Skip to main content
Erschienen in: Cancer Microenvironment 2-3/2019

Open Access 22.11.2019 | Original Article

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

verfasst von: Shilpa Ravindran, Saad Rasool, Cristina Maccalli

Erschienen in: Cancer Microenvironment | Ausgabe 2-3/2019

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 modulating 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.
Hinweise
Shilpa Ravindran and Saad Rasool contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ALDH
Aldehyde dehydrogenase
APC
Antigen presenting cells
APM
Antigen processing machinery
CAR
Chimeric antigen receptor
CIC
Cancer initiating cell
CRC
Colorectal cancer
CT
Cancer testis
CTLA-4
Cytotoxic lymphocyte antigen-4
CSPG4
Chondroitin sulphate protidoglycan 4
HLA
Human leukocyte antigen
IDO
Indoleamine 2,3-dioxygenase
GBM
Glioblastoma multiforme
GDF-15
Growth differentiation factor-15
IFN
Interferon
IL-4
Interleukin 4
IL-10
Interleukin 10
IL-13
Interleukin 13
IL-13α2
α2 chain of IL-13 receptor
mAb
Monoclonal antibody;
MDSC
Myeloid derived suppressor cell
NSCLC
Non-small cell lung cancer
PD-1
Programmed death 1
PD-L1
Programmed death ligand 1
RCC
Renal cell carcinoma
STAT3
Signal transducer and activator of transcription 3
TGFB
Transforming growth factor beta
TAA
Tumor associated antigen
Treg
T regulatory cell.

Introduction

Tumors are composed by heterogeneous cellular components including a rare subpopulation bearing “stemness properties” and being responsible of tumor initiation and progression. These cells have been denominated cancer stem cells (CSCs) or cancer initiating cells (CICs) [16]. CSCs/CICs share several characteristics with normal stem cells, such as the ability to self-renew and to give rise to differentiated progeny and the resistance to DNA damage-induced cell death [3, 512]. CSCs/CICs, through the cycling from proliferation to quiescence, expression of ABC drug pumps, high levels of anti-apoptotic proteins and resistance to DNA damage, are resistant to radiation and chemotherapy and play an important role in disease relapse and tumor progression [13, 14].
CSCs/CICs have been isolated from both hematological and solid tumors and they represent a rare subpopulation, comprising 0.01–10% of cells within the tumor [15]. They can be ex vivo identified based on their “stem cell-like” characteristics and the expression of certain cell surface and functional markers [16]. The identification of CSCs/CICs was first reported in leukemia, showing a hierarchical organization of tumor cells [17]. The leukemic cells were able to be engrafted upon transplantation of CD34+CD38 cells into severe combined immune-deficient (SCID) mice, which eventually led to the identification of the hierarchical organization of tumors with few cells endowed with stemness and tumorigenic properties [17]. Since then, a variety of studies highlighted the existence of “stem-like” cancer cells in solid tumors with different histological origins [5, 1823]. Multiple molecules (e.g., ALDH-1, CD133, CD44, CD24, CBX3, ABCA5, LGR5, etc) have been identified as CSC/CIC-associated markers with differential expression depending on the tissues of derivation, highlighting the high grade of heterogeneity of these cells [16] (Table 1). Most of these molecules are over-expressed by CSCs/CICs but are also shared with either differentiated tumor cells or normal stem cells [4, 34]. As a result, detecting the presence of these cells within tumor lesions though probing for CSC/CIC- associated markers has not provided conclusive results. The xenotransplantation in immune deficient mice represents a useful tool to demonstrate in vivo the tumorigenic properties CSCs/CICs [35]. Xenograft models have contributed to prove the existence within tumor lesions of cell population endowed with stemness properties that upon serial transplantation could propagate both tumorigenic CSCs/CICs and malignant cells with differentiated phenotype without tumorigenic properties [18]. These subpopulations can be identified only through transplantation in immune deficient mice [4, 3638].
Table 1
Markers expressed by CSCs/CICs isolated from solid tumors and their role as TAAs
Markera
Tumor type
Recognition by T cellsb
Reference
ALDH1
CRC; breast and gastric cancer; melanoma
[24]
CD133
GBM, pancreas, lung, ovarian, prostate, and gastric cancer
[2527]
CD44
CRC, head and neck cancer
  
EpCAM
CRC, Retinoblastoma
 
[28]
EpCAM CD44 CD24
Pancreatic cancer
  
CD24
CRC
[29, 30]
SOX2
GBM
[31]
CBX3
Ostocarcinoma
  
LGR5
CRC
  
ABCB5
Melanoma
  
CD90
Liver cancer
  
HSP DNAJB8
RCC
[32, 33]
CD166
CRC; NSCLC
  
aMarkers commonly identified as associated with CSCs/CICs; b 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
Nevertheless, the available CSC/CIC-associated markers are dependent on spatial and temporal features, with their modulation occurring in relation to their inoculation in immunodeficient mice, proving the high level of plasticity of these cells and that none of the available markers can be exploited to monitor the in vivo fate of these cells [1, 39, 40] CSCs/CICs, similarly to normal stem cells, require a “niche” to allow the survival of these cells and their cycling from quiescence to proliferation and to maintain stemness and multipotency [4143]. The “niche” is represented by the tumor microenvironment (TME), which is composed of multicellular and dynamic compartments that include fibroblasts, endothelial, stromal, mesenchymal and immune cells [41]. The interaction of TME with stem-like cancer cells can regulate the fate of these cells through modulating the proliferation, differentiation, immunological properties and resistance to therapies [4450].
The high grade of heterogeneity and plasticity of CSCs/CICs can depend on their tissue of derivation and, importantly, on their cross-talk with TME [4, 16, 5153]. Limiting the isolation and the functional characterization of CSCs/CICs to the usage of phenotypic markers is unsatisfactory and do not consider the possibility that “stemness” function of tumor cells can be reversible, as shown by Quintana et al. for melanoma [1, 39]. Moreover, xenotransplantation of these cells in immune deficient mice is lacking the important variable of the TME and its role in affecting the fate of CSCs/CICs [1]. Therefore, the lack of standardized methods to isolate CSCs/CICs and of in vivo models allowing to monitor the cross-talk of these cells with TME can lead to the high extent of variability in assessing the functional properties of these cells and in preventing to accurately determine their fate and role in the tissue of origins and in the clinical outcome of cancer patients [54, 55]. The tool of sphere forming assay to propagate in vitro CSCs/CICs is too simplified, lacking the important component of TME and of the “niche”, preventing the constant monitoring of plasticity and heterogeneity of these cells (Figs. 1, 2).
Therefore, the combination of deep genomic, molecular and functional profiling of CSCs/CICs could represent a relevant method to achieve a comprehensive functional characterization of these cells and, possibly, of their role in tumor outcome [56].
CSCs/CICs have been identified as the tumor components responsible for resistance to standard therapy, as well as immunotherapy [10, 5760]. Although clinical responses in cancer patients are observed following treatments, these cells can remain in the minimal residual disease and upon changes in the environment they can exit the quiescence status and give rise to novel malignant lesion(s) or even initiate the metastatic colonization [8, 6163].
The extensive molecular and immunological characterization of CSCs/CICs is warranted in order to understand the mechanisms regulating their plasticity, quiescence, interaction with the TME and resistance to therapies and to immune responses.

Immunological Profile of CSCs

HLA Molecules and APM Components

The expression of HLA class I and class II molecules and APM has been investigated in CSCs/CICs isolated from colorectal cancer (CRC) and glioblastoma multiforme (GBM) showing an overall aberrant expression of these molecules, with, in some cases, failure in their modulation by the pre-treatment with IFNs (both alpha and gamma) or DNA demethylating agent (5-Aza CdR) [64, 65]. This impairment in antigen processing and presentation by CSCs/CICs lead, upon co-culture of these cells with autologous T cells, to a preferential selection and differentiation of TH2 type T cells and failure in eliciting effector functions [64, 65]. The suboptimal expression of HLA class I molecules and APM components was also reported in CSCs/CICs isolated from different type of solid tumors [6469]. These peculiar observations suggested that the defective expression of HLA molecules could represent a tool for the identification of CSCs/CICs [70]. On the contrary, the side population (SP) cells derived from CRC and endowed with stemness properties, showed detectable level of HLA class I molecules as well susceptibility to antigen-specific cytotoxic T lymphocytes (CTLs) [71], however this study has been performed using long-term in vitro established cell lines, that could have lost phenotypic properties of primary CSCs/CICs. Indeed, stem-like cells isolated by sphere-forming assay displayed aberrant expression of HLA class I and APM components [16, 65]. Contradictory results were obtained also in glioblastoma multiforme (GBM); CSCs/CICs isolated as sphere forming cells from this tumor have been shown to exhibit the expression of HLA class I molecules [72] while, when applying these analyses to primary GBM-derived sphere forming cells, defective expression of HLA class I and APM molecules was detected [64]. Stem-like cells expressing ABCB5 and isolated from melanoma were found to express suboptimal levels of HLA class I molecules while they were positive for HLA class II (45). APM components (e.g., LMP2, LMP7 MECL-1, TAP1 and TAP2) detected through mRNA analyses were found to be expressed in tumor sphere-models from different solid malignancies, representing a tool for in vitro enrichment of stem-like cells and for the investigation of micro-metastasis [73]. However, HLA class I and class II molecules were down-modulated in these cells as compared to differentiated tumor cells, also following their pre-treatment in vitro with IFN-γ, highlighting an impairment of antigen presentation by these cells [64]. It needs to be considered that this study did not analyze the expression at protein levels of APM molecules, therefore post-transcriptional mechanism could affect their expression. Moreover, long term in vitro established cell lines were used to isolate tumor cell spheres while in other studies reporting defective expression of APM components, primary CSCs/CICs have been investigated. These examples highlight that an overall suboptimal immunogenic potency by CSCs/CICs resulting in low or impaired susceptibility to T cell mediated immune responses (Fig. 1). This represents a mechanisms of evasion by immune responses that is shared with normal stem cells, that could represent a typical feature of cells with stemness properties [74]. Importantly, the failure in the expression of HLA molecules by tumor cells was found as one of the mechanisms of failure of the clinical activity of immune checkpoint blockade agents in cancer patients [75, 76], indicating that either CSCs/CICs can display immune evasion mechanisms shared by differentiated tumor cells or that indeed the suboptimal expression of HLA molecules of these cells and their resistance to T cell recognition can protect these cells from immunotherapy interventions, leading to tumor recurrence or progression. However, the lack of standardization in methods for both, the isolation of cells with stemness properties and to analyze HLA and APM molecules, represents a limitation in providing conclusive results. Nevertheless, detailed analysis to identify the molecular mechanisms that lead to aberrant expression of HLA molecules and APM components are warranted.
The suboptimal expression of HLA class I molecules if associated with detectable NKG2D ligands, can drive the increased susceptibility of CSCs/CICs to Natural Killer (NK) cells. This phenomenon has been observed in CSCs/CICs from glioma, melanoma, and CRC [68, 7779]. However, down-modulation of NKG2D ligands on CSCs/CICs has been documented, e.g., in GBM patients [64] suggesting that the expression of low levels of NK cell activating ligands can result in the impairment of anti-CSCs/CICs innate immune responses (Fig. 1). The expression profile of molecules activating innate immune responses on CSCs/CICs can be affected by their crosstalk with TME, and thus, by their plasticity that can influence the fate in vivo of these cells.

Tumor Associated Antigens and Adaptive Immune Responses against CSCs/CICs

Tumor associated antigens (TAAs) can be recognized by T lymphocytes when exposed on the surface of tumor cells in the form of peptide/HLA complexes [80, 81]. They are categorized into three groups; (i) the overexpressed/self-antigens that are expressed at high levels by tumor cells and detectable, although at lower levels, on normal tissues (e.g., MART-1/Melan-A, hTERT, EGFR, survivin). (ii) Cancer testis (CT) antigens that are detectable on tumor cells and not on normal cells, except for testis and trophoblast (e.g., NY-ESO1, MAGE A3-A4, PRAME, CT83, SSX2). (iii) Neoantigens or mutated antigens derived by non-synonymous mutations in cancer cells (e.g., MUM-1, CDK4, ME1, ACTN4, HLA-A2) [82]. The neoantigens are higher immunogenic compared to differentiation/self TAAs since are tumor specific and do not induce tolerogenic mechanisms in immune cells [8385]. Neoantigens have been shown to drive immune responses and to mediate efficient T cell recognition of tumor cells, leading to cancer eradication in patients treated with either mutanome based vaccines or adoptive cell therapy (ACT) with tumor infiltrating lymphocytes [8486]. Notably, CSCs/CICs bearing a somatic mutation in the CRC-associated “driver” gene SMAD4, could elicit antigen-specific T cell responses directed to both stemness and differentiated components of tumor [87].
A transcriptome analysis of the SP cells and main population (MP) derived from CRC, breast and lung cancer revealed a preferential expression of 18 CT antigens (MAGEA2, MAGEA3, MAGEA4, MAGEA6, MAGEA12, MAGEB2, GAGE1, GAGE8, SPANXA1, SPANXB1, SPANXC, XAGE2, SPA17, BORIS, PLU-1, SGY-1, TEX15 and CT45A1) in CSCs/CICs [32]. The TAA DNAJB8, that is a member of the heat shock protein (HSP) 40 family, was found to be preferentially expressed in renal cell carcinoma (RCC); interestingly this protein played an important role in the maintenance of CSCs/CICs. DNAJB8-specific immune responses could be detected in a mice model study of DNA vaccination for RCC, rendering this molecule appealing for targeting CSCs/CICs by the immune system [32, 33] (Table 1). Recently a new antigen, Ankyrin repeat and SOCS box protein 4 (ASB4), was described as target molecule of CTLs recognizing CSCs/CICs and not the differentiated cellular components of the tumor [88]. Suboptimal expression of TAAs (MART-1, ML-IAP, NY-ESO-1, and MAGE-A) was reported in melanoma-derived CSCs/CICs (Fig. 1)[89]. Similar results were obtained in CSCs/CICs isolated from GBM and CRC (Fig. 1 and Table 1) [64, 65]. On the other hand, CD133+ CSCs/CICs isolated from melanoma were shown to express either NY-ESO-1 or DEAD/H (Asp-Glu-Ala-Asp/His) box polypeptide 3, X-linked (DDX3X) representing target of tumor-specific T cells [90, 91]. Other studies have described the isolation of T lymphocytes recognizing TAAs expressed by CSCs/CICs such as IL-13Rα2, SOX2 and CD133 in GBM, CEP55 and COA-1 in CRC and EpCAM in retinoblastoma (Table 1) [28, 65, 71, 92].

