Get 20M+ Full-Text Papers For Less Than $1.50/day. Start a 14-Day Trial for You or Your Team.

Learn More →

The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing Piece of the Puzzle for the Efficient Targeting of these Cells with Immunotherapy

The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The... Cancer Stem Cells/Cancer Initiating Cells (CSCs/CICs) is a rare sub-population within a tumor that is responsible for tumor formation, progression and resistance to therapies. The interaction between CSCs/CICs and tumor microenvironment (TME) can sustain “stemness” properties and promote their survival and plasticity. This cross-talk is also pivotal in regulating and modu- lating CSC/CIC properties. This review will provide an overview of the mechanisms underlying the mutual interaction between CSCs/CICs and TME. Particular focus will be dedicated to the immunological profile of CSCs/CICs and its role in orchestrating cancer immunosurveillance. Moreover, the available immunotherapy strategies that can target CSCs/CICs and of their possible implementation will be discussed. Overall, the dissection of the mechanisms regulating the CSC/CIC-TME interaction is warranted to understand the plasticity and immunoregulatory properties of stem-like tumor cells and to achieve complete eradications of tumors through the optimization of immunotherapy. . . . Keywords Cancer stem cells/Cancer initiating cells Immunosurveillance Adaptive immune responses Innate immune . . responses Tumor microenvironment Immunotherapy Abbreviations IL-4 Interleukin 4 ALDH Aldehyde dehydrogenase IL-10 Interleukin 10 APC Antigen presenting cells IL-13 Interleukin 13 APM Antigen processing machinery IL-13α2 α2 chain of IL-13 receptor CAR Chimeric antigen receptor mAb Monoclonal antibody; CIC Cancer initiating cell MDSC Myeloid derived suppressor cell CRC Colorectal cancer NSCLC Non-small cell lung cancer CT Cancer testis PD-1 Programmed death 1 CTLA-4 Cytotoxic lymphocyte antigen-4 PD-L1 Programmed death ligand 1 CSPG4 Chondroitin sulphate protidoglycan 4 RCC Renal cell carcinoma HLA Human leukocyte antigen STAT3 Signal transducer and activator of transcription 3 IDO Indoleamine 2,3-dioxygenase TGFB Transforming growth factor beta GBM Glioblastoma multiforme TAA Tumor associated antigen GDF-15 Growth differentiation factor-15 Treg T regulatory cell. IFN Interferon Introduction Shilpa Ravindran and Saad Rasool contributed equally to this work. Tumors are composed by heterogeneous cellular components * Cristina Maccalli including a rare subpopulation bearing “stemness properties” cmaccalli@sidra.org and being responsible of tumor initiation and progression. These cells have been denominated cancer stem cells Research Department, Sidra Medicine, Al Luqta Street, PO Box 26999, Doha, Qatar (CSCs) or cancer initiating cells (CICs) [1–6]. CSCs/CICs 134 Ravindran S. et al. share several characteristics with normal stem cells, such as detecting the presence of these cells within tumor lesions the ability to self-renew and to give rise to differentiated prog- though probing for CSC/CIC- associated markers has not pro- eny and the resistance to DNA damage-induced cell death [3, vided conclusive results. The xenotransplantation in immune 5–12]. CSCs/CICs, through the cycling from proliferation to deficient mice represents a useful tool to demonstrate in vivo quiescence, expression of ABC drug pumps, high levels of the tumorigenic properties CSCs/CICs [35]. Xenograft anti-apoptotic proteins and resistance to DNA damage, are models have contributed to prove the existence within tumor resistant to radiation and chemotherapy and play an important lesions of cell population endowed with stemness properties role in disease relapse and tumor progression [13, 14]. that upon serial transplantation could propagate both tumori- CSCs/CICs have been isolated from both hematological genic CSCs/CICs and malignant cells with differentiated phe- and solid tumors and they represent a rare subpopulation, notype without tumorigenic properties [18]. These subpopu- comprising 0.01–10% of cells within the tumor [15]. They lations can be identified only through transplantation in im- can be ex vivo identified based on their “stem cell-like” char- mune deficient mice [4, 36–38]. acteristics and the expression of certain cell surface and func- Nevertheless, the available CSC/CIC-associated tional markers [16]. The identification of CSCs/CICs was first markers are dependent on spatial and temporal features, reported in leukemia, showing a hierarchical organization of with their modulation occurring in relation to their inocu- tumor cells [17]. The leukemic cells were able to be engrafted lation in immunodeficient mice, proving the high level of + − upon transplantation of CD34 CD38 cells into severe com- plasticity of these cells and that none of the available bined immune-deficient (SCID) mice, which eventually led to markers can be exploited to monitor the in vivo fate of the identification of the hierarchical organization of tumors these cells [1, 39, 40] CSCs/CICs, similarly to normal stem with few cells endowed with stemness and tumorigenic prop- cells, require a “niche” to allow the survival of these cells erties [17]. Since then, a variety of studies highlighted the and their cycling from quiescence to proliferation and to existence of “stem-like” cancer cells in solid tumors with dif- maintain stemness and multipotency [41–43]. The “niche” ferent histological origins [5, 18–23]. Multiple molecules is represented by the tumor microenvironment (TME), (e.g., ALDH-1, CD133, CD44, CD24, CBX3, ABCA5, which is composed of multicellular and dynamic compart- LGR5, etc) have been identified as CSC/CIC-associated ments that include fibroblasts, endothelial, stromal, mesen- markers with differential expression depending on the tissues chymal and immune cells [41]. The interaction of TME of derivation, highlighting the high grade of heterogeneity of with stem-like cancer cells can regulate the fate of these these cells [16](Table 1). Most of these molecules are over- cells through modulating the proliferation, differentiation, expressed by CSCs/CICs but are also shared with either dif- immunological properties and resistance to therapies ferentiated tumor cells or normal stem cells [4, 34]. As a result, [44–50]. Table 1 Markers expressed by Marker Tumor type Recognition by T Reference CSCs/CICs isolated from solid cells tumors and their role as TAAs ALDH1 CRC; breast and gastric cancer; melanoma √ [24] CD133 GBM, pancreas, lung, ovarian, prostate, and gastric √ [25–27] cancer CD44 CRC, head and neck cancer EpCAM CRC, Retinoblastoma [28] EpCAM CD44 Pancreatic cancer CD24 CD24 CRC √ [29, 30] SOX2 GBM √ [31] CBX3 Ostocarcinoma LGR5 CRC ABCB5 Melanoma CD90 Liver cancer HSP DNAJB8 RCC √ [32, 33] CD166 CRC; NSCLC a b Markers commonly identified as associated with CSCs/CICs; the role of these antigens in eliciting T cell- mediated immune responses against CSCs/CICs CRC: colorectal cancer; GBM: glioblastoma multiforme; NSCLC: non-small cell lung cancer; RCC: renal cell carcinoma The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing... 135 The high grade of heterogeneity and plasticity of and of the “niche”, preventing the constant monitoring of CSCs/CICs can depend on their tissue of derivation and, plasticity and heterogeneity of these cells (Figs. 1, 2). importantly, on their cross-talk with TME [4, 16, 51–53]. Therefore, the combination of deep genomic, molecular Limiting the isolation and the functional characterization and functional profiling of CSCs/CICs could represent a rele- of CSCs/CICs to the usage of phenotypic markers is un- vant method to achieve a comprehensive functional character- satisfactory and do not consider the possibility that ization of these cells and, possibly, of their role in tumor out- “stemness” function of tumor cells can be reversible, as come [56]. shown by Quintana et al. for melanoma [1, 39]. Moreover, CSCs/CICs have been identified as the tumor compo- xenotransplantation of these cells in immune deficient nents responsible for resistance to standard therapy, as mice is lacking the important variable of the TME and well as immunotherapy [10, 57–60]. Although clinical its role in affecting the fate of CSCs/CICs [1]. responses in cancer patients are observed following Therefore, the lack of standardized methods to isolate treatments, these cells can remain in the minimal resid- CSCs/CICs and of in vivo models allowing to monitor ual disease and upon changes in the environment they the cross-talk of these cells with TME can lead to the high can exit the quiescence status and give rise to novel extent of variability in assessing the functional properties malignant lesion(s) or even initiate the metastatic colo- of thesecells andinpreventingtoaccuratelydetermine nization [8, 61–63]. their fate and role in the tissue of origins and in the clin- The extensive molecular and immunological characterization ical outcome of cancer patients [54, 55]. The tool of of CSCs/CICs is warranted in order to understand the mecha- sphere forming assay to propagate in vitro CSCs/CICs is nisms regulating their plasticity, quiescence, interaction with the too simplified, lacking the important component of TME TME and resistance to therapies and to immune responses. Fig. 1 Differential immunogenic profile by CSCs/CICs vs. bulk tumor by CSCs/CICs. Neoantigens, generated by somatic mutation bearing tu- cells. CSCs/CICs can express defective levels of HLA molecules and mor cells are equally expressed by both CSCs/CICs and differentiated APM components leading to low immunogenicity and escape from im- tumor cells. The latest TAAs represent highly immunogenic target mole- mune responses. In the presence of efficient expression of ligands of NK- cules, since they are not expressed by normal cells. APM: antigen pro- associated activatory receptors, these cells can become susceptible to NK cessing machinery; CSCs/CICs: cancer stem cells/cancer initiating cells; cell recognition. Moreover, TAAs can be expressed at suboptimal levels NK: natural killer cells 136 Ravindran S. et al. Fig. 2 Immunotherapy strategies to target CSCs/CICs. An overview of multiple immunotherapy approaches or with standard therapies warrant immunotherapy approaches including adoptive cell therapy with either 1. further investigation to assess the efficacy in increasing the immunoge- TCR or CAR engineered T lymphocytes; 2. Immune check point nicity of CSCs/CICs and to implement the targeting of these cells by blockade with mAbs; 3. Cancer vaccination with TAAs expressed by immune responses. CSCs/CICs: cancer stem cells/cancer initiating cells; both CSCs/CICs and differentiated tumor cells; 4. Innate immune re- TAA: tumor associated antigen sponse or 5. γδ T cell recognition of tumor cells. Combination of either Immunological Profile of CSCs from CRC and endowed with stemness properties, showed detectable level of HLA class I molecules as well susceptibil- HLA Molecules and APM Components ity to antigen-specific cytotoxic T lymphocytes (CTLs) [71], however this study has been performed using long-term The expression of HLA class I and class II molecules in vitro established cell lines, that could have lost phenotypic and APM has been investigated in CSCs/CICs isolated from properties of primary CSCs/CICs. Indeed, stem-like cells iso- colorectal cancer (CRC) and glioblastoma multiforme (GBM) lated by sphere-forming assay displayed aberrant expression showing an overall aberrant expression of these molecules, of HLA class I and APM components [16, 65]. Contradictory with, in some cases, failure in their modulation by the pre- results were obtained also in glioblastoma multiforme (GBM); treatment with IFNs (both alpha and gamma) or DNA CSCs/CICs isolated as sphere forming cells from this tumor demethylating agent (5-Aza CdR) [64, 65]. This impairment have been shown to exhibit the expression of HLA class I in antigen processing and presentation by CSCs/CICs lead, molecules [72] while, when applying these analyses to prima- upon co-culture of these cells with autologous T cells, to a ry GBM-derived sphere forming cells, defective expression of preferential selection and differentiation of TH2 type T cells HLA class I and APM molecules was detected [64]. Stem-like and failure in eliciting effector functions [64, 65]. The subop- cells expressing ABCB5 and isolated from melanoma were timal expression of HLA class I molecules and APM compo- found to express suboptimal levels of HLA class I molecules nents was also reported in CSCs/CICs isolated from different while they were positive for HLA class II (45). APM compo- type of solid tumors [64–69]. These peculiar observations nents (e.g., LMP2, LMP7 MECL-1, TAP1 and TAP2) detect- suggested that the defective expression of HLA molecules ed through mRNA analyses were found to be expressed in could represent a tool for the identification of CSCs/CICs tumor sphere-models from different solid malignancies, [70]. On the contrary, the side population (SP) cells derived representing a tool for in vitro enrichment of stem-like cells The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing... 137 and for the investigation of micro-metastasis [73]. However, They are categorized into three groups; (i) the HLA class I and class II molecules were down-modulated in overexpressed/self-antigens that are expressed at high these cells as compared to differentiated tumor cells, also fol- levels by tumor cells and detectable, although at lower lowing their pre-treatment in vitro with IFN-γ, highlighting an levels, on normal tissues (e.g., MART-1/Melan-A, impairment of antigen presentation by these cells [64]. It hTERT, EGFR, survivin). (ii) Cancer testis (CT) antigens needs to be considered that this study did not analyze the that are detectable on tumor cells and not on normal cells, expression at protein levels of APM molecules, therefore except for testis and trophoblast (e.g., NY-ESO1, MAGE post-transcriptional mechanism could affect their expression. A3-A4, PRAME, CT83, SSX2). (iii) Neoantigens or mu- Moreover, long term in vitro established cell lines were used tated antigens derived by non-synonymous mutations in to isolate tumor cell spheres while in other studies reporting cancer cells (e.g., MUM-1, CDK4, ME1, ACTN4, HLA- defective expression of APM components, primary CSCs/ A2) [82]. The neoantigens are higher immunogenic com- CICs have been investigated. These examples highlight that pared to differentiation/self TAAs since are tumor specific an overall suboptimal immunogenic potency by CSCs/CICs and do not induce tolerogenic mechanisms in immune resulting in low or impaired susceptibility to T cell mediated cells [83–85]. Neoantigens have been shown to drive im- immune responses (Fig. 1). This represents a mechanisms of mune responses and to mediate efficient T cell recognition evasion by immune responses that is shared with normal stem of tumor cells, leading to cancer eradication in patients cells, that could represent a typical feature of cells with treated with either mutanome based vaccines or adoptive stemness properties [74]. Importantly, the failure in the ex- cell therapy (ACT) with tumor infiltrating lymphocytes pression of HLA molecules by tumor cells was found as one [84–86]. Notably, CSCs/CICs bearing a somatic mutation of the mechanisms of failure of the clinical activity of immune in the CRC-associated “driver” gene SMAD4, could elicit checkpoint blockade agents in cancer patients [75, 76], indi- antigen-specific T cell responses directed to both stemness cating that either CSCs/CICs can display immune evasion and differentiated components of tumor [87]. mechanisms shared by differentiated tumor cells or that in- A transcriptome analysis of the SP cells and main popu- deed the suboptimal expression of HLA molecules of these lation (MP) derived from CRC, breast and lung cancer re- cells and their resistance to T cell recognition can protect these vealed a preferential expression of 18 CT antigens cells from immunotherapy interventions, leading to tumor re- (MAGEA2, MAGEA3, MAGEA4, MAGEA6, MAGEA12, currence or progression. However, the lack of standardization MAGEB2, GAGE1, GAGE8, SPANXA1, SPANXB1, in methods for both, the isolation of cells with stemness prop- SPANXC, XAGE2, SPA17, BORIS, PLU-1, SGY-1, erties and to analyze HLA and APM molecules, represents a TEX15 and CT45A1) in CSCs/CICs [32]. The TAA limitation in providing conclusive results. Nevertheless, de- DNAJB8, that is a member of the heat shock protein tailed analysis to identify the molecular mechanisms that lead (HSP) 40 family, was found to be preferentially expressed to aberrant expression of HLA molecules and APM compo- in renal cell carcinoma (RCC); interestingly this protein nents are warranted. played an important role in the maintenance of The suboptimal expression of HLA class I molecules if CSCs/CICs. DNAJB8-specific immune responses could be associated with detectable NKG2D ligands, can drive the in- detected in a mice model study of DNA vaccination for creased susceptibility of CSCs/CICs to Natural Killer (NK) RCC, rendering this molecule appealing for targeting cells. This phenomenon has been observed in CSCs/CICs CSCs/CICs by the immune system [32, 33](Table 1). from glioma, melanoma, and CRC [68, 77–79]. However, Recently a new antigen, Ankyrin repeat and SOCS box pro- down-modulation of NKG2D ligands on CSCs/CICs has been tein 4 (ASB4), was described as target molecule of CTLs documented, e.g., in GBM patients [64]suggesting thatthe recognizing CSCs/CICs and not the differentiated cellular expression of low levels of NK cell activating ligands can components of the tumor [88]. Suboptimal expression of result in the impairment of anti-CSCs/CICs innate immune TAAs (MART-1, ML-IAP, NY-ESO-1, and MAGE-A) was responses (Fig. 1). The expression profile of molecules acti- reported in melanoma-derived CSCs/CICs (Fig. 1)[89]. vating innate immune responses on CSCs/CICs can be affect- Similar results were obtained in CSCs/CICs isolated from ed by their crosstalk with TME, and thus, by their plasticity GBM and CRC (Fig. 1 and Table 1)[64, 65]. On the other that can influence the fate in vivo of these cells. hand, CD133 CSCs/CICs isolated from melanoma were shown to express either NY-ESO-1 or DEAD/H (Asp-Glu- Tumor Associated Antigens and Adaptive Immune Ala-Asp/His) box polypeptide 3, X-linked (DDX3X) Responses against CSCs/CICs representing target of tumor-specific T cells [90, 91]. Other studies have described the isolation of T lymphocytes rec- Tumor associated antigens (TAAs) can be recognized by ognizing TAAs expressed by CSCs/CICs such as IL-13Rα2, T lymphocytes when exposed on the surface of tumor SOX2 and CD133 in GBM, CEP55 and COA-1 in CRC and cells in the form of peptide/HLA complexes [80, 81]. EpCAM in retinoblastoma (Table 1)[28, 65, 71, 92]. 138 Ravindran S. et al. Innate Immune Responses and their Relationship characterized by elevated pro-inflammatory cytokines, such with CSCs/CICs as IL-12, IL-1β, IL-6, and tumor necrosis factor α (TNF-α), increased expression of HLA class II molecules, generation of Natural killer (NK) cells are the first line of defense against reactive oxygen and nitrogen intermediates and ability to in- cancer development and metastasis. NK cells have been de- duce TH1-type T cell responses [106]. 