Next Article in Journal
Boron Neutron Capture Therapy and Photodynamic Therapy for High-Grade Meningiomas
Next Article in Special Issue
Capture-Based Next-Generation Sequencing Improves the Identification of Immunoglobulin/T-Cell Receptor Clonal Markers and Gene Mutations in Adult Acute Lymphoblastic Leukemia Patients Lacking Molecular Probes
Previous Article in Journal
Application of Next-Generation Sequencing for the Genomic Characterization of Patients with Smoldering Myeloma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Bird’s-Eye View of Cell Sources for Cell-Based Therapies in Blood Cancers

by
Benjamin Motais
1,2,
Sandra Charvátová
1,2,
Matouš Hrdinka
1,3,
Michal Šimíček
1,2,3,
Tomáš Jelínek
1,2,3,
Tereza Ševčíková
1,2,3,
Zdeněk Kořístek
1,3,
Roman Hájek
1,3 and
Juli R. Bagó
1,3,*
1
Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic
2
Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
3
Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
*
Author to whom correspondence should be addressed.
Submission received: 14 April 2020 / Revised: 17 May 2020 / Accepted: 20 May 2020 / Published: 23 May 2020
(This article belongs to the Special Issue Hematologic Malignancy)

Abstract

:
Hematological malignancies comprise over a hundred different types of cancers and account for around 6.5% of all cancers. Despite the significant improvements in diagnosis and treatment, many of those cancers remain incurable. In recent years, cancer cell-based therapy has become a promising approach to treat those incurable hematological malignancies with striking results in different clinical trials. The most investigated, and the one that has advanced the most, is the cell-based therapy with T lymphocytes modified with chimeric antigen receptors. Those promising initial results prepared the ground to explore other cell-based therapies to treat patients with blood cancer. In this review, we want to provide an overview of the different types of cell-based therapies in blood cancer, describing them according to the cell source.

1. Introduction

Hematologic malignancies are the fourth-most common type of cancer, with a higher incidence in older adults [1]. The number of cases is expected to rise over the years because of the increase in life expectancy [2]. Traditionally, the treatment regimens have included chemo and radiotherapy and stem cell transplantation. In the past decade, substantial improvements in patients’ outcomes have been achieved through advances in diagnosis and treatment. Some of those new therapeutic approaches include the administration of monoclonal antibodies [3,4,5,6,7] and immunomodulatory drugs [8]. Unfortunately, some types of blood cancers remain incurable. In a rapid-fire series of breakthroughs in recent years, cancer cell-based therapy is flourishing as a novel and promising approach to combat otherwise incurable hematologic malignancies [9,10,11]. The cancer patients undergoing such cell-based therapies are administrated a “living drug” in the form of modified or unmodified living cells from the patient or a suitable donor, which are able to specifically recognize and destroy malignant cells. In practical clinical development, cell-based therapies with T lymphocytes modified with chimeric antigen receptors (CARs) have advanced the most, with striking results in the treatment of different types of hematological cancers [12,13]. In this review, we aim to give an overview of the different types of cancer cell-based therapies to treat hematological malignancies (Figure 1), organizing them according to the cell type and source used as a therapeutic vehicle and highlighting their main benefits and remaining challenges.

2. T Lymphocytes

T cells are apparent candidates for cancer cell-based immunotherapy due to their inherent activity against cancer. T lymphocytes kill tumor cells upon T cell receptor (TCR) recognition of cancer-specific antigens presented by the major histocompatibility complex (MHC). When activated by an antigen, the intracellular domains of CD3ζ in the TCR complex become phosphorylated on their immunoreceptor tyrosine-based activation motifs and triggers a signaling cascade resulting in the expression of transcription factors such as AP-1, NF-κB, or NFAT. In this activated state, the T cells express and release cytolytic enzymes such as granzymes and perforins, thus inducing apoptosis in the target tumor cells. Harnessing the natural recognition of cancer antigens, immunotherapies based on activated T cell transplantation have shown positive results in the past [14].
More recently, the introduction of novel T cell therapies based on CAR-T technology has genuinely revolutionized the battle against cancer, particularly in hematological malignancies. This technology was first described by Gross and colleagues at the Weizmann Institute of Science in Israel in the 1980s [15]. With the CAR approach, immune T cells are armed with artificial receptors that directly recognize specific epitopes present on the surface of tumors cells, bypassing the need for the classical antigen presentation process. The CARs typically consist of an extracellular antibody-derived single-chain variable fragment (scFv) that recognizes specific tumor-related antigens, a single transmembrane domain, and an intracellular signal domain responsible for triggering the cellular immune responses [16]. Over the years, several generations of CARs have been engineered in order to improve the efficacy of CAR-T cells. The first CAR generation consists of a single intracellular signaling domain of CD3ζ derived from the natural TCR complex. With the intention to boost the T cell response after antigen recognition, an addition of one or two costimulatory domains, such as signaling domains CD28 [17] and 4-1BB [18], were added to the first generation of CAR, leading to the second (one additional domain) and third (two additional domains) generations of CARs. The fourth generation of CARs, also referred to as “TRUCKs” (T cells Redirected for Universal Cytokine-mediated Killing), enables the expression of immune-stimulatory cytokines, such as IL-2, to enhance CAR-T cell persistence, expansion, and resistance to immunosuppression [19]. Over the years, rapid advances in genetic engineering have allowed the creation of more sophisticated CAR-T designs to be tested. For example, dual-target CAR-T cells have been developed to provide T-cells with two different scFvs, increasing the killing efficiency and reducing tumor escape in heterogeneous tumors [20]. Furthermore, the efforts to produce more flexible and modulated CAR-T cells in terms of antigen specificity and activity has resulted in the generation of SUPRA (Split Universal Programmable) CARs. The intracellular parts of SUPRA CARs are based on traditional signaling domains, but instead of an extracellular antigen-binding scFv, they display a high-affinity binding domain of leucine zippers [21]. The fully functional CAR is then formed when leucine zippers in the cell membrane match other leucine zippers attached to a specific soluble scFv. This elegant solution simultaneously addressed both the regulation of CAR specificity (targeting various antigens) and activity (creating an active CAR on demand). Other varieties of switchable CARs have been proposed, such as the one with the biotin-avidin system [22]. Those and other switchable designs are flexible systems, allowing an easy and quick reprogram of the CAR, expanding the possible applications and targets and reducing the cost of CAR-T manufacturing. To sum up, T cells can be armed with many viable CAR options, all of which possess unique advantages and disadvantages that will be further defined in the following preclinical and clinical studies.
In most of the cases, before treatment with CAR-T cells, patients with hematological cancers receive lymphodepleting chemotherapy, which permits an appropriate immune environment for the CAR-T cell transfer, improving their in vivo function, progression, and persistence [23].
As of 2019, according to the CARGlobalTrials online database [24], there are a total of 353 CAR-T cell clinical trials involving 16,232 patients with hematological cancers, with 52% trials in phase I. So far, two anti-CD19 CAR-T products, Kymriah (Novartis) and Yescarta (Gilead), have received marketing authorization from the US Food and Drug Administration and the European Medicine Agency. Kymriah is indicated for the treatment of young adults and pediatric patients with refractory or relapsed acute lymphoblastic leukemia (ALL), whereas Yescarta is for the treatment of large B cell lymphoma in adult patients. Kymriah was the very first CAR-T product approved, after a successful phase I trial with the second generation of CAR (with 4-1BB as a costimulatory domain) targeting CD19. Out of the 30 patients tested with B- and T-cell ALL, 19 had a sustained complete remission and successfully recovered from cytokine release syndromes triggered by therapy [25]. A second-phase clinical trial is currently being performed in 101 patients with non-Hodgkin’s lymphoma (NHL), and thus far, 54% of patients are showing a complete remission [26]. Many other clinical trials are ongoing, most of them against CD19 and others targeting BCMA (B-cell maturation antigen) [27], CD20 [28], CD22 [29], CD30 [30], or LeY [31].
The engineering of T cells to express the CAR transgene is mainly accomplished using lentiviral [32] and retroviral vectors [33]. However, due to safety concerns inherent to lentiviral transduction [34], nonviral delivery vectors are being intensively studied. Nonviral delivery systems, such as the transposition method with Sleeping Beauty [35] and PiggyBac [36] or based on CRISPR-Cas9 technology [37], have already proved to achieve a stable integration and expression of CAR in T cells.
New studies point out the importance of the T cell ratio between cytotoxic CD8+ T cells and helper CD4+ T cells [38] and a subset selection of those T cells depending on their state of differentiation [39,40,41] to achieve efficacy in CAR-T cell immunotherapy. Recent clinical trials, such as JCAR017 from JUNO Therapeutics [42], are delivering a particular ratio of CD4+ and CD8+ CAR-T cells to reduce T cell malfunction and apoptosis.
While CAR-T cells are a huge step towards a cancer cure, they are not without drawbacks. One major concern is the manufacturing costs of autologous T cells for each patient, resulting in expensive treatment. The overall costs could be reduced by using an allogeneic T cell source, which is, however, burdened with life-threatening complications such as Graft-versus-host-disease (GvHD), as well as graft rejection by the host immune system. GvHD occurs when infused T cells from a donor get activated via TCR after recognizing the human leukocyte antigen (HLA) mismatch from the patient, resulting in donor T cells attacking the healthy patient’s tissues. Graft rejection is caused by the patient’s immune system, which attacks donor T cells after recognizing the HLA mismatch. To circumvent these problems and open the way to the use of allogenic T cells, new lines of research aimed at the generation of universal, off-the-shelf T cells with missing TCR and HLA molecules to avoid GvHD and graft rejection, respectively [43]. Another shortcoming of CAR-T cell therapy is the toxicity linked to the application of CAR-T cells. The side effects observed are neurologic toxicity, cytokine release syndrome, “on-target/off-tumor” recognition, and anaphylaxis [44]. These symptoms usually appear in clinical trials at different grades and may cause minor-to-severe side effects. To mitigate those undesired secondary effects, researchers have designed different strategies to eliminate or control CAR-T cell activity in case of severe toxicity [45]. These strategies include the expression of inducible suicide genes such as the Herpes simplex virus thymidine kinase [46] and caspase 9 [47], the expression of CD20 [48] and the epidermal growth factor receptor [49] to mediate a suicide switch with antibodies, CAR activation that requires the recognition of two different tumor antigens [50], the expression of a synthetic Notch that regulates the transcription of the CAR [51], the expression of immune inhibitory receptors [52], and on-switch CARs activated by a small molecule [53]. The most employed so far in clinical trials to treat hematological malignancies has been the suicide switch with antibodies, with a significant impact on the control of the undesired side effects. The major downside of that approach is the possible injury to healthy tissues that express the same antigen as the one recognized by the antibody. In consequence, this may restrict its future development, facilitating its substitution by the other approaches, where the damage to healthy cells is avoided.