Innate Immune Responses and their Relationship with CSCs/CICs

Natural killer (NK) cells are the first line of defense against cancer development and metastasis. NK cells have been described to efficiently recognize and kill in vitro CSCs/CICs isolated from CRC, melanoma and glioblastoma [68, 78, 93, 94]. The efficiency of NK cell-mediated lysis of CSCs/CICs was dependent on the expression of NCR ligands (NKp30 and NKp44), NKG2D ligands and when suboptimal or negative expression of HLA class I molecules were found on the surface of CSCs/CICs (Fig 1) [68, 78, 9395]. Tallerico et al. found that CSC/CIC but not their differentiated counterpart of CRC is susceptible to NK cells [77]. Similar results have been reported in GBM and melanoma, highlighting that the amount of ligands of activatory NK receptors on CSCs/CICs was determinant for efficient innate immune responses [68, 77, 78]. In patients with acute myeloid leukemia (AML), the suboptimal expression of NKG2D ligands has been described as a mecahnisms of escape by tumor cells from NK cell recognition [96], confirming that these molecules can affect the susceptibility of cancer cells to innate responses. The observations that NKG2D ligands could represent as biomarkers for prediction of clinical responses to immune checkpoint blockade in melanoma highlight that the pattern of NKG2D ligands expression by tumor cells can affect the type and efficiency of elicited anti-tumor immune responses [97]. Therefore, the levels and pattern of expression of NKG2D ligands by tumor cells, including CSCs/CICs could be a predictive marker for the choice of the type of immunotherapy interventions.
Dendritic cells (DCs) are antigen presenting cells (APCs) that can activate either innate or adaptive immune responses [98]. In addition, they play an important role in the formation of anti-tumor T- and B cell immunologic memories [99]. Immature DCs can capture the tumor-derived antigens by phagocytosis or pinocytosis and then migrate to lymphoid organs where they present these TAAs in the form of HLA/peptide complexes to T cells, resulting in antigen-specific immune responses [100102]. However, DCs depending on their morphological and phenotypic subtypes can either induce anti-tumor immune responses or promote tumor growth and progression [103]. The crosstalk of tumor with their TME is a crucial factor which results in the development of cancer [104]. Along this line, it has been described that high extent of 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 with suppressive functions, that could correlate tumor progression and poor survival of patients [104].
Macrophages represent important players for innate immune responses and can act as APCs similarly to DCs [105]. Based on their phenotype and functions they can be distinguished in two subpopulations: 1. The M1 subtype that are characterized by elevated pro-inflammatory cytokines, such as IL-12, IL-1β, IL-6, and tumor necrosis factor α (TNF-α), increased expression of HLA class II molecules, generation of reactive oxygen and nitrogen intermediates and ability to induce TH1-type T cell responses [106]. 2. In the presence IL-4, IL-10, and IL-13, macrophages can polarize towards M2 phenotype. These cells express scavenging, mannose and galactose receptors, IL-10, vascular endothelial growth factor (VEGF), matrix metalloproteinases (MMPs) and activation of the arginase pathway, leading to pro-tumoral effects [105107, 109]. The cross-talk between CSCs/CICs and TAM is orchestrated by STAT3 signaling [102, 103]. Upon iper-modulation of STAT3 in TAM, they can promote stemness, survival and proliferation in cancer cells while the latest cells can induce the immunosuppressive properties of TAM, leading to the impairment of cancer immune-surveillance [110].
Myeloid derived suppressor cells (MDSCs) are immune cells endowed with suppressive functions that can inhibit the effector functions of immune responses [111]. The frequency of these cells either at tumor site or in the circulation has been described as a prognostic factor for patients’ survival as well as of responsiveness to immunotherapy [112]. Interestingly, STAT3 can lead the differentiation of monocytes towards MDSCs in pancreatic tumors [113] regulating also the development of CSCs/CICs [113]. The secretion of pro-inflammatory cytokines and chemokines by tumor cells can induce the differentiation and recruitment of immunosuppressive cells that can also contribute to sustain the inflammatory TME and to the interaction and reciprocal influence of CSCs/CICs and their niche [110, 113115]. Another key regulator of the cross-talk between CSCs/CICs and TAM and DCs is represented by CD47 [116]. This molecule is over-expressed by CSCs/CICs of B cell malignancies. The binding of this molecule to the signal regulatory protein alpha (SIRPα), that mediates phagocytic functions in DCs and macrophages, has been shown to mediate the impairment of innate responses [116]. The cross-talk between CSCs/CICs and myeloid cells can affect both the fate and immunological profile of these cells, with implications for their susceptibilities to immune responses. Further studies should be designed to dissect the interactions of CSCs/CICs with different immune cells, although the major limitation is represented by the lack of in vivo models to monitor the interaction of these different immune cell population in the context of TME.

Immunomodulatory Properties of CSCs/CICs

The immunological profiling of CSCs/CICs has revealed that they share some characteristics with embryonic, hematopoietic and mesenchymal stem cells displaying immunoregulatory functions that render these cells invisible to immune responses and able to escape from tumor immune responses [4, 74, 117]. The principle mechanisms governing the immunomodulation of CSCs/CICs are described below.

Cytokines, Chemokines, Growth Factors & Immune Checkpoint Molecules

The observations that CSCs/CICs isolated from different tumor types can secrete soluble factors, such as Galectin-3, GDF-15, IL-10, IL-13, PGE2 and TGFb, or express immune checkpoint molecules with immunosuppressive functions, have suggested that these cells can regulate the impairment of immune responses as well as regulating a pro-tumoral TME (Table 2) [4, 51, 18130]. These immunosuppressive factors have been described to induce the differentiation of regulatory T cells (Tregs) or MDSCs and M2 macrophages, resulting in the impairment of effector functions of innate and adaptive responses [4, 51, 130, 131]. Moreover, pro-inflammatory cytokines such as IL-6, IL-8, IL-10 and IL-13, released by CSCs/CICs can contribute to maintain an inflammatory and suppressive TME representing the “niche” sustaining cellular stemness (Table 2) [132, 133]. Indoleamine 2,3-dioxigenase (IDO), that mediates the catabolism of tryptophan, has been shown to be expressed by CSCs/CICs, contributing to the differentiation of Tregs, skewing the cytokine profiling of T cells toward TH2 -type and inhibiting the survival and proliferation of CTL (Table 2) [133, 134].
Table 2
Immunomodulatory molecules detected in CSCs/CICs
Molecule
Functiona
Activity in CSCs/CICsb
Reference
IL-4
Cytokine involved in differentiation of naïve T cells to Th2.
Inhibition of TH1 cell mediated immune responses.
[65, 118]
IL-10, IL-13
Anti-inflammatory cytokines
Suppression of CTL functions; differentiation of Tregs and MDSCs
[89, 119]
TGFB
Growth factor with potent inhibitory function
Tregs differentiation, inhibition of TH1 responses
[89, 120]
STAT3
Transcription factor with a potential anti-inflammatory function
Maintenance and proliferation of CSCs/CICs; differentiation of MDSCs, iDCs, M2
[115, 121]
GDF15
Growth and differentiation factor related to cellular stress.
Inhibition of anti-tumor immune responses
[122]
Galectin-3
Protein with important role in cell-cell adhesion and interactions with the extracellular environment.
Inhibitor of T cell mediated immune responses
[89, 123]
IDO
Enzyme involved in tryptophan catabolism
Suppression of TH1 type immune responses and differentiation of Tregs.
[34, 51, 64, 65, 123]
CD200
A glycoprotein that regulates myeloid cell activity and inhibits macrophage lineages.
Immune suppression and regulation of anti-tumor activity.
[124, 125]
PD-L1
Ligand of PD-1 and Immune checkpoint molecule
Inhibition of CTL immune responses.
[64, 65, 126, 127]
B7-H3 and B7-H4
Immune checkpoint molecules
Immunomodulation of cellular immune responses
[34, 64, 65, 128, 129]
a: function of the molecules listed in the Table
b: 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
In addition, the CSC/CIC-associated expression of IL-4 and CD200, through cell-to-cell interaction, lead to the inhibition of T cell effector functions (Table 2) [65, 135]. It 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]. Interestingly, the neutralization of this cytokine by mAb could overcome the T cell mediated anti-tumor impairment and induce antigen-specific recognition of both CSCs/CICs and differentiated tumor cells [65]. This study showed that, upon up-regulation of HLA class I and APM expression through IFN-γ treatment of CSCs/CICs, T cells could specifically recognize a neoantigen, SMAD4, generated by a non-synonymous mutation bearing stem-like cells and bulk tumor cells [65]. Thus, CTL reactivity against CSCs/CICs and the TAA-specific immunosurveillance could be improved by the usage of strategies to correct the low immunogenic profile of these cells.
The immune suppressive profile of CSCs/CICs has been also confirmed by evidences describing the expression by these cells of immune checkpoint molecules (e.g. CTLA-4, PD-L1, B7-H3 or B7-H4) (Table 2) [4, 34, 64, 65, 136]. These observations highlighted the similarities between CSCs/CICs and normal stem cells in terms of the immune profile [34, 137, 138]. Moreover, altered expression of STAT3 pathway in CSCs/CICs can also affect their immune suppressive activity through inhibiting T cell proliferation and activation, inducing the differentiation of Tregs and triggering T cell apoptosis [121]. The observations reported above show that multiple mechanisms and molecular pathways are either up-regulated or aberrantly activated in CSCs/CICs resulting in their immune suppressive properties, therefore the blockade of these signaling through the combination of inhibitory agents should be considered in order to rescue the tumor-specific immune responses.

MicroRNAs

miRNAs are non-coding RNAs regulating at post-transcriptional levels, through complementary binding to target mRNA, the expression of genes [139]. The altered regulation of gene expression in tumor cells can occur by both up- or down regulation of miRNA [139]. The most common activity of miRNAs in CSCs/CICs is represented by the control of the expression of either oncogenes (e.g., MiR34a, MiR31 or MiR205) or tumor suppressor genes [139]. The aberrant expression of few miRNAs, such as miRNAs 451and 199b-5p, has been shown to affect stem-like cell properties isolated from different type of tumors (e.g., GBM, breast cancer and medulloblastoma) [139143]. Of note, miRNAs displaying 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]. The level of miRNA-124, through regulating the expression of STAT3, can affect the efficiency of anti-CSC/CIC T cell responses in GBM [145]. Along this line, miR203 and miR92 can control the stemness and immunological profiles of melanoma cells [146, 147].

Immune Evasion and Tumor Dormancy

Tumor dormancy is represented by quiescent cells that can remain occult and undetectable by regular diagnostic methods for long intervals of time, even after initial clinical responses to therapies [148]. Quiescence of cells is the ability to exit cell cycle and remain in G0 phase until permissive environmental condition will lead to enter back into the cycling phase. This is considered one of the principle mechanisms underlying tumor dormancy. CSCs/CICs display the ability to cycle between quiescence and proliferation and together with their resistance to therapies represent the link between these cells and tumor dormancy [2, 5, 8, 9, 149151]. Furthermore, the immune suppressive mechanisms associated with CSCs/CICs can orchestrate the evasion of these cells from immune recognition and immunosurveillance, and could be considered additional factors responsible of tumor dormancy [152].
An important mechanisms of immune-surveillance is the homing of immune cells to the tumor site, which ultimately form the immune infiltrate. Tumors arising from epithelial breast cancer are known to possess high levels MHC class I molecules and of infiltrating T effector cells and M1 macrophages. The immune infiltrate from mesenchymal like breast cancer tumors exhibit low levels of MHC class I molecules, high levels of PD-L1 and contain Tregs, M2 like macrophages as well as exhausted T cells [153]. CSCs/CICs that are considered the architects of their own microenvironment [154], as well as generated by epithelial-to-mesenchymal transition (EMT) can be potentially responsible for the type of immune infiltration depending of their pattern of immune profile.
Common gene expression patterns have been found in normal mammary stem cells and dormant tumor cells from breast cancer suggesting the possible presence of stem-like cells in dormant tumors [151]. In addition, different cell sub-populations could be isolated from relapsed AML endowed with differential tumorigenic ability depending on their up-regulation of stemness signaling [155].
A better understand of the relationship between stemness properties, immunological profile of CSCs/CICs and tumor dormancy will provide insights on the mechanisms of therapeutic resistance of these cells and will allow to identify strategies for complete tumor eradication.

Immunological Targeting of CSCs/CICs

Cancer Vaccines

The recognition of TAAs expressed by CSCs/CICs by T cells 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 cancer vaccines in cancer patients [81]. However, the principle limiting factor of the clinical efficacy of this strategy is represented by the usage of “self”/tolerogenic TAAs, shared with normal tissues [81]. The low or negative expression of these categories of antigens and of CT-TAAs by CSCs/CICs can represent an additional reason of failure of high rate and long duration of clinical responses observed in cancer patients treated with cancer vaccines [4, 34]. In addition, the sub-optimal levels of HLA class I molecules and APM by stem-like cells can drive the failure in targeting CSCs/CICs by cancer vaccines leading to the development of tumor dormancy and tumor recurrence, although the observance of initial clinical efficacy of these therapeutic interventions [4, 34].
DC-based vaccines, exploiting these cells as APC to present TAAs to T cell-mediated responses, represent also a therapeutic strategy for cancer patients showing encouraging clinical activity [102, 156162]. DCs loaded with either CSC/CIC-lysates or mRNA isolated from these cells represented source of antigens for vaccination in the context of Phase I/II clinical trials of GBM patients [163, 164]. These studies provided proof of principle of improved overall survival of cancer patients treated with CSC/CIC targeted immunotherapy [163165]. Immune responses, with, in some cases increased frequency of circulating NK cells, were detected in patients showing clinical benefit from these treatments [163, 165]. Of note, these therapeutic interventions could overcome the failure of CSCs/CICs in expressing efficient levels of HLA class I molecules and in presenting TAAs to T cells, documenting for the first time, that cancer vaccine, if eliciting NK cell-mediated responses, could target stem-like tumor cells [158, 163165]. Tumor cell clones expressing immunogenic neoantigens can undergo immune selection due to the recognition and elimination by T lymphocytes, leading to the survival of tumor cell clones not expressing strong immunogenic antigens and maintaining the expression of low immunogenic TAAs [166] (and see https://​www.​biorxiv.​org/​content/​10.​1101/​536433v1). This process is also associated with immune evasions mechanisms developed by both tumor cells and TME [166].
In some tumors, the decrease in antigen presentation is a result of epigenetic silencing of the genes involved in antigen presentation machinery. The usage of demethylating agents such as 5-Aza-2′-deoxycytidine to reduce the methylation of genes involved in antigen presentation, is a potential strategy to increase the antigen presentation in these CSC/CICs. The effect of demethylation has been shown in CSCs/CICs from breast cancer, where it resulted in high expression levels of TAP1, which is involved in antigen presentation [167].
In addition, increased antigen presentation also improves the potential of discovering novel antigens, which can then be helpful in development of new anti-cancer vaccines (Fig. 2).

Immune Checkpoint Blockade

Immune checkpoints, including CTLA-4, PD-1 and PD-L1, are important physiological regulators of innate and adaptive immune responses [168]. Biological inhibitory agents have been clinically developed, revealing striking therapeutically success [169175]. However, a significant proportion of cancer patients failed to benefit from these therapies.
The effectiveness of immune checkpoint blockade (ICB) is largely dependent on the tumor microenvironment [176]. Tumors such as melanoma, bladder cancer and non-small cell lung cancer (NSCLC) can be characterized as “hot” tumors due to their inflamed TME, high levels of and neo-antigen expression and of T cell infiltration and detection of PD-L1. These tumors have been reported to be associated with higher frequency of susceptibility to immune checkpoint treatments. On the other hand, prostate cancer is considered to be a “cold” tumor, due to minimal level of T cell infiltration, and limited response to single agent checkpoint inhibition [176].
Expression of immune checkpoint molecules has been observed in CSCs/CICs from different histological origins [51]. PD-L1 expression was detected at high levels in CSCs/CICs isolated from primary human head and neck squamous cell carcinoma (HNSCC), gastric and breast cancer, CRC and GBM [34, 64, 65, 177179]. ,CSCs/CICs could theoretically be targeted in vivo by immune checkpoint blockade agents, enhancing the clinical efficacy of cancer vaccines, as demonstrated in a mouse model [180]. However, recent reports describing that clinical failure of these therapies was associated with defective expression of HLA class I molecules by tumor cells [75, 76], suggest that these cells might evade from the ICB-mediated unleash of immune responses (Fig. 2).