2. In the presence IL-4, scribed to efficiently recognize and kill in vitro CSCs/CICs iso- IL-10, and IL-13, macrophages can polarize towards M2 phe- lated from CRC, melanoma and glioblastoma [68, 78, 93, 94]. notype. These cells express scavenging, mannose and galac- The efficiency of NK cell-mediated lysis of CSCs/CICs was tose receptors, IL-10, vascular endothelial growth factor dependent on the expression of NCR ligands (NKp30 and (VEGF), matrix metalloproteinases (MMPs) and activation NKp44), NKG2D ligands and when suboptimal or negative ex- of the arginase pathway, leading to pro-tumoral effects pression of HLA class I molecules were found on the surface of [105–107, 109]. The cross-talk between CSCs/CICs and CSCs/CICs (Fig 1)[68, 78, 93–95]. Tallerico et al. found that TAM is orchestrated by STAT3 signaling [102, 103]. Upon CSC/CIC but not their differentiated counterpart of CRC is sus- iper-modulation of STAT3 in TAM, they can promote ceptible to NK cells [77]. Similar results have been reported in stemness, survival and proliferation in cancer cells while the GBM and melanoma, highlighting that the amount of ligands of latest cells can induce the immunosuppressive properties of activatory NK receptors on CSCs/CICs was determinant for ef- TAM, leading to the impairment of cancer immune- ficient innate immune responses [68, 77, 78]. In patients with surveillance [110]. acute myeloid leukemia (AML), the suboptimal expression of Myeloid derived suppressor cells (MDSCs) are immune cells NKG2D ligands has been described as a mecahnisms of escape endowed with suppressive functions that can inhibit the effector by tumor cells from NK cell recognition [96], confirming that functions of immune responses [111]. The frequency of these these molecules can affect the susceptibility of cancer cells to cells either at tumor site or in the circulation has been described innate responses. The observations that NKG2D ligands could as a prognostic factor for patients’ survival as well as of respon- represent as biomarkers for prediction of clinical responses to siveness to immunotherapy [112]. Interestingly, STAT3 can lead immune checkpoint blockade in melanoma highlight that the the differentiation of monocytes towards MDSCs in pancreatic pattern of NKG2D ligands expression by tumor cells can affect tumors [113] regulating also the development of CSCs/CICs the type and efficiency of elicited anti-tumor immune responses [113]. The secretion of pro-inflammatory cytokines and [97]. Therefore, the levels and pattern of expression of NKG2D chemokines by tumor cells can induce the differentiation and ligands by tumor cells, including CSCs/CICs could be a predic- recruitment of immunosuppressive cells that can also contribute tive marker for the choice of the type of immunotherapy to sustain the inflammatory TME and to the interaction and re- interventions. ciprocal influence of CSCs/CICs and their niche [110, 113–115]. Dendritic cells (DCs) are antigen presenting cells (APCs) Another key regulator of the cross-talk between CSCs/CICs and that can activate either innate or adaptive immune responses TAM and DCs is represented by CD47 [116]. This molecule is [98]. In addition, they play an important role in the formation over-expressed by CSCs/CICs of B cell malignancies. The bind- of anti-tumor T- and B cell immunologic memories [99]. ing of this molecule to the signal regulatory protein alpha Immature DCs can capture the tumor-derived antigens by (SIRPα), that mediates phagocytic functions in DCs and macro- phagocytosis or pinocytosis and then migrate to lymphoid phages, has been shown to mediate the impairment of innate organs where they present these TAAs in the form of HLA/ responses [116]. The cross-talk between CSCs/CICs and mye- peptide complexes to T cells, resulting in antigen-specific im- loid cells can affect both the fate and immunological profile of mune responses [100–102]. However, DCs depending on these cells, with implications for their susceptibilities to immune their morphological and phenotypic subtypes can either in- responses. Further studies should be designed to dissect the in- duce anti-tumor immune responses or promote tumor growth teractions of CSCs/CICs with different immune cells, although and progression [103]. The crosstalk of tumor with their TME the major limitation is represented by the lack of in vivo models is a crucial factor which results in the development of cancer to monitor the interaction of these different immune cell popula- [104]. Along this line, it has been described that high extent of tion in the context of TME. expression of the chemokine (C-X-C motif) ligand 1 (CXCL1) by tumor and stromal cells can promote CSCs/ CICs survival and proliferation and attract at tumor site DCs Immunomodulatory Properties of CSCs/CICs with suppressive functions, that could correlate tumor pro- gression and poor survival of patients [104]. The immunological profiling of CSCs/CICs has revealed that Macrophages represent important players for innate im- they share some characteristics with embryonic, hematopoiet- mune responses and can act as APCs similarly to DCs [105]. ic and mesenchymal stem cells displaying immunoregulatory Based on their phenotype and functions they can be distin- functions that render these cells invisible to immune responses guished in two subpopulations: 1. The M1 subtype that are and able to escape from tumor immune responses [4, 74, 117]. The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing... 139 The principle mechanisms governing the immunomodulation inhibition of T cell effector functions (Table 2)[65, 135]. It of CSCs/CICs are described below. has been demonstrated that the over-expression of IL-4 by CRC-derived CSCs/CICs has led to inefficient TCR- mediated proliferation and antigen recognition of CTLs [65]. Cytokines, Chemokines, Growth Factors & Immune Interestingly, the neutralization of this cytokine by mAb could Checkpoint Molecules overcome the T cell mediated anti-tumor impairment and in- duce antigen-specific recognition of both CSCs/CICs and dif- The observations that CSCs/CICs isolated from different tu- ferentiated tumor cells [65]. This study showed that, upon up- mor types can secrete soluble factors, such as Galectin-3, regulation of HLA class I and APM expression through IFN-γ GDF-15, IL-10, IL-13, PGE2 and TGFb, or express treatment of CSCs/CICs, T cells could specifically recognize a immune checkpoint molecules with immunosuppressive func- neoantigen, SMAD4, generated by a non-synonymous muta- tions, have suggested that these cells can regulate the impair- tion bearing stem-like cells and bulk tumor cells [65]. Thus, ment of immune responses as well as regulating a pro-tumoral CTL reactivity against CSCs/CICs and the TAA-specific TME (Table 2)[4, 51, 18–130]. These immunosuppressive immunosurveillance could be improved by the usage of strat- factors have been described to induce the differentiation of egies to correct the low immunogenic profile of these cells. regulatory T cells (Tregs) or MDSCs and M2 macrophages, The immune suppressive profile of CSCs/CICs has been resulting in the impairment of effector functions of innate and also confirmed by evidences describing the expression by adaptive responses [4, 51, 130, 131]. Moreover, pro- these cells of immune checkpoint molecules (e.g. CTLA-4, inflammatory cytokines such as IL-6, IL-8, IL-10 and IL-13, PD-L1, B7-H3 or B7-H4) (Table 2)[4, 34, 64, 65, 136]. released by CSCs/CICs can contribute to maintain an inflam- These observations highlighted the similarities between matory and suppressive TME representing the “niche” sus- CSCs/CICs and normal stem cells in terms of the immune taining cellular stemness (Table 2)[132, 133]. Indoleamine profile [34, 137, 138]. Moreover, altered expression of 2,3-dioxigenase (IDO), that mediates the catabolism of tryp- STAT3 pathway in CSCs/CICs can also affect their immune tophan, has been shown to be expressed by CSCs/CICs, con- suppressive activity through inhibiting T cell proliferation and tributing to the differentiation of Tregs, skewing the cytokine activation, inducing the differentiation of Tregs and triggering profiling of T cells toward TH2 -type and inhibiting the sur- Tcell apoptosis [121]. The observations reported above show vival and proliferation of CTL (Table 2)[133, 134]. that multiple mechanisms and molecular pathways are either In addition, the CSC/CIC-associated expression of IL-4 up-regulated or aberrantly activated in CSCs/CICs resulting in and CD200, through cell-to-cell interaction, lead to the Table 2 Immunomodulatory molecules detected in CSCs/CICs a b Molecule Function Activity in CSCs/CICs Reference IL-4 Cytokine involved in differentiation of Inhibition of TH1 cell mediated immune responses. [65, 118] naïve T cells to Th2. IL-10, IL-13 Anti-inflammatory cytokines Suppression of CTL functions; differentiation of [89, 119] Tregs and MDSCs TGFB Growth factor with potent inhibitory function Tregs differentiation, inhibition of TH1 responses [89, 120] STAT3 Transcription factor with a potential Maintenance and proliferation of CSCs/CICs; differ- [115, 121] anti-inflammatory function entiation of MDSCs, iDCs, M2 GDF15 Growth and differentiation factor Inhibition of anti-tumor immune responses [122] related to cellular stress. Galectin-3 Protein with important role in cell-cell Inhibitor of T cell mediated immune responses [89, 123] adhesion and interactions with the extracellular envi- ronment. IDO Enzyme involved in tryptophan catabolism Suppression of TH1 type immune responses and [34, 51, 64, 65, 123] differentiation of Tregs. CD200 A glycoprotein that regulates myeloid cell activity and Immune suppression and regulation of anti-tumor [124, 125] inhibits macrophage lineages. activity. PD-L1 Ligand of PD-1 and Immune checkpoint molecule Inhibition of CTL immune responses. [64, 65, 126, 127] B7-H3 and Immune checkpoint molecules Immunomodulation of cellular immune responses [34, 64, 65, 128, 129] B7-H4 : function of the molecules listed in the Table : Activity of these molecules when expressed by CSCs/CICs iDC: suppressive dendritic cell; MDSC: Myeloid derived suppressive cells; M2: M2 phenotype of monocytes/macrophages; TH1: T helper type 1; TH2: T helper type 2 140 Ravindran S. et al. their immune suppressive properties, therefore the blockade of high levels of PD-L1 and contain Tregs, M2 like macrophages these signaling through the combination of inhibitory agents as well as exhausted T cells [153]. CSCs/CICs that are con- should be considered in order to rescue the tumor-specific sidered the architects of their own microenvironment [154], as immune responses. well as generated by epithelial-to-mesenchymal transition (EMT) can be potentially responsible for the type of immune MicroRNAs infiltration depending of their pattern of immune profile. Common gene expression patterns have been found in miRNAs are non-coding RNAs regulating at post- normal mammary stem cells and dormant tumor cells transcriptional levels, through complementary binding to tar- from breast cancer suggesting the possible presence get mRNA, the expression of genes [139]. The altered regu- of stem-like cells in dormant tumors [151]. In addition, lation of gene expression in tumor cells can occur by both up- different cell sub-populations could be isolated from re- or down regulation of miRNA [139]. The most common ac- lapsed AML endowed with differential tumorigenic abili- tivity of miRNAs in CSCs/CICs is represented by the control ty depending on their up-regulation of stemness signaling of the expression of either oncogenes (e.g., MiR34a, MiR31 [155]. or MiR205) or tumor suppressor genes [139]. The aberrant A better understand of the relationship between stemness expression of few miRNAs, such as miRNAs 451and 199b- properties, immunological profile of CSCs/CICs and tumor 5p, has been shown to affect stem-like cell properties isolated dormancy will provide insights on the mechanisms of thera- from different type of tumors (e.g., GBM, breast cancer and peutic resistance of these cells and will allow to identify strat- medulloblastoma) [139–143]. Of note, miRNAs displaying egies for complete tumor eradication. regulatory activity on immune-related genes (e.g, miRNA- 199a that can regulate the IFN-mediated responses) can play a role in the differentiation of mammalian CSCs/CICs [144]. Immunological Targeting of CSCs/CICs The level of miRNA-124, through regulating the expression of STAT3, can affect the efficiency of anti-CSC/CIC T cell Cancer Vaccines responses in GBM [145]. Along this line, miR203 and miR92 can control the stemness and immunological profiles The recognition of TAAs expressed by CSCs/CICs by T cells of melanoma cells [146, 147]. have been documented (see Table 1). These in vitro or in vivo models were based on the usage of TAAs that represented sources of antigens for the therapeutic administration of can- Immune Evasion and Tumor Dormancy cer vaccines in cancer patients [81]. However, the principle limiting factor of the clinical efficacy of this strategy is repre- Tumor dormancy is represented by quiescent cells that can sented by the usage of “self”/tolerogenic TAAs, shared with remain occult and undetectable by regular diagnostic methods normal tissues [81]. The low or negative expression of these for long intervals of time, even after initial clinical responses categories of antigens and of CT-TAAs by CSCs/CICs can to therapies [148]. Quiescence of cells is the ability to exit cell represent an additional reason of failure of high rate and long cycle and remain in G0 phase until permissive environmental duration of clinical responses observed in cancer patients condition will lead to enter back into the cycling phase. This is treated with cancer vaccines [4, 34]. In addition, the sub- considered one of the principle mechanisms underlying tumor optimal levels of HLA class I molecules and APM by stem- dormancy. CSCs/CICs display the ability to cycle between like cells can drive the failure in targeting CSCs/CICs by quiescence and proliferation and together with their resistance cancer vaccines leading to the development of tumor dorman- to therapies represent the link between these cells and tumor cy and tumor recurrence, although the observance of initial dormancy [2, 5, 8, 9, 149–151]. Furthermore, the immune clinical efficacy of these therapeutic interventions [4, 34]. suppressive mechanisms associated with CSCs/CICs can or- DC-based vaccines, exploiting these cells as APC to pres- chestrate the evasion of these cells from immune recognition ent TAAs to T cell-mediated responses, represent also a ther- and immunosurveillance, and could be considered additional apeutic strategy for cancer patients showing encouraging clin- factors responsible of tumor dormancy [152]. ical activity [102, 156–162]. DCs loaded with either CSC/ An important mechanisms of immune-surveillance is the CIC-lysates or mRNA isolated from these cells represented homing of immune cells to the tumor site, which ultimately source of antigens for vaccination in the context of Phase I/ form the immune infiltrate. Tumors arising from epithelial II clinical trials of GBM patients [163, 164]. These studies breast cancer are known to possess high levels MHC class I provided proof of principle of improved overall survival of molecules and of infiltrating T effector cells and M1 macro- cancer patients treated with CSC/CIC targeted immunothera- phages. The immune infiltrate from mesenchymal like breast py [163–165]. Immune responses, with, in some cases in- cancer tumors exhibit low levels of MHC class I molecules, creased frequency of circulating NK cells, were detected in The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing... patients showing clinical benefit from these treatments [163, GBM [34, 64, 65, 177–179]. ,CSCs/CICs could theoretically 165]. Of note, these therapeutic interventions could overcome be targeted in vivo by immune checkpoint blockade agents, the failure of CSCs/CICs in expressing efficient levels of HLA enhancing the clinical efficacy of cancer vaccines, as demon- class I molecules and in presenting TAAs to T cells, strated in a mouse model [180]. However, recent reports de- documenting for the first time, that cancer vaccine, if eliciting scribing that clinical failure of these therapies was associated NK cell-mediated responses, could target stem-like with defective expression of HLA class I molecules by tumor tumor cells [158, 163–165]. Tumor cell clones expressing cells [75, 76], suggest that these cells might evade from the immunogenic neoantigens can undergo immune selection ICB-mediated unleash of immune responses (Fig. 2). due to the recognition and elimination by T lymphocytes, leading to the survival of tumor cell clones not expressing Adoptive Cell Therapy strong immunogenic antigens and maintaining the expression of low immunogenic TAAs [166] (and see https://www. ACT is represented by the isolation of T lymphocytes from biorxiv.org/content/10.1101/536433v1). This process is also cancer patients, their ex vivo expansion, and the infusion back associated with immune evasions mechanisms developed by into patients [181–183]. In addition, T lymphocytes both tumor cells and TME [166]. engineered to express TCR with high affinity for a cognate In some tumors, the decrease in antigen presentation is a TAA could be exploited for ACT studies [183]. Highly en- result of epigenetic silencing of the genes involved in antigen couraging and sustained responses mediated by adoptively presentation machinery. The usage of demethylating agents transferred TCR targeting the TAA NY-ESO-1 have recently such as 5-Aza-2′-deoxycytidine to reduce the methylation of been reported in different tumor types, such as breast cancer genes involved in antigen presentation, is a potential strategy and myeloma [86, 184]. Nevertheless, the antigen choice is to increase the antigen presentation in these CSC/CICs. The highly relevant to prevent severe toxicities due to “off-target” effect of demethylation has been shown in CSCs/CICs from cross-reaction with normal tissues sharing the same antigens breast cancer, where it resulted in high expression levels of or expressing molecules mimicking the TAAs [182]. TAP1, which is involved in antigen presentation [167]. Neoantigens have been described as candidate TAAs effi- In addition, increased antigen presentation also improves ciently recognized by T cells that can be exploited for ACT of the potential of discovering novel antigens, which can then be cancer patients and, interestingly, CTL targeting these anti- helpful in development of new anti-cancer vaccines (Fig. 2). gens could be isolated from tumor infiltrating lymphocytes (TILs) of melanoma and other type of malignant lesions Immune Checkpoint Blockade [183, 185, 186]. Nevertheless, ACT to target neoantigens can represent a promising approach for treatment of cancer Immune checkpoints, including CTLA-4, PD-1 and PD-L1, patients upon assessment of HLA expression by both CSCs/ are important physiological regulators of innate and adaptive CICs and differentiated tumor cells and, in case of suboptimal immune responses [168]. Biological inhibitory agents have levels of expression, the achievement of their up-regulation by been clinically developed, revealing striking therapeutically pre-treatment with immunomodulating agents [4, 34, 51]. success [169–175]. However, a significant proportion of can- T cell can be genetically modified to express a chimeric cer patients failed to benefit from these therapies. antigen receptor (CAR) that is composed of epitope-specific The effectiveness of immune checkpoint blockade domains isolated from mAbs linked to T cell-derived (ICB) is largely dependent on the tumor microenviron- activatory/costimulatory molecules [182, 187–189]. CAR-T ment [176]. Tumors such as melanoma, bladder cancer cells can recognize TAAs independently on the expression and non-small cell lung cancer (NSCLC) can be charac- of HLA molecules and APM components [189]. CAR-T cell terized as “hot” tumors due to their inflamed TME, high therapy for some subgroups of hematological malignancies levels of and neo-antigen expression and of T cell infil- represent the salvage intervention leading to stable clinical tration and detection of PD-L1. These tumors have been responses and improved overall survival of patients refractory reported to be associated with higher frequency of suscep- to standard therapies or with recurrences [187, 189–192]. The tibility to immune checkpoint treatments. On the other usage of CAR-T cell therapy for solid tumors is currently hand, prostate cancer is considered to be a “cold” tumor, under investigation, showing encouraging results in cancer due to minimal level of T cell infiltration, and limited patients with aggressive tumor types, including malignant me- response to single agent checkpoint inhibition [176]. sothelioma, pancreatic cancer and GBM [193–197]. Expression of immune checkpoint molecules has been ob- CAR-T cells targeting TAAs, such as CD133, EGFRvIII, served in CSCs/CICs from different histological origins [51]. EpCAM, CSPG4 and B7-H3, expressed by different type of PD-L1 expression was detected at high levels in CSCs/CICs solid tumors, including CSC/CIC components, have been de- isolated from primary human head and neck squamous cell veloped in pre-clinical studies [196–201]. These studies have shown that the targeting of TAAs that are expressed only by carcinoma (HNSCC), gastric and breast cancer, CRC and Ravindran S. et al. tumor cells, including CSCs/CICs, and not by normal cells, human immune system is desirable to allow the monitoring could provide the rational for safe and efficient clinical devel- of the interaction of CSCs/CICs with TME. opment of CAR-T cells therapy for these tumors(Fig. 2)[200, The targeting of CSCs/CICs by immunotherapy could re- 202]. Moreover, the combination of CAR-T cells targeting sult in the complete tumor eradication and stable clinical re- dual TAAs, EGFRvIII and CD133, has been used for the sponses in cancer patients. This goal could be achieved by the successful therapeutic treatment of a patient with advanced design of combination of strategies based on innate and/or cholangiocarcinoma [203]. antigen-specific T cell responses with immunoregulatory CAR-T cells targeting NKG2D ligands on CSCs/CICs agents that can render CSCs/CICs susceptible to cell- have been investigated and tested both in vitro and in vivo mediated immunosurveillance. In cases where epigenetic fac- [204, 205]. CSCs/CICs from glioblastoma expressing detect- tors are responsible for low antigen presentation, the usage of able NKG2D ligands could be efficiently targeted by CAR- T demethylating agents could represent a potential strategy to cells. These tools showed to efficintly eliminate also xenograft overcome the low expression of HLA molecules. tumors [205]. Nevertheless, a major limitation associated with Molecular approaches dissecting the fate of CSCs/CICs the use of