zhang K, Li J, Xie X, Wen Q, Sui Z, et al. Changes of migration, immunoregulation and osteogenic differentiation of mesenchymal stem cells in different stages of inflammation. International Journal of Medical Sciences. 2022;19:25-33
  66. 66. Liu J, Gao J, Liang Z, Gao C, Niu Q, Wu F, et al. Mesenchymal stem cells and their microenvironment. Stem Cell Research & Therapy. 20 Aug 2022;13(1):429
  67. 67. Ponte AL, Marais E, Gallay N, Langonné A, Delorme B, Hérault O, et al. The in vitro migration capacity of human bone marrow mesenchymal stem cells: Comparison of chemokine and growth factor chemotactic activities. Stem Cells. 2007;25:1737-1745
  68. 68. Yang N, Wang G, Hu C, Shi Y, Liao L, Shi S, et al. Tumor necrosis factor α suppresses the mesenchymal stem cell osteogenesis promoter miR-21 in estrogen deficiency-induced osteoporosis. Journal of Bone and Mineral Research. 2013;28:559-573
  69. 69. Durand N, Russell A, Zubair AC. Effect of comedications and endotoxins on mesenchymal stem cell secretomes, migratory and immunomodulatory capacity. Journal of Clinical Medicine. 11 Apr 2019;8(4):497
  70. 70. Li Y, Zhong X, Zhang Y, Lu X. Mesenchymal stem cells in gastric cancer: Vicious but hopeful. Frontiers in Oncology. 11 May 2021;11:617677
  71. 71. Le Blanc K, Rasmusson I, Götherström C, Seidel C, Sundberg B, Sundin M, et al. Mesenchymal stem cells inhibit the expression of CD25 (interleukin-2 receptor) and CD38 on phytohaemagglutinin-activated lymphocytes. Scandinavian Journal of Immunology. 2004;60:307-315
  72. 72. Pradier A, Passweg J, Villard J, Kindler V. Human bone marrow stromal cells and skin fibroblasts inhibit natural killer cell proliferation and cytotoxic activity. Cell Transplantation. 2011;20:681-691
  73. 73. Petinati NA, Kapranov NM, Bigil’deev AE, Popova MD, Davydova YO, Gal’tseva IV, et al. Changing the properties of multipotent mesenchymal stromal cells by IFNγ administration. Bulletin of Experimental Biology and Medicine. 2017;163:230-234
  74. 74. López-García L, Castro-Manrreza ME. Tnf-α and ifn-γ participate in improving the immunoregulatory capacity of mesenchymal stem/stromal cells: Importance of cell-cell contact and extracellular vesicles. International Journal of Molecular Sciences. 2 Sep 2021;22(17):9531
  75. 75. Liu H, Li R, Liu T, Yang L, Yin G, Xie Q. Immunomodulatory effects of mesenchymal stem cells and mesenchymal stem cell-derived extracellular vesicles in rheumatoid arthritis. Frontiers in Immunology. 20 Aug 2020;11:1912
  76. 76. Mendt M, Rezvani K, Shpall E. Mesenchymal stem cell-derived exosomes for clinical use. Bone Marrow Transplantation. 2019;542(54):789-792
  77. 77. Berglund AK, Fortier LA, Antczak DF, Schnabel LV. Immunoprivileged no more: Measuring the immunogenicity of allogeneic adult mesenchymal stem cells. Stem Cell Research & Therapy. 22 Dec 2017;8(1):288
  78. 78. Ankrum JA, Ong JF, Karp JM. Mesenchymal stem cells: Immune evasive, not immune privileged. Nature Biotechnology. 2014;32:252-260
  79. 79. Petinati N, Drize N, Sats N, Risinskaya N, Sudarikov A, Drokov M, et al. Recovery of donor hematopoiesis after graft failure and second hematopoietic stem cell transplantation with intraosseous administration of mesenchymal stromal cells. Stem Cells International. 10 Apr 2018;2018:6495018