Adoptive Cell Therapy

ACT is represented by the isolation of T lymphocytes from cancer patients, their ex vivo expansion, and the infusion back into patients [181183]. In addition, T lymphocytes engineered to express TCR with high affinity for a cognate TAA could be exploited for ACT studies [183]. Highly encouraging and sustained responses mediated by adoptively transferred TCR targeting the TAA NY-ESO-1 have recently been reported in different tumor types, such as breast cancer and myeloma [86, 184]. Nevertheless, the antigen choice is highly relevant to prevent severe toxicities due to “off-target” cross-reaction with normal tissues sharing the same antigens or expressing molecules mimicking the TAAs [182].
Neoantigens have been described as candidate TAAs efficiently recognized by T cells that can be exploited for ACT of cancer patients and, interestingly, CTL targeting these antigens could be isolated from tumor infiltrating lymphocytes (TILs) of melanoma and other type of malignant lesions [183, 185, 186]. Nevertheless, ACT to target neoantigens can represent a promising approach for treatment of cancer patients upon assessment of HLA expression by both CSCs/CICs and differentiated tumor cells and, in case of suboptimal levels of expression, the achievement of their up-regulation by pre-treatment with immunomodulating agents [4, 34, 51].
T cell can be genetically modified to express a chimeric antigen receptor (CAR) that is composed of epitope-specific domains isolated from mAbs linked to T cell-derived activatory/costimulatory molecules [182, 187189]. CAR-T cells can recognize TAAs independently on the expression of HLA molecules and APM components [189]. CAR-T cell therapy for some subgroups of hematological malignancies represent the salvage intervention leading to stable clinical responses and improved overall survival of patients refractory to standard therapies or with recurrences [187, 189192]. The usage of CAR-T cell therapy for solid tumors is currently under investigation, showing encouraging results in cancer patients with aggressive tumor types, including malignant mesothelioma, pancreatic cancer and GBM [193197].
CAR-T cells targeting TAAs, such as CD133, EGFRvIII, EpCAM, CSPG4 and B7-H3, expressed by different type of solid tumors, including CSC/CIC components, have been developed in pre-clinical studies [196201]. These studies have shown that the targeting of TAAs that are expressed only by tumor cells, including CSCs/CICs, and not by normal cells, could provide the rational for safe and efficient clinical development of CAR-T cells therapy for these tumors(Fig. 2) [200, 202]. Moreover, the combination of CAR-T cells targeting dual TAAs, EGFRvIII and CD133, has been used for the successful therapeutic treatment of a patient with advanced cholangiocarcinoma [203].
CAR-T cells targeting NKG2D ligands on CSCs/CICs have been investigated and tested both in vitro and in vivo [204, 205]. CSCs/CICs from glioblastoma expressing detectable NKG2D ligands could be efficiently targeted by CAR- T cells. These tools showed to efficintly eliminate also xenograft tumors [205]. Nevertheless, a major limitation associated with the use of these CAR-T cells is represented by the variability of the levels of NKG2D ligands on the surface of CSCs/CICs, depending on their origin and methodology used for their ex vivo isolation.
Nevertheless, further investigations aimed at a comprehensive genomic and immunological characterization CSCs/CICs are warranted to implement the efficiency and safety of ACT strategies.

Conclusions

Recent advances in the genomic, molecular and immunological profiling of CSCs/CICs have contributed to the identification of dysregulated molecular pathways that orchestrate stem-like cancer cells and their interaction with TME. The heterogeneity and plasticity of these cells and the mutual effect of TME and CSCs/CICs on the resulting anti-tumoral or 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 development of their dormancy. The down-modulation of HLA molecules and NK activatory ligands on CSCs/CICs, through decreasing their susceptibility to T or NK cell targeting, might represent one of the principle factors leading to resistance to immunotherapy. Although, the mechanisms regulating the levels of HLA molecules and NKG2D ligands on CSCs/CICs are yet to be dissected. Due to the complexity of the cross-talk between CSCs/CICs and TME and the high plasticity of these cells, it is difficult to predict what type of immune cells could play a relevant role in targeting CSCs/CICs. Multifactorial investigations, including the immunological profile, immunomodulating molecules, the interaction with TME and the type of immune cell infiltration will allow to provide insights. Nevertheless, the available tools to isolate and characterize CSCs/CICs, such as spheroids, immunodeficient mice, antigenic profile, are unsatisfactory to dissect the cross-talk of these cells with TME. Moreover, the development of pre-clinical in vivo models engrafted with human immune system is desirable to allow the monitoring of the interaction of CSCs/CICs with TME.
The targeting of CSCs/CICs by immunotherapy could result in the complete tumor eradication and stable clinical responses in cancer patients. This goal could be achieved by the design of combination of strategies based on innate and/or antigen-specific T cell responses with immunoregulatory agents that can render CSCs/CICs susceptible to cell-mediated immunosurveillance. In cases where epigenetic factors are responsible for low antigen presentation, the usage of demethylating agents could represent a potential strategy to overcome the low expression of HLA molecules.
Molecular approaches dissecting the fate of CSCs/CICs within tumor tissues will allow to develop immune-based precision medicine approaches and to identify biomarkers predictive of patients’ responsiveness to therapies.

Acknowledgements

This study was supported by Qatar National Research Fund, grant no. NPRP10-0129-170277.