these CAR-T cells is represented by the variability within tumor tissues will allow to develop immune-based pre- of the levels of NKG2D ligands on the surface of CSCs/CICs, cision medicine approaches and to identify biomarkers predic- depending on their origin and methodology used for their tive of patients’ responsiveness to therapies. ex vivo isolation. Acknowledgements This study was supported by Qatar National Nevertheless, further investigations aimed at a comprehen- Research Fund, grant no. NPRP10-0129-170277. sive genomic and immunological characterization CSCs/CICs are warranted to implement the efficiency and safety of ACT Funding Information Open Access funding provided by the Qatar strategies. National Library. Compliance with Ethical Standards Conclusions Conflict of Interest All the authors have no conflict of interest to disclose. Recent advances in the genomic, molecular and immunolog- ical profiling of CSCs/CICs have contributed to the identifi- Open Access This article is distributed under the terms of the Creative cation of dysregulated molecular pathways that orchestrate Commons Attribution 4.0 International License (http:// stem-like cancer cells and their interaction with TME. The creativecommons.org/licenses/by/4.0/), which permits unrestricted use, heterogeneity and plasticity of these cells and the mutual ef- distribution, and reproduction in any medium, provided you give appro- priate credit to the original author(s) and the source, provide a link to the fect of TME and CSCs/CICs on the resulting anti-tumoral or Creative Commons license, and indicate if changes were made. pro-tumoral environment, represent the principle limitations in predicting the fate of these cells and their role in cancer patients’ outcome. In addition, these cells have been identified as key players in therapeutic resistance of tumors and in the References development of their dormancy. The down-modulation of HLA molecules and NK activatory ligands on CSCs/CICs, 1. Shackleton M, Quintana E, Fearon ER, Morrison SJ (2009) through decreasing their susceptibility to T or NK cell Heterogeneity in cancer: cancer stem cells versus clonal evolution. targeting, might represent one of the principle factors leading Cell. 138(5):822–829 2. Clarke MF, Dick JE, Dirks PB et al (2006) Cancer stem cells– to resistance to immunotherapy. Although, the mechanisms perspectives on current status and future directions: AACR work- regulating the levels of HLA molecules and NKG2D ligands shop on cancer stem cells. Cancer Res 66(19):9339–9344 on CSCs/CICs are yet to be dissected. Due to the complexity 3. Li Y, Laterra J (2012) Cancer stem cells: distinct entities or dy- of the cross-talk between CSCs/CICs and TME and the high namically regulated phenotypes? Cancer Res 72(3):576–580 plasticity of these cells, it is difficult to predict what type of 4. Maccalli C, De Maria R (2015) Cancer stem cells: perspectives for therapeutic targeting. Cancer Immunol Immunother 64(1):91–97 immune cells could play a relevant role in targeting 5. Reya T, Morrison SJ, Clarke MF, Weissman IL (2001) Stem cells, CSCs/CICs. Multifactorial investigations, including the im- cancer, and cancer stem cells. Nature. 414(6859):105–111 munological profile, immunomodulating molecules, the inter- 6. Wicha MS, Liu S, Dontu G (2006) Cancer stem cells: an old idea– action with TME and the type of immune cell infiltration will a paradigm shift. Cancer Res 66(4):1883–1890 discussion 1895- allow to provide insights. Nevertheless, the available tools to 7. Tang DG (2012) Understanding cancer stem cell heterogeneity isolate and characterize CSCs/CICs, such as spheroids, immu- and plasticity. Cell Res 22(3):457–472 nodeficient mice, antigenic profile, are unsatisfactory to dis- 8. Eyler CE, Rich JN (2008) Survival of the fittest: cancer stem cells sect the cross-talk of these cells with TME. Moreover, the in therapeutic resistance and angiogenesis. J Clin Oncol 26(17): development of pre-clinical in vivo models engrafted with 2839–2845 The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing... 143 9. Hadjimichael C, Chanoumidou K, Papadopoulou N, Arampatzi P, for immunotherapy in vitro and in vivo. J Biol Chem 286(47): Papamatheakis J, Kretsovali A (2015) Common stemness regula- 40548–40555 tors of embryonic and cancer stem cells. World J Stem Cells 7(9): 31. Schmitz M, Temme A, Senner V et al (2007) Identification of 1150–1184 SOX2 as a novel glioma-associated antigen and potential target 10. Maugeri-Sacca M, Vigneri P, De Maria R (2011) Cancer stem cells for T cell-based immunotherapy. Br J Cancer 96(8):1293–1301 and chemosensitivity. Clin Cancer Res 17(15):4942–4947 32. Yamada R, Takahashi A, Torigoe T, Morita R, Tamura Y, 11. Soltanian S, Matin MM (2011) Cancer stem cells and cancer ther- Tsukahara T, Kanaseki T, Kubo T, Watarai K, Kondo T, apy. Tumour Biol 32(3):425–440 Hirohashi Y, Sato N (2013) Preferential expression of cancer/ testis genes in cancer stem-like cells: proposal of a novel sub- 12. Steinbichler TB, Dudas J, Skvortsov S, Ganswindt U, Riechelmann H, Skvortsova II. (2018) Therapy resistance medi- category, cancer/testis/stem gene. Tissue Antigens 81(6):428–434 ated by cancer stem cells. Semin Cancer Biol 33. Nishizawa S, Hirohashi Y, Torigoe T, Takahashi A, Tamura Y, Mori T, Kanaseki T, Kamiguchi K, Asanuma H, Morita R, 13. Ricci-Vitiani L, Fabrizi E, Palio E, De Maria R (2009) Colon cancer stem cells. J Mol Med (Berl) 87(11):1097–1104 Sokolovskaya A, Matsuzaki J, Yamada R, Fujii R, Kampinga HH, Kondo T, Hasegawa T, Hara I, Sato N (2012) HSP 14. Vezzoni L, Parmiani G (2008) Limitations of the cancer stem cell DNAJB8 controls tumor-initiating ability in renal cancer stem- theory. Cytotechnology. 58(1):3–9 like cells. Cancer Res 72(11):2844–2854 15. Morrison BJ, Steel JC, Morris JC (2018) Reduction of MHC-I 34. Maccalli C, Rasul KI, Elawad M, Ferrone S (2018) The role of expression limits T-lymphocyte-mediated killing of Cancer- initiating cells. BMC Cancer 18(1):469 cancer stem cells in the modulation of anti-tumor immune re- sponses. Semin Cancer Biol. 16. Maccalli C, Volonte A, Cimminiello C, Parmiani G (2014) 35. Clevers H (2011) The cancer stem cell: premises, promises and Immunology of cancer stem cells in solid tumours. A review. Eur J Cancer 50(3):649–655 challenges. Nat Med 17(3):313–319 36. Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, di Virgilio A, 17. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres- Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE (1994) A Conticello C, Ruco L, Peschle C, de Maria R (2008) Identification and expansion of the tumorigenic lung cancer stem cell popula- cell initiating human acute myeloid leukaemia after transplanta- tion. Cell Death Differ 15(3):504–514 tion into SCID mice. Nature. 367(6464):645–648 37. Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, 18. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke Peschle C, de Maria R (2007) Identification and expansion of MF (2003) Prospective identification of tumorigenic breast cancer human colon-cancer-initiating cells. Nature. 445(7123):111–115 cells. Proc Natl Acad Sci U S A 100(7):3983–3988 38. Todaro M, Gaggianesi M, Catalano V, Benfante A, Iovino F, 19. Clarke MF, Becker MW (2006) Stem cells: the real culprits in Biffoni M, Apuzzo T, Sperduti I, Volpe S, Cocorullo G, Gulotta cancer? Sci Am 295(1):52–59 G, Dieli F, de Maria R, Stassi G (2014) CD44v6 is a marker of 20. Singh SK, Clarke ID, Hide T, Dirks PB (2004) Cancer stem cells constitutive and reprogrammed cancer stem cells driving colon in nervous system tumors. Oncogene. 23(43):7267–7273 cancer metastasis. Cell Stem Cell 14(3):342–356 21. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, 39. Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM, Henkelman RM, Cusimano MD, Dirks PB (2004) Identification Morrison SJ (2008) Efficient tumour formation by single human of human brain tumour initiating cells. Nature. 432(7015):396– 401 melanoma cells. Nature. 456(7222):593–598 40. Roesch A, Fukunaga-Kalabis M, Schmidt EC, Zabierowski SE, 22. Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha Brafford PA, Vultur A, Basu D, Gimotty P, Vogt T, Herlyn M M, Clarke MF, Simeone DM (2007) Identification of pancreatic (2010) A temporarily distinct subpopulation of slow-cycling mel- cancer stem cells. Cancer Res 67(3):1030–1037 anoma cells is required for continuous tumor growth. Cell. 141(4): 23. Ricci-Vitiani L, Pagliuca A, Palio E, Zeuner A, De Maria R (2008) 583–594 Colon cancer stem cells. Gut. 57(4):538–548 41. Prieto-Vila M, Takahashi RU, Usuba W, Kohama I, Ochiya T 24. Visus C, Wang Y, Lozano-Leon A, Ferris RL, Silver S, (2017) Drug resistance driven by cancer stem cells and their niche. Szczepanski MJ, Brand RE, Ferrone CR, Whiteside TL, Ferrone Int J Mol Sci 18(12) S, DeLeo A, Wang X (2011) Targeting ALDH(bright) human 42. Fuchs E, Tumbar T, Guasch G (2004) Socializing with the neigh- carcinoma-initiating cells with ALDH1A1-specific CD8(+) T bors: stem cells and their niche. Cell. 116(6):769–778 cells. Clin Cancer Res 17(19):6174–6184 43. Xie T, Li L (2007) Stem cells and their niche: an inseparable 25. Brown CE, Starr R, Martinez C, Aguilar B, D'Apuzzo M, Todorov relationship. Development. 134(11):2001–2006 I, Shih CC, Badie B, Hudecek M, Riddell SR, Jensen MC (2009) 44. Ungefroren H, Sebens S, Seidl D, Lehnert H, Hass R (2011) Recognition and killing of brain tumor stem-like initiating cells by Interaction of tumor cells with the microenvironment. Cell CD8+ cytolytic T cells. Cancer Res 69(23):8886–8893 Commun Signal 9:18 26. Glumac PM, LeBeau AM (2018) The role of CD133 in cancer: a 45. Whiteside TL (2008) The tumor microenvironment and its role in concise review. Clin Transl Med 7(1):18 promoting tumor growth. Oncogene. 27(45):5904–5912 27. Zinzi L, Contino M, Cantore M, Capparelli E, Leopoldo M, 46. Cabarcas SM, Mathews LA, Farrar WL (2011) The cancer stem Colabufo NA (2014) ABC transporters in CSCs membranes as a cell niche–there goes the neighborhood? Int J Cancer 129(10): novel target for treating tumor relapse. Front Pharmacol 5:163 28. Mitra M, Kandalam M, Harilal A, Verma RS, Krishnan UM, 2315–2327 47. Cully M (2018) Tumour microenvironment: fibroblast subtype Swaminathan S, Krishnakumar S (2012) EpCAM is a putative stem marker in retinoblastoma and an effective target for T-cell- provides niche for cancer stem cells. Nat Rev Cancer 18(3):136 mediated immunotherapy. Mol Vis 18:290–308 48. Cully M (2018) Cancer: fibroblast subtype provides niche for 29. Choi D, Lee HW, Hur KY et al (2009) Cancer stem cell markers cancer stem cells. Nat Rev Drug Discov 17(3):163 CD133 and CD24 correlate with invasiveness and differentiation 49. Malanchi I, Santamaria-Martinez A, Susanto E et al (2012) in colorectal adenocarcinoma. World J Gastroenterol 15(18): Interactions between cancer stem cells and their niche govern 2258–2264 metastatic colonization. Nature. 481(7379):85–89 30. Shapira S, Kazanov D, Weisblatt S, Starr A, Arber N, Kraus S 50. Seidel S, Garvalov BK, Wirta V et al (2010) A hypoxic niche (2011) The CD24 protein inducible expression system is an ideal regulates glioblastoma stem cells through hypoxia inducible factor tool to explore the potential of CD24 as an oncogene and a target 2 alpha. Brain. 133(Pt 4):983–995 Ravindran S. et al. 51. Maccalli C, Parmiani G, Ferrone S (2017) Immunomodulating human melanoma cells with characteristics of cancer stem cells. and Immunoresistance properties of Cancer-initiating cells: impli- Int Immunol 21(7):793–801 cations for the clinical success of immunotherapy. Immunol 69. Zhang D, Tang DG, Rycaj K (2018) Cancer stem cells: regulation Investig 46(3):221–238 programs, immunological properties and immunotherapy. Semin 52. Easwaran H, Tsai HC, Baylin SB (2014) Cancer epigenetics: tu- Cancer Biol 52(Pt 2):94–106 mor heterogeneity, plasticity of stem-like states, and drug resis- 70. Grau JJ, Mesia R, de la Iglesia-Vicente M et al (2016) Enrichment tance. Mol Cell 54(5):716–727 of cells with Cancer stem cell-like markers in relapses of Chemoresistant patients with locally advanced head and neck 53. Dick JE (2008) Stem cell concepts renew cancer research. Blood. squamous cell carcinoma. Oncology. 90(5):267–272 112(13):4793–4807 71. Inoda S, Hirohashi Y, Torigoe T, Morita R, Takahashi A, Asanuma 54. Bar EE, Chaudhry A, Lin A, Fan X, Schreck K, Matsui W, H, Nakatsugawa M, Nishizawa S, Tamura Y, Tsuruma T, Terui T, Piccirillo S, Vescovi AL, DiMeco F, Olivi A, Eberhart CG Kondo T, Ishitani K, Hasegawa T, Hirata K, Sato N (2011) (2007) Cyclopamine-mediated hedgehog pathway inhibition de- Cytotoxic T lymphocytes efficiently recognize human colon can- pletes stem-like cancer cells in glioblastoma. Stem Cells 25(10): cer stem-like cells. Am J Pathol 178(4):1805–1813 2524–2533 72. Wei J, Barr J, Kong LY, Wang Y, Wu A, Sharma AK, Gumin J, 55. Sakariassen PO, Prestegarden L, Wang J, Skaftnesmo KO, Henry V, Colman H, Sawaya R, Lang FF, Heimberger AB (2010) Mahesparan R, Molthoff C, Sminia P, Sundlisaeter E, Misra A, Glioma-associated cancer-initiating cells induce immunosuppres- Tysnes BB, Chekenya M, Peters H, Lende G, Kalland KH, Øyan sion. Clin Cancer Res 16(2):461–473 AM, Petersen K, Jonassen I, van der Kogel A, Feuerstein BG, 73. Busse A, Letsch A, Fusi A, Nonnenmacher A, Stather D, Terzis AJ, Bjerkvig R, Enger PØ (2006) Angiogenesis- Ochsenreither S, Regenbrecht CR, Keilholz U (2013) independent tumor growth mediated by stem-like cancer cells. Characterization of small spheres derived from various solid tu- Proc Natl Acad Sci U S A 103(44):16466–16471 mor cell lines: are they suitable targets for T cells? Clin Exp 56. Eppert K, Takenaka K, Lechman ER, Waldron L, Nilsson B, van Metastasis 30(6):781–791 Galen P, Metzeler KH, Poeppl A, Ling V, Beyene J, Canty AJ, 74. Ichiryu N, Fairchild PJ (2013) Immune privilege of stem cells. Danska JS, Bohlander SK, Buske C, Minden MD, Golub TR, Methods Mol Biol 1029:1–16 Jurisica I, Ebert BL, Dick JE (2011) Stem cell gene expression 75. Gettinger S, Choi J, Hastings K, Truini A, Datar I, Sowell R, programs influence clinical outcome in human leukemia. Nat Med Wurtz A, Dong W, Cai G, Melnick MA, du VY, Schlessinger J, 17(9):1086–1093 Goldberg SB, Chiang A, Sanmamed MF, Melero I, Agorreta J, 57. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Montuenga LM, Lifton R, Ferrone S, Kavathas P, Rimm DL, Dewhirst MW, Bigner DD, Rich JN (2006) Glioma stem cells Kaech SM, Schalper K, Herbst RS, Politi K (2017) Impaired promote radioresistance by preferential activation of the DNA HLA class I antigen processing and presentation as a mechanism damage response. Nature. 444(7120):756–760 of acquired resistance to immune checkpoint inhibitors in lung 58. Dean M, Fojo T, Bates S (2005) Tumour stem cells and drug Cancer. Cancer Discov 7(12):1420–1435 resistance. Nat Rev Cancer 5(4):275–284 76. Chowell D, Morris LGT, Grigg CM et al (2018) Patient HLA class 59. Diehn M, Clarke MF (2006) Cancer stem cells and radiotherapy: I genotype influences cancer response to checkpoint blockade new insights into tumor radioresistance. J Natl Cancer Inst 98(24): immunotherapy. Science. 359(6375):582–587 1755–1757 77. Tallerico R, Todaro M, Di Franco S et al (2013) Human NK cells 60. Francipane MG, Alea MP, Lombardo Y, Todaro M, Medema JP, selective targeting of colon cancer-initiating cells: a role for natural Stassi G (2008) Crucial role of interleukin-4 in the survival of cytotoxicity receptors and MHC class I molecules. J Immunol colon cancer stem cells. Cancer Res 68(11):4022–4025 190(5):2381–2390 61. Frank NY, Schatton T, Frank MH (2010) The therapeutic promise 78. Castriconi R, Daga A, Dondero A et al (2009) NK cells recognize of the cancer stem cell concept. J Clin Invest 120(1):41–50 and kill human glioblastoma cells with stem cell-like properties. J 62. Li X, Lewis MT, Huang J, Gutierrez C, Osborne CK, Wu MF, Immunol 182(6):3530–3539 Hilsenbeck SG, Pavlick A, Zhang X, Chamness GC, Wong H, 79. Tseng HC, Arasteh A, Paranjpe A et al (2010) Increased lysis of Rosen J, Chang JC (2008) Intrinsic resistance of tumorigenic stem cells but not their differentiated cells by natural killer cells; breast cancer cells to chemotherapy. J Natl Cancer Inst 100(9): de-differentiation or reprogramming activates NK cells. PLoS One 672–679 5(7):e11590 63. Wilson BJ, Schatton T, Zhan Q, Gasser M, Ma J, Saab KR, 80. Maccalli C, Rasul KI, Elawad M, Ferrone S (2018) The role of Schanche R, Waaga-Gasser AM, Gold JS, Huang Q, Murphy cancer stem cells in the modulation of anti-tumor immune re- GF, Frank MH, Frank NY (2011) ABCB5 identifies a therapy- sponses. Semin Cancer Biol 53:189–200 refractory tumor cell population in colorectal cancer patients. 81. Parmiani G, Russo V, Maccalli C, Parolini D, Rizzo N, Maio M Cancer Res 71(15):5307–5316 (2014) Peptide-based vaccines for cancer therapy. Hum Vaccin 64. Di Tomaso T, Mazzoleni S, Wang E et al (2010) Immunother 10(11):3175–3178 Immunobiological characterization of cancer stem cells isolated 82. Lu YC, Robbins PF (2016) Cancer immunotherapy targeting from glioblastoma patients. Clin Cancer Res 16(3):800–813 neoantigens. Semin Immunol 28(1):22–27 65. Volonte A, Di Tomaso T, Spinelli M et al (2014) Cancer-initiating 83. Forshew T, Murtaza M, Parkinson C et al (2012) Noninvasive cells from colorectal cancer patients escape from T cell-mediated identification and monitoring of cancer mutations by targeted deep immunosurveillance in vitro through membrane-bound IL-4. J sequencing of plasma DNA. Sci Transl Med 4(136):136ra168 Immunol 192(1):523–532 84. Robbins PF (2017) Tumor-infiltrating lymphocyte therapy and 66. Catalano V (2015) Resistance of cancer stem cells to cell- medi- Neoantigens. Cancer J 23(2):138–143 ated immune responses. Vol 7: Springer 85. Sahin U, Derhovanessian E, Miller M et al (2017) Personalized 67. Codony-Servat J, Rosell R (2015) Cancer stem cells and RNA mutanome vaccines mobilize poly-specific therapeutic im- immunoresistance: clinical implications and solutions. Transl munity against cancer. Nature. 547(7662):222–226 Lung Cancer Res 4(6):689–703 86. Zacharakis N, Chinnasamy H, Black M et al (2018) Immune rec- 68. Pietra G, Manzini C, Vitale M, Balsamo M, Ognio E, Boitano M, ognition of somatic mutations leading to complete durable regres- Queirolo P, Moretta L, Mingari MC (2009) Natural killer cells kill sion in metastatic breast cancer. Nat Med 24(6):724–730 The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing... 145 87. Mennonna D, Maccalli C, Romano MC et al (2017) T cell 106. Sica A, Mantovani A (2012) Macrophage plasticity and polariza- neoepitope discovery in colorectal cancer by high throughput pro- tion: in vivo veritas. J Clin Invest 122(3):787–795 filing of somatic mutations in expressed genes. Gut. 66(3):454– 107. Gordon S, Martinez FO (2010) Alternative activation of macro- phages: mechanism and functions. Immunity. 32(5):593–604 108. Jinushi M, Chiba S, Yoshiyama H, Masutomi K, Kinoshita I, 88. Miyamoto S, Kochin V, Kanaseki T et al (2018) The antigen Dosaka-Akita H, Yagita H, Takaoka A, Tahara H (2011) Tumor- ASB4 on Cancer stem cells serves as a target for CTL immuno- therapy of colorectal Cancer. Cancer Immunol Res. associated macrophages regulate tumorigenicity and anticancer drug responses of cancer stem/initiating cells. Proc Natl Acad 89. Schatton T, Schutte U, Frank NYet al (2010) Modulation of T-cell Sci U S A 108(30):12425–12430 activation by malignant melanoma initiating cells. Cancer Res 109. Mantovani A, Sica A, Locati M (2005) Macrophage polarization 70(2):697–708 comes of age. Immunity. 23(4):344–346 90. Gedye C, Quirk J, Browning J, Svobodová S, John T, Sluka P, 110. Mitchem JB, Brennan DJ, Knolhoff BL, Belt BA, Zhu Y, Sanford Dunbar PR, Corbeil D, Cebon J, Davis ID (2009) Cancer/testis DE, Belaygorod L, Carpenter D, Collins L, Piwnica-Worms D, antigens can be immunological targets in clonogenic CD133+ Hewitt S, Udupi GM, Gallagher WM, Wegner C, West BL, melanoma cells. Cancer Immunol Immunother 58(10):1635–1646 Wang-Gillam A, Goedegebuure P, Linehan DC, DeNardo D 91. Koshio J, Kagamu H, Nozaki K, Saida Y, Tanaka T, Shoji S, (2013) Targeting tumor-infiltrating macrophages decreases Igarashi N, Miura S, Okajima M, Watanabe S, Yoshizawa H, tumor-initiating cells, relieves immunosuppression, and improves Narita I (2013) DEAD/H (asp-Glu-Ala-asp/his) box polypeptide chemotherapeutic responses. Cancer Res 73(3):1128–1141 3, X-linked is an immunogenic target of cancer stem cells. Cancer 111. Lindau D, Gielen P, Kroesen M, Wesseling P, Adema GJ (2013) Immunol Immunother 62(10):1619–1628 The immunosuppressive tumour network: myeloid-derived sup- 92. Brown CE, Starr R, Aguilar B, Shami AF, Martinez C, D'Apuzzo pressor cells, regulatory T cells and natural killer T cells. M, Barish ME, Forman SJ, Jensen MC (2012) Stem-like tumor- Immunology. 138(2):105–115 initiating cells isolated from IL13Ralpha2 expressing gliomas are 112. Filipazzi P, Huber V, Rivoltini L (2012) Phenotype, function and targeted and killed by IL13-zetakine-redirected T cells. Clin clinical implications of myeloid-derived suppressor cells in cancer Cancer Res 18(8):2199–2209 patients. Cancer Immunol Immunother 61(2):255–263 93. Tallerico R, Garofalo C, Carbone E (2016) A new biological fea- 113. Panni RZ, Sanford DE, Belt BA et al (2014) Tumor-induced ture of natural killer cells: the recognition of solid tumor-derived STAT3 activation in monocytic myeloid-derived suppressor cells Cancer stem cells. Front Immunol 7:179 enhances stemness and mesenchymal properties in human pancre- 94. Parmiani G (2016) Melanoma cancer stem cells: markers and atic Cancer. Cancer Immunol Immunother 63(5):513–528 functions. Cancers (Basel) 8(3) 114. Sherry MM, Reeves A, Wu JK, Cochran BH (2009) STAT3 is 95. Ames E, Canter RJ, Grossenbacher SK et al (2015) NK cells required for proliferation and maintenance of multipotency in glio- preferentially target tumor cells with a Cancer stem cell pheno- blastoma stem cells. Stem Cells 27(10):2383–2392 type. J Immunol 195(8):4010–4019 115. Zhou J, Wulfkuhle J, Zhang H, Gu P, Yang Y, Deng J, Margolick 96. Paczulla AM, Rothfelder K, Raffel S et al (2019) Absence of JB, Liotta LA, Petricoin E 3rd, Zhang Y (2007) Activation of the NKG2D ligands defines leukaemia stem cells and mediates their PTEN/mTOR/STAT3 pathway in breast cancer stem-like cells is immune evasion. Nature. 572(7768):254–259 required for viability and maintenance. Proc Natl Acad Sci U S A 97. Maccalli C, Giannarelli D, Chiarucci C, Cutaia O, Giacobini G, 104(41):16158–16163 Hendrickx W, Amato G, Annesi D, Bedognetti D, Altomonte M, 116. Majeti R, Chao MP, Alizadeh AA, Pang WW, Jaiswal S, Gibbs Danielli R, Calabrò L, di Giacomo AM, Marincola FM, Parmiani KD Jr, van Rooijen N, Weissman IL (2009) CD47 is an adverse G, Maio M (2017) Soluble NKG2D ligands are biomarkers asso- prognostic factor and therapeutic antibody target on human acute ciated with the clinical outcome to immune checkpoint blockade myeloid leukemia stem cells. Cell. 138(2):286–299 therapy of metastatic melanoma patients. Oncoimmunology. 6(7): 117. Ankrum JA, Ong JF, Karp JM (2014) Mesenchymal stem cells: e1323618 immune evasive, not immune privileged. Nat Biotechnol 32(3): 98. Cella M, Sallusto F, Lanzavecchia A (1997) Origin, maturation 252–260 and antigen presenting function of dendritic cells. Curr Opin 118. Brown MA, Hural J (2017) Functions of IL-4 and control of its Immunol 9(1):10–16 expression. Crit Rev Immunol 37(2–6):181–212 99. Keller AM, Xiao Y, Peperzak V, Naik SH, Borst J (2009) 119. Terabe M, Park JM, Berzofsky JA (2004) Role of IL-13 in regu- Costimulatory ligand CD70 allows induction of CD8+ T-cell im- lation of anti-tumor immunity and tumor growth. Cancer Immunol munity by immature dendritic cells in a vaccination setting. Immunother 53(2):79–85 Blood. 113(21):5167–5175 120. Miyazono K (2000) Positive and negative regulation of TGF-beta 100. Hanke N, Alizadeh D, Katsanis E, Larmonier N (2013) Dendritic signaling. J Cell Sci 113(Pt 7):1101–1109 cell tumor killing activity and its potential applications in cancer 121. Wei J, Barr J, Kong LYet al (2010) Glioblastoma cancer-initiating immunotherapy. Crit Rev Immunol 33(1):1–21 cells inhibit T-cell proliferation and effector responses by the sig- 101. Palucka K, Banchereau J (2013) Dendritic-cell-based therapeutic nal transducers and activators of transcription 3 pathway. Mol cancer vaccines. Immunity. 39(1):38–48 Cancer Ther 9(1):67–78 102. Nouri-Shirazi M, Banchereau J, Bell D et al (2000) Dendritic cells 122. Emmerson PJ, Duffin KL, Chintharlapalli S, Wu X (2018) GDF15 capture killed tumor cells and present their antigens to elicit tumor- and growth control. Front Physiol 9:1712 specific immune responses. J Immunol 165(7):3797–3803 123. Dumic J, Dabelic S, Flogel M (2006) Galectin-3: an open-ended 103. Grivennikov SI, Greten FR, Karin M (2010) Immunity, inflamma- story. Biochim Biophys Acta 1760(4):616–635 tion, and cancer. Cell. 140(6):883–899 124. Colmont CS, Benketah A, Reed SH et al (2013) CD200- 104. Hsu YL, Chen YJ, Chang WA et al. (2018) Interaction between expressing human basal cell carcinoma cells initiate tumor Tumor-Associated Dendritic Cells and Colon Cancer Cells growth. Proc Natl Acad Sci U S A 110(4):1434–1439 Contributes to Tumor Progression via CXCL1. Int J Mol Sci 19(8) 125. Wright GJ, Puklavec MJ, Willis AC et al (2000) Lymphoid/ 105. Condeelis J, Pollard JW (2006) Macrophages: obligate partners neuronal cell surface OX2 glycoprotein recognizes a novel recep- for tumor cell migration, invasion, and metastasis. Cell. 124(2): tor on macrophages implicated in the control of their function. 263–266 Immunity. 13(2):233–242 146 Ravindran S. et al. 126. Schatton T, Frank MH (2009) Antitumor immunity and cancer inhibits STAT3 signaling to enhance T cell-mediated immune stem cells. Ann N Y Acad Sci 1176:154–169 clearance of glioma. Cancer Res 73(13):3913–3926 127. Salmaninejad A, Khoramshahi V, Azani A, Soltaninejad E, Aslani 146. Sahranavardfard P, Firouzi J, Azimi M, et al. (2019) MicroRNA- S, Zamani MR, Zal M, Nesaei A, Hosseini SM (2018) PD-1 and 203 reinforces stemness properties in melanoma and augments cancer: molecular mechanisms and polymorphisms. tumorigenesis in vivo. J Cell Physiol Immunogenetics. 70(2):73–86 147. Shidal C, Singh NP, Nagarkatti P, Nagarkatti M (2019) 128. Yao Y, Ye H, Qi Z et al (2016) B7-H4(B7x)-mediated cross-talk MicroRNA-92 expression in CD133+ melanoma stem cells reg- between Glioma-initiating cells and macrophages via the IL6/ ulates immunosuppression in the tumor microenvironment via JAK/STAT3 pathway Lead to poor prognosis in Glioma patients. integrin-dependent activation of TGF-beta. Cancer Res Clin Cancer Res 22(11):2778–2790 148. Wang SH, Lin SY (2013) Tumor dormancy: potential therapeutic 129. Harris SG, Padilla J, Koumas L, Ray D, Phipps RP (2002) target in tumor recurrence and metastasis prevention. Exp Hematol Prostaglandins as modulators of immunity. Trends Immunol Oncol 2(1):29 23(3):144–150 149. Costello RT, Mallet F, Gaugler B et al (2000) Human acute mye- 130. Yoshimura A, Muto G (2011) TGF-beta function in immune sup- loid leukemia CD34+/CD38- progenitor cells have decreased sen- pression. Curr Top Microbiol Immunol 350:127–147 sitivity to chemotherapy and Fas-induced apoptosis, reduced im- munogenicity, and impaired dendritic cell transformation capaci- 131. Wan YY, Flavell RA (2007) 'Yin-Yang' functions of transforming growth factor-beta and T regulatory cells in immune regulation. ties. Cancer Res 60(16):4403–4411 Immunol Rev 220:199–213 150. Ishikawa F, Yoshida S, Saito Y, Hijikata A, Kitamura H, Tanaka S, 132. Shacter E, Weitzman SA (2002) Chronic inflammation and cancer. Nakamura R, Tanaka T, Tomiyama H, Saito N, Fukata M, Oncology (Williston Park) 16(2):217–226 229; discussion 230- Miyamoto T, Lyons B, Ohshima K, Uchida N, Taniguchi S, 212 Ohara O, Akashi K, Harada M, Shultz LD (2007) Chemotherapy-resistant human AML stem cells home to and en- 133. Parker KH, Beury DW, Ostrand-Rosenberg S (2015) Myeloid- derived suppressor cells: critical cells driving immune suppression graft within the bone-marrow endosteal region. Nat Biotechnol 25(11):1315–1321 in the tumor microenvironment. Adv Cancer Res 128:95–139 134. Smith C, Chang MY, Parker KH et al (2012) IDO is a nodal 151. Pece S, Tosoni D, Confalonieri S, Mazzarol G, Vecchi M, Ronzoni S, Bernard L, Viale G, Pelicci PG, di Fiore PP (2010) Biological pathogenic driver of lung cancer and metastasis development. and molecular heterogeneity of breast cancers correlates with their Cancer Discov 2(8):722–735 cancer stem cell content. Cell. 140(1):62–73 135. Kawasaki BT, Mistree T, Hurt EM, Kalathur M, Farrar WL (2007) 152. Steinbichler TB, Dudas J, Skvortsov S, Ganswindt U, Co-expression of the toleragenic glycoprotein, CD200, with Riechelmann H, Skvortsova II (2018) Therapy resistance medi- markers for cancer stem cells. Biochem Biophys Res Commun 364(4):778–782 ated by cancer stem cells. Semin Cancer Biol 53:156–167 136. Hirohashi Y, Torigoe T, Tsukahara T, Kanaseki T, Kochin V, Sato 153. Dongre A, Rashidian M, Reinhardt F, Bagnato A, Keckesova Z, N (2016) Immune responses to human cancer stem-like cells/ Ploegh HL, Weinberg RA (2017) Epithelial-to-Mesenchymal tran- sition contributes to immunosuppression in breast carcinomas. cancer-initiating cells. Cancer Sci 107(1):12–17 Cancer Res 77(15):3982–3989 137. Kim SY, Cho HS, Yang SH, Shin JY, Kim JS, Lee ST, Chu K, Roh 154. Prager BC, Xie Q, Bao S, Rich JN (2019) Cancer stem cells: the JK, Kim SU, Park CG (2009) Soluble mediators from human architects of the tumor ecosystem. Cell Stem Cell 24(1):41–53 neural stem cells play a critical role in suppression of T-cell acti- vation and proliferation. J Neurosci Res 87(10):2264–2272 155. Shlush LI, Mitchell A, Heisler L et al (2017) Tracing the origins of relapse in acute myeloid leukaemia to stem cells. Nature. 138. Ljujic B, Milovanovic M, Volarevic V et al (2013) Human mes- enchymal stem cells creating an immunosuppressive environment 547(7661):104–108 156. Encabo A, Solves P, Mateu E, Sepulveda P, Carbonell-Uberos F, and promote breast cancer in mice. Sci Rep 3:2298 139. Chhabra R, Saini N (2014) MicroRNAs in cancer stem cells: cur- Minana MD (2004) Selective generation of different dendritic cell rent status and future directions. Tumour Biol 35(9):8395–8405 precursors from CD34+ cells by interleukin-6 and interleukin-3. Stem Cells 22(5):725–740 140. Garzia L, Andolfo I, Cusanelli E et al (2009) MicroRNA-199b-5p impairs cancer stem cells through negative regulation of HES1 in 157. Hutten T, Thordardottir S, Hobo W, Hübel J, van der Waart A, medulloblastoma. PLoS One 4(3):e4998 Cany J, Dolstra H, Hangalapura BN (2014) Ex vivo generation of 141. Gasparini P, Lovat F, Fassan M, Casadei L, Cascione L, Jacob NK, interstitial and Langerhans cell-like dendritic cell subset-based Carasi S, Palmieri D, Costinean S, Shapiro CL, Huebner K, Croce vaccines for hematological malignancies. J Immunother 37(5): 267–277 CM (2014) Protective role of miR-155 in breast cancer through RAD51 targeting impairs homologous recombination after irradi- 158. Cornel AM, van Til NP, Boelens JJ, Nierkens S (2018) Strategies ation. Proc Natl Acad Sci U S A 111(12):4536–4541 to genetically modulate dendritic cells to potentiate anti-tumor responses in hematologic malignancies. Front Immunol 9:982 142. Petrelli A, Carollo R, Cargnelutti M et al (2015) By promoting cell differentiation, miR-100 sensitizes basal-like breast cancer stem 159. Banchereau J, Steinman RM (1998) Dendritic cells and the control of immunity. Nature. 392(6673):245–252 cells to hormonal therapy. Oncotarget. 6(4):2315–2330 143. Roscigno G, Quintavalle C, Donnarumma E, Puoti I, Diaz- 160. Kirk CJ, Mule JJ (2000) Gene-modified dendritic cells for use in Lagares A, Iaboni M, Fiore D, Russo V, Todaro M, Romano G, tumor vaccines. Hum Gene Ther 11(6):797–806 Thomas R, Cortino G, Gaggianesi M, Esteller M, Croce CM, 161. Kranz LM, Diken M, Haas H, Kreiter S, Loquai C, Reuter KC, Condorelli G (2016) MiR-221 promotes stemness of breast cancer Meng M, Fritz D, Vascotto F, Hefesha H, Grunwitz C, Vormehr cells by targeting DNMT3b. Oncotarget. 7(1):580–592 M, Hüsemann Y, Selmi A, Kuhn AN, Buck J, Derhovanessian E, 144. Celia-Terrassa T, Liu DD, Choudhury A et al (2017) Normal and Rae R, Attig S, Diekmann J, Jabulowsky RA, Heesch S, Hassel J, cancerous mammary stem cells evade interferon-induced con- Langguth P, Grabbe S, Huber C, Türeci Ö, Sahin U (2016) straint through the miR-199a-LCOR axis. Nat Cell Biol 19(6): Systemic RNA delivery to dendritic cells exploits antiviral defence 711–723 for cancer immunotherapy. Nature. 534(7607):396–401 145. Wei J, Wang F, Kong LY, Xu S, Doucette T, Ferguson SD, Yang Y, 162. Salcedo M, Bercovici N, Taylor R, Vereecken P, Massicard S, McEnery K, Jethwa K, Gjyshi O, Qiao W, Levine NB, Lang FF, Duriau D, Vernel-Pauillac F, Boyer A, Baron-Bodo V, Mallard Rao G, Fuller GN, Calin GA, Heimberger AB (2013) miR-124 E, Bartholeyns J, Goxe B, Latour N, Leroy S, Prigent D, Martiat The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing... P, Sales F, Laporte M, Bruyns C, Romet-Lemonne JL, Abastado Rogers LJ, Gracia GJ, Jones SA, Mangiameli DP, Pelletier MM, JP, Lehmann F, Velu T (2006) Vaccination of melanoma patients Gea-Banacloche J, Robinson MR, Berman DM, Filie AC, Abati using dendritic cells loaded with an allogeneic tumor cell lysate. A, Rosenberg SA (2005) Adoptive cell transfer therapy following Cancer Immunol Immunother 55(7):819–829 non-myeloablative but lymphodepleting chemotherapy for the 163. Finocchiaro G, Pellegatta S (2016) Immunotherapy with dendritic treatment of patients with refractory metastatic melanoma. J Clin cells loaded with glioblastoma stem cells: from preclinical to clin- Oncol 23(10):2346–2357 ical studies. Cancer Immunol Immunother 65(1):101–109 182. Hinrichs CS, Rosenberg SA (2014) Exploiting the curative poten- 164. Vik-Mo EO, Nyakas M, Mikkelsen BV, Moe MC, Due-Tønnesen tial of adoptive T-cell therapy for cancer. Immunol Rev 257(1):56– P, Suso EM, Sæbøe-Larssen S, Sandberg C, Brinchmann JE, 71 Helseth E, Rasmussen AM, Lote K, Aamdal S, Gaudernack G, 183. Rosenberg SA, Restifo NP (2015) Adoptive cell transfer as per- Kvalheim G, Langmoen IA (2013) Therapeutic vaccination sonalized immunotherapy for human cancer. Science. 348(6230): against autologous cancer stem cells with mRNA-transfected den- 62–68 dritic cells in patients with glioblastoma. Cancer Immunol 184. Rapoport AP, Stadtmauer EA, Binder-Scholl GK, Goloubeva O, Immunother 62(9):1499–1509 Vogl DT, Lacey SF, Badros AZ, Garfall A, Weiss B, Finklestein J, 165. Pellegatta S, Eoli M, Frigerio S, Antozzi C, Bruzzone MG, Cantini Kulikovskaya I, Sinha SK, Kronsberg S, Gupta M, Bond S, G, Nava S, Anghileri E, Cuppini L, Cuccarini V, Ciusani E, Melchiori L, Brewer JE, Bennett AD, Gerry AB, Pumphrey NJ, Dossena M, Pollo B, Mantegazza R, Parati EA, Finocchiaro G Williams D, Tayton-Martin HK, Ribeiro L, Holdich T, Yanovich (2013) The natural killer cell response and tumor debulking are S, Hardy N, Yared J, Kerr N, Philip S, Westphal S, Siegel DL, associated with prolonged survival in recurrent glioblastoma pa- Levine BL, Jakobsen BK, Kalos M, June CH (2015) NY-ESO-1- tients receiving dendritic cells loaded with autologous tumor ly- specific TCR-engineered T cells mediate sustained antigen- sates. Oncoimmunology. 2(3):e23401 specific antitumor effects in myeloma. Nat Med 21(8):914–921 166. Vinay DS, Ryan EP, Pawelec G et al (2015) Immune evasion in 185. Robbins PF, Lu YC, El-Gamil M et al (2013) Mining exomic cancer: mechanistic basis and therapeutic strategies. Semin Cancer sequencing data to identify mutated antigens recognized by adop- Biol 35(Suppl):S185–S198 tively transferred tumor-reactive T cells. Nat Med 19(6):747–752 167. Sultan M, Vidovic D, Paine AS et al (2018) Epigenetic silencing 186. Tran E, Rosenberg SA (2014) T-cell therapy against cancer muta- of TAP1 in Aldefluor(+) breast Cancer stem cells contributes to tions. Oncotarget. 5(13):4579–4580 their enhanced immune evasion. Stem Cells 36(5):641–654 187. Boyiadzis MM, Dhodapkar MV, Brentjens RJ et al (2018) 168. Pardoll DM (2012) The blockade of immune checkpoints in can- Chimeric antigen receptor (CAR) T therapies for the treatment cer immunotherapy. Nat Rev Cancer 12(4):252–264 of hematologic malignancies: clinical perspective and signifi- 169. Adachi K, Tamada K (2015) Immune checkpoint blockade opens cance. J Immunother Cancer. 6(1):137 an avenue of cancer immunotherapy with a potent clinical effica- 188. Guedan S, Posey AD Jr., Shaw C et al. (2018) Enhancing CAR T cy. Cancer Sci 106(8):945–950 cell persistence through ICOS and 4-1BB costimulation. JCI 170. Haanen JB, Robert C (2015) Immune checkpoint inhibitors. Prog Insight 3(1) Tumor Res 42:55–66 189. June CH, O'Connor RS, Kawalekar OU, Ghassemi S, Milone MC 171. Kyi C, Postow MA (2016) Immune checkpoint inhibitor combi- (2018) CAR T cell immunotherapy for human cancer. Science. nations in solid tumors: opportunities and challenges. 359(6382):1361–1365 Immunotherapy. 8(7):821–837 190. Alcantara M, Tesio M, June CH, Houot R (2018) CAR T-cells for 172. Postow MA, Callahan MK, Wolchok JD (2015) Immune check- T-cell malignancies: challenges in distinguishing between thera- point blockade in Cancer therapy. J Clin Oncol 33(17):1974–1982 peutic, normal, and neoplastic T-cells. Leukemia. 32(11):2307– 173. Rotte A, Jin JY, Lemaire V (2018) Mechanistic overview of im- 191. Hay KA, Turtle CJ (2017) Chimeric antigen receptor (CAR) T mune checkpoints to support the rational design of their combina- tions in cancer immunotherapy. Ann Oncol 29(1):71–83 cells: lessons learned from targeting of CD19 in B-cell malignan- 174. Wilden SM, Lang BM, Mohr P, Grabbe S (2016) Immune check- cies.Drugs.77(3):237–245 point inhibitors: a milestone in the treatment of melanoma. J Dtsch 192. Kohn DB, Dotti G, Brentjens R, Savoldo B, Jensen M, Cooper LJ, Dermatol Ges 14(7):685–695 June CH, Rosenberg S, Sadelain M, Heslop HE (2011) CARs on 175. Xia Y, Medeiros LJ, Young KH (2016) Immune checkpoint block- track in the clinic. Mol Ther 19(3):432–438 ade: releasing the brake towards hematological malignancies. 193. Watanabe K, Kuramitsu S, Posey AD Jr, June CH (2018) Blood Rev 30(3):189–200 Expanding the therapeutic window for CARTcell therapy in solid 176. Gajewski TF (2015) The next hurdle in Cancer immunotherapy: tumors: the Knowns and unknowns of CAR T cell biology. Front overcoming the non-T-cell-inflamed tumor microenvironment. Immunol 9:2486 Semin Oncol 42(4):663–671 194. DeRenzo C, Krenciute G, Gottschalk S (2018) The landscape of 177. Zhi Y, Mou Z, Chen J, He Y, Dong H, Fu X, Wu Y (2015) B7H1 CAR T cells beyond acute lymphoblastic leukemia for pediatric expression and epithelial-to-Mesenchymal transition phenotypes solid tumors. Am Soc Clin Oncol Educ Book 38:830–837 on colorectal Cancer stem-like cells. PLoS One 10(8):e0135528 195. Ma Q, Garber HR, Lu S, He H, Tallis E, Ding X, Sergeeva A, 178. Yang Y, Wu KE, Zhao E et al (2015) B7-H1 enhances proliferation Wood MS, Dotti G, Salvado B, Ruisaard K, Clise-Dwyer K, John ability of gastric cancer stem-like cells as a receptor. Oncol Lett LS, Rezvani K, Alatrash G, Shpall EJ, Molldrem JJ (2016) A 9(4):1833–1838 novel TCR-like CAR with specificity for PR1/HLA-A2 effective- 179. Wu Y, Chen M, Wu P, Chen C, Xu ZP, Gu W (2017) Increased ly targets myeloid leukemia in vitro when expressed in human PD-L1 expression in breast and colon cancer stem cells. Clin Exp adult peripheral blood and cord blood T cells. Cytotherapy. 18(8):985–994 Pharmacol Physiol 44(5):602–604 180. Shi X, Zhang X, Li J, Mo L, Zhao H, Zhu Y, Hu Z, Gao J, Tan W 196. Zhu X, Prasad S, Gaedicke S, Hettich M, Firat E, Niedermann G (2015) Patient-derived glioblastoma stem cells are killed by (2018) PD-1 blockade enhances the antitumor efficacy of GM- CSF surface-modified bladder cancer stem cells vaccine. Int J CD133-specific CAR T cells but induce the T cell aging marker Cancer 142(10):2106–2117 CD57. Oncotarget. 6(1):171–184 181. Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL, 197. Morgan RA, Johnson LA, Davis JL et al (2012) Recognition of Restifo NP, Royal RE, Kammula U, White DE, Mavroukakis SA, glioma stem cells by genetically modified T cells targeting 148 Ravindran S. et al. EGFRvIII and development of adoptive cell therapy for glioma. cell immunotherapy of solid tumors. Expert Opin Ther Targets Hum Gene Ther 23(10):1043–1053 19(10):1339–1350 198. Beard RE, Zheng Z, Lagisetty KH et al (2014) Multiple chimeric 203. Feng KC, Guo YL, Liu Y et al (2017) Cocktail treatment with antigen receptors successfully target chondroitin sulfate proteogly- EGFR-specific and CD133-specific chimeric antigen receptor- can 4 in several different cancer histologies and cancer stem cells. modified T cells in a patient with advanced cholangiocarcinoma. J Immunother Cancer. 2:25 J Hematol Oncol 10(1):4 199. Pellegatta S, Savoldo B, Di Ianni N, et al. (2018) Constitutive and 204. Sun B, Yang D, Dai H, et al. (2019) Eradication of hepatocellular TNFalpha-inducible expression of chondroitin sulfate proteogly- carcinoma by NKG2D-specific CAR T-cells. Cancer Immunol can 4 in glioblastoma and neurospheres: Implications for CAR-T Res cell therapy. Sci Transl Med 10(430) 205. Yang D, Sun B, Dai H, Li W, Shi L, Zhang P, Li S, Zhao X (2019) 200. Du H, Hirabayashi K, Ahn S et al (2019) Antitumor responses in T cells expressing NKG2D chimeric antigen receptors efficiently the absence of toxicity in solid tumors by targeting B7-H3 via eliminate glioblastoma and cancer stem cells. J Immunother chimeric antigen receptor Tcells. Cancer Cell 35(2):221–237 e228 Cancer 7(1):171 201. Deng Z, Wu Y, Ma W, Zhang S, Zhang YQ (2015) Adoptive T-cell therapy of prostate cancer targeting the cancer stem cell antigen Publisher’sNote Springer Nature remains neutral with regard to jurisdic- EpCAM. BMC Immunol 16:1 tional claims in published maps and institutional affiliations. 202. Wang Y, Geldres C, Ferrone S, Dotti G (2015) Chondroitin sulfate proteoglycan 4 as a target for chimeric antigen receptor-based T- http://www.deepdyve.com/assets/images/DeepDyve-Logo-lg.png Cancer Microenvironment Springer Journals

The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing Piece of the Puzzle for the Efficient Targeting of these Cells with Immunotherapy

Loading next page...
 
/lp/springer-journals/the-cross-talk-between-cancer-stem-cells-cancer-initiating-cells-and-4qHmZvi9a0

References (209)

Publisher
Springer Journals
Copyright
Copyright © 2019 by The Author(s)
Subject
Biomedicine; Cancer Research; Oncology; Immunology; Cell Biology; Biochemistry, general; Biomedicine, general
ISSN
1875-2292
eISSN
1875-2284
DOI
10.1007/s12307-019-00233-1
Publisher site
See Article on Publisher Site

Abstract

Cancer Stem Cells/Cancer Initiating Cells (CSCs/CICs) is a rare sub-population within a tumor that is responsible for tumor formation, progression and resistance to therapies. The interaction between CSCs/CICs and tumor microenvironment (TME) can sustain “stemness” properties and promote their survival and plasticity. This cross-talk is also pivotal in regulating and modu- lating CSC/CIC properties. This review will provide an overview of the mechanisms underlying the mutual interaction between CSCs/CICs and TME. Particular focus will be dedicated to the immunological profile of CSCs/CICs and its role in orchestrating cancer immunosurveillance. Moreover, the available immunotherapy strategies that can target CSCs/CICs and of their possible implementation will be discussed. Overall, the dissection of the mechanisms regulating the CSC/CIC-TME interaction is warranted to understand the plasticity and immunoregulatory properties of stem-like tumor cells and to achieve complete eradications of tumors through the optimization of immunotherapy. . . . Keywords Cancer stem cells/Cancer initiating cells Immunosurveillance Adaptive immune responses Innate immune . . responses Tumor microenvironment Immunotherapy Abbreviations IL-4 Interleukin 4 ALDH Aldehyde dehydrogenase IL-10 Interleukin 10 APC Antigen presenting cells IL-13 Interleukin 13 APM Antigen processing machinery IL-13α2 α2 chain of IL-13 receptor CAR Chimeric antigen receptor mAb Monoclonal antibody; CIC Cancer initiating cell MDSC Myeloid derived suppressor cell CRC Colorectal cancer NSCLC Non-small cell lung cancer CT Cancer testis PD-1 Programmed death 1 CTLA-4 Cytotoxic lymphocyte antigen-4 PD-L1 Programmed death ligand 1 CSPG4 Chondroitin sulphate protidoglycan 4 RCC Renal cell carcinoma HLA Human leukocyte antigen STAT3 Signal transducer and activator of transcription 3 IDO Indoleamine 2,3-dioxygenase TGFB Transforming growth factor beta GBM Glioblastoma multiforme TAA Tumor associated antigen GDF-15 Growth differentiation factor-15 Treg T regulatory cell. IFN Interferon Introduction Shilpa Ravindran and Saad Rasool contributed equally to this work. Tumors are composed by heterogeneous cellular components * Cristina Maccalli including a rare subpopulation bearing “stemness properties” cmaccalli@sidra.org and being responsible of tumor initiation and progression. These cells have been denominated cancer stem cells Research Department, Sidra Medicine, Al Luqta Street, PO Box 26999, Doha, Qatar (CSCs) or cancer initiating cells (CICs) [1–6]. CSCs/CICs 134 Ravindran S. et al. share several characteristics with normal stem cells, such as detecting the presence of these cells within tumor lesions the ability to self-renew and to give rise to differentiated prog- though probing for CSC/CIC- associated markers has not pro- eny and the resistance to DNA damage-induced cell death [3, vided conclusive results. The xenotransplantation in immune 5–12]. CSCs/CICs, through the cycling from proliferation to deficient mice represents a useful tool to demonstrate in vivo quiescence, expression of ABC drug pumps, high levels of the tumorigenic properties CSCs/CICs [35]. Xenograft anti-apoptotic proteins and resistance to DNA damage, are models have contributed to prove the existence within tumor resistant to radiation and chemotherapy and play an important lesions of cell population endowed with stemness properties role in disease relapse and tumor progression [13, 14]. that upon serial transplantation could propagate both tumori- CSCs/CICs have been isolated from both hematological genic CSCs/CICs and malignant cells with differentiated phe- and solid tumors and they represent a rare subpopulation, notype without tumorigenic properties [18]. These subpopu- comprising 0.01–10% of cells within the tumor [15]. They lations can be identified only through transplantation in im- can be ex vivo identified based on their “stem cell-like” char- mune deficient mice [4, 36–38]. acteristics and the expression of certain cell surface and func- Nevertheless, the available CSC/CIC-associated tional markers [16]. The identification of CSCs/CICs was first markers are dependent on spatial and temporal features, reported in leukemia, showing a hierarchical organization of with their modulation occurring in relation to their inocu- tumor cells [17]. The leukemic cells were able to be engrafted lation in immunodeficient mice, proving the high level of + − upon transplantation of CD34 CD38 cells into severe com- plasticity of these cells and that none of the available bined immune-deficient (SCID) mice, which eventually led to markers can be exploited to monitor the in vivo fate of the identification of the hierarchical organization of tumors these cells [1, 39, 40] CSCs/CICs, similarly to normal stem with few cells endowed with stemness and tumorigenic prop- cells, require a “niche” to allow the survival of these cells erties [17]. Since then, a variety of studies highlighted the and their cycling from quiescence to proliferation and to existence of “stem-like” cancer cells in solid tumors with dif- maintain stemness and multipotency [41–43]. The “niche” ferent histological origins [5, 18–23]. Multiple molecules is represented by the tumor microenvironment (TME), (e.g., ALDH-1, CD133, CD44, CD24, CBX3, ABCA5, which is composed of multicellular and dynamic compart- LGR5, etc) have been identified as CSC/CIC-associated ments that include fibroblasts, endothelial, stromal, mesen- markers with differential expression depending on the tissues chymal and immune cells [41]. The interaction of TME of derivation, highlighting the high grade of heterogeneity of with stem-like cancer cells can regulate the fate of these these cells [16](Table 1). Most of these molecules are over- cells through modulating the proliferation, differentiation, expressed by CSCs/CICs but are also shared with either dif- immunological properties and resistance to therapies ferentiated tumor cells or normal stem cells [4, 34]. As a result, [44–50]. Table 1 Markers expressed by Marker Tumor type Recognition by T Reference CSCs/CICs isolated from solid cells tumors and their role as TAAs ALDH1 CRC; breast and gastric cancer; melanoma √ [24] CD133 GBM, pancreas, lung, ovarian, prostate, and gastric √ [25–27] cancer CD44 CRC, head and neck cancer EpCAM CRC, Retinoblastoma [28] EpCAM CD44 Pancreatic cancer CD24 CD24 CRC √ [29, 30] SOX2 GBM √ [31] CBX3 Ostocarcinoma LGR5 CRC ABCB5 Melanoma CD90 Liver cancer HSP DNAJB8 RCC √ [32, 33] CD166 CRC; NSCLC a b Markers commonly identified as associated with CSCs/CICs; the role of these antigens in eliciting T cell- mediated immune responses against CSCs/CICs CRC: colorectal cancer; GBM: glioblastoma multiforme; NSCLC: non-small cell lung cancer; RCC: renal cell carcinoma The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing... 135 The high grade of heterogeneity and plasticity of and of the “niche”, preventing the constant monitoring of CSCs/CICs can depend on their tissue of derivation and, plasticity and heterogeneity of these cells (Figs. 1, 2). importantly, on their cross-talk with TME [4, 16, 51–53]. Therefore, the combination of deep genomic, molecular Limiting the isolation and the functional characterization and functional profiling of CSCs/CICs could represent a rele- of CSCs/CICs to the usage of phenotypic markers is un- vant method to achieve a comprehensive functional character- satisfactory and do not consider the possibility that ization of these cells and, possibly, of their role in tumor out- “stemness” function of tumor cells can be reversible, as come [56]. shown by Quintana et al. for melanoma [1, 39]. Moreover, CSCs/CICs have been identified as the tumor compo- xenotransplantation of these cells in immune deficient nents responsible for resistance to standard therapy, as mice is lacking the important variable of the TME and well as immunotherapy [10, 57–60]. Although clinical its role in affecting the fate of CSCs/CICs [1]. responses in cancer patients are observed following Therefore, the lack of standardized methods to isolate treatments, these cells can remain in the minimal resid- CSCs/CICs and of in vivo models allowing to monitor ual disease and upon changes in the environment they the cross-talk of these cells with TME can lead to the high can exit the quiescence status and give rise to novel extent of variability in assessing the functional properties malignant lesion(s) or even initiate the metastatic colo- of thesecells andinpreventingtoaccuratelydetermine nization [8, 61–63]. their fate and role in the tissue of origins and in the clin- The extensive molecular and immunological characterization ical outcome of cancer patients [54, 55]. The tool of of CSCs/CICs is warranted in order to understand the mecha- sphere forming assay to propagate in vitro CSCs/CICs is nisms regulating their plasticity, quiescence, interaction with the too simplified, lacking the important component of TME TME and resistance to therapies and to immune responses. Fig. 1 Differential immunogenic profile by CSCs/CICs vs. bulk tumor by CSCs/CICs. Neoantigens, generated by somatic mutation bearing tu- cells. CSCs/CICs can express defective levels of HLA molecules and mor cells are equally expressed by both CSCs/CICs and differentiated APM components leading to low immunogenicity and escape from im- tumor cells. The latest TAAs represent highly immunogenic target mole- mune responses. In the presence of efficient expression of ligands of NK- cules, since they are not expressed by normal cells. APM: antigen pro- associated activatory receptors, these cells can become susceptible to NK cessing machinery; CSCs/CICs: cancer stem cells/cancer initiating cells; cell recognition. Moreover, TAAs can be expressed at suboptimal levels NK: natural killer cells 136 Ravindran S. et al. Fig. 2 Immunotherapy strategies to target CSCs/CICs. An overview of multiple immunotherapy approaches or with standard therapies warrant immunotherapy approaches including adoptive cell therapy with either 1. further investigation to assess the efficacy in increasing the immunoge- TCR or CAR engineered T lymphocytes; 2. Immune check point nicity of CSCs/CICs and to implement the targeting of these cells by blockade with mAbs; 3. Cancer vaccination with TAAs expressed by immune responses. CSCs/CICs: cancer stem cells/cancer initiating cells; both CSCs/CICs and differentiated tumor cells; 4. Innate immune re- TAA: tumor associated antigen sponse or 5. γδ T cell recognition of tumor cells. Combination of either Immunological Profile of CSCs from CRC and endowed with stemness properties, showed detectable level of HLA class I molecules as well susceptibil- HLA Molecules and APM Components ity to antigen-specific cytotoxic T lymphocytes (CTLs) [71], however this study has been performed using long-term The expression of HLA class I and class II molecules in vitro established cell lines, that could have lost phenotypic and APM has been investigated in CSCs/CICs isolated from properties of primary CSCs/CICs. Indeed, stem-like cells iso- colorectal cancer (CRC) and glioblastoma multiforme (GBM) lated by sphere-forming assay displayed aberrant expression showing an overall aberrant expression of these molecules, of HLA class I and APM components [16, 65]. Contradictory with, in some cases, failure in their modulation by the pre- results were obtained also in glioblastoma multiforme (GBM); treatment with IFNs (both alpha and gamma) or DNA CSCs/CICs isolated as sphere forming cells from this tumor demethylating agent (5-Aza CdR) [64, 65]. This impairment have been shown to exhibit the expression of HLA class I in antigen processing and presentation by CSCs/CICs lead, molecules [72] while, when applying these analyses to prima- upon co-culture of these cells with autologous T cells, to a ry GBM-derived sphere forming cells, defective expression of preferential selection and differentiation of TH2 type T cells HLA class I and APM molecules was detected [64]. Stem-like and failure in eliciting effector functions [64, 65]. The subop- cells expressing ABCB5 and isolated from melanoma were timal expression of HLA class I molecules and APM compo- found to express suboptimal levels of HLA class I molecules nents was also reported in CSCs/CICs isolated from different while they were positive for HLA class II (45). APM compo- type of solid tumors [64–69]. These peculiar observations nents (e.g., LMP2, LMP7 MECL-1, TAP1 and TAP2) detect- suggested that the defective expression of HLA molecules ed through mRNA analyses were found to be expressed in could represent a tool for the identification of CSCs/CICs tumor sphere-models from different solid malignancies, [70]. On the contrary, the side population (SP) cells derived representing a tool for in vitro enrichment of stem-like cells The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing... 137 and for the investigation of micro-metastasis [73]. However, They are categorized into three groups; (i) the HLA class I and class II molecules were down-modulated in overexpressed/self-antigens that are expressed at high these cells as compared to differentiated tumor cells, also fol- levels by tumor cells and detectable, although at lower lowing their pre-treatment in vitro with IFN-γ, highlighting an levels, on normal tissues (e.g., MART-1/Melan-A, impairment of antigen presentation by these cells [64]. It hTERT, EGFR, survivin). (ii) Cancer testis (CT) antigens needs to be considered that this study did not analyze the that are detectable on tumor cells and not on normal cells, expression at protein levels of APM molecules, therefore except for testis and trophoblast (e.g., NY-ESO1, MAGE post-transcriptional mechanism could affect their expression. A3-A4, PRAME, CT83, SSX2). (iii) Neoantigens or mu- Moreover, long term in vitro established cell lines were used tated antigens derived by non-synonymous mutations in to isolate tumor cell spheres while in other studies reporting cancer cells (e.g., MUM-1, CDK4, ME1, ACTN4, HLA- defective expression of APM components, primary CSCs/ A2) [82]. The neoantigens are higher immunogenic com- CICs have been investigated. These examples highlight that pared to differentiation/self TAAs since are tumor specific an overall suboptimal immunogenic potency by CSCs/CICs and do not induce tolerogenic mechanisms in immune resulting in low or impaired susceptibility to T cell mediated cells [83–85]. Neoantigens have been shown to drive im- immune responses (Fig. 1). This represents a mechanisms of mune responses and to mediate efficient T cell recognition evasion by immune responses that is shared with normal stem of tumor cells, leading to cancer eradication in patients cells, that could represent a typical feature of cells with treated with either mutanome based vaccines or adoptive stemness properties [74]. Importantly, the failure in the ex- cell therapy (ACT) with tumor infiltrating lymphocytes pression of HLA molecules by tumor cells was found as one [84–86]. Notably, CSCs/CICs bearing a somatic mutation of the mechanisms of failure of the clinical activity of immune in the CRC-associated “driver” gene SMAD4, could elicit checkpoint blockade agents in cancer patients [75, 76], indi- antigen-specific T cell responses directed to both stemness cating that either CSCs/CICs can display immune evasion and differentiated components of tumor [87]. mechanisms shared by differentiated tumor cells or that in- A transcriptome analysis of the SP cells and main popu- deed the suboptimal expression of HLA molecules of these lation (MP) derived from CRC, breast and lung cancer re- cells and their resistance to T cell recognition can protect these vealed a preferential expression of 18 CT antigens cells from immunotherapy interventions, leading to tumor re- (MAGEA2, MAGEA3, MAGEA4, MAGEA6, MAGEA12, currence or progression. However, the lack of standardization MAGEB2, GAGE1, GAGE8, SPANXA1, SPANXB1, in methods for both, the isolation of cells with stemness prop- SPANXC, XAGE2, SPA17, BORIS, PLU-1, SGY-1, erties and to analyze HLA and APM molecules, represents a TEX15 and CT45A1) in CSCs/CICs [32]. The TAA limitation in providing conclusive results. Nevertheless, de- DNAJB8, that is a member of the heat shock protein tailed analysis to identify the molecular mechanisms that lead (HSP) 40 family, was found to be preferentially expressed to aberrant expression of HLA molecules and APM compo- in renal cell carcinoma (RCC); interestingly this protein nents are warranted. played an important role in the maintenance of The suboptimal expression of HLA class I molecules if CSCs/CICs. DNAJB8-specific immune responses could be associated with detectable NKG2D ligands, can drive the in- detected in a mice model study of DNA vaccination for creased susceptibility of CSCs/CICs to Natural Killer (NK) RCC, rendering this molecule appealing for targeting cells. This phenomenon has been observed in CSCs/CICs CSCs/CICs by the immune system [32, 33](Table 1). from glioma, melanoma, and CRC [68, 77–79]. However, Recently a new antigen, Ankyrin repeat and SOCS box pro- down-modulation of NKG2D ligands on CSCs/CICs has been tein 4 (ASB4), was described as target molecule of CTLs documented, e.g., in GBM patients [64]suggesting thatthe recognizing CSCs/CICs and not the differentiated cellular expression of low levels of NK cell activating ligands can components of the tumor [88]. Suboptimal expression of result in the impairment of anti-CSCs/CICs innate immune TAAs (MART-1, ML-IAP, NY-ESO-1, and MAGE-A) was responses (Fig. 1). The expression profile of molecules acti- reported in melanoma-derived CSCs/CICs (Fig. 1)[89]. vating innate immune responses on CSCs/CICs can be affect- Similar results were obtained in CSCs/CICs isolated from ed by their crosstalk with TME, and thus, by their plasticity GBM and CRC (Fig. 1 and Table 1)[64, 65]. On the other that can influence the fate in vivo of these cells. hand, CD133 CSCs/CICs isolated from melanoma were shown to express either NY-ESO-1 or DEAD/H (Asp-Glu- Tumor Associated Antigens and Adaptive Immune Ala-Asp/His) box polypeptide 3, X-linked (DDX3X) Responses against CSCs/CICs representing target of tumor-specific T cells [90, 91]. Other studies have described the isolation of T lymphocytes rec- Tumor associated antigens (TAAs) can be recognized by ognizing TAAs expressed by CSCs/CICs such as IL-13Rα2, T lymphocytes when exposed on the surface of tumor SOX2 and CD133 in GBM, CEP55 and COA-1 in CRC and cells in the form of peptide/HLA complexes [80, 81]. EpCAM in retinoblastoma (Table 1)[28, 65, 71, 92]. 138 Ravindran S. et al. Innate Immune Responses and their Relationship characterized by elevated pro-inflammatory cytokines, such with CSCs/CICs as IL-12, IL-1β, IL-6, and tumor necrosis factor α (TNF-α), increased expression of HLA class II molecules, generation of Natural killer (NK) cells are the first line of defense against reactive oxygen and nitrogen intermediates and ability to in- cancer development and metastasis. NK cells have been de- duce TH1-type T cell responses [106]. 2. In the presence IL-4, scribed to efficiently recognize and kill in vitro CSCs/CICs iso- IL-10, and IL-13, macrophages can polarize towards M2 phe- lated from CRC, melanoma and glioblastoma [68, 78, 93, 94]. notype. These cells express scavenging, mannose and galac- The efficiency of NK cell-mediated lysis of CSCs/CICs was tose receptors, IL-10, vascular endothelial growth factor dependent on the expression of NCR ligands (NKp30 and (VEGF), matrix metalloproteinases (MMPs) and activation NKp44), NKG2D ligands and when suboptimal or negative ex- of the arginase pathway, leading to pro-tumoral effects pression of HLA class I molecules were found on the surface of [105–107, 109]. The cross-talk between CSCs/CICs and CSCs/CICs (Fig 1)[68, 78, 93–95]. Tallerico et al. found that TAM is orchestrated by STAT3 signaling [102, 103]. Upon CSC/CIC but not their differentiated counterpart of CRC is sus- iper-modulation of STAT3 in TAM, they can promote ceptible to NK cells [77]. Similar results have been reported in stemness, survival and proliferation in cancer cells while the GBM and melanoma, highlighting that the amount of ligands of latest cells can induce the immunosuppressive properties of activatory NK receptors on CSCs/CICs was determinant for ef- TAM, leading to the impairment of cancer immune- ficient innate immune responses [68, 77, 78]. In patients with surveillance [110]. acute myeloid leukemia (AML), the suboptimal expression of Myeloid derived suppressor cells (MDSCs) are immune cells NKG2D ligands has been described as a mecahnisms of escape endowed with suppressive functions that can inhibit the effector by tumor cells from NK cell recognition [96], confirming that functions of immune responses [111]. The frequency of these these molecules can affect the susceptibility of cancer cells to cells either at tumor site or in the circulation has been described innate responses. The observations that NKG2D ligands could as a prognostic factor for patients’ survival as well as of respon- represent as biomarkers for prediction of clinical responses to siveness to immunotherapy [112]. Interestingly, STAT3 can lead immune checkpoint blockade in melanoma highlight that the the differentiation of monocytes towards MDSCs in pancreatic pattern of NKG2D ligands expression by tumor cells can affect tumors [113] regulating also the development of CSCs/CICs the type and efficiency of elicited anti-tumor immune responses [113]. The secretion of pro-inflammatory cytokines and [97]. Therefore, the levels and pattern of expression of NKG2D chemokines by tumor cells can induce the differentiation and ligands by tumor cells, including CSCs/CICs could be a predic- recruitment of immunosuppressive cells that can also contribute tive marker for the choice of the type of immunotherapy to sustain the inflammatory TME and to the interaction and re- interventions. ciprocal influence of CSCs/CICs and their niche [110, 113–115]. Dendritic cells (DCs) are antigen presenting cells (APCs) Another key regulator of the cross-talk between CSCs/CICs and that can activate either innate or adaptive immune responses TAM and DCs is represented by CD47 [116]. This molecule is [98]. In addition, they play an important role in the formation over-expressed by CSCs/CICs of B cell malignancies. The bind- of anti-tumor T- and B cell immunologic memories [99]. ing of this molecule to the signal regulatory protein alpha Immature DCs can capture the tumor-derived antigens by (SIRPα), that mediates phagocytic functions in DCs and macro- phagocytosis or pinocytosis and then migrate to lymphoid phages, has been shown to mediate the impairment of innate organs where they present these TAAs in the form of HLA/ responses [116]. The cross-talk between CSCs/CICs and mye- peptide complexes to T cells, resulting in antigen-specific im- loid cells can affect both the fate and immunological profile of mune responses [100–102]. However, DCs depending on these cells, with implications for their susceptibilities to immune their morphological and phenotypic subtypes can either in- responses. Further studies should be designed to dissect the in- duce anti-tumor immune responses or promote tumor growth teractions of CSCs/CICs with different immune cells, although and progression [103]. The crosstalk of tumor with their TME the major limitation is represented by the lack of in vivo models is a crucial factor which results in the development of cancer to monitor the interaction of these different immune cell popula- [104]. Along this line, it has been described that high extent of tion in the context of TME. expression of the chemokine (C-X-C motif) ligand 1 (CXCL1) by tumor and stromal cells can promote CSCs/ CICs survival and proliferation and attract at tumor site DCs Immunomodulatory Properties of CSCs/CICs with suppressive functions, that could correlate tumor pro- gression and poor survival of patients [104]. The immunological profiling of CSCs/CICs has revealed that Macrophages represent important players for innate im- they share some characteristics with embryonic, hematopoiet- mune responses and can act as APCs similarly to DCs [105]. ic and mesenchymal stem cells displaying immunoregulatory Based on their phenotype and functions they can be distin- functions that render these cells invisible to immune responses guished in two subpopulations: 1. The M1 subtype that are and able to escape from tumor immune responses [4, 74, 117]. The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing... 139 The principle mechanisms governing the immunomodulation inhibition of T cell effector functions (Table 2)[65, 135]. It of CSCs/CICs are described below. has been demonstrated that the over-expression of IL-4 by CRC-derived CSCs/CICs has led to inefficient TCR- mediated proliferation and antigen recognition of CTLs [65]. Cytokines, Chemokines, Growth Factors & Immune Interestingly, the neutralization of this cytokine by mAb could Checkpoint Molecules overcome the T cell mediated anti-tumor impairment and in- duce antigen-specific recognition of both CSCs/CICs and dif- The observations that CSCs/CICs isolated from different tu- ferentiated tumor cells [65]. This study showed that, upon up- mor types can secrete soluble factors, such as Galectin-3, regulation of HLA class I and APM expression through IFN-γ GDF-15, IL-10, IL-13, PGE2 and TGFb, or express treatment of CSCs/CICs, T cells could specifically recognize a immune checkpoint molecules with immunosuppressive func- neoantigen, SMAD4, generated by a non-synonymous muta- tions, have suggested that these cells can regulate the impair- tion bearing stem-like cells and bulk tumor cells [65]. Thus, ment of immune responses as well as regulating a pro-tumoral CTL reactivity against CSCs/CICs and the TAA-specific TME (Table 2)[4, 51, 18–130]. These immunosuppressive immunosurveillance could be improved by the usage of strat- factors have been described to induce the differentiation of egies to correct the low immunogenic profile of these cells. regulatory T cells (Tregs) or MDSCs and M2 macrophages, The immune suppressive profile of CSCs/CICs has been resulting in the impairment of effector functions of innate and also confirmed by evidences describing the expression by adaptive responses [4, 51, 130, 131]. Moreover, pro- these cells of immune checkpoint molecules (e.g. CTLA-4, inflammatory cytokines such as IL-6, IL-8, IL-10 and IL-13, PD-L1, B7-H3 or B7-H4) (Table 2)[4, 34, 64, 65, 136]. released by CSCs/CICs can contribute to maintain an inflam- These observations highlighted the similarities between matory and suppressive TME representing the “niche” sus- CSCs/CICs and normal stem cells in terms of the immune taining cellular stemness (Table 2)[132, 133]. Indoleamine profile [34, 137, 138]. Moreover, altered expression of 2,3-dioxigenase (IDO), that mediates the catabolism of tryp- STAT3 pathway in CSCs/CICs can also affect their immune tophan, has been shown to be expressed by CSCs/CICs, con- suppressive activity through inhibiting T cell proliferation and tributing to the differentiation of Tregs, skewing the cytokine activation, inducing the differentiation of Tregs and triggering profiling of T cells toward TH2 -type and inhibiting the sur- Tcell apoptosis [121]. The observations reported above show vival and proliferation of CTL (Table 2)[133, 134]. that multiple mechanisms and molecular pathways are either In addition, the CSC/CIC-associated expression of IL-4 up-regulated or aberrantly activated in CSCs/CICs resulting in and CD200, through cell-to-cell interaction, lead to the Table 2 Immunomodulatory molecules detected in CSCs/CICs a b Molecule Function Activity in CSCs/CICs Reference IL-4 Cytokine involved in differentiation of Inhibition of TH1 cell mediated immune responses. [65, 118] naïve T cells to Th2. IL-10, IL-13 Anti-inflammatory cytokines Suppression of CTL functions; differentiation of [89, 119] Tregs and MDSCs TGFB Growth factor with potent inhibitory function Tregs differentiation, inhibition of TH1 responses [89, 120] STAT3 Transcription factor with a potential Maintenance and proliferation of CSCs/CICs; differ- [115, 121] anti-inflammatory function entiation of MDSCs, iDCs, M2 GDF15 Growth and differentiation factor Inhibition of anti-tumor immune responses [122] related to cellular stress. Galectin-3 Protein with important role in cell-cell Inhibitor of T cell mediated immune responses [89, 123] adhesion and interactions with the extracellular envi- ronment. IDO Enzyme involved in tryptophan catabolism Suppression of TH1 type immune responses and [34, 51, 64, 65, 123] differentiation of Tregs. CD200 A glycoprotein that regulates myeloid cell activity and Immune suppression and regulation of anti-tumor [124, 125] inhibits macrophage lineages. activity. PD-L1 Ligand of PD-1 and Immune checkpoint molecule Inhibition of CTL immune responses. [64, 65, 126, 127] B7-H3 and Immune checkpoint molecules Immunomodulation of cellular immune responses [34, 64, 65, 128, 129] B7-H4 : function of the molecules listed in the Table : Activity of these molecules when expressed by CSCs/CICs iDC: suppressive dendritic cell; MDSC: Myeloid derived suppressive cells; M2: M2 phenotype of monocytes/macrophages; TH1: T helper type 1; TH2: T helper type 2 140 Ravindran S. et al. their immune suppressive properties, therefore the blockade of high levels of PD-L1 and contain Tregs, M2 like macrophages these signaling through the combination of inhibitory agents as well as exhausted T cells [153]. CSCs/CICs that are con- should be considered in order to rescue the tumor-specific sidered the architects of their own microenvironment [154], as immune responses. well as generated by epithelial-to-mesenchymal transition (EMT) can be potentially responsible for the type of immune MicroRNAs infiltration depending of their pattern of immune profile. Common gene expression patterns have been found in miRNAs are non-coding RNAs regulating at post- normal mammary stem cells and dormant tumor cells transcriptional levels, through complementary binding to tar- from breast cancer suggesting the possible presence get mRNA, the expression of genes [139]. The altered regu- of stem-like cells in dormant tumors [151]. In addition, lation of gene expression in tumor cells can occur by both up- different cell sub-populations could be isolated from re- or down regulation of miRNA [139]. The most common ac- lapsed AML endowed with differential tumorigenic abili- tivity of miRNAs in CSCs/CICs is represented by the control ty depending on their up-regulation of stemness signaling of the expression of either oncogenes (e.g., MiR34a, MiR31 [155]. or MiR205) or tumor suppressor genes [139]. The aberrant A better understand of the relationship between stemness expression of few miRNAs, such as miRNAs 451and 199b- properties, immunological profile of CSCs/CICs and tumor 5p, has been shown to affect stem-like cell properties isolated dormancy will provide insights on the mechanisms of thera- from different type of tumors (e.g., GBM, breast cancer and peutic resistance of these cells and will allow to identify strat- medulloblastoma) [139–143]. Of note, miRNAs displaying egies for complete tumor eradication. regulatory activity on immune-related genes (e.g, miRNA- 199a that can regulate the IFN-mediated responses) can play a role in the differentiation of mammalian CSCs/CICs [144]. Immunological Targeting of CSCs/CICs The level of miRNA-124, through regulating the expression of STAT3, can affect the efficiency of anti-CSC/CIC T cell Cancer Vaccines responses in GBM [145]. Along this line, miR203 and miR92 can control the stemness and immunological profiles The recognition of TAAs expressed by CSCs/CICs by T cells of melanoma cells [146, 147]. have been documented (see Table 1). These in vitro or in vivo models were based on the usage of TAAs that represented sources of antigens for the therapeutic administration of can- Immune Evasion and Tumor Dormancy cer vaccines in cancer patients [81]. However, the principle limiting factor of the clinical efficacy of this strategy is repre- Tumor dormancy is represented by quiescent cells that can sented by the usage of “self”/tolerogenic TAAs, shared with remain occult and undetectable by regular diagnostic methods normal tissues [81]. The low or negative expression of these for long intervals of time, even after initial clinical responses categories of antigens and of CT-TAAs by CSCs/CICs can to therapies [148]. Quiescence of cells is the ability to exit cell represent an additional reason of failure of high rate and long cycle and remain in G0 phase until permissive environmental duration of clinical responses observed in cancer patients condition will lead to enter back into the cycling phase. This is treated with cancer vaccines [4, 34]. In addition, the sub- considered one of the principle mechanisms underlying tumor optimal levels of HLA class I molecules and APM by stem- dormancy. CSCs/CICs display the ability to cycle between like cells can drive the failure in targeting CSCs/CICs by quiescence and proliferation and together with their resistance cancer vaccines leading to the development of tumor dorman- to therapies represent the link between these cells and tumor cy and tumor recurrence, although the observance of initial dormancy [2, 5, 8, 9, 149–151]. Furthermore, the immune clinical efficacy of these therapeutic interventions [4, 34]. suppressive mechanisms associated with CSCs/CICs can or- DC-based vaccines, exploiting these cells as APC to pres- chestrate the evasion of these cells from immune recognition ent TAAs to T cell-mediated responses, represent also a ther- and immunosurveillance, and could be considered additional apeutic strategy for cancer patients showing encouraging clin- factors responsible of tumor dormancy [152]. ical activity [102, 156–162]. DCs loaded with either CSC/ An important mechanisms of immune-surveillance is the CIC-lysates or mRNA isolated from these cells represented homing of immune cells to the tumor site, which ultimately source of antigens for vaccination in the context of Phase I/ form the immune infiltrate. Tumors arising from epithelial II clinical trials of GBM patients [163, 164]. These studies breast cancer are known to possess high levels MHC class I provided proof of principle of improved overall survival of molecules and of infiltrating T effector cells and M1 macro- cancer patients treated with CSC/CIC targeted immunothera- phages. The immune infiltrate from mesenchymal like breast py [163–165]. Immune responses, with, in some cases in- cancer tumors exhibit low levels of MHC class I molecules, creased frequency of circulating NK cells, were detected in The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing... patients showing clinical benefit from these treatments [163, GBM [34, 64, 65, 177–179]. ,CSCs/CICs could theoretically 165]. Of note, these therapeutic interventions could overcome be targeted in vivo by immune checkpoint blockade agents, the failure of CSCs/CICs in expressing efficient levels of HLA enhancing the clinical efficacy of cancer vaccines, as demon- class I molecules and in presenting TAAs to T cells, strated in a mouse model [180]. However, recent reports de- documenting for the first time, that cancer vaccine, if eliciting scribing that clinical failure of these therapies was associated NK cell-mediated responses, could target stem-like with defective expression of HLA class I molecules by tumor tumor cells [158, 163–165]. Tumor cell clones expressing cells [75, 76], suggest that these cells might evade from the immunogenic neoantigens can undergo immune selection ICB-mediated unleash of immune responses (Fig. 2). due to the recognition and elimination by T lymphocytes, leading to the survival of tumor cell clones not expressing Adoptive Cell Therapy strong immunogenic antigens and maintaining the expression of low immunogenic TAAs [166] (and see https://www. ACT is represented by the isolation of T lymphocytes from biorxiv.org/content/10.1101/536433v1). This process is also cancer patients, their ex vivo expansion, and the infusion back associated with immune evasions mechanisms developed by into patients [181–183]. In addition, T lymphocytes both tumor cells and TME [166]. engineered to express TCR with high affinity for a cognate In some tumors, the decrease in antigen presentation is a TAA could be exploited for ACT studies [183]. Highly en- result of epigenetic silencing of the genes involved in antigen couraging and sustained responses mediated by adoptively presentation machinery. The usage of demethylating agents transferred TCR targeting the TAA NY-ESO-1 have recently such as 5-Aza-2′-deoxycytidine to reduce the methylation of been reported in different tumor types, such as breast cancer genes involved in antigen presentation, is a potential strategy and myeloma [86, 184]. Nevertheless, the antigen choice is to increase the antigen presentation in these CSC/CICs. The highly relevant to prevent severe toxicities due to “off-target” effect of demethylation has been shown in CSCs/CICs from cross-reaction with normal tissues sharing the same antigens breast cancer, where it resulted in high expression levels of or expressing molecules mimicking the TAAs [182]. TAP1, which is involved in antigen presentation [167]. Neoantigens have been described as candidate TAAs effi- In addition, increased antigen presentation also improves ciently recognized by T cells that can be exploited for ACT of the potential of discovering novel antigens, which can then be cancer patients and, interestingly, CTL targeting these anti- helpful in development of new anti-cancer vaccines (Fig. 2). gens could be isolated from tumor infiltrating lymphocytes (TILs) of melanoma and other type of malignant lesions Immune Checkpoint Blockade [183, 185, 186]. Nevertheless, ACT to target neoantigens can represent a promising approach for treatment of cancer Immune checkpoints, including CTLA-4, PD-1 and PD-L1, patients upon assessment of HLA expression by both CSCs/ are important physiological regulators of innate and adaptive CICs and differentiated tumor cells and, in case of suboptimal immune responses [168]. Biological inhibitory agents have levels of expression, the achievement of their up-regulation by been clinically developed, revealing striking therapeutically pre-treatment with immunomodulating agents [4, 34, 51]. success [169–175]. However, a significant proportion of can- T cell can be genetically modified to express a chimeric cer patients failed to benefit from these therapies. antigen receptor (CAR) that is composed of epitope-specific The effectiveness of immune checkpoint blockade domains isolated from mAbs linked to T cell-derived (ICB) is largely dependent on the tumor microenviron- activatory/costimulatory molecules [182, 187–189]. CAR-T ment [176]. Tumors such as melanoma, bladder cancer cells can recognize TAAs independently on the expression and non-small cell lung cancer (NSCLC) can be charac- of HLA molecules and APM components [189]. CAR-T cell terized as “hot” tumors due to their inflamed TME, high therapy for some subgroups of hematological malignancies levels of and neo-antigen expression and of T cell infil- represent the salvage intervention leading to stable clinical tration and detection of PD-L1. These tumors have been responses and improved overall survival of patients refractory reported to be associated with higher frequency of suscep- to standard therapies or with recurrences [187, 189–192]. The tibility to immune checkpoint treatments. On the other usage of CAR-T cell therapy for solid tumors is currently hand, prostate cancer is considered to be a “cold” tumor, under investigation, showing encouraging results in cancer due to minimal level of T cell infiltration, and limited patients with aggressive tumor types, including malignant me- response to single agent checkpoint inhibition [176]. sothelioma, pancreatic cancer and GBM [193–197]. Expression of immune checkpoint molecules has been ob- CAR-T cells targeting TAAs, such as CD133, EGFRvIII, served in CSCs/CICs from different histological origins [51]. EpCAM, CSPG4 and B7-H3, expressed by different type of PD-L1 expression was detected at high levels in CSCs/CICs solid tumors, including CSC/CIC components, have been de- isolated from primary human head and neck squamous cell veloped in pre-clinical studies [196–201]. These studies have shown that the targeting of TAAs that are expressed only by carcinoma (HNSCC), gastric and breast cancer, CRC and Ravindran S. et al. tumor cells, including CSCs/CICs, and not by normal cells, human immune system is desirable to allow the monitoring could provide the rational for safe and efficient clinical devel- of the interaction of CSCs/CICs with TME. opment of CAR-T cells therapy for these tumors(Fig. 2)[200, The targeting of CSCs/CICs by immunotherapy could re- 202]. Moreover, the combination of CAR-T cells targeting sult in the complete tumor eradication and stable clinical re- dual TAAs, EGFRvIII and CD133, has been used for the sponses in cancer patients. This goal could be achieved by the successful therapeutic treatment of a patient with advanced design of combination of strategies based on innate and/or cholangiocarcinoma [203]. antigen-specific T cell responses with immunoregulatory CAR-T cells targeting NKG2D ligands on CSCs/CICs agents that can render CSCs/CICs susceptible to cell- have been investigated and tested both in vitro and in vivo mediated immunosurveillance. In cases where epigenetic fac- [204, 205]. CSCs/CICs from glioblastoma expressing detect- tors are responsible for low antigen presentation, the usage of able NKG2D ligands could be efficiently targeted by CAR- T demethylating agents could represent a potential strategy to cells. These tools showed to efficintly eliminate also xenograft overcome the low expression of HLA molecules. tumors [205]. Nevertheless, a major limitation associated with Molecular approaches dissecting the fate of CSCs/CICs the use of these CAR-T cells is represented by the variability within tumor tissues will allow to develop immune-based pre- of the levels of NKG2D ligands on the surface of CSCs/CICs, cision medicine approaches and to identify biomarkers predic- depending on their origin and methodology used for their tive of patients’ responsiveness to therapies. ex vivo isolation. Acknowledgements This study was supported by Qatar National Nevertheless, further investigations aimed at a comprehen- Research Fund, grant no. NPRP10-0129-170277. sive genomic and immunological characterization CSCs/CICs are warranted to implement the efficiency and safety of ACT Funding Information Open Access funding provided by the Qatar strategies. National Library. Compliance with Ethical Standards Conclusions Conflict of Interest All the authors have no conflict of interest to disclose. Recent advances in the genomic, molecular and immunolog- ical profiling of CSCs/CICs have contributed to the identifi- Open Access This article is distributed under the terms of the Creative cation of dysregulated molecular pathways that orchestrate Commons Attribution 4.0 International License (http:// stem-like cancer cells and their interaction with TME. The creativecommons.org/licenses/by/4.0/), which permits unrestricted use, heterogeneity and plasticity of these cells and the mutual ef- distribution, and reproduction in any medium, provided you give appro- priate credit to the original author(s) and the source, provide a link to the fect of TME and CSCs/CICs on the resulting anti-tumoral or Creative Commons license, and indicate if changes were made. pro-tumoral environment, represent the principle limitations in predicting the fate of these cells and their role in cancer patients’ outcome. In addition, these cells have been identified as key players in therapeutic resistance of tumors and in the References development of their dormancy. The down-modulation of HLA molecules and NK activatory ligands on CSCs/CICs, 1. Shackleton M, Quintana E, Fearon ER, Morrison SJ (2009) through decreasing their susceptibility to T or NK cell Heterogeneity in cancer: cancer stem cells versus clonal evolution. targeting, might represent one of the principle factors leading Cell. 138(5):822–829 2. Clarke MF, Dick JE, Dirks PB et al (2006) Cancer stem cells– to resistance to immunotherapy. Although, the mechanisms perspectives on current status and future directions: AACR work- regulating the levels of HLA molecules and NKG2D ligands shop on cancer stem cells. Cancer Res 66(19):9339–9344 on CSCs/CICs are yet to be dissected. Due to the complexity 3. Li Y, Laterra J (2012) Cancer stem cells: distinct entities or dy- of the cross-talk between CSCs/CICs and TME and the high namically regulated phenotypes? Cancer Res 72(3):576–580 plasticity of these cells, it is difficult to predict what type of 4. Maccalli C, De Maria R (2015) Cancer stem cells: perspectives for therapeutic targeting. Cancer Immunol Immunother 64(1):91–97 immune cells could play a relevant role in targeting 5. Reya T, Morrison SJ, Clarke MF, Weissman IL (2001) Stem cells, CSCs/CICs. Multifactorial investigations, including the im- cancer, and cancer stem cells. Nature. 414(6859):105–111 munological profile, immunomodulating molecules, the inter- 6. Wicha MS, Liu S, Dontu G (2006) Cancer stem cells: an old idea– action with TME and the type of immune cell infiltration will a paradigm shift. Cancer Res 66(4):1883–1890 discussion 1895- allow to provide insights. Nevertheless, the available tools to 7. Tang DG (2012) Understanding cancer stem cell heterogeneity isolate and characterize CSCs/CICs, such as spheroids, immu- and plasticity. Cell Res 22(3):457–472 nodeficient mice, antigenic profile, are unsatisfactory to dis- 8. Eyler CE, Rich JN (2008) Survival of the fittest: cancer stem cells sect the cross-talk of these cells with TME. Moreover, the in therapeutic resistance and angiogenesis. J Clin Oncol 26(17): development of pre-clinical in vivo models engrafted with 2839–2845 The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing... 143 9. Hadjimichael C, Chanoumidou K, Papadopoulou N, Arampatzi P, for immunotherapy in vitro and in vivo. J Biol Chem 286(47): Papamatheakis J, Kretsovali A (2015) Common stemness regula- 40548–40555 tors of embryonic and cancer stem cells. World J Stem Cells 7(9): 31. Schmitz M, Temme A, Senner V et al (2007) Identification of 1150–1184 SOX2 as a novel glioma-associated antigen and potential target 10. Maugeri-Sacca M, Vigneri P, De Maria R (2011) Cancer stem cells for T cell-based immunotherapy. Br J Cancer 96(8):1293–1301 and chemosensitivity. Clin Cancer Res 17(15):4942–4947 32. Yamada R, Takahashi A, Torigoe T, Morita R, Tamura Y, 11. Soltanian S, Matin MM (2011) Cancer stem cells and cancer ther- Tsukahara T, Kanaseki T, Kubo T, Watarai K, Kondo T, apy. Tumour Biol 32(3):425–440 Hirohashi Y, Sato N (2013) Preferential expression of cancer/ testis genes in cancer stem-like cells: proposal of a novel sub- 12. Steinbichler TB, Dudas J, Skvortsov S, Ganswindt U, Riechelmann H, Skvortsova II. (2018) Therapy resistance medi- category, cancer/testis/stem gene. Tissue Antigens 81(6):428–434 ated by cancer stem cells. Semin Cancer Biol 33. Nishizawa S, Hirohashi Y, Torigoe T, Takahashi A, Tamura Y, Mori T, Kanaseki T, Kamiguchi K, Asanuma H, Morita R, 13. Ricci-Vitiani L, Fabrizi E, Palio E, De Maria R (2009) Colon cancer stem cells. J Mol Med (Berl) 87(11):1097–1104 Sokolovskaya A, Matsuzaki J, Yamada R, Fujii R, Kampinga HH, Kondo T, Hasegawa T, Hara I, Sato N (2012) HSP 14. Vezzoni L, Parmiani G (2008) Limitations of the cancer stem cell DNAJB8 controls tumor-initiating ability in renal cancer stem- theory. Cytotechnology. 58(1):3–9 like cells. Cancer Res 72(11):2844–2854 15. Morrison BJ, Steel JC, Morris JC (2018) Reduction of MHC-I 34. Maccalli C, Rasul KI, Elawad M, Ferrone S (2018) The role of expression limits T-lymphocyte-mediated killing of Cancer- initiating cells. BMC Cancer 18(1):469 cancer stem cells in the modulation of anti-tumor immune re- sponses. Semin Cancer Biol. 16. Maccalli C, Volonte A, Cimminiello C, Parmiani G (2014) 35. Clevers H (2011) The cancer stem cell: premises, promises and Immunology of cancer stem cells in solid tumours. A review. Eur J Cancer 50(3):649–655 challenges. Nat Med 17(3):313–319 36. Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, di Virgilio A, 17. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres- Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE (1994) A Conticello C, Ruco L, Peschle C, de Maria R (2008) Identification and expansion of the tumorigenic lung cancer stem cell popula- cell initiating human acute myeloid leukaemia after transplanta- tion. Cell Death Differ 15(3):504–514 tion into SCID mice. Nature. 367(6464):645–648 37. Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, 18. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke Peschle C, de Maria R (2007) Identification and expansion of MF (2003) Prospective identification of tumorigenic breast cancer human colon-cancer-initiating cells. Nature. 445(7123):111–115 cells. Proc Natl Acad Sci U S A 100(7):3983–3988 38. Todaro M, Gaggianesi M, Catalano V, Benfante A, Iovino F, 19. Clarke MF, Becker MW (2006) Stem cells: the real culprits in Biffoni M, Apuzzo T, Sperduti I, Volpe S, Cocorullo G, Gulotta cancer? Sci Am 295(1):52–59 G, Dieli F, de Maria R, Stassi G (2014) CD44v6 is a marker of 20. Singh SK, Clarke ID, Hide T, Dirks PB (2004) Cancer stem cells constitutive and reprogrammed cancer stem cells driving colon in nervous system tumors. Oncogene. 23(43):7267–7273 cancer metastasis. Cell Stem Cell 14(3):342–356 21. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, 39. Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM, Henkelman RM, Cusimano MD, Dirks PB (2004) Identification Morrison SJ (2008) Efficient tumour formation by single human of human brain tumour initiating cells. Nature. 432(7015):396– 401 melanoma cells. Nature. 456(7222):593–598 40. Roesch A, Fukunaga-Kalabis M, Schmidt EC, Zabierowski SE, 22. Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha Brafford PA, Vultur A, Basu D, Gimotty P, Vogt T, Herlyn M M, Clarke MF, Simeone DM (2007) Identification of pancreatic (2010) A temporarily distinct subpopulation of slow-cycling mel- cancer stem cells. Cancer Res 67(3):1030–1037 anoma cells is required for continuous tumor growth. Cell. 141(4): 23. Ricci-Vitiani L, Pagliuca A, Palio E, Zeuner A, De Maria R (2008) 583–594 Colon cancer stem cells. Gut. 57(4):538–548 41. Prieto-Vila M, Takahashi RU, Usuba W, Kohama I, Ochiya T 24. Visus C, Wang Y, Lozano-Leon A, Ferris RL, Silver S, (2017) Drug resistance driven by cancer stem cells and their niche. Szczepanski MJ, Brand RE, Ferrone CR, Whiteside TL, Ferrone Int J Mol Sci 18(12) S, DeLeo A, Wang X (2011) Targeting ALDH(bright) human 42. Fuchs E, Tumbar T, Guasch G (2004) Socializing with the neigh- carcinoma-initiating cells with ALDH1A1-specific CD8(+) T bors: stem cells and their niche. Cell. 116(6):769–778 cells. Clin Cancer Res 17(19):6174–6184 43. Xie T, Li L (2007) Stem cells and their niche: an inseparable 25. Brown CE, Starr R, Martinez C, Aguilar B, D'Apuzzo M, Todorov relationship. Development. 134(11):2001–2006 I, Shih CC, Badie B, Hudecek M, Riddell SR, Jensen MC (2009) 44. Ungefroren H, Sebens S, Seidl D, Lehnert H, Hass R (2011) Recognition and killing of brain tumor stem-like initiating cells by Interaction of tumor cells with the microenvironment. Cell CD8+ cytolytic T cells. Cancer Res 69(23):8886–8893 Commun Signal 9:18 26. Glumac PM, LeBeau AM (2018) The role of CD133 in cancer: a 45. Whiteside TL (2008) The tumor microenvironment and its role in concise review. Clin Transl Med 7(1):18 promoting tumor growth. Oncogene. 27(45):5904–5912 27. Zinzi L, Contino M, Cantore M, Capparelli E, Leopoldo M, 46. Cabarcas SM, Mathews LA, Farrar WL (2011) The cancer stem Colabufo NA (2014) ABC transporters in CSCs membranes as a cell niche–there goes the neighborhood? Int J Cancer 129(10): novel target for treating tumor relapse. Front Pharmacol 5:163 28. Mitra M, Kandalam M, Harilal A, Verma RS, Krishnan UM, 2315–2327 47. Cully M (2018) Tumour microenvironment: fibroblast subtype Swaminathan S, Krishnakumar S (2012) EpCAM is a putative stem marker in retinoblastoma and an effective target for T-cell- provides niche for cancer stem cells. Nat Rev Cancer 18(3):136 mediated immunotherapy. Mol Vis 18:290–308 48. Cully M (2018) Cancer: fibroblast subtype provides niche for 29. Choi D, Lee HW, Hur KY et al (2009) Cancer stem cell markers cancer stem cells. Nat Rev Drug Discov 17(3):163 CD133 and CD24 correlate with invasiveness and differentiation 49. Malanchi I, Santamaria-Martinez A, Susanto E et al (2012) in colorectal adenocarcinoma. World J Gastroenterol 15(18): Interactions between cancer stem cells and their niche govern 2258–2264 metastatic colonization. Nature. 481(7379):85–89 30. Shapira S, Kazanov D, Weisblatt S, Starr A, Arber N, Kraus S 50. Seidel S, Garvalov BK, Wirta V et al (2010) A hypoxic niche (2011) The CD24 protein inducible expression system is an ideal regulates glioblastoma stem cells through hypoxia inducible factor tool to explore the potential of CD24 as an oncogene and a target 2 alpha. Brain. 133(Pt 4):983–995 Ravindran S. et al. 51. Maccalli C, Parmiani G, Ferrone S (2017) Immunomodulating human melanoma cells with characteristics of cancer stem cells. and Immunoresistance properties of Cancer-initiating cells: impli- Int Immunol 21(7):793–801 cations for the clinical success of immunotherapy. Immunol 69. Zhang D, Tang DG, Rycaj K (2018) Cancer stem cells: regulation Investig 46(3):221–238 programs, immunological properties and immunotherapy. Semin 52. Easwaran H, Tsai HC, Baylin SB (2014) Cancer epigenetics: tu- Cancer Biol 52(Pt 2):94–106 mor heterogeneity, plasticity of stem-like states, and drug resis- 70. Grau JJ, Mesia R, de la Iglesia-Vicente M et al (2016) Enrichment tance. Mol Cell 54(5):716–727 of cells with Cancer stem cell-like markers in relapses of Chemoresistant patients with locally advanced head and neck 53. Dick JE (2008) Stem cell concepts renew cancer research. Blood. squamous cell carcinoma. Oncology. 90(5):267–272 112(13):4793–4807 71. Inoda S, Hirohashi Y, Torigoe T, Morita R, Takahashi A, Asanuma 54. Bar EE, Chaudhry A, Lin A, Fan X, Schreck K, Matsui W, H, Nakatsugawa M, Nishizawa S, Tamura Y, Tsuruma T, Terui T, Piccirillo S, Vescovi AL, DiMeco F, Olivi A, Eberhart CG Kondo T, Ishitani K, Hasegawa T, Hirata K, Sato N (2011) (2007) Cyclopamine-mediated hedgehog pathway inhibition de- Cytotoxic T lymphocytes efficiently recognize human colon can- pletes stem-like cancer cells in glioblastoma. Stem Cells 25(10): cer stem-like cells. Am J Pathol 178(4):1805–1813 2524–2533 72. Wei J, Barr J, Kong LY, Wang Y, Wu A, Sharma AK, Gumin J, 55. Sakariassen PO, Prestegarden L, Wang J, Skaftnesmo KO, Henry V, Colman H, Sawaya R, Lang FF, Heimberger AB (2010) Mahesparan R, Molthoff C, Sminia P, Sundlisaeter E, Misra A, Glioma-associated cancer-initiating cells induce immunosuppres- Tysnes BB, Chekenya M, Peters H, Lende G, Kalland KH, Øyan sion. Clin Cancer Res 16(2):461–473 AM, Petersen K, Jonassen I, van der Kogel A, Feuerstein BG, 73. Busse A, Letsch A, Fusi A, Nonnenmacher A, Stather D, Terzis AJ, Bjerkvig R, Enger PØ (2006) Angiogenesis- Ochsenreither S, Regenbrecht CR, Keilholz U (2013) independent tumor growth mediated by stem-like cancer cells. Characterization of small spheres derived from various solid tu- Proc Natl Acad Sci U S A 103(44):16466–16471 mor cell lines: are they suitable targets for T cells? Clin Exp 56. Eppert K, Takenaka K, Lechman ER, Waldron L, Nilsson B, van Metastasis 30(6):781–791 Galen P, Metzeler KH, Poeppl A, Ling V, Beyene J, Canty AJ, 74. Ichiryu N, Fairchild PJ (2013) Immune privilege of stem cells. Danska JS, Bohlander SK, Buske C, Minden MD, Golub TR, Methods Mol Biol 1029:1–16 Jurisica I, Ebert BL, Dick JE (2011) Stem cell gene expression 75. Gettinger S, Choi J, Hastings K, Truini A, Datar I, Sowell R, programs influence clinical outcome in human leukemia. Nat Med Wurtz A, Dong W, Cai G, Melnick MA, du VY, Schlessinger J, 17(9):1086–1093 Goldberg SB, Chiang A, Sanmamed MF, Melero I, Agorreta J, 57. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Montuenga LM, Lifton R, Ferrone S, Kavathas P, Rimm DL, Dewhirst MW, Bigner DD, Rich JN (2006) Glioma stem cells Kaech SM, Schalper K, Herbst RS, Politi K (2017) Impaired promote radioresistance by preferential activation of the DNA HLA class I antigen processing and presentation as a mechanism damage response. Nature. 444(7120):756–760 of acquired resistance to immune checkpoint inhibitors in lung 58. Dean M, Fojo T, Bates S (2005) Tumour stem cells and drug Cancer. Cancer Discov 7(12):1420–1435 resistance. Nat Rev Cancer 5(4):275–284 76. Chowell D, Morris LGT, Grigg CM et al (2018) Patient HLA class 59. Diehn M, Clarke MF (2006) Cancer stem cells and radiotherapy: I genotype influences cancer response to checkpoint blockade new insights into tumor radioresistance. J Natl Cancer Inst 98(24): immunotherapy. Science. 359(6375):582–587 1755–1757 77. Tallerico R, Todaro M, Di Franco S et al (2013) Human NK cells 60. Francipane MG, Alea MP, Lombardo Y, Todaro M, Medema JP, selective targeting of colon cancer-initiating cells: a role for natural Stassi G (2008) Crucial role of interleukin-4 in the survival of cytotoxicity receptors and MHC class I molecules. J Immunol colon cancer stem cells. Cancer Res 68(11):4022–4025 190(5):2381–2390 61. Frank NY, Schatton T, Frank MH (2010) The therapeutic promise 78. Castriconi R, Daga A, Dondero A et al (2009) NK cells recognize of the cancer stem cell concept. J Clin Invest 120(1):41–50 and kill human glioblastoma cells with stem cell-like properties. J 62. Li X, Lewis MT, Huang J, Gutierrez C, Osborne CK, Wu MF, Immunol 182(6):3530–3539 Hilsenbeck SG, Pavlick A, Zhang X, Chamness GC, Wong H, 79. Tseng HC, Arasteh A, Paranjpe A et al (2010) Increased lysis of Rosen J, Chang JC (2008) Intrinsic resistance of tumorigenic stem cells but not their differentiated cells by natural killer cells; breast cancer cells to chemotherapy. J Natl Cancer Inst 100(9): de-differentiation or reprogramming activates NK cells. PLoS One 672–679 5(7):e11590 63. Wilson BJ, Schatton T, Zhan Q, Gasser M, Ma J, Saab KR, 80. Maccalli C, Rasul KI, Elawad M, Ferrone S (2018) The role of Schanche R, Waaga-Gasser AM, Gold JS, Huang Q, Murphy cancer stem cells in the modulation of anti-tumor immune re- GF, Frank MH, Frank NY (2011) ABCB5 identifies a therapy- sponses. Semin Cancer Biol 53:189–200 refractory tumor cell population in colorectal cancer patients. 81. Parmiani G, Russo V, Maccalli C, Parolini D, Rizzo N, Maio M Cancer Res 71(15):5307–5316 (2014) Peptide-based vaccines for cancer therapy. Hum Vaccin 64. Di Tomaso T, Mazzoleni S, Wang E et al (2010) Immunother 10(11):3175–3178 Immunobiological characterization of cancer stem cells isolated 82. Lu YC, Robbins PF (2016) Cancer immunotherapy targeting from glioblastoma patients. Clin Cancer Res 16(3):800–813 neoantigens. Semin Immunol 28(1):22–27 65. Volonte A, Di Tomaso T, Spinelli M et al (2014) Cancer-initiating 83. Forshew T, Murtaza M, Parkinson C et al (2012) Noninvasive cells from colorectal cancer patients escape from T cell-mediated identification and monitoring of cancer mutations by targeted deep immunosurveillance in vitro through membrane-bound IL-4. J sequencing of plasma DNA. Sci Transl Med 4(136):136ra168 Immunol 192(1):523–532 84. Robbins PF (2017) Tumor-infiltrating lymphocyte therapy and 66. Catalano V (2015) Resistance of cancer stem cells to cell- medi- Neoantigens. Cancer J 23(2):138–143 ated immune responses. Vol 7: Springer 85. Sahin U, Derhovanessian E, Miller M et al (2017) Personalized 67. Codony-Servat J, Rosell R (2015) Cancer stem cells and RNA mutanome vaccines mobilize poly-specific therapeutic im- immunoresistance: clinical implications and solutions. Transl munity against cancer. Nature. 547(7662):222–226 Lung Cancer Res 4(6):689–703 86. Zacharakis N, Chinnasamy H, Black M et al (2018) Immune rec- 68. Pietra G, Manzini C, Vitale M, Balsamo M, Ognio E, Boitano M, ognition of somatic mutations leading to complete durable regres- Queirolo P, Moretta L, Mingari MC (2009) Natural killer cells kill sion in metastatic breast cancer. Nat Med 24(6):724–730 The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing... 145 87. Mennonna D, Maccalli C, Romano MC et al (2017) T cell 106. Sica A, Mantovani A (2012) Macrophage plasticity and polariza- neoepitope discovery in colorectal cancer by high throughput pro- tion: in vivo veritas. J Clin Invest 122(3):787–795 filing of somatic mutations in expressed genes. Gut. 66(3):454– 107. Gordon S, Martinez FO (2010) Alternative activation of macro- phages: mechanism and functions. Immunity. 32(5):593–604 108. Jinushi M, Chiba S, Yoshiyama H, Masutomi K, Kinoshita I, 88. Miyamoto S, Kochin V, Kanaseki T et al (2018) The antigen Dosaka-Akita H, Yagita H, Takaoka A, Tahara H (2011) Tumor- ASB4 on Cancer stem cells serves as a target for CTL immuno- therapy of colorectal Cancer. Cancer Immunol Res. associated macrophages regulate tumorigenicity and anticancer drug responses of cancer stem/initiating cells. Proc Natl Acad 89. Schatton T, Schutte U, Frank NYet al (2010) Modulation of T-cell Sci U S A 108(30):12425–12430 activation by malignant melanoma initiating cells. Cancer Res 109. Mantovani A, Sica A, Locati M (2005) Macrophage polarization 70(2):697–708 comes of age. Immunity. 23(4):344–346 90. Gedye C, Quirk J, Browning J, Svobodová S, John T, Sluka P, 110. Mitchem JB, Brennan DJ, Knolhoff BL, Belt BA, Zhu Y, Sanford Dunbar PR, Corbeil D, Cebon J, Davis ID (2009) Cancer/testis DE, Belaygorod L, Carpenter D, Collins L, Piwnica-Worms D, antigens can be immunological targets in clonogenic CD133+ Hewitt S, Udupi GM, Gallagher WM, Wegner C, West BL, melanoma cells. Cancer Immunol Immunother 58(10):1635–1646 Wang-Gillam A, Goedegebuure P, Linehan DC, DeNardo D 91. Koshio J, Kagamu H, Nozaki K, Saida Y, Tanaka T, Shoji S, (2013) Targeting tumor-infiltrating macrophages decreases Igarashi N, Miura S, Okajima M, Watanabe S, Yoshizawa H, tumor-initiating cells, relieves immunosuppression, and improves Narita I (2013) DEAD/H (asp-Glu-Ala-asp/his) box polypeptide chemotherapeutic responses. Cancer Res 73(3):1128–1141 3, X-linked is an immunogenic target of cancer stem cells. Cancer 111. Lindau D, Gielen P, Kroesen M, Wesseling P, Adema GJ (2013) Immunol Immunother 62(10):1619–1628 The immunosuppressive tumour network: myeloid-derived sup- 92. Brown CE, Starr R, Aguilar B, Shami AF, Martinez C, D'Apuzzo pressor cells, regulatory T cells and natural killer T cells. M, Barish ME, Forman SJ, Jensen MC (2012) Stem-like tumor- Immunology. 