3. Natural Killer (NK) Cells

Cancer immunotherapy based on NK cells has gradually risen over the past few years as an attractive and promising alternative to CAR-T cell therapy. The unique biological characteristics of NK cells allow us to circumvent two main limitations observed in the CAR-T cells. First, the tumor cells escape from T cell surveillance. The full immune response against cancer cells exerted by T cells relies on MHC-I recognition. Unfortunately, tumors have a propensity to downregulate MHC-I, leading to the escape of T cell antitumor actions [54]. In contrast to T cells, NK cells can exhibit a direct cytotoxic effect against tumor tissues lacking the expression of MHC-I [55]. NK cells express different activating and inhibitory receptors that, upon binding to specific ligands, govern the cytotoxic response. Some examples of activating receptors expressed by NK cells are NKp46, NK1.1, NKG2D, CD16, and CD244. NK cell inhibitory receptors fall into two groups: the monomeric type I glycoproteins of the immunoglobulin superfamily and the type II glycoproteins with a C-type lectin-like scaffold. Examples of type I are killer cell immunoglobulin-like receptors and leukocyte immunoglobulin-like receptors that recognize specific MHC-I molecules. Second, as the function of NK cells relies on the balance of signals from inhibitory and activating receptors that recognize a specific pattern of ligands in healthy cells and diseased ones, the infusion of allogeneic NK cells is safe and does not cause unwanted and deleterious GvHD [56], thus laying the foundation for the development of another universal, off-the-shelf cancer cell-based immunotherapy.
Different cell sources have been proposed for NK cell-based cancer therapy. Interestingly, one approach utilizes established NK cell lines, such as the NK-92 cell line, obtained from a patient with clonal NK cell lymphoma. The main advantage is that the NK-92 cell line can be very easily expanded in vitro and retains high antitumor activity [57]. This high activity is due to a consequence of their unique expression profile of receptors, with few inhibitory receptors and a relatively large amount of activating receptors [58]. A significant disadvantage, however, is the need to irradiate the therapeutic cells before the infusion to completely abrogate their proliferation, reducing, in consequence, the in vivo persistence of the effector cells.
Traditionally, NK cells are obtained from donor peripheral blood mononuclear cells [59] and umbilical cord blood [60] but, also, from stem cell sources, such as umbilical cord blood hematopoietic stem cells [61] and human pluripotent stem cells [62]. Each cell source has its inherent advantages and disadvantages, and more preclinical studies will be needed to untangle what the best source of NK cells is.
Similar to T cell-based therapies, NK cells have been armed with different CARs to boost their antitumor efficacy. The CAR structure is analogous to CARs used in T cells. In some cases, however, the signaling domain is slightly modified to be more adaptive to the characteristics of NK cells [63]. Several preclinical studies have already demonstrated the feasibility and benefits of CAR-NK technology. For example, the NK-92 cell line expressing an anti-CD138 CAR [64] showed a significantly enhanced cytotoxicity when compared to control NK-92 cells. In vivo experiments highlighted a significant reduction of the multiple myeloma (MM) tumor volume and increased the survival rate in xenograft mice models treated with anti-CD138 CAR NK-92 cells.
In 2019, the CARGlobalTrials database registered 11 clinical trials utilizing CAR-NK cells, with 174 patients enrolled. To date, only two studies have disclosed their results. The first disclosed phase I clinical trial was performed in three patients with acute myeloid leukemia (AML) with an infusion of NK-92 cells targeting CD33+ tumor cells [65]. The transplanted NK 92 cells did not produce any adverse effects on patients but also did not demonstrate any clinical efficacy. The second disclosed phase I/IIa clinical trial was conducted at the University of Texas MD Anderson Cancer Center and used CD19 CAR-NK cells derived from cord blood to treat 11 patients with relapsed or refractory NHL and chronic lymphocytic leukemia (CLL) [66]. In this case, the study revealed a favorable clinical outcome, with a 73% response rate without significant toxicities.
Overall, CAR-NK cells have demonstrated a great potential to overcome CAR-T cell limitations. However, the CAR-NK cell approach is a recent concept and, therefore, needs to be further improved to surpass its challenges. One of them is the resilience of the NK cells to be genetically engineered by viral and conventional nonviral gene-editing techniques [67]. The use of the endogenous baboon virus was recently found to significantly increase the transduction efficiency of NK cells as compared to VSV-G or RD114 pseudotyped lentiviruses [68]. Electroporation or use of cationic polymers on NK cells have also shown some limitations, even though optimized protocols have been published [69,70].

4. Dendritic Cells (DCs)

In 1973, DCs were discovered in mice by Ralph M. Steinman [71], who also elucidated their essential role in adaptive immunity. For his discoveries, he was awarded the Nobel Prize in Physiology or Medicine in 2011. DCs are part of the innate immune system and play a key role in antigen presentation. DCs have no direct killing activity against cancer cells but can present tumor-associated antigens (TAAs) to naïve T cells, leading to their activation. They can also activate other immune cells, such as NK cells. Therefore, the infusion of dendritic cells, called DC vaccines, has been proposed as a way of immune system reactivation in cancer patients by overcoming endogenous DC malfunctions [72,73] and, in turn, enhancing T cell responses against tumor cells.
Several clinical trials employing different approaches have been performed with DCs to treat patients with hematological cancers. In one approach, DCs were loaded in vitro with specific TAAs, applying diverse methods such as incubation with peptides derived from tumors [74]; tumor apoptotic bodies [75]; or whole tumor cells (lysed, heat-killed, or irradiated) [76]. Other teams directly engineered the DCs to express specific antigens [77] or even used DCs derived from leukemia patients naturally displaying TAAs on their surface [78]. Another approach selected a specific subset of cytotoxic DCs (killer DCs), which show the particularity to secrete cytotoxic molecules such as soluble TNF-related apoptosis-inducing ligand (sTRAIL), granzymes, or TNF-α [79]. The majority of these clinical trials reported safety, with mild side effects and an overall survival advantage.
Since DCs represent less than 1% of peripheral blood mononuclear cells, obtaining therapeutically meaningful numbers for vaccination purposes is one of the main problems. This problem can be bypassed by an allogeneic source of DCs, as they do not cause GvHD, or, alternatively, by the ex-vivo differentiation of CD14+ monocytes to DCs [80].
Overall, DC vaccines hold potential in blood cancer cell therapy, as the preclinical and clinical results are promising. Nevertheless, this field is still in its infancy, and more studies need to be carried out to improve the efficacy and elucidate the exact working mechanisms.

5. Macrophages

Macrophages are a type of specialized innate immune cells implicated in the detection, phagocytosis, and elimination of cellular debris, pathogens, and cancer cells. They are also involved in antigen presentation to T cells.
The commencement of macrophage-based therapy in cancer can be traced back to the work of Dr. Isaiah Fidler, who used macrophages previously cultured in vitro with conditioned media from B16 tumor cells (mouse melanoma cells) and sensitized lymphocytes. Such stimulated macrophages were then infused in mice with subcutaneous B16 tumors and achieved a significant reduction in pulmonary metastases [81]. From 1987 to 2010, autologous macrophages were employed in clinical trials to treat different types of solid tumors, with nonserious side effects in various dose-escalation regimens but with moderate therapeutic efficacy and no long-term remissions [82]. Monocytes from peripheral blood were the cell source in most of the cases. Once isolated, they were expanded ex vivo and differentiated to the cytotoxic phenotype (M1 macrophages) with 100–1000 U/mL human interferon-gamma (IFN-γ) before infusion [83].
Today, we have better understanding of the biodistribution and mechanism of action of macrophages. Two main reasons could be used to explain its failure in clinical trials, the tissue distribution of infused macrophages being the first. The tissue distribution of labeled macrophages with 111indium after intravenous infusion in patients with renal carcinoma shows infiltration in the lungs, liver, and spleen but with a lack of trafficking into the tumor [84]. The second is the tumor microenvironment’s capacity to polarize M1 macrophages towards an M2 macrophage’s phenotype, which is related to wound healing and tissue repair, thus boosting the tumor’s malignancy [85,86].
Recent preclinical studies have been focused on surpassing those pitfalls and increasing the macrophages’ efficacy as effector cells by genetic modifications [87,88,89] or by loading them with anticancer drugs or nanoparticles for photothermal therapies [90].
To date, no clinical trials with macrophages have been performed in hematological malignancies, and only a few preclinical studies have been reported. One of those preclinical approaches engineered macrophages to express a new type of CAR that activates their phagocytic mechanism after the recognition of CD19 in Burkitt’s lymphoma cell line Raji [91]. This novel CAR has an extracellular scFv that recognizes the antigen CD19 and an intracellular domain composed of Megf10 or FcRɣ capable of triggering phagocytosis after antigen recognition. The researchers observed that the engineered macrophages cocultured in vitro with the Raji cell line were able to engulf six cancer cells per 100 macrophages, reducing the number of cancer cells by 40%.
The majority of the recent striking trials in blood cancer cell-based therapy have been performed using the immune cells described above and summarized in Table 1.

6. Platelets

Platelets, or thrombocytes, are non-nucleated cell fragments. Their primary function is the formation of clots after blood vessel injury to stop bleeding and to maintain vascular integrity. Harnessing their capacity to adhere to tumor cells [97], platelets have been studied as a vehicle to deliver therapeutic drugs to cancer cells [98]. In a recent preclinical study, platelets were loaded with doxorubicin to deliver the cytotoxic agent to lymphoma cells [99]. The study proved the localized delivery of the drug to the tumor, increasing the therapeutic efficacy and reducing the cardiotoxicity associated with the chemotherapeutic agent.
Platelets are a promising drug delivery vehicle for cancer treatment because of their abundance [100] and their capacity to adhere to cancer cells. However, platelets can be a double-edged sword, as they also play an essential role in the stimulation of tumor dissemination and proliferation by acting on tumor angiogenesis [101], metastasis [102], growth [103], and, in chemotherapy, resistance [104].

7. Stem Cells and Progenitor Cells

Stem cells (SCs) are a particular type of cells with a high self-renewal capacity and exceptional ability to differentiate into many specialized cell types. Depending on their differentiation capacity, they can be classified as pluripotent or multipotent. Among the pluripotent SCs, we have the embryonic SCs and the induced pluripotent SCs, with the ability to differentiate into all three germ layers: mesoderm, endoderm, and ectoderm. In the group of multipotent SCs, we have the adult SCs that can be found throughout the body, with a more limited capacity to differentiate to diverse cell types. Examples of adult SCs are the hematopoietic SCs, the mesenchymal SCs, and the endothelial progenitor cells. They play the central role in the homeostasis and regeneration of all body tissues. SCs exhibit inherent tropism for cancer cells, which makes them an ideal therapeutic vehicle in anticancer therapy [105,106] (Table 2).

7.1. Hematopoietic Stem Cells (HSCs)

HSCs are adult multipotent stem cells responsible for generating all the blood cell types in the bone marrow by a process called hematopoiesis. The use of HSCs for cancer treatment is not new. The first successful HSC transplantation was accomplished by Dr. E. Donnall Thomas in 1957 in a patient with acute leukemia [112]. He received the Nobel Prize in Physiology or Medicine for this groundbreaking work. Nowadays, HSC transplantation is an established treatment for leukemia and other hematological malignancies [113,114], to overcome hematopoietic failure during the high doses of chemotherapy. HSC transplantation can be autologous, allogeneic, or syngeneic, and the cell source can be peripheral blood, bone marrow, or umbilical cord blood [115]. The selection of the source depends on the transplantation indication [116], as well as donor availability. In the allogeneic transplantation of HSCs, the immune cells in the graft can exert an immune response against residual malignant cells, in a process known as the graft versus tumor effect (GvT). An interesting recent approach used the ex vivo gene modification of those HSCs to boost the GvT by the expression of CARs or by the prearranging of the TCR. Thus, the modified HSC population transplanted into the patient can become a long-lasting source of T or NK cells engineered to recognize specific tumor antigens [117]. A recent preclinical study in 2019 has engineered HSC to express NY-ESO-1 TCR. They show how these cells can differentiate into all blood lineages, along with persistence and safety after transplantation to myelo-depleted HLA-A2/Kb mice. This preclinical study was performed to authorize an investigational new drug application for a clinical trial that is currently being conducted at the University of California, Los Angeles, to treat patients in stage IV or locally advanced unresectable cancers [118]. In the future, this approach could be applied in patients with a hematological cancer, as the NY-ESO-1 TCR can recognize the immunogenic cancer-testis antigen NY-ESO-1 expressed in more than 60% of advanced MM [119].
Whether these new approaches will lead to an increase in efficacy compared to the traditional HSC transplantation remains to be proven in clinical trials. Regardless, the use of genetically modified HSCs is an appealing approach, as it can provide a life-long source of effector immune cells engineered against the specific antigen and can continuously replenish exhausted immune cells.