  80. 80. Kuzmina LA, Petinati NA, Sats NV, Drize NJ, Risinskaya NV, Sudarikov AB, et al. Long-term survival of donor bone marrow multipotent mesenchymal stromal cells implanted into the periosteum of patients with allogeneic graft failure. International Journal of Hematology. 2016;104:403-407
  81. 81. Liechty KW, Mackenzie TC, Shaaban AF, Radu A, Moseley AMB, Deans R, et al. Human mesenchymal stem cells engraft and demonstrate site-specific differentiation after in utero transplantation in sheep. Nature Medicine. 2000;6:1282-1286
  82. 82. Karpenko D, Kapranov N, Bigildeev A. Nestin-GFP transgene labels immunoprivileged bone marrow mesenchymal stem cells in the model of ectopic foci formation. Frontiers in Cell and Developmental Biology. 5 Sep 2022;10:993056
  83. 83. Karpenko D, Kapranov N, Bigildeev A. Strong Immune Privileges of MSC and Other Nes-GFP+ Progenitors in Bone Marrow of Transgenic Mice. Immune Network. Apr 2025;25:e20
  84. 84. Petinati N, Kapranov N, Davydova Y, Bigildeev A, Pshenichnikova O, Karpenko D, et al. Immunophenotypic characteristics of multipotent mesenchymal stromal cells that affect the efficacy of their use in the prevention of acute graft vs host disease. World Journal of Stem Cells. 2020;12:1377-1395
  85. 85. Metwally AM, Li H, Houghton J. Alterations of epigenetic regulators and P53 mutations in murine mesenchymal stem cell cultures: A possible mechanism of spontaneous transformation. Cancer Biomarkers. 2021;32:327-337
  86. 86. Saidi N, Ghalavand M, Hashemzadeh MS, Dorostkar R, Mohammadi H, Mahdian-shakib A. Dynamic changes of epigenetic signatures during chondrogenic and adipogenic differentiation of mesenchymal stem cells. Biomedicine & Pharmacotherapy. 2017;89:719-731
  87. 87. Keating A, Singer JW, Killen PD, Striker GE, Salo AC, Sanders J, et al. Donor origin of the in vitro haematopoietic microenvironment after marrow transplantation in man. Nature. 1982;298:280-283
  88. 88. Marshall MJ, Nisbet NW, Evans S. Donor origin of the in vitro hematopoietic microenvironment after marrow transplantation in mice. Experientia. 1984;40:385-386
  89. 89. Sale G, Storb R. Bilateral diffuse pulmonary ectopic ossification after marrow allograft in a dog. Evidence for allotransplantation of hemopoietic and mesenchymal stem cells. Experimental Hematology. 1983;11:961-966
  90. 90. Nilsson SK, Dooner MS, Weier HU, Frenkel B, Lian JB, Stein GS, et al. Cells capable of bone production engraft from whole bone marrow transplants in nonablated mice. The Journal of Experimental Medicine. 1999;189:729-734
  91. 91. Goan SR, Junghahn I, Wissler M, Becker M, Aumann J, Just U, et al. Donor stromal cells from human blood engraft in NOD/SCID mice. Blood. 2000;96:3971-3978
  92. 92. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143-147
  93. 93. Méndez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, MacArthur BD, Lira SA, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466:829-834
  94. 94. Kfoury Y, Scadden DT. Mesenchymal cell contributions to the stem cell niche. Cell Stem Cell. 2015;16:239-253
  95. 95. Chertkov JL, Gurevitch OA, Udalov GA. Role of bone marrow stroma in hemopoietic stem cell regulation. Experimental Hematology. 1980;8:770-778
  96. 96. Nicolay NH, Lopez Perez R, Debus J, Huber PE. Mesenchymal stem cells - A new hope for radiotherapy-induced tissue damage? Cancer Letters. 2015;366:133-140