Compliance with Ethical Standards

Conflict of Interest

All the authors have no conflict of interest to disclose.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Publisher’s Note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Shackleton M, Quintana E, Fearon ER, Morrison SJ (2009) Heterogeneity in cancer: cancer stem cells versus clonal evolution. Cell. 138(5):822–829PubMedCrossRef Shackleton M, Quintana E, Fearon ER, Morrison SJ (2009) Heterogeneity in cancer: cancer stem cells versus clonal evolution. Cell. 138(5):822–829PubMedCrossRef
2.
Zurück zum Zitat Clarke MF, Dick JE, Dirks PB et al (2006) Cancer stem cells--perspectives on current status and future directions: AACR workshop on cancer stem cells. Cancer Res 66(19):9339–9344PubMedCrossRef Clarke MF, Dick JE, Dirks PB et al (2006) Cancer stem cells--perspectives on current status and future directions: AACR workshop on cancer stem cells. Cancer Res 66(19):9339–9344PubMedCrossRef
4.
Zurück zum Zitat Maccalli C, De Maria R (2015) Cancer stem cells: perspectives for therapeutic targeting. Cancer Immunol Immunother 64(1):91–97PubMedCrossRef Maccalli C, De Maria R (2015) Cancer stem cells: perspectives for therapeutic targeting. Cancer Immunol Immunother 64(1):91–97PubMedCrossRef
5.
Zurück zum Zitat Reya T, Morrison SJ, Clarke MF, Weissman IL (2001) Stem cells, cancer, and cancer stem cells. Nature. 414(6859):105–111PubMedCrossRef Reya T, Morrison SJ, Clarke MF, Weissman IL (2001) Stem cells, cancer, and cancer stem cells. Nature. 414(6859):105–111PubMedCrossRef
6.
Zurück zum Zitat Wicha MS, Liu S, Dontu G (2006) Cancer stem cells: an old idea--a paradigm shift. Cancer Res 66(4):1883–1890 discussion 1895-1886PubMedCrossRef Wicha MS, Liu S, Dontu G (2006) Cancer stem cells: an old idea--a paradigm shift. Cancer Res 66(4):1883–1890 discussion 1895-1886PubMedCrossRef
8.
Zurück zum Zitat Eyler CE, Rich JN (2008) Survival of the fittest: cancer stem cells in therapeutic resistance and angiogenesis. J Clin Oncol 26(17):2839–2845PubMedCrossRef Eyler CE, Rich JN (2008) Survival of the fittest: cancer stem cells in therapeutic resistance and angiogenesis. J Clin Oncol 26(17):2839–2845PubMedCrossRef
9.
Zurück zum Zitat Hadjimichael C, Chanoumidou K, Papadopoulou N, Arampatzi P, Papamatheakis J, Kretsovali A (2015) Common stemness regulators of embryonic and cancer stem cells. World J Stem Cells 7(9):1150–1184PubMedPubMedCentral Hadjimichael C, Chanoumidou K, Papadopoulou N, Arampatzi P, Papamatheakis J, Kretsovali A (2015) Common stemness regulators of embryonic and cancer stem cells. World J Stem Cells 7(9):1150–1184PubMedPubMedCentral
10.
Zurück zum Zitat Maugeri-Sacca M, Vigneri P, De Maria R (2011) Cancer stem cells and chemosensitivity. Clin Cancer Res 17(15):4942–4947PubMedCrossRef Maugeri-Sacca M, Vigneri P, De Maria R (2011) Cancer stem cells and chemosensitivity. Clin Cancer Res 17(15):4942–4947PubMedCrossRef
11.
Zurück zum Zitat Soltanian S, Matin MM (2011) Cancer stem cells and cancer therapy. Tumour Biol 32(3):425–440PubMedCrossRef Soltanian S, Matin MM (2011) Cancer stem cells and cancer therapy. Tumour Biol 32(3):425–440PubMedCrossRef
12.
Zurück zum Zitat Steinbichler TB, Dudas J, Skvortsov S, Ganswindt U, Riechelmann H, Skvortsova II. (2018) Therapy resistance mediated by cancer stem cells. Semin Cancer Biol Steinbichler TB, Dudas J, Skvortsov S, Ganswindt U, Riechelmann H, Skvortsova II. (2018) Therapy resistance mediated by cancer stem cells. Semin Cancer Biol
13.
Zurück zum Zitat Ricci-Vitiani L, Fabrizi E, Palio E, De Maria R (2009) Colon cancer stem cells. J Mol Med (Berl) 87(11):1097–1104CrossRef Ricci-Vitiani L, Fabrizi E, Palio E, De Maria R (2009) Colon cancer stem cells. J Mol Med (Berl) 87(11):1097–1104CrossRef
15.
Zurück zum Zitat Morrison BJ, Steel JC, Morris JC (2018) Reduction of MHC-I expression limits T-lymphocyte-mediated killing of Cancer-initiating cells. BMC Cancer 18(1):469PubMedPubMedCentralCrossRef Morrison BJ, Steel JC, Morris JC (2018) Reduction of MHC-I expression limits T-lymphocyte-mediated killing of Cancer-initiating cells. BMC Cancer 18(1):469PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Maccalli C, Volonte A, Cimminiello C, Parmiani G (2014) Immunology of cancer stem cells in solid tumours. A review. Eur J Cancer 50(3):649–655PubMedCrossRef Maccalli C, Volonte A, Cimminiello C, Parmiani G (2014) Immunology of cancer stem cells in solid tumours. A review. Eur J Cancer 50(3):649–655PubMedCrossRef
17.
Zurück zum Zitat Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE (1994) A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 367(6464):645–648PubMedCrossRef Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE (1994) A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 367(6464):645–648PubMedCrossRef
18.
Zurück zum Zitat Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF (2003) Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 100(7):3983–3988PubMedPubMedCentralCrossRef Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF (2003) Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 100(7):3983–3988PubMedPubMedCentralCrossRef
19.
20.
Zurück zum Zitat Singh SK, Clarke ID, Hide T, Dirks PB (2004) Cancer stem cells in nervous system tumors. Oncogene. 23(43):7267–7273PubMedCrossRef Singh SK, Clarke ID, Hide T, Dirks PB (2004) Cancer stem cells in nervous system tumors. Oncogene. 23(43):7267–7273PubMedCrossRef
21.
Zurück zum Zitat Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB (2004) Identification of human brain tumour initiating cells. Nature. 432(7015):396–401PubMedCrossRef Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB (2004) Identification of human brain tumour initiating cells. Nature. 432(7015):396–401PubMedCrossRef
22.
Zurück zum Zitat Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha M, Clarke MF, Simeone DM (2007) Identification of pancreatic cancer stem cells. Cancer Res 67(3):1030–1037PubMedCrossRef Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha M, Clarke MF, Simeone DM (2007) Identification of pancreatic cancer stem cells. Cancer Res 67(3):1030–1037PubMedCrossRef
23.
Zurück zum Zitat Ricci-Vitiani L, Pagliuca A, Palio E, Zeuner A, De Maria R (2008) Colon cancer stem cells. Gut. 57(4):538–548PubMedCrossRef Ricci-Vitiani L, Pagliuca A, Palio E, Zeuner A, De Maria R (2008) Colon cancer stem cells. Gut. 57(4):538–548PubMedCrossRef
24.
Zurück zum Zitat Visus C, Wang Y, Lozano-Leon A, Ferris RL, Silver S, Szczepanski MJ, Brand RE, Ferrone CR, Whiteside TL, Ferrone S, DeLeo A, Wang X (2011) Targeting ALDH(bright) human carcinoma-initiating cells with ALDH1A1-specific CD8(+) T cells. Clin Cancer Res 17(19):6174–6184PubMedPubMedCentralCrossRef Visus C, Wang Y, Lozano-Leon A, Ferris RL, Silver S, Szczepanski MJ, Brand RE, Ferrone CR, Whiteside TL, Ferrone S, DeLeo A, Wang X (2011) Targeting ALDH(bright) human carcinoma-initiating cells with ALDH1A1-specific CD8(+) T cells. Clin Cancer Res 17(19):6174–6184PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Brown CE, Starr R, Martinez C, Aguilar B, D'Apuzzo M, Todorov I, Shih CC, Badie B, Hudecek M, Riddell SR, Jensen MC (2009) Recognition and killing of brain tumor stem-like initiating cells by CD8+ cytolytic T cells. Cancer Res 69(23):8886–8893PubMedPubMedCentralCrossRef Brown CE, Starr R, Martinez C, Aguilar B, D'Apuzzo M, Todorov I, Shih CC, Badie B, Hudecek M, Riddell SR, Jensen MC (2009) Recognition and killing of brain tumor stem-like initiating cells by CD8+ cytolytic T cells. Cancer Res 69(23):8886–8893PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Zinzi L, Contino M, Cantore M, Capparelli E, Leopoldo M, Colabufo NA (2014) ABC transporters in CSCs membranes as a novel target for treating tumor relapse. Front Pharmacol 5:163PubMedPubMedCentralCrossRef Zinzi L, Contino M, Cantore M, Capparelli E, Leopoldo M, Colabufo NA (2014) ABC transporters in CSCs membranes as a novel target for treating tumor relapse. Front Pharmacol 5:163PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Mitra M, Kandalam M, Harilal A, Verma RS, Krishnan UM, Swaminathan S, Krishnakumar S (2012) EpCAM is a putative stem marker in retinoblastoma and an effective target for T-cell-mediated immunotherapy. Mol Vis 18:290–308PubMedPubMedCentral Mitra M, Kandalam M, Harilal A, Verma RS, Krishnan UM, Swaminathan S, Krishnakumar S (2012) EpCAM is a putative stem marker in retinoblastoma and an effective target for T-cell-mediated immunotherapy. Mol Vis 18:290–308PubMedPubMedCentral
29.
Zurück zum Zitat Choi D, Lee HW, Hur KY et al (2009) Cancer stem cell markers CD133 and CD24 correlate with invasiveness and differentiation in colorectal adenocarcinoma. World J Gastroenterol 15(18):2258–2264PubMedPubMedCentralCrossRef Choi D, Lee HW, Hur KY et al (2009) Cancer stem cell markers CD133 and CD24 correlate with invasiveness and differentiation in colorectal adenocarcinoma. World J Gastroenterol 15(18):2258–2264PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Shapira S, Kazanov D, Weisblatt S, Starr A, Arber N, Kraus S (2011) The CD24 protein inducible expression system is an ideal tool to explore the potential of CD24 as an oncogene and a target for immunotherapy in vitro and in vivo. J Biol Chem 286(47):40548–40555PubMedPubMedCentralCrossRef Shapira S, Kazanov D, Weisblatt S, Starr A, Arber N, Kraus S (2011) The CD24 protein inducible expression system is an ideal tool to explore the potential of CD24 as an oncogene and a target for immunotherapy in vitro and in vivo. J Biol Chem 286(47):40548–40555PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Schmitz M, Temme A, Senner V et al (2007) Identification of SOX2 as a novel glioma-associated antigen and potential target for T cell-based immunotherapy. Br J Cancer 96(8):1293–1301PubMedPubMedCentralCrossRef Schmitz M, Temme A, Senner V et al (2007) Identification of SOX2 as a novel glioma-associated antigen and potential target for T cell-based immunotherapy. Br J Cancer 96(8):1293–1301PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Yamada R, Takahashi A, Torigoe T, Morita R, Tamura Y, Tsukahara T, Kanaseki T, Kubo T, Watarai K, Kondo T, Hirohashi Y, Sato N (2013) Preferential expression of cancer/testis genes in cancer stem-like cells: proposal of a novel sub-category, cancer/testis/stem gene. Tissue Antigens 81(6):428–434PubMedCrossRef Yamada R, Takahashi A, Torigoe T, Morita R, Tamura Y, Tsukahara T, Kanaseki T, Kubo T, Watarai K, Kondo T, Hirohashi Y, Sato N (2013) Preferential expression of cancer/testis genes in cancer stem-like cells: proposal of a novel sub-category, cancer/testis/stem gene. Tissue Antigens 81(6):428–434PubMedCrossRef
33.
Zurück zum Zitat Nishizawa S, Hirohashi Y, Torigoe T, Takahashi A, Tamura Y, Mori T, Kanaseki T, Kamiguchi K, Asanuma H, Morita R, Sokolovskaya A, Matsuzaki J, Yamada R, Fujii R, Kampinga HH, Kondo T, Hasegawa T, Hara I, Sato N (2012) HSP DNAJB8 controls tumor-initiating ability in renal cancer stem-like cells. Cancer Res 72(11):2844–2854PubMedCrossRef Nishizawa S, Hirohashi Y, Torigoe T, Takahashi A, Tamura Y, Mori T, Kanaseki T, Kamiguchi K, Asanuma H, Morita R, Sokolovskaya A, Matsuzaki J, Yamada R, Fujii R, Kampinga HH, Kondo T, Hasegawa T, Hara I, Sato N (2012) HSP DNAJB8 controls tumor-initiating ability in renal cancer stem-like cells. Cancer Res 72(11):2844–2854PubMedCrossRef
34.
Zurück zum Zitat Maccalli C, Rasul KI, Elawad M, Ferrone S (2018) The role of cancer stem cells in the modulation of anti-tumor immune responses. Semin Cancer Biol. Maccalli C, Rasul KI, Elawad M, Ferrone S (2018) The role of cancer stem cells in the modulation of anti-tumor immune responses. Semin Cancer Biol.
35.
Zurück zum Zitat Clevers H (2011) The cancer stem cell: premises, promises and challenges. Nat Med 17(3):313–319PubMedCrossRef Clevers H (2011) The cancer stem cell: premises, promises and challenges. Nat Med 17(3):313–319PubMedCrossRef
36.
Zurück zum Zitat Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, di Virgilio A, Conticello C, Ruco L, Peschle C, de Maria R (2008) Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ 15(3):504–514PubMedCrossRef Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, di Virgilio A, Conticello C, Ruco L, Peschle C, de Maria R (2008) Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ 15(3):504–514PubMedCrossRef
37.
Zurück zum Zitat Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, de Maria R (2007) Identification and expansion of human colon-cancer-initiating cells. Nature. 445(7123):111–115PubMedCrossRef Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, de Maria R (2007) Identification and expansion of human colon-cancer-initiating cells. Nature. 445(7123):111–115PubMedCrossRef
38.
Zurück zum Zitat Todaro M, Gaggianesi M, Catalano V, Benfante A, Iovino F, Biffoni M, Apuzzo T, Sperduti I, Volpe S, Cocorullo G, Gulotta G, Dieli F, de Maria R, Stassi G (2014) CD44v6 is a marker of constitutive and reprogrammed cancer stem cells driving colon cancer metastasis. Cell Stem Cell 14(3):342–356PubMedCrossRef Todaro M, Gaggianesi M, Catalano V, Benfante A, Iovino F, Biffoni M, Apuzzo T, Sperduti I, Volpe S, Cocorullo G, Gulotta G, Dieli F, de Maria R, Stassi G (2014) CD44v6 is a marker of constitutive and reprogrammed cancer stem cells driving colon cancer metastasis. Cell Stem Cell 14(3):342–356PubMedCrossRef
39.
Zurück zum Zitat Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM, Morrison SJ (2008) Efficient tumour formation by single human melanoma cells. Nature. 456(7222):593–598PubMedPubMedCentralCrossRef Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM, Morrison SJ (2008) Efficient tumour formation by single human melanoma cells. Nature. 456(7222):593–598PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Roesch A, Fukunaga-Kalabis M, Schmidt EC, Zabierowski SE, Brafford PA, Vultur A, Basu D, Gimotty P, Vogt T, Herlyn M (2010) A temporarily distinct subpopulation of slow-cycling melanoma cells is required for continuous tumor growth. Cell. 141(4):583–594PubMedPubMedCentralCrossRef Roesch A, Fukunaga-Kalabis M, Schmidt EC, Zabierowski SE, Brafford PA, Vultur A, Basu D, Gimotty P, Vogt T, Herlyn M (2010) A temporarily distinct subpopulation of slow-cycling melanoma cells is required for continuous tumor growth. Cell. 141(4):583–594PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Prieto-Vila M, Takahashi RU, Usuba W, Kohama I, Ochiya T (2017) Drug resistance driven by cancer stem cells and their niche. Int J Mol Sci 18(12)PubMedCentralCrossRef Prieto-Vila M, Takahashi RU, Usuba W, Kohama I, Ochiya T (2017) Drug resistance driven by cancer stem cells and their niche. Int J Mol Sci 18(12)PubMedCentralCrossRef
42.
Zurück zum Zitat Fuchs E, Tumbar T, Guasch G (2004) Socializing with the neighbors: stem cells and their niche. Cell. 116(6):769–778PubMedCrossRef Fuchs E, Tumbar T, Guasch G (2004) Socializing with the neighbors: stem cells and their niche. Cell. 116(6):769–778PubMedCrossRef
43.
Zurück zum Zitat Xie T, Li L (2007) Stem cells and their niche: an inseparable relationship. Development. 134(11):2001–2006PubMedCrossRef Xie T, Li L (2007) Stem cells and their niche: an inseparable relationship. Development. 134(11):2001–2006PubMedCrossRef
44.
47.
Zurück zum Zitat Cully M (2018) Tumour microenvironment: fibroblast subtype provides niche for cancer stem cells. Nat Rev Cancer 18(3):136PubMedCrossRef Cully M (2018) Tumour microenvironment: fibroblast subtype provides niche for cancer stem cells. Nat Rev Cancer 18(3):136PubMedCrossRef
48.
Zurück zum Zitat Cully M (2018) Cancer: fibroblast subtype provides niche for cancer stem cells. Nat Rev Drug Discov 17(3):163PubMedCrossRef Cully M (2018) Cancer: fibroblast subtype provides niche for cancer stem cells. Nat Rev Drug Discov 17(3):163PubMedCrossRef