7.2. Mesenchymal Stem Cells (MSCs)

MSCs are multipotent stromal cells that can differentiate to many cell types, such as osteoblasts, chondrocytes, and myocytes [120]. As a consequence of this unique capacity, MSCs have been extensively studied for tissue regeneration purposes [121,122,123,124]. More recently, MSCs have been proposed as a vehicle for targeted tumor therapy because of their tumor-tropic properties and relative resistance to chemotherapeutic drugs [125,126]. Tumor cells and tumor microenvironments secrete chemoattractant factors that induce MSC homing [127]. For instance, the expression of CCL25 by MM cells has been proved to attract MSCs after interaction with their receptor CCR9 [128]. The migration capacity of MSCs has also been studied in the context of radiation therapy, increasing their interest as a therapeutic vehicle for cancer treatment. Indeed, Klopp et al. [129] showed that MSCs have a better migration towards irradiated mouse mammary tumor cells (4T1) compared to nonirradiated ones. The migration enhancement after the irradiation of tumor cells was a consequence of the upregulation of cytokines involved in MSC migration, such as VEGF, PDGF-BB, TGF-β, and SDF-1. Besides, they observed that MSCs in the presence of irradiated tumor cells upregulated the expression of the chemokine receptor CCR2. Harnessing the tumor-tropic capacity of MSCs, Li et al. [130] and Ciavarella et al. [109] demonstrated the killing capacity of MSC towards leukemia and myeloma cells after engineering them for the expression of human interferon-gamma (IFN-γ) and TRAIL, respectively. Bonomi et al. [131] and Pessina et al. [108] took advantage of the resistance to chemotherapeutic drugs and tumor-tropic properties of MSCs and loaded them with paclitaxel (10,000 ng/mL) to suppress proliferation of the human MM cell line RPMI8226 and the leukemia cell line MOLT-4 in different preclinical studies. This capacity to absorb and release paclitaxel in tumors has also been proved in other chemotherapeutic drugs, such as doxorubicin and gentamicin, with an in vitro inhibition of cell growth on tongue squamous cell carcinoma [132].
Other assets of the MSCs are their relatively easy isolation, expansion, and genetic modification [133]. Besides, they have many possible tissue sources, such as bone marrow; peripheral blood; adipose tissue; or neonatal birth-associated tissues such as the cord blood, umbilical cord, or placenta [134]. The function of MSCs in hematological cancers is less-known than in solid tumors, but they share the same controversy. Probably because of the heterogeneity in the MSC population, there are studies that claim their antitumor properties [135]; others show the opposite due to evidence that they favor tumor growth [136]. The antitumor effect of MSCs in blood cancers has been reported primarily as a consequence of the suppression in the proliferation of malignant cells by the cell cycle arrest [137,138,139]. On the other hand, the MSCs have been stated to promote different hematological malignancies by activating metastasis/recurrence [140], suppressing apoptosis [141,142], involvement in the immunomodulation of cancer cells [143,144], supporting tumor vasculature [145], and inducing drug resistance [146,147]. More conclusive and standardized studies need to be performed in this matter before MSCs can be considered as an efficient therapeutic vehicle in hematological malignancies [148].

7.3. Endothelial Progenitor Cells (EPCs)

The definition of EPCs is still problematic due to their lack of specific markers. At the moment, they are characterized according to their capacity to differentiate to mature endothelial cells, to proliferate and migrate, and by functional parameters such as the ability to form vessels in vivo and tubular-like structures in vitro [149]. EPCs can be obtained from peripheral blood, bone marrow, cord blood, and other tissues such as adipose tissue [150].
Due to their tumor-homing properties, EPCs have been studied as a vehicle to deliver different therapeutic agents to tumors by the transgene expression of antiangiogenic agents, suicide genes, immune stimulators, or even employed as a virus and nanoparticle carrier to increase the primary therapeutic efficacy [151]. So far, no clinical trials with EPCs as a cancer therapeutic vehicle have been performed. The major hindrances in the translation of EPCs into the clinics are the absence of standardization in isolation and expansion, low numbers of EPCs after isolation [152], the immunogenicity in allogeneic sources, and their inherent protumor proliferation properties [153].

7.4. Induced Pluripotent Stem Cells (iPSCs)

iPSCs are derived from somatic cells by the expression of key transcription factors (Myc, Oct3/4, Sox2, and Klf4). The expression of those transcription factors reprograms the somatic cells into an embryonic-like pluripotent nature that permits the generation of an unlimited source of a specific cell type after the induction of differentiation. This revolutionary discovery was made by Takahashi and Yamanaka in 2006 [154], who were subsequently awarded the Nobel Prize in 2012. iPSCs derived from fibroblasts have been successfully differentiated into functional tumor-targeting T and NK cells. In 2009, Lei et al. [155] were the first to state that the differentiation of iPSCs to T lymphocytes is possible through the coculture of iPSCs (iPS-MEF-Ng-20D-17 cell line) with OP9 stromal cells expressing the Notch ligand Delta-like 1. Transfer of these cells into Rag-deficient mice restored T cell pools and generated mature T lymphocytes. A year later, Watari et al. [156] successfully obtained fully functional natural killer T cells (NKT) from iPSCs derived from murine embryonic fibroblasts. These NKT cells derived from iPSCs were able to suppress EG7 (murine T cell lymphoma) tumor growth in vivo in α18–/– mice. In 2013, Knorr et al. [110] were able to obtain a large number of cytotoxic NK cells from iPSCs derived from hematopoietic progenitor cells in a feeder-free system. In the same year, Themeli et al. [111] successfully combined iPSCs and CAR technologies to produce human T cells that target CD19 in malignant B cells. A recent publication by Li Zhang et al. [157] obtained functional macrophages of iPSC derived from peripheral blood mononuclear cells. These macrophages were engineered for the expression of CD19 CAR to trigger phagocytosis after tumor antigen recognition in leukemia cell lines Nalm6 and K562. Though some anticancer activity was observed in mouse models of leukemia, the results are not conclusive as a consequence of high variability, requiring further development.
At the moment, the trend is leading towards the development of T and NK cells derived from iPSCs that can be delivered off-the-shelf, simplifying the manufacturing process and reducing the overall costs compared to traditional approaches using autologous cells. A significant drawback in cells derived from iPSCs is the potential risk of teratoma formation due to the activation of pluripotency genes [158]. In the future, inducible CRISPR-Cas9 technology could be used to permanently turn off or even delete these genes.

8. Conclusions

As a consequence of the promising results obtained in the recent clinical trials, cancer cell-based therapy is flourishing as a new pillar in cancer treatment and is likely to become the cornerstone in future blood cancer treatments. However, for the time being, cancer cell-based therapy is a fledgling, and therefore, there is still a long road ahead (Figure 2). To consolidate this novel approach will require more basic and translational research to solve roadblocks such as effector toxicity, persistence, homing, tumor escape, and universal access. A better understanding of the different cell sources available may help to improve the future cell-based therapeutic approaches to treat hematological malignancies by selecting the proper cell type to increase the efficacy and to reduce toxicity and the cost of production. In saying that, a universal effector cell source for different cancers may not exist, and perhaps, it will be required to identify the best cell source for each type of cancer or the best combination of different effector cells to tackle specific cancer cell types.

Author Contributions

B.M.; S.C.; J.R.B. wrote the original draft and prepare the figures. M.H.; M.Š.; T.J.; T.Š.; Z.K.; R.H. reviewed and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This review was funded by the project “Cell Coolab Ostrava—Research and Development Center for Cell Therapy in Hematology and Oncology” (No. CZ.02.1.01/0.0/0.0/17_049/0008440), New Directions of Biomedical Research in the Ostrava Region (No. CZ.02.1.01/0.0/0.0/18_069/0010060), and the Institutional Development Plan of University of Ostrava, allocated by The Ministry of Education, Youth and Sports (project no. IRP03_2018-2020).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ALLAcute lymphoblastic leukemia
AMLAcute myeloid leukemia
AP-1Activator protein 1
B16Name of murine melanoma cell line
BCLB cell lymphoma
BCMAB-cell maturation antigen
CAR-TChimeric antigen receptor T-cell
CARsChimeric antigen receptors
Cas9CRISPR associated protein 9
CCL25C-C motif chemokine ligand 25
CCR2C-C motif chemokine receptor 2; cluster of differentiation 192
CCR9C-C motif chemokine receptor 9
CD137 ζCluster of differentiation 137 ζ
CD138Syndecan-1; Cluster of differentiation 138
CD14Cluster of differentiation 14
CD16Cluster of differentiation 16
CD19Cluster of differentiation 19
CD20Cluster of differentiation 20
CD22Cluster of differentiation 22
CD244Cluster of differentiation 244
CD28Cluster of differentiation 28
CD3Cluster of differentiation 3
CD30Tumor necrosis factor receptor superfamily member 8; Cluster of differentiation 30
CD33Cluster of differentiation
CD3ζCluster of differentiation 3ζ
CD4Cluster of differentiation 4
CD8Cluster of differentiation 8
CLLChronic lymphocytic leukemia
CMLChronic myelogenous leukemia
CRComplete response
CRISPRClustered regularly interspaced palindromic repeats
CRSCytokine release syndrome
DCsDendritic cells
DLBCLDiffuse large B-cell lymphoma
EG7Name of cell line derived from murine T-cell lymphoma
EGFRtEpidermal growth factor receptor
EPCsEndothelial progenitor cells
FcRγFc receptor gamma
FDAFood and Drug Administration
GvHDGraft versus host disease
GvTGraft versus tumor effect
HLAHuman leukocyte antigen
HSCsHematopoietic stem cells
IFN-γHuman interferon gamma
IL-2Interleukin 2
iPS-MEF-Ng-20D-17Name of mouse induced pluripotent stem cell line
iPSCsInduced pluripotent stem cells
K562Human erytroleukemic cell line
Klf4Kruppel-like factor 4
L1210Name of murine cancer cell line derived from lymphoblasts
LeYLewis Y antigen
M1Macrophage phenotype 1
M2Macrophage phenotype 2
MDSMyelodysplastic syndrome
Megf10Multiple epidermal growth factor-like domains 10
MHCMajor histocompatibility complex
MM Multiple myeloma
MRDMinimal residual disease
mRNAMessenger ribonucleic acid
MSCsMesenchymal stem cells
MycMaster regulator of cell cycle entry and proliferative metabolism
Nalm6Human B cell precursor leukemia cell line
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
NFATNuclear factor of activated T-cells
NHLNon-Hodgkin’s lymphoma
NKNatural killer
NK-92Name of human NK cell line
NK1.1NK cell-associated marker 1.1; cluster of differentiation 161
NKG2DNatural killer group 2D receptor
NKp46Natural cytotoxicity triggering receptor 1; cluster of differentiation 335
NKTNatural killer T cells
NTNeurotoxicity
NY-ESO-1Cancer-testis antigen
Oct3/4Octamer-binding transcription factor 4
OP9Name of murine embryonic cell line
ORObjective response
PDProgressive disease
PDGF-BBPlatelet-derived growth factor BB
PRPartial response
RD114Name of envelope glycoprotein of lentiviral vectors
RPMI8226Name of multiple myeloma human cancer cell line derived from B lymphocytes
scFvSingle-chain variable fragment
SCRSustained complete response
SCsStem cells
SDStable disease
SDF-1Stromal cell derived factor 1
Sox2Sex determining region Y-box 2
sTRAILSoluble TNF-related apoptosis-inducing ligand
SUPRASplit Universal Programmable
TAAsTumor-associated antigens
TCRT cell receptor
TGF-βTransforming growth factor β
TNF-αTumor necrosis factor alpha
TRAILTumor necrosis factor-related apoptosis-inducing ligand
TRUCKsT cells redirected for universal cytokine-mediated killing
U-266Name of human cancer cell line derived from B lymphocyte
VEGFVascular endothelial growth factor
VGPRVery good partial remission
VSV-GVesicular stomatitis virus G
WT1Wilms’ tumor 1
4T1Murine mammary tumor cell line