  97. 97. Bigildeev AE, Bigildeev EA, Boulygina ES, Tsygankova SV, Gusakova MS, Illarionova OI. Successful bone marrow stroma transplantation is enabled by preliminary recipient’s stromal compartment injury. Scientific Reports. 2025. DOI: 10.20944/preprints202502.1215.v1
  98. 98. Chertkov J, Gurevitch O. Radiosensitivity of progenitor cells of the hematopoietic microenvironment. Radiation Research. 1979;79:177-186
  99. 99. Valiev AK, Konev AA, Kurilchik AA, Machak GN, Musaev ER, Rogozhin DV, et al. Malignant bone tumors. Malignant Tumors. 2023;13:335-355
  100. 100. National Comprehensive Cancer Network. Bone Cancer (Version 2.2025). 2025. p. 2
  101. 101. Yoshida H, Hayashi SI, Kunisada T, Ogawa M, Nishikawa S, Okamura H, et al. The murine mutation osteopetrosis is in the coding region of the macrophage colony stimulating factor gene. Nature. 1990;345:442-444
  102. 102. Nakano T, Kodama H, Honjo T. Generation of lymphohematopoietic cells from embryonic stem cells in culture. Science. 1994;265:1098-1101
  103. 103. Globig P, Willumeit-Römer R, Martini F, Mazzoni E, Luthringer-Feyerabend BJC. Optimizing an osteosarcoma-fibroblast coculture model to study antitumoral activity of magnesium-based biomaterials. International Journal of Molecular Sciences. 2020;21:1-18
  104. 104. Minguell JJ, Erices A, Conget P. Mesenchymal stem cells. Experimental Biology and Medicine (Maywood, N.J.). 2001;226:507-520
  105. 105. Costela-ruiz VJ, Melguizo-rodríguez L, Bellotti C, Illescas-montes R, Stanco D, Arciola CR, et al. Different sources of mesenchymal stem cells for tissue regeneration: A guide to identifying the most favorable one in orthopedics and dentistry applications. International Journal of Molecular Sciences. 6 Jun 2022;23(11):6356
  106. 106. Yang D, Liu J, Qian H, Zhuang Q. Cancer-associated fibroblasts: From basic science to anticancer therapy. Experimental & Molecular Medicine. 2023;55:1322-1332
  107. 107. Baryawno N, Przybylski D, Kowalczyk MS, Kfoury Y, Severe N, Gustafsson K, et al. A cellular taxonomy of the bone marrow stroma in homeostasis and leukemia. Cell. 2019;177:1915-1932.e16
  108. 108. Li H, Ghazanfari R, Zacharaki D, Lim HC, Scheding S. Isolation and characterization of primary bone marrow mesenchymal stromal cells. Annals of the New York Academy of Sciences. 2016;1370:109-118
  109. 109. Watson JT, Foo T, Wu J, Moed BR, Thorpe M, Schon L, et al. CD271 as a marker for mesenchymal stem cells in bone marrow versus umbilical cord blood. Cells, Tissues, Organs. 2013;197:496-504
  110. 110. Armulik A, Genové G, Betsholtz C. Pericytes: Developmental, physiological, and pathological perspectives, problems, and promises. Developmental Cell. 2011;21:193-215
  111. 111. Kunisaki Y, Bruns I, Scheiermann C, Ahmed J, Pinho S, Zhang D, et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature. 2013;502:637
  112. 112. Wakao S, Kitada M, Kuroda Y, Shigemoto T, Matsuse D, Akashi H, et al. Multilineage-differentiating stress-enduring (muse) cells are a primary source of induced pluripotent stem cells in human fibroblasts. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:9875-9880
  113. 113. Morrison SJ, Scadden DT. The bone marrow niche for haematopoietic stem cells. Nature. 2014;505:327-334
  114. 114. Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, Knight MC, et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003;425:841-846