138(2):105–115 initiating cells isolated from IL13Ralpha2 expressing gliomas are 112. Filipazzi P, Huber V, Rivoltini L (2012) Phenotype, function and targeted and killed by IL13-zetakine-redirected T cells. Clin clinical implications of myeloid-derived suppressor cells in cancer Cancer Res 18(8):2199–2209 patients. Cancer Immunol Immunother 61(2):255–263 93. Tallerico R, Garofalo C, Carbone E (2016) A new biological fea- 113. Panni RZ, Sanford DE, Belt BA et al (2014) Tumor-induced ture of natural killer cells: the recognition of solid tumor-derived STAT3 activation in monocytic myeloid-derived suppressor cells Cancer stem cells. Front Immunol 7:179 enhances stemness and mesenchymal properties in human pancre- 94. Parmiani G (2016) Melanoma cancer stem cells: markers and atic Cancer. Cancer Immunol Immunother 63(5):513–528 functions. Cancers (Basel) 8(3) 114. Sherry MM, Reeves A, Wu JK, Cochran BH (2009) STAT3 is 95. Ames E, Canter RJ, Grossenbacher SK et al (2015) NK cells required for proliferation and maintenance of multipotency in glio- preferentially target tumor cells with a Cancer stem cell pheno- blastoma stem cells. Stem Cells 27(10):2383–2392 type. J Immunol 195(8):4010–4019 115. Zhou J, Wulfkuhle J, Zhang H, Gu P, Yang Y, Deng J, Margolick 96. Paczulla AM, Rothfelder K, Raffel S et al (2019) Absence of JB, Liotta LA, Petricoin E 3rd, Zhang Y (2007) Activation of the NKG2D ligands defines leukaemia stem cells and mediates their PTEN/mTOR/STAT3 pathway in breast cancer stem-like cells is immune evasion. Nature. 572(7768):254–259 required for viability and maintenance. Proc Natl Acad Sci U S A 97. Maccalli C, Giannarelli D, Chiarucci C, Cutaia O, Giacobini G, 104(41):16158–16163 Hendrickx W, Amato G, Annesi D, Bedognetti D, Altomonte M, 116. Majeti R, Chao MP, Alizadeh AA, Pang WW, Jaiswal S, Gibbs Danielli R, Calabrò L, di Giacomo AM, Marincola FM, Parmiani KD Jr, van Rooijen N, Weissman IL (2009) CD47 is an adverse G, Maio M (2017) Soluble NKG2D ligands are biomarkers asso- prognostic factor and therapeutic antibody target on human acute ciated with the clinical outcome to immune checkpoint blockade myeloid leukemia stem cells. Cell. 138(2):286–299 therapy of metastatic melanoma patients. Oncoimmunology. 6(7): 117. Ankrum JA, Ong JF, Karp JM (2014) Mesenchymal stem cells: e1323618 immune evasive, not immune privileged. Nat Biotechnol 32(3): 98. Cella M, Sallusto F, Lanzavecchia A (1997) Origin, maturation 252–260 and antigen presenting function of dendritic cells. Curr Opin 118. Brown MA, Hural J (2017) Functions of IL-4 and control of its Immunol 9(1):10–16 expression. Crit Rev Immunol 37(2–6):181–212 99. Keller AM, Xiao Y, Peperzak V, Naik SH, Borst J (2009) 119. Terabe M, Park JM, Berzofsky JA (2004) Role of IL-13 in regu- Costimulatory ligand CD70 allows induction of CD8+ T-cell im- lation of anti-tumor immunity and tumor growth. Cancer Immunol munity by immature dendritic cells in a vaccination setting. Immunother 53(2):79–85 Blood. 113(21):5167–5175 120. Miyazono K (2000) Positive and negative regulation of TGF-beta 100. Hanke N, Alizadeh D, Katsanis E, Larmonier N (2013) Dendritic signaling. J Cell Sci 113(Pt 7):1101–1109 cell tumor killing activity and its potential applications in cancer 121. Wei J, Barr J, Kong LYet al (2010) Glioblastoma cancer-initiating immunotherapy. Crit Rev Immunol 33(1):1–21 cells inhibit T-cell proliferation and effector responses by the sig- 101. Palucka K, Banchereau J (2013) Dendritic-cell-based therapeutic nal transducers and activators of transcription 3 pathway. Mol cancer vaccines. Immunity. 39(1):38–48 Cancer Ther 9(1):67–78 102. Nouri-Shirazi M, Banchereau J, Bell D et al (2000) Dendritic cells 122. Emmerson PJ, Duffin KL, Chintharlapalli S, Wu X (2018) GDF15 capture killed tumor cells and present their antigens to elicit tumor- and growth control. Front Physiol 9:1712 specific immune responses. J Immunol 165(7):3797–3803 123. Dumic J, Dabelic S, Flogel M (2006) Galectin-3: an open-ended 103. Grivennikov SI, Greten FR, Karin M (2010) Immunity, inflamma- story. Biochim Biophys Acta 1760(4):616–635 tion, and cancer. Cell. 140(6):883–899 124. Colmont CS, Benketah A, Reed SH et al (2013) CD200- 104. Hsu YL, Chen YJ, Chang WA et al. (2018) Interaction between expressing human basal cell carcinoma cells initiate tumor Tumor-Associated Dendritic Cells and Colon Cancer Cells growth. Proc Natl Acad Sci U S A 110(4):1434–1439 Contributes to Tumor Progression via CXCL1. Int J Mol Sci 19(8) 125. Wright GJ, Puklavec MJ, Willis AC et al (2000) Lymphoid/ 105. Condeelis J, Pollard JW (2006) Macrophages: obligate partners neuronal cell surface OX2 glycoprotein recognizes a novel recep- for tumor cell migration, invasion, and metastasis. Cell. 124(2): tor on macrophages implicated in the control of their function. 263–266 Immunity. 13(2):233–242 146 Ravindran S. et al. 126. Schatton T, Frank MH (2009) Antitumor immunity and cancer inhibits STAT3 signaling to enhance T cell-mediated immune stem cells. Ann N Y Acad Sci 1176:154–169 clearance of glioma. Cancer Res 73(13):3913–3926 127. Salmaninejad A, Khoramshahi V, Azani A, Soltaninejad E, Aslani 146. Sahranavardfard P, Firouzi J, Azimi M, et al. (2019) MicroRNA- S, Zamani MR, Zal M, Nesaei A, Hosseini SM (2018) PD-1 and 203 reinforces stemness properties in melanoma and augments cancer: molecular mechanisms and polymorphisms. tumorigenesis in vivo. J Cell Physiol Immunogenetics. 70(2):73–86 147. Shidal C, Singh NP, Nagarkatti P, Nagarkatti M (2019) 128. Yao Y, Ye H, Qi Z et al (2016) B7-H4(B7x)-mediated cross-talk MicroRNA-92 expression in CD133+ melanoma stem cells reg- between Glioma-initiating cells and macrophages via the IL6/ ulates immunosuppression in the tumor microenvironment via JAK/STAT3 pathway Lead to poor prognosis in Glioma patients. integrin-dependent activation of TGF-beta. Cancer Res Clin Cancer Res 22(11):2778–2790 148. Wang SH, Lin SY (2013) Tumor dormancy: potential therapeutic 129. Harris SG, Padilla J, Koumas L, Ray D, Phipps RP (2002) target in tumor recurrence and metastasis prevention. Exp Hematol Prostaglandins as modulators of immunity. Trends Immunol Oncol 2(1):29 23(3):144–150 149. Costello RT, Mallet F, Gaugler B et al (2000) Human acute mye- 130. Yoshimura A, Muto G (2011) TGF-beta function in immune sup- loid leukemia CD34+/CD38- progenitor cells have decreased sen- pression. Curr Top Microbiol Immunol 350:127–147 sitivity to chemotherapy and Fas-induced apoptosis, reduced im- munogenicity, and impaired dendritic cell transformation capaci- 131. Wan YY, Flavell RA (2007) 'Yin-Yang' functions of transforming growth factor-beta and T regulatory cells in immune regulation. ties. Cancer Res 60(16):4403–4411 Immunol Rev 220:199–213 150. Ishikawa F, Yoshida S, Saito Y, Hijikata A, Kitamura H, Tanaka S, 132. Shacter E, Weitzman SA (2002) Chronic inflammation and cancer. Nakamura R, Tanaka T, Tomiyama H, Saito N, Fukata M, Oncology (Williston Park) 16(2):217–226 229; discussion 230- Miyamoto T, Lyons B, Ohshima K, Uchida N, Taniguchi S, 212 Ohara O, Akashi K, Harada M, Shultz LD (2007) Chemotherapy-resistant human AML stem cells home to and en- 133. Parker KH, Beury DW, Ostrand-Rosenberg S (2015) Myeloid- derived suppressor cells: critical cells driving immune suppression graft within the bone-marrow endosteal region. Nat Biotechnol 25(11):1315–1321 in the tumor microenvironment. Adv Cancer Res 128:95–139 134. Smith C, Chang MY, Parker KH et al (2012) IDO is a nodal 151. Pece S, Tosoni D, Confalonieri S, Mazzarol G, Vecchi M, Ronzoni S, Bernard L, Viale G, Pelicci PG, di Fiore PP (2010) Biological pathogenic driver of lung cancer and metastasis development. and molecular heterogeneity of breast cancers correlates with their Cancer Discov 2(8):722–735 cancer stem cell content. Cell. 140(1):62–73 135. Kawasaki BT, Mistree T, Hurt EM, Kalathur M, Farrar WL (2007) 152. Steinbichler TB, Dudas J, Skvortsov S, Ganswindt U, Co-expression of the toleragenic glycoprotein, CD200, with Riechelmann H, Skvortsova II (2018) Therapy resistance medi- markers for cancer stem cells. Biochem Biophys Res Commun 364(4):778–782 ated by cancer stem cells. Semin Cancer Biol 53:156–167 136. Hirohashi Y, Torigoe T, Tsukahara T, Kanaseki T, Kochin V, Sato 153. Dongre A, Rashidian M, Reinhardt F, Bagnato A, Keckesova Z, N (2016) Immune responses to human cancer stem-like cells/ Ploegh HL, Weinberg RA (2017) Epithelial-to-Mesenchymal tran- sition contributes to immunosuppression in breast carcinomas. cancer-initiating cells. Cancer Sci 107(1):12–17 Cancer Res 77(15):3982–3989 137. Kim SY, Cho HS, Yang SH, Shin JY, Kim JS, Lee ST, Chu K, Roh 154. Prager BC, Xie Q, Bao S, Rich JN (2019) Cancer stem cells: the JK, Kim SU, Park CG (2009) Soluble mediators from human architects of the tumor ecosystem. Cell Stem Cell 24(1):41–53 neural stem cells play a critical role in suppression of T-cell acti- vation and proliferation. J Neurosci Res 87(10):2264–2272 155. Shlush LI, Mitchell A, Heisler L et al (2017) Tracing the origins of relapse in acute myeloid leukaemia to stem cells. Nature. 138. Ljujic B, Milovanovic M, Volarevic V et al (2013) Human mes- enchymal stem cells creating an immunosuppressive environment 547(7661):104–108 156. Encabo A, Solves P, Mateu E, Sepulveda P, Carbonell-Uberos F, and promote breast cancer in mice. Sci Rep 3:2298 139. Chhabra R, Saini N (2014) MicroRNAs in cancer stem cells: cur- Minana MD (2004) Selective generation of different dendritic cell rent status and future directions. Tumour Biol 35(9):8395–8405 precursors from CD34+ cells by interleukin-6 and interleukin-3. Stem Cells 22(5):725–740 140. Garzia L, Andolfo I, Cusanelli E et al (2009) MicroRNA-199b-5p impairs cancer stem cells through negative regulation of HES1 in 157. Hutten T, Thordardottir S, Hobo W, Hübel J, van der Waart A, medulloblastoma. PLoS One 4(3):e4998 Cany J, Dolstra H, Hangalapura BN (2014) Ex vivo generation of 141. Gasparini P, Lovat F, Fassan M, Casadei L, Cascione L, Jacob NK, interstitial and Langerhans cell-like dendritic cell subset-based Carasi S, Palmieri D, Costinean S, Shapiro CL, Huebner K, Croce vaccines for hematological malignancies. J Immunother 37(5): 267–277 CM (2014) Protective role of miR-155 in breast cancer through RAD51 targeting impairs homologous recombination after irradi- 158. Cornel AM, van Til NP, Boelens JJ, Nierkens S (2018) Strategies ation. Proc Natl Acad Sci U S A 111(12):4536–4541 to genetically modulate dendritic cells to potentiate anti-tumor responses in hematologic malignancies. Front Immunol 9:982 142. Petrelli A, Carollo R, Cargnelutti M et al (2015) By promoting cell differentiation, miR-100 sensitizes basal-like breast cancer stem 159. Banchereau J, Steinman RM (1998) Dendritic cells and the control of immunity. Nature. 392(6673):245–252 cells to hormonal therapy. Oncotarget. 6(4):2315–2330 143. Roscigno G, Quintavalle C, Donnarumma E, Puoti I, Diaz- 160. Kirk CJ, Mule JJ (2000) Gene-modified dendritic cells for use in Lagares A, Iaboni M, Fiore D, Russo V, Todaro M, Romano G, tumor vaccines. Hum Gene Ther 11(6):797–806 Thomas R, Cortino G, Gaggianesi M, Esteller M, Croce CM, 161. Kranz LM, Diken M, Haas H, Kreiter S, Loquai C, Reuter KC, Condorelli G (2016) MiR-221 promotes stemness of breast cancer Meng M, Fritz D, Vascotto F, Hefesha H, Grunwitz C, Vormehr cells by targeting DNMT3b. Oncotarget. 7(1):580–592 M, Hüsemann Y, Selmi A, Kuhn AN, Buck J, Derhovanessian E, 144. Celia-Terrassa T, Liu DD, Choudhury A et al (2017) Normal and Rae R, Attig S, Diekmann J, Jabulowsky RA, Heesch S, Hassel J, cancerous mammary stem cells evade interferon-induced con- Langguth P, Grabbe S, Huber C, Türeci Ö, Sahin U (2016) straint through the miR-199a-LCOR axis. Nat Cell Biol 19(6): Systemic RNA delivery to dendritic cells exploits antiviral defence 711–723 for cancer immunotherapy. Nature. 534(7607):396–401 145. Wei J, Wang F, Kong LY, Xu S, Doucette T, Ferguson SD, Yang Y, 162. Salcedo M, Bercovici N, Taylor R, Vereecken P, Massicard S, McEnery K, Jethwa K, Gjyshi O, Qiao W, Levine NB, Lang FF, Duriau D, Vernel-Pauillac F, Boyer A, Baron-Bodo V, Mallard Rao G, Fuller GN, Calin GA, Heimberger AB (2013) miR-124 E, Bartholeyns J, Goxe B, Latour N, Leroy S, Prigent D, Martiat The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing... P, Sales F, Laporte M, Bruyns C, Romet-Lemonne JL, Abastado Rogers LJ, Gracia GJ, Jones SA, Mangiameli DP, Pelletier MM, JP, Lehmann F, Velu T (2006) Vaccination of melanoma patients Gea-Banacloche J, Robinson MR, Berman DM, Filie AC, Abati using dendritic cells loaded with an allogeneic tumor cell lysate. A, Rosenberg SA (2005) Adoptive cell transfer therapy following Cancer Immunol Immunother 55(7):819–829 non-myeloablative but lymphodepleting chemotherapy for the 163. Finocchiaro G, Pellegatta S (2016) Immunotherapy with dendritic treatment of patients with refractory metastatic melanoma. J Clin cells loaded with glioblastoma stem cells: from preclinical to clin- Oncol 23(10):2346–2357 ical studies. Cancer Immunol Immunother 65(1):101–109 182. Hinrichs CS, Rosenberg SA (2014) Exploiting the curative poten- 164. Vik-Mo EO, Nyakas M, Mikkelsen BV, Moe MC, Due-Tønnesen tial of adoptive T-cell therapy for cancer. Immunol Rev 257(1):56– P, Suso EM, Sæbøe-Larssen S, Sandberg C, Brinchmann JE, 71 Helseth E, Rasmussen AM, Lote K, Aamdal S, Gaudernack G, 183. Rosenberg SA, Restifo NP (2015) Adoptive cell transfer as per- Kvalheim G, Langmoen IA (2013) Therapeutic vaccination sonalized immunotherapy for human cancer. Science. 348(6230): against autologous cancer stem cells with mRNA-transfected den- 62–68 dritic cells in patients with glioblastoma. Cancer Immunol 184. Rapoport AP, Stadtmauer EA, Binder-Scholl GK, Goloubeva O, Immunother 62(9):1499–1509 Vogl DT, Lacey SF, Badros AZ, Garfall A, Weiss B, Finklestein J, 165. Pellegatta S, Eoli M, Frigerio S, Antozzi C, Bruzzone MG, Cantini Kulikovskaya I, Sinha SK, Kronsberg S, Gupta M, Bond S, G, Nava S, Anghileri E, Cuppini L, Cuccarini V, Ciusani E, Melchiori L, Brewer JE, Bennett AD, Gerry AB, Pumphrey NJ, Dossena M, Pollo B, Mantegazza R, Parati EA, Finocchiaro G Williams D, Tayton-Martin HK, Ribeiro L, Holdich T, Yanovich (2013) The natural killer cell response and tumor debulking are S, Hardy N, Yared J, Kerr N, Philip S, Westphal S, Siegel DL, associated with prolonged survival in recurrent glioblastoma pa- Levine BL, Jakobsen BK, Kalos M, June CH (2015) NY-ESO-1- tients receiving dendritic cells loaded with autologous tumor ly- specific TCR-engineered T cells mediate sustained antigen- sates. Oncoimmunology. 2(3):e23401 specific antitumor effects in myeloma. Nat Med 21(8):914–921 166. Vinay DS, Ryan EP, Pawelec G et al (2015) Immune evasion in 185. Robbins PF, Lu YC, El-Gamil M et al (2013) Mining exomic cancer: mechanistic basis and therapeutic strategies. Semin Cancer sequencing data to identify mutated antigens recognized by adop- Biol 35(Suppl):S185–S198 tively transferred tumor-reactive T cells. Nat Med 19(6):747–752 167. Sultan M, Vidovic D, Paine AS et al (2018) Epigenetic silencing 186. Tran E, Rosenberg SA (2014) T-cell therapy against cancer muta- of TAP1 in Aldefluor(+) breast Cancer stem cells contributes to tions. Oncotarget. 5(13):4579–4580 their enhanced immune evasion. Stem Cells 36(5):641–654 187. Boyiadzis MM, Dhodapkar MV, Brentjens RJ et al (2018) 168. Pardoll DM (2012) The blockade of immune checkpoints in can- Chimeric antigen receptor (CAR) T therapies for the treatment cer immunotherapy. Nat Rev Cancer 12(4):252–264 of hematologic malignancies: clinical perspective and signifi- 169. Adachi K, Tamada K (2015) Immune checkpoint blockade opens cance. J Immunother Cancer. 6(1):137 an avenue of cancer immunotherapy with a potent clinical effica- 188. Guedan S, Posey AD Jr., Shaw C et al. (2018) Enhancing CAR T cy. Cancer Sci 106(8):945–950 cell persistence through ICOS and 4-1BB costimulation. JCI 170. Haanen JB, Robert C (2015) Immune checkpoint inhibitors. Prog Insight 3(1) Tumor Res 42:55–66 189. June CH, O'Connor RS, Kawalekar OU, Ghassemi S, Milone MC 171. Kyi C, Postow MA (2016) Immune checkpoint inhibitor combi- (2018) CAR T cell immunotherapy for human cancer. Science. nations in solid tumors: opportunities and challenges. 359(6382):1361–1365 Immunotherapy. 8(7):821–837 190. Alcantara M, Tesio M, June CH, Houot R (2018) CAR T-cells for 172. Postow MA, Callahan MK, Wolchok JD (2015) Immune check- T-cell malignancies: challenges in distinguishing between thera- point blockade in Cancer therapy. J Clin Oncol 33(17):1974–1982 peutic, normal, and neoplastic T-cells. Leukemia. 32(11):2307– 173. Rotte A, Jin JY, Lemaire V (2018) Mechanistic overview of im- 191. Hay KA, Turtle CJ (2017) Chimeric antigen receptor (CAR) T mune checkpoints to support the rational design of their combina- tions in cancer immunotherapy. Ann Oncol 29(1):71–83 cells: lessons learned from targeting of CD19 in B-cell malignan- 174. Wilden SM, Lang BM, Mohr P, Grabbe S (2016) Immune check- cies.Drugs.77(3):237–245 point inhibitors: a milestone in the treatment of melanoma. J Dtsch 192. Kohn DB, Dotti G, Brentjens R, Savoldo B, Jensen M, Cooper LJ, Dermatol Ges 14(7):685–695 June CH, Rosenberg S, Sadelain M, Heslop HE (2011) CARs on 175. Xia Y, Medeiros LJ, Young KH (2016) Immune checkpoint block- track in the clinic. Mol Ther 19(3):432–438 ade: releasing the brake towards hematological malignancies. 193. Watanabe K, Kuramitsu S, Posey AD Jr, June CH (2018) Blood Rev 30(3):189–200 Expanding the therapeutic window for CARTcell therapy in solid 176. Gajewski TF (2015) The next hurdle in Cancer immunotherapy: tumors: the Knowns and unknowns of CAR T cell biology. Front overcoming the non-T-cell-inflamed tumor microenvironment. Immunol 9:2486 Semin Oncol 42(4):663–671 194. DeRenzo C, Krenciute G, Gottschalk S (2018) The landscape of 177. Zhi Y, Mou Z, Chen J, He Y, Dong H, Fu X, Wu Y (2015) B7H1 CAR T cells beyond acute lymphoblastic leukemia for pediatric expression and epithelial-to-Mesenchymal transition phenotypes solid tumors. Am Soc Clin Oncol Educ Book 38:830–837 on colorectal Cancer stem-like cells. PLoS One 10(8):e0135528 195. Ma Q, Garber HR, Lu S, He H, Tallis E, Ding X, Sergeeva A, 178. Yang Y, Wu KE, Zhao E et al (2015) B7-H1 enhances proliferation Wood MS, Dotti G, Salvado B, Ruisaard K, Clise-Dwyer K, John ability of gastric cancer stem-like cells as a receptor. Oncol Lett LS, Rezvani K, Alatrash G, Shpall EJ, Molldrem JJ (2016) A 9(4):1833–1838 novel TCR-like CAR with specificity for PR1/HLA-A2 effective- 179. Wu Y, Chen M, Wu P, Chen C, Xu ZP, Gu W (2017) Increased ly targets myeloid leukemia in vitro when expressed in human PD-L1 expression in breast and colon cancer stem cells. Clin Exp adult peripheral blood and cord blood T cells. Cytotherapy. 18(8):985–994 Pharmacol Physiol 44(5):602–604 180. Shi X, Zhang X, Li J, Mo L, Zhao H, Zhu Y, Hu Z, Gao J, Tan W 196. Zhu X, Prasad S, Gaedicke S, Hettich M, Firat E, Niedermann G (2015) Patient-derived glioblastoma stem cells are killed by (2018) PD-1 blockade enhances the antitumor efficacy of GM- CSF surface-modified bladder cancer stem cells vaccine. Int J CD133-specific CAR T cells but induce the T cell aging marker Cancer 142(10):2106–2117 CD57. Oncotarget. 6(1):171–184 181. Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL, 197. Morgan RA, Johnson LA, Davis JL et al (2012) Recognition of Restifo NP, Royal RE, Kammula U, White DE, Mavroukakis SA, glioma stem cells by genetically modified T cells targeting 148 Ravindran S. et al. EGFRvIII and development of adoptive cell therapy for glioma. cell immunotherapy of solid tumors. Expert Opin Ther Targets Hum Gene Ther 23(10):1043–1053 19(10):1339–1350 198. Beard RE, Zheng Z, Lagisetty KH et al (2014) Multiple chimeric 203. Feng KC, Guo YL, Liu Y et al (2017) Cocktail treatment with antigen receptors successfully target chondroitin sulfate proteogly- EGFR-specific and CD133-specific chimeric antigen receptor- can 4 in several different cancer histologies and cancer stem cells. modified T cells in a patient with advanced cholangiocarcinoma. J Immunother Cancer. 2:25 J Hematol Oncol 10(1):4 199. Pellegatta S, Savoldo B, Di Ianni N, et al. (2018) Constitutive and 204. Sun B, Yang D, Dai H, et al. (2019) Eradication of hepatocellular TNFalpha-inducible expression of chondroitin sulfate proteogly- carcinoma by NKG2D-specific CAR T-cells. Cancer Immunol can 4 in glioblastoma and neurospheres: Implications for CAR-T Res cell therapy. Sci Transl Med 10(430) 205. Yang D, Sun B, Dai H, Li W, Shi L, Zhang P, Li S, Zhao X (2019) 200. Du H, Hirabayashi K, Ahn S et al (2019) Antitumor responses in T cells expressing NKG2D chimeric antigen receptors efficiently the absence of toxicity in solid tumors by targeting B7-H3 via eliminate glioblastoma and cancer stem cells. J Immunother chimeric antigen receptor Tcells. Cancer Cell 35(2):221–237 e228 Cancer 7(1):171 201. Deng Z, Wu Y, Ma W, Zhang S, Zhang YQ (2015) Adoptive T-cell therapy of prostate cancer targeting the cancer stem cell antigen Publisher’sNote Springer Nature remains neutral with regard to jurisdic- EpCAM. BMC Immunol 16:1 tional claims in published maps and institutional affiliations. 202. Wang Y, Geldres C, Ferrone S, Dotti G (2015) Chondroitin sulfate proteoglycan 4 as a target for chimeric antigen receptor-based T-

Journal

Cancer MicroenvironmentSpringer Journals

Published: Nov 22, 2019

There are no references for this article.