References

  1. SEER Cancer Statistics Review (CSR) 1975-2014. Available online: https://seer.cancer.gov (accessed on 22 May 2020).
  2. DeSantis, C.E.; Miller, K.D.; Dale, W.; Mohile, S.G.; Cohen, H.J.; Leach, C.R.; Goding Sauer, A.; Jemal, A.; Siegel, R.L. Cancer statistics for adults aged 85 years and older, 2019. CA Cancer J. Clin. 2019, 69, 452–467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Oostindie, S.C.; van der Horst, H.J.; Kil, L.P.; Strumane, K.; Overdijk, M.B.; van den Brink, E.N.; van den Brakel, J.H.N.; Rademaker, H.J.; van Kessel, B.; van den Noort, J.; et al. DuoHexaBody-CD37((R)), a novel biparatopic CD37 antibody with enhanced Fc-mediated hexamerization as a potential therapy for B-cell malignancies. Blood Cancer J. 2020, 10, 30. [Google Scholar] [CrossRef] [PubMed]
  4. Bonello, F.; D’Agostino, M.; Moscvin, M.; Cerrato, C.; Boccadoro, M.; Gay, F. CD38 as an immunotherapeutic target in multiple myeloma. Expert Opin. Biol. Ther. 2018, 18, 1209–1221. [Google Scholar] [CrossRef] [PubMed]
  5. Salles, G.; Barrett, M.; Foa, R.; Maurer, J.; O’Brien, S.; Valente, N.; Wenger, M.; Maloney, D.G. Rituximab in B-Cell Hematologic Malignancies: A Review of 20 Years of Clinical Experience. Adv. Ther. 2017, 34, 2232–2273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Gottardi, M.; Mosna, F.; de Angeli, S.; Papayannidis, C.; Candoni, A.; Clavio, M.; Tecchio, C.; Piccin, A.; dell’Orto, M.C.; Benedetti, F.; et al. Clinical and experimental efficacy of gemtuzumab ozogamicin in core binding factor acute myeloid leukemia. Hematol. Rep. 2017, 9, 7029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Abdallah, N.; Kumar, S.K. Daratumumab in untreated newly diagnosed multiple myeloma. Ther. Adv. Hematol. 2019, 10, 2040620719894871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Mori, Y.; Choi, I.; Yoshimoto, G.; Muta, T.; Yamasaki, S.; Tanimoto, K.; Kamimura, T.; Iwasaki, H.; Ogawa, R.; Akashi, K.; et al. Phase I/II study of bortezomib, lenalidomide, and dexamethasone treatment for relapsed and refractory multiple myeloma. Int. J. Hematol. 2020, 111, 673–680. [Google Scholar] [CrossRef]
  9. Fathi, E.; Farahzadi, R.; Sheervalilou, R.; Sanaat, Z.; Vietor, I. A general view of CD33(+) leukemic stem cells and CAR-T cells as interesting targets in acute myeloblatsic leukemia therapy. Blood Res. 2020, 55, 10–16. [Google Scholar] [CrossRef] [Green Version]
  10. Lee, H.R.; Baek, K.H. Role of natural killer cells for immunotherapy in chronic myeloid leukemia (Review). Oncol. Rep. 2019, 41, 2625–2635. [Google Scholar] [CrossRef]
  11. Van Acker, H.H.; Versteven, M.; Lichtenegger, F.S.; Roex, G.; Campillo-Davo, D.; Lion, E.; Subklewe, M.; Van Tendeloo, V.F.; Berneman, Z.N.; Anguille, S. Dendritic Cell-Based Immunotherapy of Acute Myeloid Leukemia. J. Clin. Med. 2019, 8, 579. [Google Scholar] [CrossRef] [Green Version]
  12. Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.; Myers, G.D.; et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 439–448. [Google Scholar] [CrossRef] [PubMed]
  13. Song, W.; Zhang, M. Use of CAR-T cell therapy, PD-1 blockade, and their combination for the treatment of hematological malignancies. Clin. Immunol. 2020, 214, 108382. [Google Scholar] [CrossRef] [PubMed]
  14. Bollard, C.M.; Gottschalk, S.; Torrano, V.; Diouf, O.; Ku, S.; Hazrat, Y.; Carrum, G.; Ramos, C.; Fayad, L.; Shpall, E.J.; et al. Sustained complete responses in patients with lymphoma receiving autologous cytotoxic T lymphocytes targeting Epstein-Barr virus latent membrane proteins. J. Clin. Oncol. 2014, 32, 798–808. [Google Scholar] [CrossRef] [PubMed]
  15. Gross, G.; Waks, T.; Eshhar, Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl. Acad. Sci. USA 1989, 86, 10024–10028. [Google Scholar] [CrossRef] [Green Version]
  16. Abate-Daga, D.; Davila, M.L. CAR models: Next-generation CAR modifications for enhanced T-cell function. Mol. Ther. Oncolytics 2016, 3, 16014. [Google Scholar] [CrossRef] [Green Version]
  17. Davila, M.L.; Riviere, I.; Wang, X.; Bartido, S.; Park, J.; Curran, K.; Chung, S.S.; Stefanski, J.; Borquez-Ojeda, O.; Olszewska, M.; et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci. Transl. Med. 2014, 6, 224ra225. [Google Scholar] [CrossRef] [Green Version]
  18. Drent, E.; Poels, R.; Ruiter, R.; van de Donk, N.; Zweegman, S.; Yuan, H.; de Bruijn, J.; Sadelain, M.; Lokhorst, H.M.; Groen, R.W.J.; et al. Combined CD28 and 4-1BB Costimulation Potentiates Affinity-tuned Chimeric Antigen Receptor-engineered T Cells. Clin. Cancer Res. 2019, 25, 4014–4025. [Google Scholar] [CrossRef] [Green Version]
  19. Chmielewski, M.; Abken, H. TRUCKs: The fourth generation of CARs. Expert Opin. Biol. Ther. 2015, 15, 1145–1154. [Google Scholar] [CrossRef]
  20. Zhao, J.; Song, Y.; Liu, D. Clinical trials of dual-target CAR T cells, donor-derived CAR T cells, and universal CAR T cells for acute lymphoid leukemia. J. Hematol. Oncol. 2019, 12, 17. [Google Scholar] [CrossRef] [Green Version]
  21. Cho, J.H.; Collins, J.J.; Wong, W.W. Universal Chimeric Antigen Receptors for Multiplexed and Logical Control of T Cell Responses. Cell 2018, 173, 1426–1438. [Google Scholar] [CrossRef] [Green Version]
  22. Lohmueller, J.J.; Ham, J.D.; Kvorjak, M.; Finn, O.J. mSA2 affinity-enhanced biotin-binding CAR T cells for universal tumor targeting. Oncoimmunology 2017, 7, e1368604. [Google Scholar] [CrossRef] [PubMed]
  23. Brentjens, R.J.; Riviere, I.; Park, J.H.; Davila, M.L.; Wang, X.; Stefanski, J.; Taylor, C.; Yeh, R.; Bartido, S.; Borquez-Ojeda, O.; et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 2011, 118, 4817–4828. [Google Scholar] [CrossRef] [PubMed]
  24. MacKay, M. CARGlobalTrials. Available online: https://carglobaltrials.com/ (accessed on 22 May 2020).
  25. Maude, S.L.; Frey, N.; Shaw, P.A.; Aplenc, R.; Barrett, D.M.; Bunin, N.J.; Chew, A.; Gonzalez, V.E.; Zheng, Z.; Lacey, S.F.; et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 2014, 371, 1507–1517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Vairy, S.; Garcia, J.L.; Teira, P.; Bittencourt, H. CTL019 (tisagenlecleucel): CAR-T therapy for relapsed and refractory B-cell acute lymphoblastic leukemia. Drug Des. Devel. Ther. 2018, 12, 3885–3898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Raje, N.; Berdeja, J.; Lin, Y.; Siegel, D.; Jagannath, S.; Madduri, D.; Liedtke, M.; Rosenblatt, J.; Maus, M.V.; Turka, A.; et al. Anti-BCMA CAR T-Cell Therapy bb2121 in Relapsed or Refractory Multiple Myeloma. N. Engl. J. Med. 2019, 380, 1726–1737. [Google Scholar] [CrossRef] [PubMed]
  28. Zhang, W.Y.; Wang, Y.; Guo, Y.L.; Dai, H.R.; Yang, Q.M.; Zhang, Y.J.; Zhang, Y.; Chen, M.X.; Wang, C.M.; Feng, K.C.; et al. Treatment of CD20-directed Chimeric Antigen Receptor-modified T cells in patients with relapsed or refractory B-cell non-Hodgkin lymphoma: An early phase IIa trial report. Signal Transduct. Target. Ther. 2016, 1, 16002. [Google Scholar] [CrossRef]
  29. Fry, T.J.; Shah, N.N.; Orentas, R.J.; Stetler-Stevenson, M.; Yuan, C.M.; Ramakrishna, S.; Wolters, P.; Martin, S.; Delbrook, C.; Yates, B.; et al. CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat. Med. 2018, 24, 20–28. [Google Scholar] [CrossRef] [Green Version]
  30. Ramos, C.A.; Ballard, B.; Zhang, H.; Dakhova, O.; Gee, A.P.; Mei, Z.; Bilgi, M.; Wu, M.F.; Liu, H.; Grilley, B.; et al. Clinical and immunological responses after CD30-specific chimeric antigen receptor-redirected lymphocytes. J. Clin. Invest. 2017, 127, 3462–3471. [Google Scholar] [CrossRef]
  31. Ritchie, D.S.; Neeson, P.J.; Khot, A.; Peinert, S.; Tai, T.; Tainton, K.; Chen, K.; Shin, M.; Wall, D.M.; Honemann, D.; et al. Persistence and efficacy of second generation CAR T cell against the LeY antigen in acute myeloid leukemia. Mol. Ther. 2013, 21, 2122–2129. [Google Scholar] [CrossRef] [Green Version]
  32. Picanco-Castro, V.; Moco, P.D.; Mizukami, A.; Vaz, L.D.; de Souza Fernandes Pereira, M.; Silvestre, R.N.; de Azevedo, J.T.C.; de Sousa Bomfim, A.; de Abreu Neto, M.S.; Malmegrim, K.C.R.; et al. Establishment of a simple and efficient platform for car-t cell generation and expansion: From lentiviral production to in vivo studies. Hematol. Transfus. Cell Ther. 2019. [Google Scholar] [CrossRef]
  33. Pampusch, M.S.; Haran, K.P.; Hart, G.T.; Rakasz, E.G.; Rendahl, A.K.; Berger, E.A.; Connick, E.; Skinner, P.J. Rapid Transduction and Expansion of Transduced T Cells with Maintenance of Central Memory Populations. Mol. Ther. Methods Clin. Dev. 2020, 16, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Schlimgen, R.; Howard, J.; Wooley, D.; Thompson, M.; Baden, L.R.; Yang, O.O.; Christiani, D.C.; Mostoslavsky, G.; Diamond, D.V.; Duane, E.G.; et al. Risks Associated With Lentiviral Vector Exposures and Prevention Strategies. J. Occup. Environ. Med. 2016, 58, 1159–1166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Kebriaei, P.; Singh, H.; Huls, M.H.; Figliola, M.J.; Bassett, R.; Olivares, S.; Jena, B.; Dawson, M.J.; Kumaresan, P.R.; Su, S.; et al. Phase I trials using Sleeping Beauty to generate CD19-specific CAR T cells. J. Clin. Invest. 2016, 126, 3363–3376. [Google Scholar] [CrossRef] [PubMed]
  36. Morita, D.; Nishio, N.; Saito, S.; Tanaka, M.; Kawashima, N.; Okuno, Y.; Suzuki, S.; Matsuda, K.; Maeda, Y.; Wilson, M.H.; et al. Enhanced Expression of Anti-CD19 Chimeric Antigen Receptor in piggyBac Transposon-Engineered T Cells. Mol. Ther. Methods Clin. Dev. 2018, 8, 131–140. [Google Scholar] [CrossRef] [Green Version]