  115. 115. Coskun S, Hirschi KK. Establishment and regulation of the HSC niche: Roles of osteoblastic and vascular compartments. Birth Defects Research. Part C, Embryo Today. 2010;90:229-242
  116. 116. Sugiyama T, Omatsu Y, Nagasawa T. Niches for hematopoietic stem cells and immune cell progenitors. International Immunology. 2019;31:5-11
  117. 117. Reyes-Botella C, Montes MJ, Abadía-Molina AC, Vallecillo-Capilla MF, Ruiz C. CD10 expression in cultured human osteoblast-like cells. Folia Biologica. 1999;45:257-260
  118. 118. Reyes-Botella C, Montes M, Vallecillo-Capilla M, Olivares E, Ruiz C. Antigenic phenotype of cultured human osteoblast-like cells. Cellular Physiology and Biochemistry. 2002;12:359-364
  119. 119. Ignatius A, Schoengraf P, Kreja L, Liedert A, Recknagel S, Kandert S, et al. Complement C3a and C5a modulate osteoclast formation and inflammatory response of osteoblasts in synergism with IL-1β. Journal of Cellular Biochemistry. 2011;112:2594-2605
  120. 120. Badawy T, Kyumoto-Nakamura Y, Uehara N, Zhang J, Sonoda S, Hiura H, et al. Osteoblast lineage-specific cell-surface antigen (A7) regulates osteoclast recruitment and calcification during bone remodeling. Laboratory Investigation. 2019;99:866-884
  121. 121. Brinkmann U, Kontermann RE. The making of bispecific antibodies. MAbs. 2017;9:182-212
  122. 122. Shim H. Bispecific antibodies and antibody-drug conjugates for cancer therapy: Technological considerations. Biomolecules. 26 Feb 2020;10(3):360
  123. 123. Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proceedings of the National Academy of Sciences of the United States of America. 1989;86:10024-10028
  124. 124. Irving BA, Weiss A. The cytoplasmic domain of the T cell receptor zeta chain is sufficient to couple to receptor-associated signal transduction pathways. Cell. 1991;64:891-901
  125. 125. Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced Leukemia. Science Translational Medicine. 2011;3:95ra73
  126. 126. Xie G, Dong H, Liang Y, Ham JD, Rizwan R, Chen J. CAR-NK cells: A promising cellular immunotherapy for cancer. eBioMedicine. Sep 2020;59:102975
  127. 127. Gourd E. CAR T-cell cocktail therapy for B-cell malignancies. The Lancet Oncology. 2019;20:e669
  128. 128. Hirata Y, Furuhashi K, Ishii H, Li HW, Pinho S, Ding L, et al. CD150 high bone marrow Tregs maintain hematopoietic stem cell quiescence and immune privilege via adenosine. Cell Stem Cell. 2018;22:445-453.e5
  129. 129. Riether C. Regulation of hematopoietic and leukemia stem cells by regulatory T cells. Frontiers in Immunology. 2 Nov 2022;13:1049301
  130. 130. Kasahara H, Kondo T, Nakatsukasa H, Chikuma S, Ito M, Ando M, et al. Generation of Allo-antigen-specific induced Treg stabilized by vitamin C treatment and its application for prevention of acute graft versus host disease model. International Immunology. 2017;29:457-469
  131. 131. Sirpilla O, Sakemura RL, Hefazi M, Kimball BL, Huynh TN, Siegler EL, et al. Chimeric antigen receptor design in human mesenchymal stromal cells (CAR-MSCs) mediates homing to target sites and downstream immunosuppression for immune disease treatment. Transplantation and Cellular Therapy. 2024;30:S11-S12
  132. 132. Yan L, Li J, Zhang C. The role of MSCs and CAR-MSCs in cellular immunotherapy. Cell Communication and Signaling: CCS. 2023;211(21):1-22
  133. 133. Kovtun I, von Bonin M, Ibneeva L, Frimmel J, Middeke JM, Kunadt D, et al. Profound sympathetic neuropathy in the bone marrow of patients with acute myeloid leukemia. Leukemia. 2024;38:393-397
  134. 134. Cai TY, Zhu W, Chen XS, Zhou SY, Jia LS, Sun YQ. Fibroblast growth factor 2 induces mesenchymal stem cells to differentiate into tenocytes through the MAPK pathway. Molecular Medicine Reports. 2013;8:1323-1328
  135. 135. Li SN, Wu JF. TGF-β/SMAD signaling regulation of mesenchymal stem cells in adipocyte commitment. Stem Cell Research & Therapy. 29 Jan 2020;11(1):41
  136. 136. Bigildeev AE, Zhironkina OA, Lubkova ON, Drize NJ. Interleukin-1 beta is an irradiation-induced stromal growth factor. Cytokine. 2013;64:131-137
  137. 137. Bigildeev AE, Zezina EA, Shipounova IN, Drize NJ. Interleukin-1 beta enhances human multipotent mesenchymal stromal cell proliferative potential and their ability to maintain hematopoietic precursor cells. Cytokine. 2015;71:246-254
  138. 138. Hackel A, Aksamit A, Bruderek K, Lang S, Brandau S. TNF-α and IL-1β sensitize human MSC for IFN-γ signaling and enhance neutrophil recruitment. European Journal of Immunology. 2021;51:319-330
  139. 139. Burnham AJ, Foppiani EM, Goss KL, Jang-Milligan F, Kamalakar A, Bradley H, et al. Differential response of mesenchymal stromal cells (MSCs) to type 1 ex vivo cytokine priming: Implications for MSC therapy. Cytotherapy. 2023;25:1277-1284
  140. 140. Leitão L, Alves CJ, Sousa DM, Neto E, Conceição F, Lamghari M. The alliance between nerve fibers and stem cell populations in bone marrow: Life partners in sickness and health. The FASEB Journal. 2019;33:8697-8710
  141. 141. Doglio M, Crossland RE, Alho AC, Penack O, Dickinson AM, Stary G, et al. Cell-based therapy in prophylaxis and treatment of chronic graft-versus-host disease. Frontiers in Immunology. 17 Nov 2022;13:1045168
  142. 142. Zaripova LN, Midgley A, Christmas SE, Beresford MW, Pain C, Baildam EM, et al. Mesenchymal stem cells in the pathogenesis and therapy of autoimmune and autoinflammatory diseases. International Journal of Molecular Sciences. 7 Nov 2023;24(22):16040
  143. 143. Vianello F, Dazzi F. Mesenchymal stem cells for graft-versus-host disease: A double edged sword? Leukemia. 2008;22:463-465
  144. 144. Forte D, García-Fernández M, Sánchez-Aguilera A, Stavropoulou V, Fielding C, Martín-Pérez D, et al. Bone marrow mesenchymal stem cells support acute myeloid Leukemia bioenergetics and enhance antioxidant Defense and escape from chemotherapy. Cell Metabolism. 2020;32:829-843.e9
  145. 145. Ramuta TŽ, Kreft ME. Mesenchymal stem/stromal cells may decrease success of cancer treatment by inducing resistance to chemotherapy in cancer cells. Cancers (Basel). 2 Aug 2022;14(15):3761
  146. 146. Antoon R, Overdevest N, Saleh AH, Keating A. Mesenchymal stromal cells as cancer promoters. Oncogene. Nov 2024;43(49):3545-3555
  147. 147. Lee MW, Ryu S, Kim DS, Lee JW, Sung KW, Koo HH, et al. Mesenchymal stem cells in suppression or progression of hematologic malignancy: Current status and challenges. Leukemia. 2019;33:597-611
  148. 148. Wong RSY. Mesenchymal stem cells: Angels or demons? Journal of Biomedicine & Biotechnology. 2011:8. Article ID: 459510. DOI: 10.1155/2011/459510
  149. 149. Qiao L, Xu ZL, Zhao TJ, Ye LH, Zhang XD. Dkk-1 secreted by mesenchymal stem cells inhibits growth of breast cancer cells via depression of Wnt signalling. Cancer Letters. 2008;269:67-77
  150. 150. Li L, Tian H, Yue W, Zhu F, Li S, Li W. Human mesenchymal stem cells play a dual role on tumor cell growth in vitro and in vivo. Journal of Cellular Physiology. 2011;226:1860-1867
  151. 151. Kidd S, Caldwell L, Dietrich M, Samudio I, Spaeth EL, Watson K, et al. Mesenchymal stromal cells alone or expressing interferon-beta suppress pancreatic tumors in vivo, an effect countered by anti-inflammatory treatment. Cytotherapy. 2010;12:615-625
  152. 152. Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, et al. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 2007;449:557-563
  153. 153. Suzuki K, Sun R, Origuch M, Kanehira M, Takahata T, Itoh J, et al. Mesenchymal stromal cells promote tumor growth through the enhancement of neovascularization. Molecular Medicine. 2011;17:579-587
  154. 154. Yu JM, Jun ES, Bae YC, Jung JS. Mesenchymal stem cells derived from human adipose tissues favor tumor cell growth in vivo. Stem Cells and Development. 2008;17:463-473
  155. 155. Lin G, Yang R, Banie L, Wang G, Ning H, Li LC, et al. Effects of transplantation of adipose tissue-derived stem cells on prostate tumor. Prostate. 2010;70:1066-1073
  156. 156. Klopp AH, Gupta A, Spaeth E, Andreeff M, Marini F. Concise review: Dissecting a discrepancy in the literature: Do mesenchymal stem cells support or suppress tumor growth? Stem Cells. 2011;29:11-19
  157. 157. Nobre AR, Risson E, Singh DK, Di Martino JS, Cheung JF, Wang J, et al. Bone marrow NG2+/nestin+ mesenchymal stem cells drive DTC dormancy via TGF-β2. Nature Cancer. 2021;23(2):327-339
  158. 158. Ning H, Yang F, Jiang M, Hu L, Feng K, Zhang J, et al. The correlation between cotransplantation of mesenchymal stem cells and higher recurrence rate in hematologic malignancy patients: Outcome of a pilot clinical study. Leukemia. 2008;22:593-599
  159. 159. Zeng Z, Shi YX, Samudio IJ, Wang RY, Ling X, Frolova O, et al. Targeting the leukemia microenvironment by CXCR4 inhibition overcomes resistance to kinase inhibitors and chemotherapy in AML. Blood. 2009;113:6215-6224
  160. 160. Konopleva M, Konoplev S, Hu W, Zaritskey AY, Afanasiev BV, Andreeff M. Stromal cells prevent apoptosis of AML cells by up-regulation of anti-apoptotic proteins. Leukemia. 2002;16:1713-1724
  161. 161. Jin L, Tabe Y, Konoplev S, Xu Y, Leysath CE, Lu H, et al. CXCR4 up-regulation by imatinib induces chronic myelogenous leukemia (CML) cell migration to bone marrow stroma and promotes survival of quiescent CML cells. Molecular Cancer Therapeutics. 2008;7:48-58
  162. 162. Vianello F, Villanova F, Tisato V, Lymperi S, Ho KK, Gomes AR, et al. Bone marrow mesenchymal stromal cells non-selectively protect chronic myeloid leukemia cells from imatinib-induced apoptosis via the CXCR4/CXCL12 axis. Haematologica. 2010;95:1081-1089
  163. 163. Zhang Y, Hu K, Hu Y, Liu L, Wang B, Huang H. Bone marrow mesenchymal stromal cells affect the cell cycle arrest effect of genotoxic agents on acute lymphocytic leukemia cells via p21 down-regulation. Annals of Hematology. 2014;93:1499-1508
  164. 164. Zhang X, Tu H, Yang Y, Wan Q, Fang L, Wu Q, et al. High IL-7 levels in the bone marrow microenvironment mediate imatinib resistance and predict disease progression in chronic myeloid leukemia. International Journal of Hematology. 2016;104:358-367