49.
Zurück zum Zitat Malanchi I, Santamaria-Martinez A, Susanto E et al (2012) Interactions between cancer stem cells and their niche govern metastatic colonization. Nature. 481(7379):85–89CrossRef Malanchi I, Santamaria-Martinez A, Susanto E et al (2012) Interactions between cancer stem cells and their niche govern metastatic colonization. Nature. 481(7379):85–89CrossRef
50.
Zurück zum Zitat Seidel S, Garvalov BK, Wirta V et al (2010) A hypoxic niche regulates glioblastoma stem cells through hypoxia inducible factor 2 alpha. Brain. 133(Pt 4):983–995PubMedCrossRef Seidel S, Garvalov BK, Wirta V et al (2010) A hypoxic niche regulates glioblastoma stem cells through hypoxia inducible factor 2 alpha. Brain. 133(Pt 4):983–995PubMedCrossRef
51.
Zurück zum Zitat Maccalli C, Parmiani G, Ferrone S (2017) Immunomodulating and Immunoresistance properties of Cancer-initiating cells: implications for the clinical success of immunotherapy. Immunol Investig 46(3):221–238CrossRef Maccalli C, Parmiani G, Ferrone S (2017) Immunomodulating and Immunoresistance properties of Cancer-initiating cells: implications for the clinical success of immunotherapy. Immunol Investig 46(3):221–238CrossRef
52.
Zurück zum Zitat Easwaran H, Tsai HC, Baylin SB (2014) Cancer epigenetics: tumor heterogeneity, plasticity of stem-like states, and drug resistance. Mol Cell 54(5):716–727PubMedPubMedCentralCrossRef Easwaran H, Tsai HC, Baylin SB (2014) Cancer epigenetics: tumor heterogeneity, plasticity of stem-like states, and drug resistance. Mol Cell 54(5):716–727PubMedPubMedCentralCrossRef
53.
54.
Zurück zum Zitat Bar EE, Chaudhry A, Lin A, Fan X, Schreck K, Matsui W, Piccirillo S, Vescovi AL, DiMeco F, Olivi A, Eberhart CG (2007) Cyclopamine-mediated hedgehog pathway inhibition depletes stem-like cancer cells in glioblastoma. Stem Cells 25(10):2524–2533PubMedPubMedCentralCrossRef Bar EE, Chaudhry A, Lin A, Fan X, Schreck K, Matsui W, Piccirillo S, Vescovi AL, DiMeco F, Olivi A, Eberhart CG (2007) Cyclopamine-mediated hedgehog pathway inhibition depletes stem-like cancer cells in glioblastoma. Stem Cells 25(10):2524–2533PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Sakariassen PO, Prestegarden L, Wang J, Skaftnesmo KO, Mahesparan R, Molthoff C, Sminia P, Sundlisaeter E, Misra A, Tysnes BB, Chekenya M, Peters H, Lende G, Kalland KH, Øyan AM, Petersen K, Jonassen I, van der Kogel A, Feuerstein BG, Terzis AJ, Bjerkvig R, Enger PØ (2006) Angiogenesis-independent tumor growth mediated by stem-like cancer cells. Proc Natl Acad Sci U S A 103(44):16466–16471PubMedPubMedCentralCrossRef Sakariassen PO, Prestegarden L, Wang J, Skaftnesmo KO, Mahesparan R, Molthoff C, Sminia P, Sundlisaeter E, Misra A, Tysnes BB, Chekenya M, Peters H, Lende G, Kalland KH, Øyan AM, Petersen K, Jonassen I, van der Kogel A, Feuerstein BG, Terzis AJ, Bjerkvig R, Enger PØ (2006) Angiogenesis-independent tumor growth mediated by stem-like cancer cells. Proc Natl Acad Sci U S A 103(44):16466–16471PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Eppert K, Takenaka K, Lechman ER, Waldron L, Nilsson B, van Galen P, Metzeler KH, Poeppl A, Ling V, Beyene J, Canty AJ, Danska JS, Bohlander SK, Buske C, Minden MD, Golub TR, Jurisica I, Ebert BL, Dick JE (2011) Stem cell gene expression programs influence clinical outcome in human leukemia. Nat Med 17(9):1086–1093PubMedCrossRef Eppert K, Takenaka K, Lechman ER, Waldron L, Nilsson B, van Galen P, Metzeler KH, Poeppl A, Ling V, Beyene J, Canty AJ, Danska JS, Bohlander SK, Buske C, Minden MD, Golub TR, Jurisica I, Ebert BL, Dick JE (2011) Stem cell gene expression programs influence clinical outcome in human leukemia. Nat Med 17(9):1086–1093PubMedCrossRef
57.
Zurück zum Zitat Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW, Bigner DD, Rich JN (2006) Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 444(7120):756–760PubMedCrossRef Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW, Bigner DD, Rich JN (2006) Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 444(7120):756–760PubMedCrossRef
58.
Zurück zum Zitat Dean M, Fojo T, Bates S (2005) Tumour stem cells and drug resistance. Nat Rev Cancer 5(4):275–284PubMedCrossRef Dean M, Fojo T, Bates S (2005) Tumour stem cells and drug resistance. Nat Rev Cancer 5(4):275–284PubMedCrossRef
59.
Zurück zum Zitat Diehn M, Clarke MF (2006) Cancer stem cells and radiotherapy: new insights into tumor radioresistance. J Natl Cancer Inst 98(24):1755–1757PubMedCrossRef Diehn M, Clarke MF (2006) Cancer stem cells and radiotherapy: new insights into tumor radioresistance. J Natl Cancer Inst 98(24):1755–1757PubMedCrossRef
60.
Zurück zum Zitat Francipane MG, Alea MP, Lombardo Y, Todaro M, Medema JP, Stassi G (2008) Crucial role of interleukin-4 in the survival of colon cancer stem cells. Cancer Res 68(11):4022–4025PubMedCrossRef Francipane MG, Alea MP, Lombardo Y, Todaro M, Medema JP, Stassi G (2008) Crucial role of interleukin-4 in the survival of colon cancer stem cells. Cancer Res 68(11):4022–4025PubMedCrossRef
62.
Zurück zum Zitat Li X, Lewis MT, Huang J, Gutierrez C, Osborne CK, Wu MF, Hilsenbeck SG, Pavlick A, Zhang X, Chamness GC, Wong H, Rosen J, Chang JC (2008) Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst 100(9):672–679PubMedCrossRef Li X, Lewis MT, Huang J, Gutierrez C, Osborne CK, Wu MF, Hilsenbeck SG, Pavlick A, Zhang X, Chamness GC, Wong H, Rosen J, Chang JC (2008) Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst 100(9):672–679PubMedCrossRef
63.
Zurück zum Zitat Wilson BJ, Schatton T, Zhan Q, Gasser M, Ma J, Saab KR, Schanche R, Waaga-Gasser AM, Gold JS, Huang Q, Murphy GF, Frank MH, Frank NY (2011) ABCB5 identifies a therapy-refractory tumor cell population in colorectal cancer patients. Cancer Res 71(15):5307–5316PubMedPubMedCentralCrossRef Wilson BJ, Schatton T, Zhan Q, Gasser M, Ma J, Saab KR, Schanche R, Waaga-Gasser AM, Gold JS, Huang Q, Murphy GF, Frank MH, Frank NY (2011) ABCB5 identifies a therapy-refractory tumor cell population in colorectal cancer patients. Cancer Res 71(15):5307–5316PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Di Tomaso T, Mazzoleni S, Wang E et al (2010) Immunobiological characterization of cancer stem cells isolated from glioblastoma patients. Clin Cancer Res 16(3):800–813PubMedPubMedCentralCrossRef Di Tomaso T, Mazzoleni S, Wang E et al (2010) Immunobiological characterization of cancer stem cells isolated from glioblastoma patients. Clin Cancer Res 16(3):800–813PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Volonte A, Di Tomaso T, Spinelli M et al (2014) Cancer-initiating cells from colorectal cancer patients escape from T cell-mediated immunosurveillance in vitro through membrane-bound IL-4. J Immunol 192(1):523–532PubMedCrossRef Volonte A, Di Tomaso T, Spinelli M et al (2014) Cancer-initiating cells from colorectal cancer patients escape from T cell-mediated immunosurveillance in vitro through membrane-bound IL-4. J Immunol 192(1):523–532PubMedCrossRef
66.
Zurück zum Zitat Catalano V (2015) Resistance of cancer stem cells to cell- mediated immune responses. Vol 7: Springer Catalano V (2015) Resistance of cancer stem cells to cell- mediated immune responses. Vol 7: Springer
67.
Zurück zum Zitat Codony-Servat J, Rosell R (2015) Cancer stem cells and immunoresistance: clinical implications and solutions. Transl Lung Cancer Res 4(6):689–703PubMedPubMedCentral Codony-Servat J, Rosell R (2015) Cancer stem cells and immunoresistance: clinical implications and solutions. Transl Lung Cancer Res 4(6):689–703PubMedPubMedCentral
68.
Zurück zum Zitat Pietra G, Manzini C, Vitale M, Balsamo M, Ognio E, Boitano M, Queirolo P, Moretta L, Mingari MC (2009) Natural killer cells kill human melanoma cells with characteristics of cancer stem cells. Int Immunol 21(7):793–801PubMedCrossRef Pietra G, Manzini C, Vitale M, Balsamo M, Ognio E, Boitano M, Queirolo P, Moretta L, Mingari MC (2009) Natural killer cells kill human melanoma cells with characteristics of cancer stem cells. Int Immunol 21(7):793–801PubMedCrossRef
69.
Zurück zum Zitat Zhang D, Tang DG, Rycaj K (2018) Cancer stem cells: regulation programs, immunological properties and immunotherapy. Semin Cancer Biol 52(Pt 2):94–106PubMedPubMedCentralCrossRef Zhang D, Tang DG, Rycaj K (2018) Cancer stem cells: regulation programs, immunological properties and immunotherapy. Semin Cancer Biol 52(Pt 2):94–106PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Grau JJ, Mesia R, de la Iglesia-Vicente M et al (2016) Enrichment of cells with Cancer stem cell-like markers in relapses of Chemoresistant patients with locally advanced head and neck squamous cell carcinoma. Oncology. 90(5):267–272PubMedCrossRef Grau JJ, Mesia R, de la Iglesia-Vicente M et al (2016) Enrichment of cells with Cancer stem cell-like markers in relapses of Chemoresistant patients with locally advanced head and neck squamous cell carcinoma. Oncology. 90(5):267–272PubMedCrossRef
71.
Zurück zum Zitat Inoda S, Hirohashi Y, Torigoe T, Morita R, Takahashi A, Asanuma H, Nakatsugawa M, Nishizawa S, Tamura Y, Tsuruma T, Terui T, Kondo T, Ishitani K, Hasegawa T, Hirata K, Sato N (2011) Cytotoxic T lymphocytes efficiently recognize human colon cancer stem-like cells. Am J Pathol 178(4):1805–1813PubMedPubMedCentralCrossRef Inoda S, Hirohashi Y, Torigoe T, Morita R, Takahashi A, Asanuma H, Nakatsugawa M, Nishizawa S, Tamura Y, Tsuruma T, Terui T, Kondo T, Ishitani K, Hasegawa T, Hirata K, Sato N (2011) Cytotoxic T lymphocytes efficiently recognize human colon cancer stem-like cells. Am J Pathol 178(4):1805–1813PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Wei J, Barr J, Kong LY, Wang Y, Wu A, Sharma AK, Gumin J, Henry V, Colman H, Sawaya R, Lang FF, Heimberger AB (2010) Glioma-associated cancer-initiating cells induce immunosuppression. Clin Cancer Res 16(2):461–473PubMedPubMedCentralCrossRef Wei J, Barr J, Kong LY, Wang Y, Wu A, Sharma AK, Gumin J, Henry V, Colman H, Sawaya R, Lang FF, Heimberger AB (2010) Glioma-associated cancer-initiating cells induce immunosuppression. Clin Cancer Res 16(2):461–473PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Busse A, Letsch A, Fusi A, Nonnenmacher A, Stather D, Ochsenreither S, Regenbrecht CR, Keilholz U (2013) Characterization of small spheres derived from various solid tumor cell lines: are they suitable targets for T cells? Clin Exp Metastasis 30(6):781–791PubMedCrossRef Busse A, Letsch A, Fusi A, Nonnenmacher A, Stather D, Ochsenreither S, Regenbrecht CR, Keilholz U (2013) Characterization of small spheres derived from various solid tumor cell lines: are they suitable targets for T cells? Clin Exp Metastasis 30(6):781–791PubMedCrossRef
74.
75.
Zurück zum Zitat Gettinger S, Choi J, Hastings K, Truini A, Datar I, Sowell R, Wurtz A, Dong W, Cai G, Melnick MA, du VY, Schlessinger J, Goldberg SB, Chiang A, Sanmamed MF, Melero I, Agorreta J, Montuenga LM, Lifton R, Ferrone S, Kavathas P, Rimm DL, Kaech SM, Schalper K, Herbst RS, Politi K (2017) Impaired HLA class I antigen processing and presentation as a mechanism of acquired resistance to immune checkpoint inhibitors in lung Cancer. Cancer Discov 7(12):1420–1435PubMedPubMedCentralCrossRef Gettinger S, Choi J, Hastings K, Truini A, Datar I, Sowell R, Wurtz A, Dong W, Cai G, Melnick MA, du VY, Schlessinger J, Goldberg SB, Chiang A, Sanmamed MF, Melero I, Agorreta J, Montuenga LM, Lifton R, Ferrone S, Kavathas P, Rimm DL, Kaech SM, Schalper K, Herbst RS, Politi K (2017) Impaired HLA class I antigen processing and presentation as a mechanism of acquired resistance to immune checkpoint inhibitors in lung Cancer. Cancer Discov 7(12):1420–1435PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Chowell D, Morris LGT, Grigg CM et al (2018) Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science. 359(6375):582–587PubMedCrossRef Chowell D, Morris LGT, Grigg CM et al (2018) Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science. 359(6375):582–587PubMedCrossRef
77.
Zurück zum Zitat Tallerico R, Todaro M, Di Franco S et al (2013) Human NK cells selective targeting of colon cancer-initiating cells: a role for natural cytotoxicity receptors and MHC class I molecules. J Immunol 190(5):2381–2390PubMedCrossRef Tallerico R, Todaro M, Di Franco S et al (2013) Human NK cells selective targeting of colon cancer-initiating cells: a role for natural cytotoxicity receptors and MHC class I molecules. J Immunol 190(5):2381–2390PubMedCrossRef
78.
Zurück zum Zitat Castriconi R, Daga A, Dondero A et al (2009) NK cells recognize and kill human glioblastoma cells with stem cell-like properties. J Immunol 182(6):3530–3539PubMedCrossRef Castriconi R, Daga A, Dondero A et al (2009) NK cells recognize and kill human glioblastoma cells with stem cell-like properties. J Immunol 182(6):3530–3539PubMedCrossRef
79.
Zurück zum Zitat Tseng HC, Arasteh A, Paranjpe A et al (2010) Increased lysis of stem cells but not their differentiated cells by natural killer cells; de-differentiation or reprogramming activates NK cells. PLoS One 5(7):e11590PubMedPubMedCentralCrossRef Tseng HC, Arasteh A, Paranjpe A et al (2010) Increased lysis of stem cells but not their differentiated cells by natural killer cells; de-differentiation or reprogramming activates NK cells. PLoS One 5(7):e11590PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Maccalli C, Rasul KI, Elawad M, Ferrone S (2018) The role of cancer stem cells in the modulation of anti-tumor immune responses. Semin Cancer Biol 53:189–200PubMedPubMedCentralCrossRef Maccalli C, Rasul KI, Elawad M, Ferrone S (2018) The role of cancer stem cells in the modulation of anti-tumor immune responses. Semin Cancer Biol 53:189–200PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Parmiani G, Russo V, Maccalli C, Parolini D, Rizzo N, Maio M (2014) Peptide-based vaccines for cancer therapy. Hum Vaccin Immunother 10(11):3175–3178PubMedPubMedCentralCrossRef Parmiani G, Russo V, Maccalli C, Parolini D, Rizzo N, Maio M (2014) Peptide-based vaccines for cancer therapy. Hum Vaccin Immunother 10(11):3175–3178PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Lu YC, Robbins PF (2016) Cancer immunotherapy targeting neoantigens. Semin Immunol 28(1):22–27PubMedCrossRef Lu YC, Robbins PF (2016) Cancer immunotherapy targeting neoantigens. Semin Immunol 28(1):22–27PubMedCrossRef
83.
Zurück zum Zitat Forshew T, Murtaza M, Parkinson C et al (2012) Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med 4(136):136ra168CrossRef Forshew T, Murtaza M, Parkinson C et al (2012) Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med 4(136):136ra168CrossRef
84.
Zurück zum Zitat Robbins PF (2017) Tumor-infiltrating lymphocyte therapy and Neoantigens. Cancer J 23(2):138–143PubMedCrossRef Robbins PF (2017) Tumor-infiltrating lymphocyte therapy and Neoantigens. Cancer J 23(2):138–143PubMedCrossRef
85.
Zurück zum Zitat Sahin U, Derhovanessian E, Miller M et al (2017) Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 547(7662):222–226PubMedCrossRef Sahin U, Derhovanessian E, Miller M et al (2017) Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 547(7662):222–226PubMedCrossRef
86.
Zurück zum Zitat Zacharakis N, Chinnasamy H, Black M et al (2018) Immune recognition of somatic mutations leading to complete durable regression in metastatic breast cancer. Nat Med 24(6):724–730PubMedPubMedCentralCrossRef Zacharakis N, Chinnasamy H, Black M et al (2018) Immune recognition of somatic mutations leading to complete durable regression in metastatic breast cancer. Nat Med 24(6):724–730PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Mennonna D, Maccalli C, Romano MC et al (2017) T cell neoepitope discovery in colorectal cancer by high throughput profiling of somatic mutations in expressed genes. Gut. 66(3):454–463PubMedCrossRef Mennonna D, Maccalli C, Romano MC et al (2017) T cell neoepitope discovery in colorectal cancer by high throughput profiling of somatic mutations in expressed genes. Gut. 66(3):454–463PubMedCrossRef