  37. Eyquem, J.; Mansilla-Soto, J.; Giavridis, T.; van der Stegen, S.J.; Hamieh, M.; Cunanan, K.M.; Odak, A.; Gonen, M.; Sadelain, M. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 2017, 543, 113–117. [Google Scholar] [CrossRef] [Green Version]
  38. Janssen, E.M.; Lemmens, E.E.; Wolfe, T.; Christen, U.; von Herrath, M.G.; Schoenberger, S.P. CD4+ T cells are required for secondary expansion and memory in CD8+ T lymphocytes. Nature 2003, 421, 852–856. [Google Scholar] [CrossRef]
  39. Berger, C.; Jensen, M.C.; Lansdorp, P.M.; Gough, M.; Elliott, C.; Riddell, S.R. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J. Clin. Invest. 2008, 118, 294–305. [Google Scholar] [CrossRef] [Green Version]
  40. Xu, Y.; Zhang, M.; Ramos, C.A.; Durett, A.; Liu, E.; Dakhova, O.; Liu, H.; Creighton, C.J.; Gee, A.P.; Heslop, H.E.; et al. Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15. Blood 2014, 123, 3750–3759. [Google Scholar] [CrossRef] [Green Version]
  41. Golubovskaya, V.; Wu, L. Different Subsets of T Cells, Memory, Effector Functions, and CAR-T Immunotherapy. Cancers (Basel) 2016, 8, 36. [Google Scholar] [CrossRef] [Green Version]
  42. Chavez, J.C.; Bachmeier, C.; Kharfan-Dabaja, M.A. CAR T-cell therapy for B-cell lymphomas: Clinical trial results of available products. Ther. Adv. Hematol. 2019, 10, 2040620719841581. [Google Scholar] [CrossRef] [Green Version]
  43. Depil, S.; Duchateau, P.; Grupp, S.A.; Mufti, G.; Poirot, L. ‘Off-the-shelf’ allogeneic CAR T cells: Development and challenges. Nat. Rev. Drug Discov. 2020, 19, 185–199. [Google Scholar] [CrossRef] [PubMed]
  44. Bonifant, C.L.; Jackson, H.J.; Brentjens, R.J.; Curran, K.J. Toxicity and management in CAR T-cell therapy. Mol. Ther. Oncolytics 2016, 3, 16011. [Google Scholar] [CrossRef] [PubMed]
  45. Yu, S.; Yi, M.; Qin, S.; Wu, K. Next generation chimeric antigen receptor T cells: Safety strategies to overcome toxicity. Mol. Cancer 2019, 18, 125. [Google Scholar] [CrossRef] [PubMed]
  46. Diaconu, I.; Ballard, B.; Zhang, M.; Chen, Y.; West, J.; Dotti, G.; Savoldo, B. Inducible Caspase-9 Selectively Modulates the Toxicities of CD19-Specific Chimeric Antigen Receptor-Modified T Cells. Mol. Ther. 2017, 25, 580–592. [Google Scholar] [CrossRef] [Green Version]
  47. Gargett, T.; Brown, M.P. The inducible caspase-9 suicide gene system as a “safety switch” to limit on-target, off-tumor toxicities of chimeric antigen receptor T cells. Front. Pharmacol. 2014, 5, 235. [Google Scholar] [CrossRef]
  48. Griffioen, M.; van Egmond, E.H.; Kester, M.G.; Willemze, R.; Falkenburg, J.H.; Heemskerk, M.H. Retroviral transfer of human CD20 as a suicide gene for adoptive T-cell therapy. Haematologica 2009, 94, 1316–1320. [Google Scholar] [CrossRef] [Green Version]
  49. Paszkiewicz, P.J.; Frassle, S.P.; Srivastava, S.; Sommermeyer, D.; Hudecek, M.; Drexler, I.; Sadelain, M.; Liu, L.; Jensen, M.C.; Riddell, S.R.; et al. Targeted antibody-mediated depletion of murine CD19 CAR T cells permanently reverses B cell aplasia. J. Clin. Invest. 2016, 126, 4262–4272. [Google Scholar] [CrossRef]
  50. Grada, Z.; Hegde, M.; Byrd, T.; Shaffer, D.R.; Ghazi, A.; Brawley, V.S.; Corder, A.; Schonfeld, K.; Koch, J.; Dotti, G.; et al. TanCAR: A Novel Bispecific Chimeric Antigen Receptor for Cancer Immunotherapy. Mol. Ther. Nucleic Acids 2013, 2, e105. [Google Scholar] [CrossRef]
  51. Yuan, X.; Wu, H.; Xu, H.; Xiong, H.; Chu, Q.; Yu, S.; Wu, G.S.; Wu, K. Notch signaling: An emerging therapeutic target for cancer treatment. Cancer Lett. 2015, 369, 20–27. [Google Scholar] [CrossRef] [Green Version]
  52. Fedorov, V.D.; Themeli, M.; Sadelain, M. PD-1- and CTLA-4-based inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy responses. Sci. Transl. Med. 2013, 5, 215ra172. [Google Scholar] [CrossRef] [Green Version]
  53. Wu, C.Y.; Roybal, K.T.; Puchner, E.M.; Onuffer, J.; Lim, W.A. Remote control of therapeutic T cells through a small molecule-gated chimeric receptor. Science 2015, 350, aab4077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Garrido, F.; Perea, F.; Bernal, M.; Sanchez-Palencia, A.; Aptsiauri, N.; Ruiz-Cabello, F. The Escape of Cancer from T Cell-Mediated Immune Surveillance: HLA Class I Loss and Tumor Tissue Architecture. Vaccines (Basel) 2017, 5, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Paul, S.; Lal, G. The Molecular Mechanism of Natural Killer Cells Function and Its Importance in Cancer Immunotherapy. Front. Immunol. 2017, 8, 1124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Jaiswal, S.R.; Zaman, S.; Nedunchezhian, M.; Chakrabarti, A.; Bhakuni, P.; Ahmed, M.; Sharma, K.; Rawat, S.; O’Donnell, P.; Chakrabarti, S. CD56-enriched donor cell infusion after post-transplantation cyclophosphamide for haploidentical transplantation of advanced myeloid malignancies is associated with prompt reconstitution of mature natural killer cells and regulatory T cells with reduced incidence of acute graft versus host disease: A pilot study. Cytotherapy 2017, 19, 531–542. [Google Scholar] [CrossRef]
  57. Williams, B.A.; Law, A.D.; Routy, B.; denHollander, N.; Gupta, V.; Wang, X.H.; Chaboureau, A.; Viswanathan, S.; Keating, A. A phase I trial of NK-92 cells for refractory hematological malignancies relapsing after autologous hematopoietic cell transplantation shows safety and evidence of efficacy. Oncotarget 2017, 8, 89256–89268. [Google Scholar] [CrossRef] [Green Version]
  58. Maki, G.; Klingemann, H.G.; Martinson, J.A.; Tam, Y.K. Factors regulating the cytotoxic activity of the human natural killer cell line, NK-92. J. Hematother. Stem Cell Res. 2001, 10, 369–383. [Google Scholar] [CrossRef]
  59. Masuyama, J.; Murakami, T.; Iwamoto, S.; Fujita, S. Ex vivo expansion of natural killer cells from human peripheral blood mononuclear cells co-stimulated with anti-CD3 and anti-CD52 monoclonal antibodies. Cytotherapy 2016, 18, 80–90. [Google Scholar] [CrossRef]
  60. Herrera, L.; Santos, S.; Vesga, M.A.; Anguita, J.; Martin-Ruiz, I.; Carrascosa, T.; Juan, M.; Eguizabal, C. Adult peripheral blood and umbilical cord blood NK cells are good sources for effective CAR therapy against CD19 positive leukemic cells. Sci. Rep. 2019, 9, 18729. [Google Scholar] [CrossRef] [Green Version]
  61. Herrera, L.; Salcedo, J.M.; Santos, S.; Vesga, M.A.; Borrego, F.; Eguizabal, C. OP9 Feeder Cells Are Superior to M2-10B4 Cells for the Generation of Mature and Functional Natural Killer Cells from Umbilical Cord Hematopoietic Progenitors. Front. Immunol. 2017, 8, 755. [Google Scholar] [CrossRef] [Green Version]
  62. Nianias, A.; Themeli, M. Induced Pluripotent Stem Cell (iPSC)-Derived Lymphocytes for Adoptive Cell Immunotherapy: Recent Advances and Challenges. Curr. Hematol. Malig. Rep. 2019, 14, 261–268. [Google Scholar] [CrossRef] [Green Version]
  63. Topfer, K.; Cartellieri, M.; Michen, S.; Wiedemuth, R.; Muller, N.; Lindemann, D.; Bachmann, M.; Fussel, M.; Schackert, G.; Temme, A. DAP12-based activating chimeric antigen receptor for NK cell tumor immunotherapy. J. Immunol. 2015, 194, 3201–3212. [Google Scholar] [CrossRef] [PubMed]
  64. Jiang, H.; Zhang, W.; Shang, P.; Zhang, H.; Fu, W.; Ye, F.; Zeng, T.; Huang, H.; Zhang, X.; Sun, W.; et al. Transfection of chimeric anti-CD138 gene enhances natural killer cell activation and killing of multiple myeloma cells. Mol. Oncol. 2014, 8, 297–310. [Google Scholar] [CrossRef] [PubMed]
  65. Tang, X.; Yang, L.; Li, Z.; Nalin, A.P.; Dai, H.; Xu, T.; Yin, J.; You, F.; Zhu, M.; Shen, W.; et al. First-in-man clinical trial of CAR NK-92 cells: Safety test of CD33-CAR NK-92 cells in patients with relapsed and refractory acute myeloid leukemia. Am. J. Cancer Res. 2018, 8, 1083–1089. [Google Scholar] [PubMed]
  66. Liu, E.; Marin, D.; Banerjee, P.; Macapinlac, H.A.; Thompson, P.; Basar, R.; Nassif Kerbauy, L.; Overman, B.; Thall, P.; Kaplan, M.; et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N. Engl. J. Med. 2020, 382, 545–553. [Google Scholar] [CrossRef] [PubMed]
  67. Carlsten, M.; Childs, R.W. Genetic Manipulation of NK Cells for Cancer Immunotherapy: Techniques and Clinical Implications. Front. Immunol. 2015, 6, 266. [Google Scholar] [CrossRef] [Green Version]
  68. Colamartino, A.B.L.; Lemieux, W.; Bifsha, P.; Nicoletti, S.; Chakravarti, N.; Sanz, J.; Romero, H.; Selleri, S.; Beland, K.; Guiot, M.; et al. Efficient and Robust NK-Cell Transduction With Baboon Envelope Pseudotyped Lentivector. Front. Immunol. 2019, 10, 2873. [Google Scholar] [CrossRef]
  69. Ingegnere, T.; Mariotti, F.R.; Pelosi, A.; Quintarelli, C.; De Angelis, B.; Tumino, N.; Besi, F.; Cantoni, C.; Locatelli, F.; Vacca, P.; et al. Human CAR NK Cells: A New Non-viral Method Allowing High Efficient Transfection and Strong Tumor Cell Killing. Front. Immunol. 2019, 10, 957. [Google Scholar] [CrossRef] [Green Version]
  70. Wang, J.; Lupo, K.B.; Chambers, A.M.; Matosevic, S. Purinergic targeting enhances immunotherapy of CD73(+) solid tumors with piggyBac-engineered chimeric antigen receptor natural killer cells. J. Immunother. Cancer 2018, 6, 136. [Google Scholar] [CrossRef]
  71. Steinman, R.M.; Cohn, Z.A. Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J. Exp. Med. 1973, 137, 1142–1162. [Google Scholar] [CrossRef]
  72. Ma, Y.; Shurin, G.V.; Peiyuan, Z.; Shurin, M.R. Dendritic cells in the cancer microenvironment. J. Cancer 2013, 4, 36–44. [Google Scholar] [CrossRef] [Green Version]
  73. Gabrilovich, D. Mechanisms and functional significance of tumour-induced dendritic-cell defects. Nat. Rev. Immunol. 2004, 4, 941–952. [Google Scholar] [CrossRef] [PubMed]