  165. 165. Cai J, Wang J, Huang Y, Wu H, Xia T, Xiao J, et al. ERK/Drp1-dependent mitochondrial fission is involved in the MSC-induced drug resistance of T-cell acute lymphoblastic leukemia cells. Cell Death & Disease. 2016;7(11):e2459, 11
  166. 166. Carter BZ, Mak PY, Chen Y, Mak DH, Mu H, Jacamo R, et al. Anti-apoptotic ARC protein confers chemoresistance by controlling leukemia-microenvironment interactions through a NFκB/IL1β signaling network. Oncotarget. 2016;7:20054-20067
  167. 167. Kamga PT, Bassi G, Cassaro A, Midolo M, Di Trapani M, Gatti A, et al. Notch signalling drives bone marrow stromal cell-mediated chemoresistance in acute myeloid leukemia. Oncotarget. 2016;7:21713-21727
  168. 168. Bernardo ME, Avanzini MA, Perotti C, Cometa AM, Moretta A, Lenta E, et al. Optimization of in vitro expansion of human multipotent mesenchymal stromal cells for cell-therapy approaches: Further insights in the search for a fetal calf serum substitute. Journal of Cellular Physiology. 2007;211:121-130
  169. 169. Kim J, Kang JW, Park JH, Choi Y, Choi KS, Park KD, et al. Biological characterization of long-term cultured human mesenchymal stem cells. Archives of Pharmacal Research. 2009;32:117-126
  170. 170. Choumerianou DM, Dimitriou H, Perdikogianni C, Martimianaki G, Riminucci M, Kalmanti M. Study of oncogenic transformation in ex vivo expanded mesenchymal cells, from paediatric bone marrow. Cell Proliferation. 2008;41:909-922
  171. 171. Wang Y, Huso DI, Harrington J, Kellner J, Jeong DK, Turney J, et al. Outgrowth of a transformed cell population derived from normal human BM mesenchymal stem cell culture. Cytotherapy. 2005;7:509-519
  172. 172. Røsland GV, Svendsen A, Torsvik A, Sobala E, McCormack E, Immervoll H, et al. Long-term cultures of bone marrow-derived human mesenchymal stem cells frequently undergo spontaneous malignant transformation. Cancer Research. 2009;69:5331-5339
  173. 173. Mutsaers AJ, Walkley CR. Cells of origin in osteosarcoma: Mesenchymal stem cells or osteoblast committed cells? Bone. 2014;62:56-63
  174. 174. Abarrategi A, Tornin J, Lucia MC, Hamilton A, Enrique MC, Rodrigo JP, et al. Osteosarcoma: Cells-of-origin, cancer stem cells, and targeted therapies. Stem Cells International. 2016;2016:3631764
  175. 175. Yamanaka R, Hayano A. Radiation-induced Schwannomas and neurofibromas: A systematic review. World Neurosurgery. 2017;104:713-722
  176. 176. Yamanaka R, Hayano A. Radiation-induced malignant peripheral nerve sheath Tumors: A systematic review. World Neurosurgery. 2017;105:961-970.e8
  177. 177. Inchaustegui ML, Kon-Liao K, Ruiz-Arellanos K, Silva GAE, Gonzalez MR, Pretell-Mazzini J. Treatment and outcomes of radiation-induced soft tissue sarcomas of the extremities and trunk—A systematic review of the literature. Cancers (Basel). 25 Nov 2023;15(23):5584
  178. 178. Kunisaki Y. Pericytes in bone marrow. Advances in Experimental Medicine and Biology. 2019;1122:101-114
  179. 179. Lebeau G, Ah-Pine F, Daniel M, Bedoui Y, Vagner D, Frumence E, et al. Perivascular mesenchymal stem/stromal cells, an immune privileged niche for viruses? International Journal of Molecular Sciences. 21 Jul 2022;23(14):8038

Written By

Aleksei E. Bigildeev

Submitted: 06 May 2025 Reviewed: 22 May 2025 Published: 09 July 2025