88.
Zurück zum Zitat Miyamoto S, Kochin V, Kanaseki T et al (2018) The antigen ASB4 on Cancer stem cells serves as a target for CTL immunotherapy of colorectal Cancer. Cancer Immunol Res. Miyamoto S, Kochin V, Kanaseki T et al (2018) The antigen ASB4 on Cancer stem cells serves as a target for CTL immunotherapy of colorectal Cancer. Cancer Immunol Res.
89.
90.
Zurück zum Zitat Gedye C, Quirk J, Browning J, Svobodová S, John T, Sluka P, Dunbar PR, Corbeil D, Cebon J, Davis ID (2009) Cancer/testis antigens can be immunological targets in clonogenic CD133+ melanoma cells. Cancer Immunol Immunother 58(10):1635–1646PubMedCrossRef Gedye C, Quirk J, Browning J, Svobodová S, John T, Sluka P, Dunbar PR, Corbeil D, Cebon J, Davis ID (2009) Cancer/testis antigens can be immunological targets in clonogenic CD133+ melanoma cells. Cancer Immunol Immunother 58(10):1635–1646PubMedCrossRef
91.
Zurück zum Zitat Koshio J, Kagamu H, Nozaki K, Saida Y, Tanaka T, Shoji S, Igarashi N, Miura S, Okajima M, Watanabe S, Yoshizawa H, Narita I (2013) DEAD/H (asp-Glu-Ala-asp/his) box polypeptide 3, X-linked is an immunogenic target of cancer stem cells. Cancer Immunol Immunother 62(10):1619–1628PubMedCrossRef Koshio J, Kagamu H, Nozaki K, Saida Y, Tanaka T, Shoji S, Igarashi N, Miura S, Okajima M, Watanabe S, Yoshizawa H, Narita I (2013) DEAD/H (asp-Glu-Ala-asp/his) box polypeptide 3, X-linked is an immunogenic target of cancer stem cells. Cancer Immunol Immunother 62(10):1619–1628PubMedCrossRef
92.
Zurück zum Zitat Brown CE, Starr R, Aguilar B, Shami AF, Martinez C, D'Apuzzo M, Barish ME, Forman SJ, Jensen MC (2012) Stem-like tumor-initiating cells isolated from IL13Ralpha2 expressing gliomas are targeted and killed by IL13-zetakine-redirected T cells. Clin Cancer Res 18(8):2199–2209PubMedPubMedCentralCrossRef Brown CE, Starr R, Aguilar B, Shami AF, Martinez C, D'Apuzzo M, Barish ME, Forman SJ, Jensen MC (2012) Stem-like tumor-initiating cells isolated from IL13Ralpha2 expressing gliomas are targeted and killed by IL13-zetakine-redirected T cells. Clin Cancer Res 18(8):2199–2209PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Tallerico R, Garofalo C, Carbone E (2016) A new biological feature of natural killer cells: the recognition of solid tumor-derived Cancer stem cells. Front Immunol 7:179PubMedPubMedCentralCrossRef Tallerico R, Garofalo C, Carbone E (2016) A new biological feature of natural killer cells: the recognition of solid tumor-derived Cancer stem cells. Front Immunol 7:179PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Ames E, Canter RJ, Grossenbacher SK et al (2015) NK cells preferentially target tumor cells with a Cancer stem cell phenotype. J Immunol 195(8):4010–4019PubMedCrossRef Ames E, Canter RJ, Grossenbacher SK et al (2015) NK cells preferentially target tumor cells with a Cancer stem cell phenotype. J Immunol 195(8):4010–4019PubMedCrossRef
96.
Zurück zum Zitat Paczulla AM, Rothfelder K, Raffel S et al (2019) Absence of NKG2D ligands defines leukaemia stem cells and mediates their immune evasion. Nature. 572(7768):254–259PubMedPubMedCentralCrossRef Paczulla AM, Rothfelder K, Raffel S et al (2019) Absence of NKG2D ligands defines leukaemia stem cells and mediates their immune evasion. Nature. 572(7768):254–259PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Maccalli C, Giannarelli D, Chiarucci C, Cutaia O, Giacobini G, Hendrickx W, Amato G, Annesi D, Bedognetti D, Altomonte M, Danielli R, Calabrò L, di Giacomo AM, Marincola FM, Parmiani G, Maio M (2017) Soluble NKG2D ligands are biomarkers associated with the clinical outcome to immune checkpoint blockade therapy of metastatic melanoma patients. Oncoimmunology. 6(7):e1323618PubMedPubMedCentralCrossRef Maccalli C, Giannarelli D, Chiarucci C, Cutaia O, Giacobini G, Hendrickx W, Amato G, Annesi D, Bedognetti D, Altomonte M, Danielli R, Calabrò L, di Giacomo AM, Marincola FM, Parmiani G, Maio M (2017) Soluble NKG2D ligands are biomarkers associated with the clinical outcome to immune checkpoint blockade therapy of metastatic melanoma patients. Oncoimmunology. 6(7):e1323618PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Cella M, Sallusto F, Lanzavecchia A (1997) Origin, maturation and antigen presenting function of dendritic cells. Curr Opin Immunol 9(1):10–16PubMedCrossRef Cella M, Sallusto F, Lanzavecchia A (1997) Origin, maturation and antigen presenting function of dendritic cells. Curr Opin Immunol 9(1):10–16PubMedCrossRef
99.
Zurück zum Zitat Keller AM, Xiao Y, Peperzak V, Naik SH, Borst J (2009) Costimulatory ligand CD70 allows induction of CD8+ T-cell immunity by immature dendritic cells in a vaccination setting. Blood. 113(21):5167–5175PubMedCrossRef Keller AM, Xiao Y, Peperzak V, Naik SH, Borst J (2009) Costimulatory ligand CD70 allows induction of CD8+ T-cell immunity by immature dendritic cells in a vaccination setting. Blood. 113(21):5167–5175PubMedCrossRef
100.
Zurück zum Zitat Hanke N, Alizadeh D, Katsanis E, Larmonier N (2013) Dendritic cell tumor killing activity and its potential applications in cancer immunotherapy. Crit Rev Immunol 33(1):1–21PubMedPubMedCentralCrossRef Hanke N, Alizadeh D, Katsanis E, Larmonier N (2013) Dendritic cell tumor killing activity and its potential applications in cancer immunotherapy. Crit Rev Immunol 33(1):1–21PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Nouri-Shirazi M, Banchereau J, Bell D et al (2000) Dendritic cells capture killed tumor cells and present their antigens to elicit tumor-specific immune responses. J Immunol 165(7):3797–3803PubMedCrossRef Nouri-Shirazi M, Banchereau J, Bell D et al (2000) Dendritic cells capture killed tumor cells and present their antigens to elicit tumor-specific immune responses. J Immunol 165(7):3797–3803PubMedCrossRef
104.
Zurück zum Zitat Hsu YL, Chen YJ, Chang WA et al. (2018) Interaction between Tumor-Associated Dendritic Cells and Colon Cancer Cells Contributes to Tumor Progression via CXCL1. Int J Mol Sci 19(8)PubMedCentralCrossRef Hsu YL, Chen YJ, Chang WA et al. (2018) Interaction between Tumor-Associated Dendritic Cells and Colon Cancer Cells Contributes to Tumor Progression via CXCL1. Int J Mol Sci 19(8)PubMedCentralCrossRef
105.
Zurück zum Zitat Condeelis J, Pollard JW (2006) Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell. 124(2):263–266PubMedCrossRef Condeelis J, Pollard JW (2006) Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell. 124(2):263–266PubMedCrossRef
107.
Zurück zum Zitat Gordon S, Martinez FO (2010) Alternative activation of macrophages: mechanism and functions. Immunity. 32(5):593–604PubMedCrossRef Gordon S, Martinez FO (2010) Alternative activation of macrophages: mechanism and functions. Immunity. 32(5):593–604PubMedCrossRef
108.
Zurück zum Zitat Jinushi M, Chiba S, Yoshiyama H, Masutomi K, Kinoshita I, Dosaka-Akita H, Yagita H, Takaoka A, Tahara H (2011) Tumor-associated macrophages regulate tumorigenicity and anticancer drug responses of cancer stem/initiating cells. Proc Natl Acad Sci U S A 108(30):12425–12430PubMedPubMedCentralCrossRef Jinushi M, Chiba S, Yoshiyama H, Masutomi K, Kinoshita I, Dosaka-Akita H, Yagita H, Takaoka A, Tahara H (2011) Tumor-associated macrophages regulate tumorigenicity and anticancer drug responses of cancer stem/initiating cells. Proc Natl Acad Sci U S A 108(30):12425–12430PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Mantovani A, Sica A, Locati M (2005) Macrophage polarization comes of age. Immunity. 23(4):344–346PubMedCrossRef Mantovani A, Sica A, Locati M (2005) Macrophage polarization comes of age. Immunity. 23(4):344–346PubMedCrossRef
110.
Zurück zum Zitat Mitchem JB, Brennan DJ, Knolhoff BL, Belt BA, Zhu Y, Sanford DE, Belaygorod L, Carpenter D, Collins L, Piwnica-Worms D, Hewitt S, Udupi GM, Gallagher WM, Wegner C, West BL, Wang-Gillam A, Goedegebuure P, Linehan DC, DeNardo D (2013) Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res 73(3):1128–1141PubMedCrossRef Mitchem JB, Brennan DJ, Knolhoff BL, Belt BA, Zhu Y, Sanford DE, Belaygorod L, Carpenter D, Collins L, Piwnica-Worms D, Hewitt S, Udupi GM, Gallagher WM, Wegner C, West BL, Wang-Gillam A, Goedegebuure P, Linehan DC, DeNardo D (2013) Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res 73(3):1128–1141PubMedCrossRef
111.
Zurück zum Zitat Lindau D, Gielen P, Kroesen M, Wesseling P, Adema GJ (2013) The immunosuppressive tumour network: myeloid-derived suppressor cells, regulatory T cells and natural killer T cells. Immunology. 138(2):105–115PubMedPubMedCentralCrossRef Lindau D, Gielen P, Kroesen M, Wesseling P, Adema GJ (2013) The immunosuppressive tumour network: myeloid-derived suppressor cells, regulatory T cells and natural killer T cells. Immunology. 138(2):105–115PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Filipazzi P, Huber V, Rivoltini L (2012) Phenotype, function and clinical implications of myeloid-derived suppressor cells in cancer patients. Cancer Immunol Immunother 61(2):255–263PubMedCrossRef Filipazzi P, Huber V, Rivoltini L (2012) Phenotype, function and clinical implications of myeloid-derived suppressor cells in cancer patients. Cancer Immunol Immunother 61(2):255–263PubMedCrossRef
113.
Zurück zum Zitat Panni RZ, Sanford DE, Belt BA et al (2014) Tumor-induced STAT3 activation in monocytic myeloid-derived suppressor cells enhances stemness and mesenchymal properties in human pancreatic Cancer. Cancer Immunol Immunother 63(5):513–528PubMedPubMedCentralCrossRef Panni RZ, Sanford DE, Belt BA et al (2014) Tumor-induced STAT3 activation in monocytic myeloid-derived suppressor cells enhances stemness and mesenchymal properties in human pancreatic Cancer. Cancer Immunol Immunother 63(5):513–528PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Sherry MM, Reeves A, Wu JK, Cochran BH (2009) STAT3 is required for proliferation and maintenance of multipotency in glioblastoma stem cells. Stem Cells 27(10):2383–2392PubMedPubMedCentralCrossRef Sherry MM, Reeves A, Wu JK, Cochran BH (2009) STAT3 is required for proliferation and maintenance of multipotency in glioblastoma stem cells. Stem Cells 27(10):2383–2392PubMedPubMedCentralCrossRef
115.
Zurück zum Zitat Zhou J, Wulfkuhle J, Zhang H, Gu P, Yang Y, Deng J, Margolick JB, Liotta LA, Petricoin E 3rd, Zhang Y (2007) Activation of the PTEN/mTOR/STAT3 pathway in breast cancer stem-like cells is required for viability and maintenance. Proc Natl Acad Sci U S A 104(41):16158–16163PubMedPubMedCentralCrossRef Zhou J, Wulfkuhle J, Zhang H, Gu P, Yang Y, Deng J, Margolick JB, Liotta LA, Petricoin E 3rd, Zhang Y (2007) Activation of the PTEN/mTOR/STAT3 pathway in breast cancer stem-like cells is required for viability and maintenance. Proc Natl Acad Sci U S A 104(41):16158–16163PubMedPubMedCentralCrossRef
116.
Zurück zum Zitat Majeti R, Chao MP, Alizadeh AA, Pang WW, Jaiswal S, Gibbs KD Jr, van Rooijen N, Weissman IL (2009) CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell. 138(2):286–299PubMedPubMedCentralCrossRef Majeti R, Chao MP, Alizadeh AA, Pang WW, Jaiswal S, Gibbs KD Jr, van Rooijen N, Weissman IL (2009) CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell. 138(2):286–299PubMedPubMedCentralCrossRef
118.
Zurück zum Zitat Brown MA, Hural J (2017) Functions of IL-4 and control of its expression. Crit Rev Immunol 37(2–6):181–212PubMedCrossRef Brown MA, Hural J (2017) Functions of IL-4 and control of its expression. Crit Rev Immunol 37(2–6):181–212PubMedCrossRef
119.
Zurück zum Zitat Terabe M, Park JM, Berzofsky JA (2004) Role of IL-13 in regulation of anti-tumor immunity and tumor growth. Cancer Immunol Immunother 53(2):79–85PubMedCrossRef Terabe M, Park JM, Berzofsky JA (2004) Role of IL-13 in regulation of anti-tumor immunity and tumor growth. Cancer Immunol Immunother 53(2):79–85PubMedCrossRef
120.
Zurück zum Zitat Miyazono K (2000) Positive and negative regulation of TGF-beta signaling. J Cell Sci 113(Pt 7):1101–1109PubMedCrossRef Miyazono K (2000) Positive and negative regulation of TGF-beta signaling. J Cell Sci 113(Pt 7):1101–1109PubMedCrossRef
121.
Zurück zum Zitat Wei J, Barr J, Kong LY et al (2010) Glioblastoma cancer-initiating cells inhibit T-cell proliferation and effector responses by the signal transducers and activators of transcription 3 pathway. Mol Cancer Ther 9(1):67–78PubMedPubMedCentralCrossRef Wei J, Barr J, Kong LY et al (2010) Glioblastoma cancer-initiating cells inhibit T-cell proliferation and effector responses by the signal transducers and activators of transcription 3 pathway. Mol Cancer Ther 9(1):67–78PubMedPubMedCentralCrossRef
123.
Zurück zum Zitat Dumic J, Dabelic S, Flogel M (2006) Galectin-3: an open-ended story. Biochim Biophys Acta 1760(4):616–635PubMedCrossRef Dumic J, Dabelic S, Flogel M (2006) Galectin-3: an open-ended story. Biochim Biophys Acta 1760(4):616–635PubMedCrossRef
124.
Zurück zum Zitat Colmont CS, Benketah A, Reed SH et al (2013) CD200-expressing human basal cell carcinoma cells initiate tumor growth. Proc Natl Acad Sci U S A 110(4):1434–1439PubMedPubMedCentralCrossRef Colmont CS, Benketah A, Reed SH et al (2013) CD200-expressing human basal cell carcinoma cells initiate tumor growth. Proc Natl Acad Sci U S A 110(4):1434–1439PubMedPubMedCentralCrossRef
125.
Zurück zum Zitat Wright GJ, Puklavec MJ, Willis AC et al (2000) Lymphoid/neuronal cell surface OX2 glycoprotein recognizes a novel receptor on macrophages implicated in the control of their function. Immunity. 13(2):233–242PubMedCrossRef Wright GJ, Puklavec MJ, Willis AC et al (2000) Lymphoid/neuronal cell surface OX2 glycoprotein recognizes a novel receptor on macrophages implicated in the control of their function. Immunity. 13(2):233–242PubMedCrossRef
127.
Zurück zum Zitat Salmaninejad A, Khoramshahi V, Azani A, Soltaninejad E, Aslani S, Zamani MR, Zal M, Nesaei A, Hosseini SM (2018) PD-1 and cancer: molecular mechanisms and polymorphisms. Immunogenetics. 70(2):73–86PubMedCrossRef Salmaninejad A, Khoramshahi V, Azani A, Soltaninejad E, Aslani S, Zamani MR, Zal M, Nesaei A, Hosseini SM (2018) PD-1 and cancer: molecular mechanisms and polymorphisms. Immunogenetics. 70(2):73–86PubMedCrossRef
128.
Zurück zum Zitat Yao Y, Ye H, Qi Z et al (2016) B7-H4(B7x)-mediated cross-talk between Glioma-initiating cells and macrophages via the IL6/JAK/STAT3 pathway Lead to poor prognosis in Glioma patients. Clin Cancer Res 22(11):2778–2790PubMedPubMedCentralCrossRef Yao Y, Ye H, Qi Z et al (2016) B7-H4(B7x)-mediated cross-talk between Glioma-initiating cells and macrophages via the IL6/JAK/STAT3 pathway Lead to poor prognosis in Glioma patients. Clin Cancer Res 22(11):2778–2790PubMedPubMedCentralCrossRef
129.
Zurück zum Zitat Harris SG, Padilla J, Koumas L, Ray D, Phipps RP (2002) Prostaglandins as modulators of immunity. Trends Immunol 23(3):144–150PubMedCrossRef Harris SG, Padilla J, Koumas L, Ray D, Phipps RP (2002) Prostaglandins as modulators of immunity. Trends Immunol 23(3):144–150PubMedCrossRef
130.
Zurück zum Zitat Yoshimura A, Muto G (2011) TGF-beta function in immune suppression. Curr Top Microbiol Immunol 350:127–147PubMed Yoshimura A, Muto G (2011) TGF-beta function in immune suppression. Curr Top Microbiol Immunol 350:127–147PubMed
131.
Zurück zum Zitat Wan YY, Flavell RA (2007) 'Yin-Yang' functions of transforming growth factor-beta and T regulatory cells in immune regulation. Immunol Rev 220:199–213PubMedPubMedCentralCrossRef Wan YY, Flavell RA (2007) 'Yin-Yang' functions of transforming growth factor-beta and T regulatory cells in immune regulation. Immunol Rev 220:199–213PubMedPubMedCentralCrossRef
132.
Zurück zum Zitat Shacter E, Weitzman SA (2002) Chronic inflammation and cancer. Oncology (Williston Park) 16(2):217–226 229; discussion 230-212 Shacter E, Weitzman SA (2002) Chronic inflammation and cancer. Oncology (Williston Park) 16(2):217–226 229; discussion 230-212
133.
Zurück zum Zitat Parker KH, Beury DW, Ostrand-Rosenberg S (2015) Myeloid-derived suppressor cells: critical cells driving immune suppression in the tumor microenvironment. Adv Cancer Res 128:95–139PubMedPubMedCentralCrossRef Parker KH, Beury DW, Ostrand-Rosenberg S (2015) Myeloid-derived suppressor cells: critical cells driving immune suppression in the tumor microenvironment. Adv Cancer Res 128:95–139PubMedPubMedCentralCrossRef
134.
Zurück zum Zitat Smith C, Chang MY, Parker KH et al (2012) IDO is a nodal pathogenic driver of lung cancer and metastasis development. Cancer Discov 2(8):722–735PubMedPubMedCentralCrossRef Smith C, Chang MY, Parker KH et al (2012) IDO is a nodal pathogenic driver of lung cancer and metastasis development. Cancer Discov 2(8):722–735PubMedPubMedCentralCrossRef
135.
Zurück zum Zitat Kawasaki BT, Mistree T, Hurt EM, Kalathur M, Farrar WL (2007) Co-expression of the toleragenic glycoprotein, CD200, with markers for cancer stem cells. Biochem Biophys Res Commun 364(4):778–782PubMedPubMedCentralCrossRef Kawasaki BT, Mistree T, Hurt EM, Kalathur M, Farrar WL (2007) Co-expression of the toleragenic glycoprotein, CD200, with markers for cancer stem cells. Biochem Biophys Res Commun 364(4):778–782PubMedPubMedCentralCrossRef
136.
Zurück zum Zitat Hirohashi Y, Torigoe T, Tsukahara T, Kanaseki T, Kochin V, Sato N (2016) Immune responses to human cancer stem-like cells/cancer-initiating cells. Cancer Sci 107(1):12–17PubMedCrossRef Hirohashi Y, Torigoe T, Tsukahara T, Kanaseki T, Kochin V, Sato N (2016) Immune responses to human cancer stem-like cells/cancer-initiating cells. Cancer Sci 107(1):12–17PubMedCrossRef
137.
Zurück zum Zitat Kim SY, Cho HS, Yang SH, Shin JY, Kim JS, Lee ST, Chu K, Roh JK, Kim SU, Park CG (2009) Soluble mediators from human neural stem cells play a critical role in suppression of T-cell activation and proliferation. J Neurosci Res 87(10):2264–2272PubMedCrossRef Kim SY, Cho HS, Yang SH, Shin JY, Kim JS, Lee ST, Chu K, Roh JK, Kim SU, Park CG (2009) Soluble mediators from human neural stem cells play a critical role in suppression of T-cell activation and proliferation. J Neurosci Res 87(10):2264–2272PubMedCrossRef
138.
Zurück zum Zitat Ljujic B, Milovanovic M, Volarevic V et al (2013) Human mesenchymal stem cells creating an immunosuppressive environment and promote breast cancer in mice. Sci Rep 3:2298PubMedPubMedCentralCrossRef Ljujic B, Milovanovic M, Volarevic V et al (2013) Human mesenchymal stem cells creating an immunosuppressive environment and promote breast cancer in mice. Sci Rep 3:2298PubMedPubMedCentralCrossRef
139.
Zurück zum Zitat Chhabra R, Saini N (2014) MicroRNAs in cancer stem cells: current status and future directions. Tumour Biol 35(9):8395–8405PubMedCrossRef Chhabra R, Saini N (2014) MicroRNAs in cancer stem cells: current status and future directions. Tumour Biol 35(9):8395–8405PubMedCrossRef
140.
Zurück zum Zitat Garzia L, Andolfo I, Cusanelli E et al (2009) MicroRNA-199b-5p impairs cancer stem cells through negative regulation of HES1 in medulloblastoma. PLoS One 4(3):e4998PubMedPubMedCentralCrossRef Garzia L, Andolfo I, Cusanelli E et al (2009) MicroRNA-199b-5p impairs cancer stem cells through negative regulation of HES1 in medulloblastoma. PLoS One 4(3):e4998PubMedPubMedCentralCrossRef
141.
Zurück zum Zitat Gasparini P, Lovat F, Fassan M, Casadei L, Cascione L, Jacob NK, Carasi S, Palmieri D, Costinean S, Shapiro CL, Huebner K, Croce CM (2014) Protective role of miR-155 in breast cancer through RAD51 targeting impairs homologous recombination after irradiation. Proc Natl Acad Sci U S A 111(12):4536–4541PubMedPubMedCentralCrossRef Gasparini P, Lovat F, Fassan M, Casadei L, Cascione L, Jacob NK, Carasi S, Palmieri D, Costinean S, Shapiro CL, Huebner K, Croce CM (2014) Protective role of miR-155 in breast cancer through RAD51 targeting impairs homologous recombination after irradiation. Proc Natl Acad Sci U S A 111(12):4536–4541PubMedPubMedCentralCrossRef
142.
Zurück zum Zitat Petrelli A, Carollo R, Cargnelutti M et al (2015) By promoting cell differentiation, miR-100 sensitizes basal-like breast cancer stem cells to hormonal therapy. Oncotarget. 6(4):2315–2330PubMedCrossRef Petrelli A, Carollo R, Cargnelutti M et al (2015) By promoting cell differentiation, miR-100 sensitizes basal-like breast cancer stem cells to hormonal therapy. Oncotarget. 6(4):2315–2330PubMedCrossRef
143.
Zurück zum Zitat Roscigno G, Quintavalle C, Donnarumma E, Puoti I, Diaz-Lagares A, Iaboni M, Fiore D, Russo V, Todaro M, Romano G, Thomas R, Cortino G, Gaggianesi M, Esteller M, Croce CM, Condorelli G (2016) MiR-221 promotes stemness of breast cancer cells by targeting DNMT3b. Oncotarget. 7(1):580–592PubMedCrossRef Roscigno G, Quintavalle C, Donnarumma E, Puoti I, Diaz-Lagares A, Iaboni M, Fiore D, Russo V, Todaro M, Romano G, Thomas R, Cortino G, Gaggianesi M, Esteller M, Croce CM, Condorelli G (2016) MiR-221 promotes stemness of breast cancer cells by targeting DNMT3b. Oncotarget. 7(1):580–592PubMedCrossRef
144.
Zurück zum Zitat Celia-Terrassa T, Liu DD, Choudhury A et al (2017) Normal and cancerous mammary stem cells evade interferon-induced constraint through the miR-199a-LCOR axis. Nat Cell Biol 19(6):711–723PubMedPubMedCentralCrossRef Celia-Terrassa T, Liu DD, Choudhury A et al (2017) Normal and cancerous mammary stem cells evade interferon-induced constraint through the miR-199a-LCOR axis. Nat Cell Biol 19(6):711–723PubMedPubMedCentralCrossRef
145.
Zurück zum Zitat Wei J, Wang F, Kong LY, Xu S, Doucette T, Ferguson SD, Yang Y, McEnery K, Jethwa K, Gjyshi O, Qiao W, Levine NB, Lang FF, Rao G, Fuller GN, Calin GA, Heimberger AB (2013) miR-124 inhibits STAT3 signaling to enhance T cell-mediated immune clearance of glioma. Cancer Res 73(13):3913–3926PubMedPubMedCentralCrossRef Wei J, Wang F, Kong LY, Xu S, Doucette T, Ferguson SD, Yang Y, McEnery K, Jethwa K, Gjyshi O, Qiao W, Levine NB, Lang FF, Rao G, Fuller GN, Calin GA, Heimberger AB (2013) miR-124 inhibits STAT3 signaling to enhance T cell-mediated immune clearance of glioma. Cancer Res 73(13):3913–3926PubMedPubMedCentralCrossRef
146.
Zurück zum Zitat Sahranavardfard P, Firouzi J, Azimi M, et al. (2019) MicroRNA-203 reinforces stemness properties in melanoma and augments tumorigenesis in vivo. J Cell Physiol Sahranavardfard P, Firouzi J, Azimi M, et al. (2019) MicroRNA-203 reinforces stemness properties in melanoma and augments tumorigenesis in vivo. J Cell Physiol
147.
Zurück zum Zitat Shidal C, Singh NP, Nagarkatti P, Nagarkatti M (2019) MicroRNA-92 expression in CD133+ melanoma stem cells regulates immunosuppression in the tumor microenvironment via integrin-dependent activation of TGF-beta. Cancer Res Shidal C, Singh NP, Nagarkatti P, Nagarkatti M (2019) MicroRNA-92 expression in CD133+ melanoma stem cells regulates immunosuppression in the tumor microenvironment via integrin-dependent activation of TGF-beta. Cancer Res
148.
149.
Zurück zum Zitat Costello RT, Mallet F, Gaugler B et al (2000) Human acute myeloid leukemia CD34+/CD38- progenitor cells have decreased sensitivity to chemotherapy and Fas-induced apoptosis, reduced immunogenicity, and impaired dendritic cell transformation capacities. Cancer Res 60(16):4403–4411PubMed Costello RT, Mallet F, Gaugler B et al (2000) Human acute myeloid leukemia CD34+/CD38- progenitor cells have decreased sensitivity to chemotherapy and Fas-induced apoptosis, reduced immunogenicity, and impaired dendritic cell transformation capacities. Cancer Res 60(16):4403–4411PubMed
150.
Zurück zum Zitat Ishikawa F, Yoshida S, Saito Y, Hijikata A, Kitamura H, Tanaka S, Nakamura R, Tanaka T, Tomiyama H, Saito N, Fukata M, Miyamoto T, Lyons B, Ohshima K, Uchida N, Taniguchi S, Ohara O, Akashi K, Harada M, Shultz LD (2007) Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat Biotechnol 25(11):1315–1321PubMedCrossRef Ishikawa F, Yoshida S, Saito Y, Hijikata A, Kitamura H, Tanaka S, Nakamura R, Tanaka T, Tomiyama H, Saito N, Fukata M, Miyamoto T, Lyons B, Ohshima K, Uchida N, Taniguchi S, Ohara O, Akashi K, Harada M, Shultz LD (2007) Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat Biotechnol 25(11):1315–1321PubMedCrossRef
151.
Zurück zum Zitat Pece S, Tosoni D, Confalonieri S, Mazzarol G, Vecchi M, Ronzoni S, Bernard L, Viale G, Pelicci PG, di Fiore PP (2010) Biological and molecular heterogeneity of breast cancers correlates with their cancer stem cell content. Cell. 140(1):62–73PubMedCrossRef Pece S, Tosoni D, Confalonieri S, Mazzarol G, Vecchi M, Ronzoni S, Bernard L, Viale G, Pelicci PG, di Fiore PP (2010) Biological and molecular heterogeneity of breast cancers correlates with their cancer stem cell content. Cell. 140(1):62–73PubMedCrossRef
152.
Zurück zum Zitat Steinbichler TB, Dudas J, Skvortsov S, Ganswindt U, Riechelmann H, Skvortsova II (2018) Therapy resistance mediated by cancer stem cells. Semin Cancer Biol 53:156–167PubMedCrossRef Steinbichler TB, Dudas J, Skvortsov S, Ganswindt U, Riechelmann H, Skvortsova II (2018) Therapy resistance mediated by cancer stem cells. Semin Cancer Biol 53:156–167PubMedCrossRef
153.
Zurück zum Zitat Dongre A, Rashidian M, Reinhardt F, Bagnato A, Keckesova Z, Ploegh HL, Weinberg RA (2017) Epithelial-to-Mesenchymal transition contributes to immunosuppression in breast carcinomas. Cancer Res 77(15):3982–3989PubMedPubMedCentralCrossRef Dongre A, Rashidian M, Reinhardt F, Bagnato A, Keckesova Z, Ploegh HL, Weinberg RA (2017) Epithelial-to-Mesenchymal transition contributes to immunosuppression in breast carcinomas. Cancer Res 77(15):3982–3989PubMedPubMedCentralCrossRef
155.
Zurück zum Zitat Shlush LI, Mitchell A, Heisler L et al (2017) Tracing the origins of relapse in acute myeloid leukaemia to stem cells. Nature. 547(7661):104–108PubMedCrossRef Shlush LI, Mitchell A, Heisler L et al (2017) Tracing the origins of relapse in acute myeloid leukaemia to stem cells. Nature. 547(7661):104–108PubMedCrossRef
156.
Zurück zum Zitat Encabo A, Solves P, Mateu E, Sepulveda P, Carbonell-Uberos F, Minana MD (2004) Selective generation of different dendritic cell precursors from CD34+ cells by interleukin-6 and interleukin-3. Stem Cells 22(5):725–740PubMedCrossRef Encabo A, Solves P, Mateu E, Sepulveda P, Carbonell-Uberos F, Minana MD (2004) Selective generation of different dendritic cell precursors from CD34+ cells by interleukin-6 and interleukin-3. Stem Cells 22(5):725–740PubMedCrossRef
157.
Zurück zum Zitat Hutten T, Thordardottir S, Hobo W, Hübel J, van der Waart A, Cany J, Dolstra H, Hangalapura BN (2014) Ex vivo generation of interstitial and Langerhans cell-like dendritic cell subset-based vaccines for hematological malignancies. J Immunother 37(5):267–277PubMedCrossRef Hutten T, Thordardottir S, Hobo W, Hübel J, van der Waart A, Cany J, Dolstra H, Hangalapura BN (2014) Ex vivo generation of interstitial and Langerhans cell-like dendritic cell subset-based vaccines for hematological malignancies. J Immunother 37(5):267–277PubMedCrossRef
158.
Zurück zum Zitat Cornel AM, van Til NP, Boelens JJ, Nierkens S (2018) Strategies to genetically modulate dendritic cells to potentiate anti-tumor responses in hematologic malignancies. Front Immunol 9:982PubMedPubMedCentralCrossRef Cornel AM, van Til NP, Boelens JJ, Nierkens S (2018) Strategies to genetically modulate dendritic cells to potentiate anti-tumor responses in hematologic malignancies. Front Immunol 9:982PubMedPubMedCentralCrossRef
159.
Zurück zum Zitat Banchereau J, Steinman RM (1998) Dendritic cells and the control of immunity. Nature. 392(6673):245–252PubMedCrossRef Banchereau J, Steinman RM (1998) Dendritic cells and the control of immunity. Nature. 392(6673):245–252PubMedCrossRef
160.
Zurück zum Zitat Kirk CJ, Mule JJ (2000) Gene-modified dendritic cells for use in tumor vaccines. Hum Gene Ther 11(6):797–806PubMedCrossRef Kirk CJ, Mule JJ (2000) Gene-modified dendritic cells for use in tumor vaccines. Hum Gene Ther 11(6):797–806PubMedCrossRef
161.
Zurück zum Zitat Kranz LM, Diken M, Haas H, Kreiter S, Loquai C, Reuter KC, Meng M, Fritz D, Vascotto F, Hefesha H, Grunwitz C, Vormehr M, Hüsemann Y, Selmi A, Kuhn AN, Buck J, Derhovanessian E, Rae R, Attig S, Diekmann J, Jabulowsky RA, Heesch S, Hassel J, Langguth P, Grabbe S, Huber C, Türeci Ö, Sahin U (2016) Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature. 534(7607):396–401PubMedCrossRef Kranz LM, Diken M, Haas H, Kreiter S, Loquai C, Reuter KC, Meng M, Fritz D, Vascotto F, Hefesha H, Grunwitz C, Vormehr M, Hüsemann Y, Selmi A, Kuhn AN, Buck J, Derhovanessian E, Rae R, Attig S, Diekmann J, Jabulowsky RA, Heesch S, Hassel J, Langguth P, Grabbe S, Huber C, Türeci Ö, Sahin U (2016) Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature. 534(7607):396–401PubMedCrossRef
162.
Zurück zum Zitat Salcedo M, Bercovici N, Taylor R, Vereecken P, Massicard S, Duriau D, Vernel-Pauillac F, Boyer A, Baron-Bodo V, Mallard E, Bartholeyns J, Goxe B, Latour N, Leroy S, Prigent D, Martiat P, Sales F, Laporte M, Bruyns C, Romet-Lemonne JL, Abastado JP, Lehmann F, Velu T (2006) Vaccination of melanoma patients using dendritic cells loaded with an allogeneic tumor cell lysate. Cancer Immunol Immunother 55(7):819–829PubMedCrossRef Salcedo M, Bercovici N, Taylor R, Vereecken P, Massicard S, Duriau D, Vernel-Pauillac F, Boyer A, Baron-Bodo V, Mallard E, Bartholeyns J, Goxe B, Latour N, Leroy S, Prigent D, Martiat P, Sales F, Laporte M, Bruyns C, Romet-Lemonne JL, Abastado JP, Lehmann F, Velu T (2006) Vaccination of melanoma patients using dendritic cells loaded with an allogeneic tumor cell lysate. Cancer Immunol Immunother 55(7):819–829PubMedCrossRef
163.
Zurück zum Zitat Finocchiaro G, Pellegatta S (2016) Immunotherapy with dendritic cells loaded with glioblastoma stem cells: from preclinical to clinical studies. Cancer Immunol Immunother 65(1):101–109PubMedCrossRef Finocchiaro G, Pellegatta S (2016) Immunotherapy with dendritic cells loaded with glioblastoma stem cells: from preclinical to clinical studies. Cancer Immunol Immunother 65(1):101–109PubMedCrossRef
164.
Zurück zum Zitat Vik-Mo EO, Nyakas M, Mikkelsen BV, Moe MC, Due-Tønnesen P, Suso EM, Sæbøe-Larssen S, Sandberg C, Brinchmann JE, Helseth E, Rasmussen AM, Lote K, Aamdal S, Gaudernack G, Kvalheim G, Langmoen IA (2013) Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol Immunother 62(9):1499–1509PubMedPubMedCentralCrossRef Vik-Mo EO, Nyakas M, Mikkelsen BV, Moe MC, Due-Tønnesen P, Suso EM, Sæbøe-Larssen S, Sandberg C, Brinchmann JE, Helseth E, Rasmussen AM, Lote K, Aamdal S, Gaudernack G, Kvalheim G, Langmoen IA (2013) Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol Immunother 62(9):1499–1509PubMedPubMedCentralCrossRef
165.
Zurück zum Zitat Pellegatta S, Eoli M, Frigerio S, Antozzi C, Bruzzone MG, Cantini G, Nava S, Anghileri E, Cuppini L, Cuccarini V, Ciusani E, Dossena M, Pollo B, Mantegazza R, Parati EA, Finocchiaro G (2013) The natural killer cell response and tumor debulking are associated with prolonged survival in recurrent glioblastoma patients receiving dendritic cells loaded with autologous tumor lysates. Oncoimmunology. 2(3):e23401PubMedPubMedCentralCrossRef Pellegatta S, Eoli M, Frigerio S, Antozzi C, Bruzzone MG, Cantini G, Nava S, Anghileri E, Cuppini L, Cuccarini V, Ciusani E, Dossena M, Pollo B, Mantegazza R, Parati EA, Finocchiaro G (2013) The natural killer cell response and tumor debulking are associated with prolonged survival in recurrent glioblastoma patients receiving dendritic cells loaded with autologous tumor lysates. Oncoimmunology. 2(3):e23401PubMedPubMedCentralCrossRef
166.
Zurück zum Zitat Vinay DS, Ryan EP, Pawelec G et al (2015) Immune evasion in cancer: mechanistic basis and therapeutic strategies. Semin Cancer Biol 35(Suppl):S185–S198PubMedCrossRef Vinay DS, Ryan EP, Pawelec G et al (2015) Immune evasion in cancer: mechanistic basis and therapeutic strategies. Semin Cancer Biol 35(Suppl):S185–S198PubMedCrossRef
167.
Zurück zum Zitat Sultan M, Vidovic D, Paine AS et al (2018) Epigenetic silencing of TAP1 in Aldefluor(+) breast Cancer stem cells contributes to their enhanced immune evasion. Stem Cells 36(5):641–654PubMedCrossRef Sultan M, Vidovic D, Paine AS et al (2018) Epigenetic silencing of TAP1 in Aldefluor(+) breast Cancer stem cells contributes to their enhanced immune evasion. Stem Cells 36(5):641–654PubMedCrossRef
169.
Zurück zum Zitat Adachi K, Tamada K (2015) Immune checkpoint blockade opens an avenue of cancer immunotherapy with a potent clinical efficacy. Cancer Sci 106(8):945–950PubMedPubMedCentralCrossRef Adachi K, Tamada K (2015) Immune checkpoint blockade opens an avenue of cancer immunotherapy with a potent clinical efficacy. Cancer Sci 106(8):945–950PubMedPubMedCentralCrossRef
170.
171.
173.
Zurück zum Zitat Rotte A, Jin JY, Lemaire V (2018) Mechanistic overview of immune checkpoints to support the rational design of their combinations in cancer immunotherapy. Ann Oncol 29(1):71–83PubMedCrossRef Rotte A, Jin JY, Lemaire V (2018) Mechanistic overview of immune checkpoints to support the rational design of their combinations in cancer immunotherapy. Ann Oncol 29(1):71–83PubMedCrossRef
174.
Zurück zum Zitat Wilden SM, Lang BM, Mohr P, Grabbe S (2016) Immune checkpoint inhibitors: a milestone in the treatment of melanoma. J Dtsch Dermatol Ges 14(7):685–695PubMed Wilden SM, Lang BM, Mohr P, Grabbe S (2016) Immune checkpoint inhibitors: a milestone in the treatment of melanoma. J Dtsch Dermatol Ges 14(7):685–695PubMed
175.
Zurück zum Zitat Xia Y, Medeiros LJ, Young KH (2016) Immune checkpoint blockade: releasing the brake towards hematological malignancies. Blood Rev 30(3):189–200PubMedCrossRef Xia Y, Medeiros LJ, Young KH (2016) Immune checkpoint blockade: releasing the brake towards hematological malignancies. Blood Rev 30(3):189–200PubMedCrossRef
176.
Zurück zum Zitat Gajewski TF (2015) The next hurdle in Cancer immunotherapy: overcoming the non-T-cell-inflamed tumor microenvironment. Semin Oncol 42(4):663–671PubMedPubMedCentralCrossRef Gajewski TF (2015) The next hurdle in Cancer immunotherapy: overcoming the non-T-cell-inflamed tumor microenvironment. Semin Oncol 42(4):663–671PubMedPubMedCentralCrossRef
177.
Zurück zum Zitat Zhi Y, Mou Z, Chen J, He Y, Dong H, Fu X, Wu Y (2015) B7H1 expression and epithelial-to-Mesenchymal transition phenotypes on colorectal Cancer stem-like cells. PLoS One 10(8):e0135528PubMedPubMedCentralCrossRef Zhi Y, Mou Z, Chen J, He Y, Dong H, Fu X, Wu Y (2015) B7H1 expression and epithelial-to-Mesenchymal transition phenotypes on colorectal Cancer stem-like cells. PLoS One 10(8):e0135528PubMedPubMedCentralCrossRef
178.
Zurück zum Zitat Yang Y, Wu KE, Zhao E et al (2015) B7-H1 enhances proliferation ability of gastric cancer stem-like cells as a receptor. Oncol Lett 9(4):1833–1838PubMedPubMedCentralCrossRef Yang Y, Wu KE, Zhao E et al (2015) B7-H1 enhances proliferation ability of gastric cancer stem-like cells as a receptor. Oncol Lett 9(4):1833–1838PubMedPubMedCentralCrossRef
179.
Zurück zum Zitat Wu Y, Chen M, Wu P, Chen C, Xu ZP, Gu W (2017) Increased PD-L1 expression in breast and colon cancer stem cells. Clin Exp Pharmacol Physiol 44(5):602–604PubMedCrossRef Wu Y, Chen M, Wu P, Chen C, Xu ZP, Gu W (2017) Increased PD-L1 expression in breast and colon cancer stem cells. Clin Exp Pharmacol Physiol 44(5):602–604PubMedCrossRef
180.
Zurück zum Zitat Shi X, Zhang X, Li J, Mo L, Zhao H, Zhu Y, Hu Z, Gao J, Tan W (2018) PD-1 blockade enhances the antitumor efficacy of GM-CSF surface-modified bladder cancer stem cells vaccine. Int J Cancer 142(10):2106–2117PubMedCrossRef Shi X, Zhang X, Li J, Mo L, Zhao H, Zhu Y, Hu Z, Gao J, Tan W (2018) PD-1 blockade enhances the antitumor efficacy of GM-CSF surface-modified bladder cancer stem cells vaccine. Int J Cancer 142(10):2106–2117PubMedCrossRef
181.
Zurück zum Zitat Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL, Restifo NP, Royal RE, Kammula U, White DE, Mavroukakis SA, Rogers LJ, Gracia GJ, Jones SA, Mangiameli DP, Pelletier MM, Gea-Banacloche J, Robinson MR, Berman DM, Filie AC, Abati A, Rosenberg SA (2005) Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol 23(10):2346–2357PubMedCrossRef Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL, Restifo NP, Royal RE, Kammula U, White DE, Mavroukakis SA, Rogers LJ, Gracia GJ, Jones SA, Mangiameli DP, Pelletier MM, Gea-Banacloche J, Robinson MR, Berman DM, Filie AC, Abati A, Rosenberg SA (2005) Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol 23(10):2346–2357PubMedCrossRef
182.
184.
Zurück zum Zitat Rapoport AP, Stadtmauer EA, Binder-Scholl GK, Goloubeva O, Vogl DT, Lacey SF, Badros AZ, Garfall A, Weiss B, Finklestein J, Kulikovskaya I, Sinha SK, Kronsberg S, Gupta M, Bond S, Melchiori L, Brewer JE, Bennett AD, Gerry AB, Pumphrey NJ, Williams D, Tayton-Martin HK, Ribeiro L, Holdich T, Yanovich S, Hardy N, Yared J, Kerr N, Philip S, Westphal S, Siegel DL, Levine BL, Jakobsen BK, Kalos M, June CH (2015) NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat Med 21(8):914–921PubMedPubMedCentralCrossRef Rapoport AP, Stadtmauer EA, Binder-Scholl GK, Goloubeva O, Vogl DT, Lacey SF, Badros AZ, Garfall A, Weiss B, Finklestein J, Kulikovskaya I, Sinha SK, Kronsberg S, Gupta M, Bond S, Melchiori L, Brewer JE, Bennett AD, Gerry AB, Pumphrey NJ, Williams D, Tayton-Martin HK, Ribeiro L, Holdich T, Yanovich S, Hardy N, Yared J, Kerr N, Philip S, Westphal S, Siegel DL, Levine BL, Jakobsen BK, Kalos M, June CH (2015) NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat Med 21(8):914–921PubMedPubMedCentralCrossRef
185.
Zurück zum Zitat Robbins PF, Lu YC, El-Gamil M et al (2013) Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med 19(6):747–752PubMedPubMedCentralCrossRef Robbins PF, Lu YC, El-Gamil M et al (2013) Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med 19(6):747–752PubMedPubMedCentralCrossRef
187.
Zurück zum Zitat Boyiadzis MM, Dhodapkar MV, Brentjens RJ et al (2018) Chimeric antigen receptor (CAR) T therapies for the treatment of hematologic malignancies: clinical perspective and significance. J Immunother Cancer. 6(1):137PubMedPubMedCentralCrossRef Boyiadzis MM, Dhodapkar MV, Brentjens RJ et al (2018) Chimeric antigen receptor (CAR) T therapies for the treatment of hematologic malignancies: clinical perspective and significance. J Immunother Cancer. 6(1):137PubMedPubMedCentralCrossRef
188.
Zurück zum Zitat Guedan S, Posey AD Jr., Shaw C et al. (2018) Enhancing CAR T cell persistence through ICOS and 4-1BB costimulation. JCI Insight 3(1) Guedan S, Posey AD Jr., Shaw C et al. (2018) Enhancing CAR T cell persistence through ICOS and 4-1BB costimulation. JCI Insight 3(1)
189.
Zurück zum Zitat June CH, O'Connor RS, Kawalekar OU, Ghassemi S, Milone MC (2018) CAR T cell immunotherapy for human cancer. Science. 359(6382):1361–1365PubMedCrossRef June CH, O'Connor RS, Kawalekar OU, Ghassemi S, Milone MC (2018) CAR T cell immunotherapy for human cancer. Science. 359(6382):1361–1365PubMedCrossRef
190.
Zurück zum Zitat Alcantara M, Tesio M, June CH, Houot R (2018) CAR T-cells for T-cell malignancies: challenges in distinguishing between therapeutic, normal, and neoplastic T-cells. Leukemia. 32(11):2307–2315PubMedPubMedCentralCrossRef Alcantara M, Tesio M, June CH, Houot R (2018) CAR T-cells for T-cell malignancies: challenges in distinguishing between therapeutic, normal, and neoplastic T-cells. Leukemia. 32(11):2307–2315PubMedPubMedCentralCrossRef
191.
Zurück zum Zitat Hay KA, Turtle CJ (2017) Chimeric antigen receptor (CAR) T cells: lessons learned from targeting of CD19 in B-cell malignancies. Drugs. 77(3):237–245PubMedPubMedCentralCrossRef Hay KA, Turtle CJ (2017) Chimeric antigen receptor (CAR) T cells: lessons learned from targeting of CD19 in B-cell malignancies. Drugs. 77(3):237–245PubMedPubMedCentralCrossRef
192.
Zurück zum Zitat Kohn DB, Dotti G, Brentjens R, Savoldo B, Jensen M, Cooper LJ, June CH, Rosenberg S, Sadelain M, Heslop HE (2011) CARs on track in the clinic. Mol Ther 19(3):432–438PubMedPubMedCentralCrossRef Kohn DB, Dotti G, Brentjens R, Savoldo B, Jensen M, Cooper LJ, June CH, Rosenberg S, Sadelain M, Heslop HE (2011) CARs on track in the clinic. Mol Ther 19(3):432–438PubMedPubMedCentralCrossRef
193.
Zurück zum Zitat Watanabe K, Kuramitsu S, Posey AD Jr, June CH (2018) Expanding the therapeutic window for CAR T cell therapy in solid tumors: the Knowns and unknowns of CAR T cell biology. Front Immunol 9:2486PubMedPubMedCentralCrossRef Watanabe K, Kuramitsu S, Posey AD Jr, June CH (2018) Expanding the therapeutic window for CAR T cell therapy in solid tumors: the Knowns and unknowns of CAR T cell biology. Front Immunol 9:2486PubMedPubMedCentralCrossRef
194.
Zurück zum Zitat DeRenzo C, Krenciute G, Gottschalk S (2018) The landscape of CAR T cells beyond acute lymphoblastic leukemia for pediatric solid tumors. Am Soc Clin Oncol Educ Book 38:830–837PubMedCrossRef DeRenzo C, Krenciute G, Gottschalk S (2018) The landscape of CAR T cells beyond acute lymphoblastic leukemia for pediatric solid tumors. Am Soc Clin Oncol Educ Book 38:830–837PubMedCrossRef
195.
Zurück zum Zitat Ma Q, Garber HR, Lu S, He H, Tallis E, Ding X, Sergeeva A, Wood MS, Dotti G, Salvado B, Ruisaard K, Clise-Dwyer K, John LS, Rezvani K, Alatrash G, Shpall EJ, Molldrem JJ (2016) A novel TCR-like CAR with specificity for PR1/HLA-A2 effectively targets myeloid leukemia in vitro when expressed in human adult peripheral blood and cord blood T cells. Cytotherapy. 18(8):985–994PubMedPubMedCentralCrossRef Ma Q, Garber HR, Lu S, He H, Tallis E, Ding X, Sergeeva A, Wood MS, Dotti G, Salvado B, Ruisaard K, Clise-Dwyer K, John LS, Rezvani K, Alatrash G, Shpall EJ, Molldrem JJ (2016) A novel TCR-like CAR with specificity for PR1/HLA-A2 effectively targets myeloid leukemia in vitro when expressed in human adult peripheral blood and cord blood T cells. Cytotherapy. 18(8):985–994PubMedPubMedCentralCrossRef
196.
Zurück zum Zitat Zhu X, Prasad S, Gaedicke S, Hettich M, Firat E, Niedermann G (2015) Patient-derived glioblastoma stem cells are killed by CD133-specific CAR T cells but induce the T cell aging marker CD57. Oncotarget. 6(1):171–184PubMedCrossRef Zhu X, Prasad S, Gaedicke S, Hettich M, Firat E, Niedermann G (2015) Patient-derived glioblastoma stem cells are killed by CD133-specific CAR T cells but induce the T cell aging marker CD57. Oncotarget. 6(1):171–184PubMedCrossRef
197.
Zurück zum Zitat Morgan RA, Johnson LA, Davis JL et al (2012) Recognition of glioma stem cells by genetically modified T cells targeting EGFRvIII and development of adoptive cell therapy for glioma. Hum Gene Ther 23(10):1043–1053PubMedPubMedCentralCrossRef Morgan RA, Johnson LA, Davis JL et al (2012) Recognition of glioma stem cells by genetically modified T cells targeting EGFRvIII and development of adoptive cell therapy for glioma. Hum Gene Ther 23(10):1043–1053PubMedPubMedCentralCrossRef
198.
Zurück zum Zitat Beard RE, Zheng Z, Lagisetty KH et al (2014) Multiple chimeric antigen receptors successfully target chondroitin sulfate proteoglycan 4 in several different cancer histologies and cancer stem cells. J Immunother Cancer. 2:25PubMedPubMedCentralCrossRef Beard RE, Zheng Z, Lagisetty KH et al (2014) Multiple chimeric antigen receptors successfully target chondroitin sulfate proteoglycan 4 in several different cancer histologies and cancer stem cells. J Immunother Cancer. 2:25PubMedPubMedCentralCrossRef
199.
Zurück zum Zitat Pellegatta S, Savoldo B, Di Ianni N, et al. (2018) Constitutive and TNFalpha-inducible expression of chondroitin sulfate proteoglycan 4 in glioblastoma and neurospheres: Implications for CAR-T cell therapy. Sci Transl Med 10(430)PubMedPubMedCentralCrossRef Pellegatta S, Savoldo B, Di Ianni N, et al. (2018) Constitutive and TNFalpha-inducible expression of chondroitin sulfate proteoglycan 4 in glioblastoma and neurospheres: Implications for CAR-T cell therapy. Sci Transl Med 10(430)PubMedPubMedCentralCrossRef
200.
Zurück zum Zitat Du H, Hirabayashi K, Ahn S et al (2019) Antitumor responses in the absence of toxicity in solid tumors by targeting B7-H3 via chimeric antigen receptor T cells. Cancer Cell 35(2):221–237 e228PubMedPubMedCentralCrossRef Du H, Hirabayashi K, Ahn S et al (2019) Antitumor responses in the absence of toxicity in solid tumors by targeting B7-H3 via chimeric antigen receptor T cells. Cancer Cell 35(2):221–237 e228PubMedPubMedCentralCrossRef
201.
Zurück zum Zitat Deng Z, Wu Y, Ma W, Zhang S, Zhang YQ (2015) Adoptive T-cell therapy of prostate cancer targeting the cancer stem cell antigen EpCAM. BMC Immunol 16:1PubMedPubMedCentralCrossRef Deng Z, Wu Y, Ma W, Zhang S, Zhang YQ (2015) Adoptive T-cell therapy of prostate cancer targeting the cancer stem cell antigen EpCAM. BMC Immunol 16:1PubMedPubMedCentralCrossRef
202.
Zurück zum Zitat Wang Y, Geldres C, Ferrone S, Dotti G (2015) Chondroitin sulfate proteoglycan 4 as a target for chimeric antigen receptor-based T-cell immunotherapy of solid tumors. Expert Opin Ther Targets 19(10):1339–1350PubMedCrossRef Wang Y, Geldres C, Ferrone S, Dotti G (2015) Chondroitin sulfate proteoglycan 4 as a target for chimeric antigen receptor-based T-cell immunotherapy of solid tumors. Expert Opin Ther Targets 19(10):1339–1350PubMedCrossRef
203.
Zurück zum Zitat Feng KC, Guo YL, Liu Y et al (2017) Cocktail treatment with EGFR-specific and CD133-specific chimeric antigen receptor-modified T cells in a patient with advanced cholangiocarcinoma. J Hematol Oncol 10(1):4PubMedPubMedCentralCrossRef Feng KC, Guo YL, Liu Y et al (2017) Cocktail treatment with EGFR-specific and CD133-specific chimeric antigen receptor-modified T cells in a patient with advanced cholangiocarcinoma. J Hematol Oncol 10(1):4PubMedPubMedCentralCrossRef
204.
Zurück zum Zitat Sun B, Yang D, Dai H, et al. (2019) Eradication of hepatocellular carcinoma by NKG2D-specific CAR T-cells. Cancer Immunol Res Sun B, Yang D, Dai H, et al. (2019) Eradication of hepatocellular carcinoma by NKG2D-specific CAR T-cells. Cancer Immunol Res
205.
Zurück zum Zitat Yang D, Sun B, Dai H, Li W, Shi L, Zhang P, Li S, Zhao X (2019) T cells expressing NKG2D chimeric antigen receptors efficiently eliminate glioblastoma and cancer stem cells. J Immunother Cancer 7(1):171PubMedPubMedCentralCrossRef Yang D, Sun B, Dai H, Li W, Shi L, Zhang P, Li S, Zhao X (2019) T cells expressing NKG2D chimeric antigen receptors efficiently eliminate glioblastoma and cancer stem cells. J Immunother Cancer 7(1):171PubMedPubMedCentralCrossRef
Metadaten
Titel
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
verfasst von
Shilpa Ravindran
Saad Rasool
Cristina Maccalli
Publikationsdatum
22.11.2019
Verlag
Springer Netherlands
Erschienen in
Cancer Microenvironment / Ausgabe 2-3/2019
Print ISSN: 1875-2292
Elektronische ISSN: 1875-2284
DOI
https://doi.org/10.1007/s12307-019-00233-1

Weitere Artikel der Ausgabe 2-3/2019

Cancer Microenvironment 2-3/2019 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.