  74. Timmerman, J.M.; Czerwinski, D.K.; Davis, T.A.; Hsu, F.J.; Benike, C.; Hao, Z.M.; Taidi, B.; Rajapaksa, R.; Caspar, C.B.; Okada, C.Y.; et al. Idiotype-pulsed dendritic cell vaccination for B-cell lymphoma: Clinical and immune responses in 35 patients. Blood 2002, 99, 1517–1526. [Google Scholar] [CrossRef] [PubMed]
  75. Kokhaei, P.; Choudhury, A.; Mahdian, R.; Lundin, J.; Moshfegh, A.; Osterborg, A.; Mellstedt, H. Apoptotic tumor cells are superior to tumor cell lysate, and tumor cell RNA in induction of autologous T cell response in B-CLL. Leukemia 2004, 18, 1810–1815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Ho, V.T.; Vanneman, M.; Kim, H.; Sasada, T.; Kang, Y.J.; Pasek, M.; Cutler, C.; Koreth, J.; Alyea, E.; Sarantopoulos, S.; et al. Biologic activity of irradiated, autologous, GM-CSF-secreting leukemia cell vaccines early after allogeneic stem cell transplantation. Proc. Natl. Acad. Sci. USA 2009, 106, 15825–15830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Van Driessche, A.; Van de Velde, A.L.; Nijs, G.; Braeckman, T.; Stein, B.; De Vries, J.M.; Berneman, Z.N.; Van Tendeloo, V.F. Clinical-grade manufacturing of autologous mature mRNA-electroporated dendritic cells and safety testing in acute myeloid leukemia patients in a phase I dose-escalation clinical trial. Cytotherapy 2009, 11, 653–668. [Google Scholar] [CrossRef]
  78. Roddie, H.; Klammer, M.; Thomas, C.; Thomson, R.; Atkinson, A.; Sproul, A.; Waterfall, M.; Samuel, K.; Yin, J.; Johnson, P.; et al. Phase I/II study of vaccination with dendritic-like leukaemia cells for the immunotherapy of acute myeloid leukaemia. Br. J. Haematol. 2006, 133, 152–157. [Google Scholar] [CrossRef]
  79. Tel, J.; Anguille, S.; Waterborg, C.E.; Smits, E.L.; Figdor, C.G.; de Vries, I.J. Tumoricidal activity of human dendritic cells. Trends Immunol. 2014, 35, 38–46. [Google Scholar] [CrossRef]
  80. Zhou, L.J.; Tedder, T.F. CD14+ blood monocytes can differentiate into functionally mature CD83+ dendritic cells. Proc. Natl. Acad. Sci. USA 1996, 93, 2588–2592. [Google Scholar] [CrossRef] [Green Version]
  81. Fidler, I.J. Inhibition of pulmonary metastasis by intravenous injection of specifically activated macrophages. Cancer Res. 1974, 34, 1074–1078. [Google Scholar]
  82. Andreesen, R.; Hennemann, B.; Krause, S.W. Adoptive immunotherapy of cancer using monocyte-derived macrophages: Rationale, current status, and perspectives. J. Leukoc. Biol. 1998, 64, 419–426. [Google Scholar] [CrossRef]
  83. Stevenson, H.C.; Keenan, A.M.; Woodhouse, C.; Ottow, R.T.; Miller, P.; Steller, E.P.; Foon, K.A.; Abrams, P.G.; Beman, J.; Larson, S.M.; et al. Fate of gamma-interferon-activated killer blood monocytes adoptively transferred into the abdominal cavity of patients with peritoneal carcinomatosis. Cancer Res. 1987, 47, 6100–6103. [Google Scholar] [PubMed]
  84. Quillien, V.; Moisan, A.; Lesimple, T.; Leberre, C.; Toujas, L. Biodistribution of 111indium-labeled macrophages infused intravenously in patients with renal carcinoma. Cancer Immunol. Immunother. 2001, 50, 477–482. [Google Scholar] [CrossRef] [PubMed]
  85. Ruffell, B.; Affara, N.I.; Coussens, L.M. Differential macrophage programming in the tumor microenvironment. Trends Immunol. 2012, 33, 119–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Squadrito, M.L.; De Palma, M. Macrophage regulation of tumor angiogenesis: Implications for cancer therapy. Mol. Aspects Med. 2011, 32, 123–145. [Google Scholar] [CrossRef] [PubMed]
  87. Kan, O.; Day, D.; Iqball, S.; Burke, F.; Grimshaw, M.J.; Naylor, S.; Binley, K. Genetically modified macrophages expressing hypoxia regulated cytochrome P450 and P450 reductase for the treatment of cancer. Int. J. Mol. Med. 2011, 27, 173–180. [Google Scholar] [CrossRef] [PubMed]
  88. Basel, M.T.; Balivada, S.; Shrestha, T.B.; Seo, G.M.; Pyle, M.M.; Tamura, M.; Bossmann, S.H.; Troyer, D.L. A cell-delivered and cell-activated SN38-dextran prodrug increases survival in a murine disseminated pancreatic cancer model. Small 2012, 8, 913–920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Hagemann, T.; Lawrence, T.; McNeish, I.; Charles, K.A.; Kulbe, H.; Thompson, R.G.; Robinson, S.C.; Balkwill, F.R. “Re-educating” tumor-associated macrophages by targeting NF-kappaB. J. Exp. Med. 2008, 205, 1261–1268. [Google Scholar] [CrossRef] [PubMed]
  90. Yang, T.D.; Choi, W.; Yoon, T.H.; Lee, K.J.; Lee, J.S.; Joo, J.H.; Lee, M.G.; Yim, H.S.; Choi, K.M.; Kim, B.; et al. In vivo photothermal treatment by the peritumoral injection of macrophages loaded with gold nanoshells. Biomed. Opt. Express 2016, 7, 185–193. [Google Scholar] [CrossRef] [Green Version]
  91. Morrissey, M.A.; Williamson, A.P.; Steinbach, A.M.; Roberts, E.W.; Kern, N.; Headley, M.B.; Vale, R.D. Chimeric antigen receptors that trigger phagocytosis. Elife 2018, 7. [Google Scholar] [CrossRef]
  92. Van Tendeloo, V.F.; Van de Velde, A.; Van Driessche, A.; Cools, N.; Anguille, S.; Ladell, K.; Gostick, E.; Vermeulen, K.; Pieters, K.; Nijs, G.; et al. Induction of complete and molecular remissions in acute myeloid leukemia by Wilms’ tumor 1 antigen-targeted dendritic cell vaccination. Proc. Natl. Acad. Sci. USA 2010, 107, 13824–13829. [Google Scholar] [CrossRef] [Green Version]
  93. Rosenblatt, J.; Avivi, I.; Vasir, B.; Uhl, L.; Munshi, N.C.; Katz, T.; Dey, B.R.; Somaiya, P.; Mills, H.; Campigotto, F.; et al. Vaccination with dendritic cell/tumor fusions following autologous stem cell transplant induces immunologic and clinical responses in multiple myeloma patients. Clin. Cancer Res. 2013, 19, 3640–3648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. van de Loosdrecht, A.A.; van Wetering, S.; Santegoets, S.; Singh, S.K.; Eeltink, C.M.; den Hartog, Y.; Koppes, M.; Kaspers, J.; Ossenkoppele, G.J.; Kruisbeek, A.M.; et al. A novel allogeneic off-the-shelf dendritic cell vaccine for post-remission treatment of elderly patients with acute myeloid leukemia. Cancer Immunol. Immunother. 2018, 67, 1505–1518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Neelapu, S.S.; Locke, F.L.; Bartlett, N.L.; Lekakis, L.J.; Miklos, D.B.; Jacobson, C.A.; Braunschweig, I.; Oluwole, O.O.; Siddiqi, T.; Lin, Y.; et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 2531–2544. [Google Scholar] [CrossRef] [PubMed]
  96. Yan, Z.; Cao, J.; Cheng, H.; Qiao, J.; Zhang, H.; Wang, Y.; Shi, M.; Lan, J.; Fei, X.; Jin, L.; et al. A combination of humanised anti-CD19 and anti-BCMA CAR T cells in patients with relapsed or refractory multiple myeloma: A single-arm, phase 2 trial. Lancet Haematol. 2019, 6, e521–e529. [Google Scholar] [CrossRef]
  97. Lavergne, M.; Janus-Bell, E.; Schaff, M.; Gachet, C.; Mangin, P.H. Platelet Integrins in Tumor Metastasis: Do They Represent a Therapeutic Target? Cancers (Basel) 2017, 9, 133. [Google Scholar] [CrossRef]
  98. Zhang, Y.; Liu, G.; Wei, J.; Nie, G. Platelet membrane-based and tumor-associated platelettargeted drug delivery systems for cancer therapy. Front. Med. 2018, 12, 667–677. [Google Scholar] [CrossRef]
  99. Xu, P.; Zuo, H.; Chen, B.; Wang, R.; Ahmed, A.; Hu, Y.; Ouyang, J. Corrigendum: Doxorubicin-loaded platelets as a smart drug delivery system: An improved therapy for lymphoma. Sci. Rep. 2017, 7, 44974. [Google Scholar] [CrossRef] [Green Version]
  100. Grozovsky, R.; Giannini, S.; Falet, H.; Hoffmeister, K.M. Regulating billions of blood platelets: Glycans and beyond. Blood 2015, 126, 1877–1884. [Google Scholar] [CrossRef] [Green Version]
  101. Janowska-Wieczorek, A.; Wysoczynski, M.; Kijowski, J.; Marquez-Curtis, L.; Machalinski, B.; Ratajczak, J.; Ratajczak, M.Z. Microvesicles derived from activated platelets induce metastasis and angiogenesis in lung cancer. Int. J. Cancer 2005, 113, 752–760. [Google Scholar] [CrossRef]
  102. Gasic, G.J.; Gasic, T.B.; Galanti, N.; Johnson, T.; Murphy, S. Platelet-tumor-cell interactions in mice. The role of platelets in the spread of malignant disease. Int. J. Cancer 1973, 11, 704–718. [Google Scholar] [CrossRef]
  103. Cho, M.S.; Bottsford-Miller, J.; Vasquez, H.G.; Stone, R.; Zand, B.; Kroll, M.H.; Sood, A.K.; Afshar-Kharghan, V. Platelets increase the proliferation of ovarian cancer cells. Blood 2012, 120, 4869–4872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Radziwon-Balicka, A.; Medina, C.; O’Driscoll, L.; Treumann, A.; Bazou, D.; Inkielewicz-Stepniak, I.; Radomski, A.; Jow, H.; Radomski, M.W. Platelets increase survival of adenocarcinoma cells challenged with anticancer drugs: Mechanisms and implications for chemoresistance. Br. J. Pharmacol. 2012, 167, 787–804. [Google Scholar] [CrossRef] [PubMed]
  105. Tran, C.; Damaser, M.S. Stem cells as drug delivery methods: Application of stem cell secretome for regeneration. Adv. Drug. Deliv. Rev. 2015, 82–83, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Kavari, S.L.; Shah, K. Engineered stem cells targeting multiple cell surface receptors in tumors. Stem Cells 2020, 38, 34–44. [Google Scholar] [CrossRef] [Green Version]
  107. van Rood, J.J.; Scaradavou, A.; Stevens, C.E. Indirect evidence that maternal microchimerism in cord blood mediates a graft-versus-leukemia effect in cord blood transplantation. Proc. Natl. Acad. Sci. USA 2012, 109, 2509–2514. [Google Scholar] [CrossRef] [Green Version]
  108. Pessina, A.; Cocce, V.; Pascucci, L.; Bonomi, A.; Cavicchini, L.; Sisto, F.; Ferrari, M.; Ciusani, E.; Crovace, A.; Falchetti, M.L.; et al. Mesenchymal stromal cells primed with Paclitaxel attract and kill leukaemia cells, inhibit angiogenesis and improve survival of leukaemia-bearing mice. Br. J. Haematol. 2013, 160, 766–778. [Google Scholar] [CrossRef]
  109. Ciavarella, S.; Grisendi, G.; Dominici, M.; Tucci, M.; Brunetti, O.; Dammacco, F.; Silvestris, F. In vitro anti-myeloma activity of TRAIL-expressing adipose-derived mesenchymal stem cells. Br. J. Haematol. 2012, 157, 586–598. [Google Scholar] [CrossRef]
  110. Bock, A.M.; Knorr, D.; Kaufman, D.S. Development, expansion, and in vivo monitoring of human NK cells from human embryonic stem cells (hESCs) and and induced pluripotent stem cells (iPSCs). J. Vis. Exp. 2013, e50337. [Google Scholar] [CrossRef] [Green Version]
  111. Themeli, M.; Kloss, C.C.; Ciriello, G.; Fedorov, V.D.; Perna, F.; Gonen, M.; Sadelain, M. Generation of tumor-targeted human T lymphocytes from induced pluripotent stem cells for cancer therapy. Nat. Biotechnol. 2013, 31, 928–933. [Google Scholar] [CrossRef]
  112. Thomas, E.D.; Lochte, H.L., Jr.; Lu, W.C.; Ferrebee, J.W. Intravenous infusion of bone marrow in patients receiving radiation and chemotherapy. N. Engl. J. Med. 1957, 257, 491–496. [Google Scholar] [CrossRef]
  113. Singh, A.K.; McGuirk, J.P. Allogeneic Stem Cell Transplantation: A Historical and Scientific Overview. Cancer Res. 2016, 76, 6445–6451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Bair, S.M.; Brandstadter, J.D.; Ayers, E.C.; Stadtmauer, E.A. Hematopoietic stem cell transplantation for blood cancers in the era of precision medicine and immunotherapy. Cancer 2020, 126, 1837–1855. [Google Scholar] [CrossRef] [PubMed]
  115. Panch, S.R.; Szymanski, J.; Savani, B.N.; Stroncek, D.F. Sources of Hematopoietic Stem and Progenitor Cells and Methods to Optimize Yields for Clinical Cell Therapy. Biol. Blood Marrow Transplant. 2017, 23, 1241–1249. [Google Scholar] [CrossRef]
  116. Khaddour, K.; Mewawalla, P. Hematopoietic Stem Cell Transplantation. In StatPearls StatPearls [Internet], Treasure Island (FL); StatPearls Publishing: Petersburg, FL, USA, 2020. [Google Scholar]
  117. Gschweng, E.; De Oliveira, S.; Kohn, D.B. Hematopoietic stem cells for cancer immunotherapy. Immunol. Rev. 2014, 257, 237–249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Puig-Saus, C.; Parisi, G.; Garcia-Diaz, A.; Krystofinski, P.E.; Sandoval, S.; Zhang, R.; Champhekar, A.S.; McCabe, J.; Cheung-Lau, G.C.; Truong, N.A.; et al. IND-Enabling Studies for a Clinical Trial to Genetically Program a Persistent Cancer-Targeted Immune System. Clin. Cancer Res. 2019, 25, 1000–1011. [Google Scholar] [CrossRef] [Green Version]
  119. Rapoport, A.P.; Stadtmauer, E.A.; Binder-Scholl, G.K.; Goloubeva, O.; Vogl, D.T.; Lacey, S.F.; Badros, A.Z.; Garfall, A.; Weiss, B.; Finklestein, J.; et al. NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat. Med. 2015, 21, 914–921. [Google Scholar] [CrossRef] [Green Version]
  120. Gimble, J.M.; Guilak, F.; Nuttall, M.E.; Sathishkumar, S.; Vidal, M.; Bunnell, B.A. In vitro Differentiation Potential of Mesenchymal Stem Cells. Transfus. Med. Hemother. 2008, 35, 228–238. [Google Scholar] [CrossRef] [Green Version]
  121. Luo, L.; Li, T.S. Mini review: Recent advances in the cell-based therapies for cardiac regeneration. Curr. Stem Cell Res. Ther. 2020. [Google Scholar] [CrossRef]
  122. Rozier, P.; Maria, A.; Goulabchand, R.; Jorgensen, C.; Guilpain, P.; Noel, D. Mesenchymal Stem Cells in Systemic Sclerosis: Allogenic or Autologous Approaches for Therapeutic Use? Front. Immunol. 2018, 9, 2938. [Google Scholar] [CrossRef] [Green Version]
  123. Conrad, S.; Weber, K.; Walliser, U.; Geburek, F.; Skutella, T. Stem Cell Therapy for Tendon Regeneration: Current Status and Future Directions. Adv. Exp. Med. Biol. 2019, 1084, 61–93. [Google Scholar] [CrossRef]
  124. Bago, J.R.; Soler-Botija, C.; Casani, L.; Aguilar, E.; Alieva, M.; Rubio, N.; Bayes-Genis, A.; Blanco, J. Bioluminescence imaging of cardiomyogenic and vascular differentiation of cardiac and subcutaneous adipose tissue-derived progenitor cells in fibrin patches in a myocardium infarct model. Int. J. Cardiol. 2013, 169, 288–295. [Google Scholar] [CrossRef] [PubMed]
  125. Chulpanova, D.S.; Kitaeva, K.V.; Tazetdinova, L.G.; James, V.; Rizvanov, A.A.; Solovyeva, V.V. Application of Mesenchymal Stem Cells for Therapeutic Agent Delivery in Anti-tumor Treatment. Front. Pharmacol. 2018, 9, 259. [Google Scholar] [CrossRef] [PubMed]
  126. Bago, J.R.; Pegna, G.J.; Okolie, O.; Hingtgen, S.D. Fibrin matrices enhance the transplant and efficacy of cytotoxic stem cell therapy for post-surgical cancer. Biomaterials 2016, 84, 42–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Sun, Z.; Wang, S.; Zhao, R.C. The roles of mesenchymal stem cells in tumor inflammatory microenvironment. J. Hematol. Oncol. 2014, 7, 14. [Google Scholar] [CrossRef] [Green Version]
  128. Xu, S.; Menu, E.; De Becker, A.; Van Camp, B.; Vanderkerken, K.; Van Riet, I. Bone marrow-derived mesenchymal stromal cells are attracted by multiple myeloma cell-produced chemokine CCL25 and favor myeloma cell growth in vitro and in vivo. Stem Cells 2012, 30, 266–279. [Google Scholar] [CrossRef]
  129. Klopp, A.H.; Spaeth, E.L.; Dembinski, J.L.; Woodward, W.A.; Munshi, A.; Meyn, R.E.; Cox, J.D.; Andreeff, M.; Marini, F.C. Tumor irradiation increases the recruitment of circulating mesenchymal stem cells into the tumor microenvironment. Cancer Res. 2007, 67, 11687–11695. [Google Scholar] [CrossRef] [Green Version]
  130. Li, X.; Lu, Y.; Huang, W.; Xu, H.; Chen, X.; Geng, Q.; Fan, H.; Tan, Y.; Xue, G.; Jiang, X. In vitro effect of adenovirus-mediated human Gamma Interferon gene transfer into human mesenchymal stem cells for chronic myelogenous leukemia. Hematol. Oncol. 2006, 24, 151–158. [Google Scholar] [CrossRef]
  131. Bonomi, A.; Steimberg, N.; Benetti, A.; Berenzi, A.; Alessandri, G.; Pascucci, L.; Boniotti, J.; Cocce, V.; Sordi, V.; Pessina, A.; et al. Paclitaxel-releasing mesenchymal stromal cells inhibit the growth of multiple myeloma cells in a dynamic 3D culture system. Hematol. Oncol. 2017, 35, 693–702. [Google Scholar] [CrossRef]
  132. Cocce, V.; Farronato, D.; Brini, A.T.; Masia, C.; Gianni, A.B.; Piovani, G.; Sisto, F.; Alessandri, G.; Angiero, F.; Pessina, A. Drug Loaded Gingival Mesenchymal Stromal Cells (GinPa-MSCs) Inhibit In Vitro Proliferation of Oral Squamous Cell Carcinoma. Sci. Rep. 2017, 7, 9376. [Google Scholar] [CrossRef]
  133. Dwyer, R.M.; Khan, S.; Barry, F.P.; O’Brien, T.; Kerin, M.J. Advances in mesenchymal stem cell-mediated gene therapy for cancer. Stem Cell Res. Ther. 2010, 1, 25. [Google Scholar] [CrossRef] [Green Version]
  134. Klingemann, H.; Matzilevich, D.; Marchand, J. Mesenchymal Stem Cells—Sources and Clinical Applications. Transfus. Med. Hemother. 2008, 35, 272–277. [Google Scholar] [CrossRef] [PubMed]
  135. Lin, H.D.; Fong, C.Y.; Biswas, A.; Choolani, M.; Bongso, A. Human Umbilical Cord Wharton’s Jelly Stem Cell Conditioned Medium Induces Tumoricidal Effects on Lymphoma Cells Through Hydrogen Peroxide Mediation. J. Cell. Biochem. 2016, 117, 2045–2055. [Google Scholar] [CrossRef] [PubMed]
  136. Lee, M.W.; Park, Y.J.; Kim, D.S.; Park, H.J.; Jung, H.L.; Lee, J.W.; Sung, K.W.; Koo, H.H.; Yoo, K.H. Human Adipose Tissue Stem Cells Promote the Growth of Acute Lymphoblastic Leukemia Cells in NOD/SCID Mice. Stem Cell Rev. Rep. 2018, 14, 451–460. [Google Scholar] [CrossRef]
  137. Song, N.; Gao, L.; Qiu, H.; Huang, C.; Cheng, H.; Zhou, H.; Lv, S.; Chen, L.; Wang, J. Mouse bone marrow-derived mesenchymal stem cells inhibit leukemia/lymphoma cell proliferation in vitro and in a mouse model of allogeneic bone marrow transplant. Int. J. Mol. Med. 2015, 36, 139–149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Ramasamy, R.; Lam, E.W.; Soeiro, I.; Tisato, V.; Bonnet, D.; Dazzi, F. Mesenchymal stem cells inhibit proliferation and apoptosis of tumor cells: Impact on in vivo tumor growth. Leukemia 2007, 21, 304–310. [Google Scholar] [CrossRef] [PubMed]
  139. Sarmadi, V.H.; Tong, C.K.; Vidyadaran, S.; Abdullah, M.; Seow, H.F.; Ramasamy, R. Mesenchymal stem cells inhibit proliferation of lymphoid origin haematopoietic tumour cells by inducing cell cycle arrest. Med. J. Malaysia 2010, 65, 209–214. [Google Scholar] [PubMed]
  140. Ning, H.; Yang, F.; Jiang, M.; Hu, L.; Feng, K.; Zhang, J.; Yu, Z.; Li, B.; Xu, C.; Li, Y.; 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. [Google Scholar] [CrossRef] [Green Version]
  141. Manabe, A.; Coustan-Smith, E.; Behm, F.G.; Raimondi, S.C.; Campana, D. Bone marrow-derived stromal cells prevent apoptotic cell death in B-lineage acute lymphoblastic leukemia. Blood 1992, 79, 2370–2377. [Google Scholar] [CrossRef] [Green Version]
  142. Nwabo Kamdje, A.H.; Mosna, F.; Bifari, F.; Lisi, V.; Bassi, G.; Malpeli, G.; Ricciardi, M.; Perbellini, O.; Scupoli, M.T.; Pizzolo, G.; et al. Notch-3 and Notch-4 signaling rescue from apoptosis human B-ALL cells in contact with human bone marrow-derived mesenchymal stromal cells. Blood 2011, 118, 380–389. [Google Scholar] [CrossRef]
  143. Di Ianni, M.; Del Papa, B.; De Ioanni, M.; Moretti, L.; Bonifacio, E.; Cecchini, D.; Sportoletti, P.; Falzetti, F.; Tabilio, A. Mesenchymal cells recruit and regulate T regulatory cells. Exp. Hematol. 2008, 36, 309–318. [Google Scholar] [CrossRef]
  144. Sotiropoulou, P.A.; Perez, S.A.; Gritzapis, A.D.; Baxevanis, C.N.; Papamichail, M. Interactions between human mesenchymal stem cells and natural killer cells. Stem Cells 2006, 24, 74–85. [Google Scholar] [CrossRef] [PubMed]
  145. Roorda, B.D.; ter Elst, A.; Kamps, W.A.; de Bont, E.S. Bone marrow-derived cells and tumor growth: Contribution of bone marrow-derived cells to tumor micro-environments with special focus on mesenchymal stem cells. Crit. Rev. Oncol. Hematol. 2009, 69, 187–198. [Google Scholar] [CrossRef] [PubMed]
  146. Xia, B.; Tian, C.; Guo, S.; Zhang, L.; Zhao, D.; Qu, F.; Zhao, W.; Wang, Y.; Wu, X.; Da, W.; et al. c-Myc plays part in drug resistance mediated by bone marrow stromal cells in acute myeloid leukemia. Leuk. Res. 2015, 39, 92–99. [Google Scholar] [CrossRef] [PubMed]
  147. Cai, J.; Wang, J.; Huang, Y.; Wu, H.; Xia, T.; Xiao, J.; Chen, X.; Li, H.; Qiu, Y.; Wang, Y.; et al. ERK/Drp1-dependent mitochondrial fission is involved in the MSC-induced drug resistance of T-cell acute lymphoblastic leukemia cells. Cell Death Dis. 2016, 7, e2459. [Google Scholar] [CrossRef]
  148. Lee, M.W.; Ryu, S.; Kim, D.S.; Lee, J.W.; Sung, K.W.; Koo, H.H.; Yoo, K.H. Mesenchymal stem cells in suppression or progression of hematologic malignancy: Current status and challenges. Leukemia 2019, 33, 597–611. [Google Scholar] [CrossRef] [Green Version]
  149. Barber, C.L.; Iruela-Arispe, M.L. The ever-elusive endothelial progenitor cell: Identities, functions and clinical implications. Pediatr. Res. 2006, 59, 26R–32R. [Google Scholar] [CrossRef]
  150. Chopra, H.; Hung, M.K.; Kwong, D.L.; Zhang, C.F.; Pow, E.H.N. Insights into Endothelial Progenitor Cells: Origin, Classification, Potentials, and Prospects. Stem Cells Int. 2018, 2018, 9847015. [Google Scholar] [CrossRef]
  151. Laurenzana, A.; Margheri, F.; Chilla, A.; Biagioni, A.; Margheri, G.; Calorini, L.; Fibbi, G.; Del Rosso, M. Endothelial Progenitor Cells as Shuttle of Anticancer Agents. Hum. Gene Ther. 2016, 27, 784–791. [Google Scholar] [CrossRef]
  152. Keighron, C.; Lyons, C.J.; Creane, M.; O’Brien, T.; Liew, A. Recent Advances in Endothelial Progenitor Cells Toward Their Use in Clinical Translation. Front. Med. (Lausanne) 2018, 5, 354. [Google Scholar] [CrossRef] [Green Version]
  153. Zhao, X.; Liu, H.Q.; Li, J.; Liu, X.L. Endothelial progenitor cells promote tumor growth and progression by enhancing new vessel formation. Oncol. Lett. 2016, 12, 793–799. [Google Scholar] [CrossRef] [Green Version]
  154. Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Lei, F.; Haque, R.; Weiler, L.; Vrana, K.E.; Song, J. T lineage differentiation from induced pluripotent stem cells. Cell Immunol. 2009, 260, 1–5. [Google Scholar] [CrossRef] [PubMed]
  156. Watarai, H.; Fujii, S.; Yama da, D.; Rybouchkin, A.; Sakata, S.; Nagata, Y.; Iida-Kobayashi, M.; Sekine-Kondo, E.; Shimizu, K.; Shozaki, Y.; et al. Murine induced pluripotent stem cells can be derived from and differentiate into natural killer T cells. J. Clin. Invest. 2010, 120, 2610–2618. [Google Scholar] [CrossRef] [PubMed]
  157. Zhang, L.; Tian, L.; Dai, X.; Yu, H.; Wang, J.; Lei, A.; Zhao, W.; Zhu, Y.; Sun, Z.; Zhang, H.; et al. Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell Functions for Liquid and Solid Tumors. bioRxiv 2020. [Google Scholar] [CrossRef]
  158. Yasuda, S.; Kusakawa, S.; Kuroda, T.; Miura, T.; Tano, K.; Takada, N.; Matsuyama, S.; Matsuyama, A.; Nasu, M.; Umezawa, A.; et al. Tumorigenicity-associated characteristics of human iPS cell lines. PLoS ONE 2018, 13, e0205022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Diagram depicting the different cell-based therapies to treat hematological malignancies with the mode of action in each case. NK: natural killer, iPSCs: induced pluripotent stem cells, HSCs: hematopoietic stem cells, MSCs: mesenchymal stem cells, and EPCs: endothelial progenitor cells.
Figure 1. Diagram depicting the different cell-based therapies to treat hematological malignancies with the mode of action in each case. NK: natural killer, iPSCs: induced pluripotent stem cells, HSCs: hematopoietic stem cells, MSCs: mesenchymal stem cells, and EPCs: endothelial progenitor cells.
Cancers 12 01333 g001
Figure 2. Timeline of cancer cell-based therapy milestones in the past 70 years in blood cancer treatments. ALL: acute lymphoblastic leukemia, BCL: B cell lymphoma, CLL: chronic lymphocytic leukemia, ESCs: embryonic stem cells, NHL: Non-Hodgkin’s lymphoma, MM: multiple myeloma.
Figure 2. Timeline of cancer cell-based therapy milestones in the past 70 years in blood cancer treatments. ALL: acute lymphoblastic leukemia, BCL: B cell lymphoma, CLL: chronic lymphocytic leukemia, ESCs: embryonic stem cells, NHL: Non-Hodgkin’s lymphoma, MM: multiple myeloma.
Cancers 12 01333 g002
Table 1. Selected cell-based clinical trials with immune cells to treat hematological malignancies featuring key aspects.
Table 1. Selected cell-based clinical trials with immune cells to treat hematological malignancies featuring key aspects.
YearCell SourceTargetEngineering MethodNo. of PatientsAge of Patients
(Mean)
DoseOutcomesCytotoxic EffectsAdditional NotesReference
2010Autologous DCsALLWT1 mRNA electroporation1031–83
(61)
4 shots
2 weeks interval
5, 10, or 20 × 106
2CR [92]
2013Autologous DCsCD38+ and CD138+ MMFusion with whole MM cell10035–703–4 shots
2 × 106/kg per shot
78% R
47% VGPR
31% CR
57 % two-year progression-free survival[93]
2018Allogenic DCsAMLDifferentiated from AML cell line1257–74
(68)
4 shots
2 weeks interval
10, 25 or 50 × 106
5 CR
4 PD
Post-remission treatment[94]
2013Autologous T cellsCD19+ ALL2nd gen CAR (+CD28)1623–74
(50)
One shot
1.5–3 × 106/kg
14 OR
10 CR
13 MRD-
2 CRS4
2 CRS3
[17]
2014Autologous T cellsCD19+ B-ALL and T-ALL2nd gen CAR
(+4-1BB)
305–60One shot
0.76–20 × 106/kg
27 CR
19 SCR
Severe in 8 patientsFDA approved for ALL and DLBCL
Produced by Novartis under the commercial name Kymriah
[25]
2016Autologous T cellsCD20+ B-cell NHL2nd gen CAR (+CD137ζ)1125–70
(59)
One shot
0.5–1.5 × 106/kg
6 CR
3 PR
2 SD
2 CRS3 [28]
2017Autologous T cellsCD19+ B-cell lymphomas2nd gen CAR (+CD28)11123–76One shot
2 × 106/kg
82% OR
54% CR
95% CRS3+FDA-approved for B-cell lymphoma Produced by Gilead under the commercial name Yescarta[95]
2017Autologous T cellsCD22+ lymphoma and leukemia2nd gen CAR217–30
(19)
One shot
0.3–3 × 106/kg
12 CR
9 MRD-
1 CRS4
1 CRS3
17 patients were resistant to CAR anti-CD19 in the past[29]
2019Autologous T cellsCD19+ and BCMA+ MM2nd gen CAR2149–61
(58)
One shot
1 × 106 + 1 × 106/kg
12 CR
8 PR
1 SD
17 MRD-
1CRS3
2 NT
[96]
2018NK92
cell line
CD33+ AML3rd gen CAR
(+CD28 and 4-1BB)
314–49Three shots
2 days interval
300, 600, 1000 × 106
No response2CRS1One patient died of GvHD after chemotherapy and donor lymphocyte infusion[65]
2020Allogenic NK cells from cord bloodCD19+ lymphoma and leukemia2nd gen CAR
(+CD28)
1123–66 (52)One shot
0.1, 1 or 10 × 106/kg
8 OR
7 CR
No GvHD
No CRS
No GvHD despite some HLA mismatch; Toxic events related to lymphodepletion[66]
Abbreviations: OR, objective response. CR, complete response. VGPR, very good partial remission. PR, partial response. SD, stable disease. PD, progressive disease. MRD, minimal residual disease negative. SCR, sustained complete response. CRS, cytokine release syndrome (+grade). NT, neurotoxicity. DCs, dendritic cells. MM, multiple myeloma. CARs, chimeric antigen receptors. AML, acute myeloid leukemia. BCMA, B-cell maturation antigen. NK, natural killer. GvHD, graft-versus-host-disease. HLA, human leukocyte antigen. NHL, non-Hodgkin’s lymphoma. ALL, acute lymphoblastic leukemia. DLBCL, diffuse large B-cell lymphoma.
Table 2. Selected cell-based clinical trials and preclinical studies with stem cells to treat hematological malignancies, featuring key aspects. iPSCs: induced pluripotent stem cells, HSCs: hematopoietic stem cells, MSCs: mesenchymal stem cells, GvT: graft versus tumor effect, TRAIL: TNF-related apoptosis-inducing ligand, and NKT: natural killer T cells.
Table 2. Selected cell-based clinical trials and preclinical studies with stem cells to treat hematological malignancies, featuring key aspects. iPSCs: induced pluripotent stem cells, HSCs: hematopoietic stem cells, MSCs: mesenchymal stem cells, GvT: graft versus tumor effect, TRAIL: TNF-related apoptosis-inducing ligand, and NKT: natural killer T cells.
Cell SourceTargetMode of ActionOutcomesReference
HSCs from cord bloodPatients with AML or ALLGvTDecrease in leukemic relapse[107]
MSCs from bone marrowMouse models with leukemia cell lines MOLT-4 and L1210Loaded with paclitaxelAntileukemia activity in vitro and in the mouse model[108]
MSCs from adipose tissueMM cell lines in vitro: RPMI-8226 U-266, MMCAR-1, LIG-1, and MCC-2 Engineered with the expression of TRAILAntimyeloma activity in vitro[109]
iPSCs from murine embryonic fibroblastsMurine T-cell lymphomaDifferentiation to NKT cellsTumor growth suppression in the mouse model[110]
iPSCs from murine embryonic fibroblastsRaji human Burkitt lymphoma cell line Differentiation to T cells expressing CD19 CARTumor growth inhibition in the mouse model [111]

Share and Cite

MDPI and ACS Style

Motais, B.; Charvátová, S.; Hrdinka, M.; Šimíček, M.; Jelínek, T.; Ševčíková, T.; Kořístek, Z.; Hájek, R.; Bagó, J.R. A Bird’s-Eye View of Cell Sources for Cell-Based Therapies in Blood Cancers. Cancers 2020, 12, 1333. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12051333

AMA Style

Motais B, Charvátová S, Hrdinka M, Šimíček M, Jelínek T, Ševčíková T, Kořístek Z, Hájek R, Bagó JR. A Bird’s-Eye View of Cell Sources for Cell-Based Therapies in Blood Cancers. Cancers. 2020; 12(5):1333. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12051333

Chicago/Turabian Style

Motais, Benjamin, Sandra Charvátová, Matouš Hrdinka, Michal Šimíček, Tomáš Jelínek, Tereza Ševčíková, Zdeněk Kořístek, Roman Hájek, and Juli R. Bagó. 2020. "A Bird’s-Eye View of Cell Sources for Cell-Based Therapies in Blood Cancers" Cancers 12, no. 5: 1333. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12051333

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop