Next Article in Journal
Prognostic Value of Measurable Residual Disease in Patients with AML Undergoing HSCT: A Multicenter Study
Previous Article in Journal
A Novel Phenazine Analog, CPUL1, Suppresses Autophagic Flux and Proliferation in Hepatocellular Carcinoma: Insight from Integrated Transcriptomic and Metabolomic Analysis
Previous Article in Special Issue
Suppression of Cancer Cell Stemness and Drug Resistance via MYC Destabilization by Deubiquitinase USP45 Inhibition with a Natural Small Molecule
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunotargeting of Cancer Stem Cells

by
Ayse Sedef Köseer
1,2,
Simona Di Gaetano
2,
Claudia Arndt
3,4,
Michael Bachmann
1,3,5 and
Anna Dubrovska
1,2,5,6,*
1
National Center for Tumor Diseases (NCT), Partner Site Dresden: German Cancer Research Center (DKFZ), Heidelberg, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01307 Dresden, Germany
2
OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01309 Dresden, Germany
3
Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
4
Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
5
German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
6
Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(5), 1608; https://doi.org/10.3390/cancers15051608
Submission received: 1 February 2023 / Revised: 24 February 2023 / Accepted: 1 March 2023 / Published: 5 March 2023
(This article belongs to the Special Issue Cancer Stem Cells and Targeted Therapy)

Abstract

:

Simple Summary

Tumor cells from the same specimen are functionally heterogeneous. Cancer stem cells (CSCs) are populations of tumor cells with self-renewal and differentiation properties. CSCs are found in nearly all solid and hematological tumors and are characterized by various surface or intracellular markers. These markers can be used to develop tumor-specific antibodies, cytotoxic immune cells, vaccines, and direct immune responses to the tumor cells, including CSC populations. This review discusses the emerging CSC-directed immunotherapies, the current state of their clinical development, the approaches to improve their safety and efficacy, and future strategies to strengthen anti-CSC immunotherapy.

Abstract

The generally accepted view is that CSCs hijack the signaling pathways attributed to normal stem cells that regulate the self-renewal and differentiation processes. Therefore, the development of selective targeting strategies for CSC, although clinically meaningful, is associated with significant challenges because CSC and normal stem cells share many important signaling mechanisms for their maintenance and survival. Furthermore, the efficacy of this therapy is opposed by tumor heterogeneity and CSC plasticity. While there have been considerable efforts to target CSC populations by the chemical inhibition of the developmental pathways such as Notch, Hedgehog (Hh), and Wnt/β-catenin, noticeably fewer attempts were focused on the stimulation of the immune response by CSC-specific antigens, including cell-surface targets. Cancer immunotherapies are based on triggering the anti-tumor immune response by specific activation and targeted redirecting of immune cells toward tumor cells. This review is focused on CSC-directed immunotherapeutic approaches such as bispecific antibodies and antibody-drug candidates, CSC-targeted cellular immunotherapies, and immune-based vaccines. We discuss the strategies to improve the safety and efficacy of the different immunotherapeutic approaches and describe the current state of their clinical development.

Graphical Abstract

1. Introduction

1.1. CSC Definition and Clinical Significance

Cancer remains a leading cause of death worldwide, despite advancements in its treatment [1]. Conventional cancer treatments such as surgery, chemotherapy, and radiotherapy may be the most effective during an earlier stage of tumor development. However, treatment efficacy might be limited by tumor genetic and epigenetic heterogeneity. Each tumor is composed of cells with different features, including therapy resistance, metastatic dissemination, differentiation potential, and potency to maintain tumor growth [2]. Cancer stem cells (CSC) are a population of tumor cells that sustain tumor growth and heterogeneity [3]. CSCs were first characterized by Dick and colleagues for acute myeloid leukemia (AML) and proven to possess two fundamental properties, such as the capacity of self-renewal (e.g., an asymmetrical division that produces an identical copy and more differentiated progeny cells) and differentiation into multiple cellular subtypes observed within tumors [4,5,6]. Because of their self-renewal and differentiation capabilities, it has been shown that leukemia-initiating stem cells could repopulate and induce AML in severe combined immunodeficient hosts (SCID) after transplantation [3]. Different studies supported the tumor-initiating and tumor-maintaining properties of CSCs in various tumor entities [7,8,9]. CSCs have been characterized by many surface or intracellular markers in solid and hematological tumors. The most used indicators for CSC identification are surface markers such as CD133, CD44, and CD123, as well as the activity of some intracellular proteins such as aldehyde dehydrogenase (ALDH), as recently reviewed [10,11,12,13,14].
As the tumor develops, the tumor microenvironment (TME) becomes progressively more crucial to maintaining the growth and functions of CSCs through the interplay with cellular components and modification of the extracellular matrix (ECM). The cellular components of TME, such as endothelial cells (ECs), mesenchymal cells (MSCs), immune cells, and cancer-associated fibroblasts (CAFs), play a role in therapeutic resistance by activating CSC-related signaling pathways such as Wnt, Notch, and nuclear factor kappa B (NF-κB) pathways [15,16,17,18]. In turn, CSCs, by secreting several signaling factors, including pro-inflammatory cytokines and chemokines, recruit and alter the functions of stromal and immune cells to facilitate tumor growth and progression, especially during and after anticancer treatments, hence compromising treatment outcomes [19]. The exosomes released from CSCs can form the premetastatic niche via upregulation of vascular endothelial growth factor (VEGF) and matrix metalloproteinase-2 (MMP-2), resulting in the activation of angiogenesis and promotion of metastatic growth [20].
Some CSCs can withstand conventional treatments such as chemo- and radiotherapy, which effectively destroys a large portion of the tumor bulk, causing tumor shrinkage. However, standard treatment often fails to prevent disease recurrence if the CSCs are not completely eradicated [21,22,23]. Some CSCs can resist the direct or indirect damages induced by ionizing irradiation. The described mechanisms of the CSCs radioresistance include the activation of DNA damage response mechanisms (e.g., ATM, ATR, and Chk1/2), the scavenging of reactive oxygen species (ROS), protection from oxidative stress, activation of the anti-apoptotic pathways, and residing in protective microenvironmental niches [6,23,24]. The chemotherapy-resistant CSCs might also exhibit enhanced expression and activity of the membrane transporters of the ATP-binding cassette (ABC) family, which are linked to multidrug resistance [17,22,23,24,25,26,27,28]. Due to their self-renewal and differentiation properties, some subpopulations of CSCs, termed metastasis-initiating cells, are capable of dissemination through the bloodstream and metastasis initiation in lymph nodes and distant organs [6,29]. Therefore, CSCs might serve as biomarkers for tumor diagnosis, prognosis, and therapy response prediction, whereas CSC-related markers can be utilized to develop more efficient targeted therapies.

1.2. CSC-Directed Therapeutic Approaches

Targeted therapies against CSCs are promising strategies to prevent cancer development and reduce the risk of recurrence [30]. Therefore, the signaling pathways regulating CSC maintenance and therapy resistance can be utilized as potential treatment targets. These pathways include, e.g., Hedgehog (Hh), Notch, JAK-STAT, PI3K/AKT/mTOR, Wnt/β-catenin, NF-κB, TGF-β, and FGF signaling [31,32,33,34,35,36,37,38,39,40,41]. Deregulation of these signaling pathways has been observed in various cancers [42,43,44,45,46]. Furthermore, these signaling axes interplay to regulate the self-renewal and differentiation properties of the CSCs, TME, and tumor development. Downstream transcription and pluripotency factors such as β-catenin, signal transducer, and activator of transcription 3 (STAT3), OCT4, Sox2, Nanog, KLF4, and MYC were also characterized as potential clinical targets [12,33,36,47,48,49,50,51]. A wide variety of inhibitors have been developed to specifically target these mechanisms, and there have been many clinical trials to test their anti-tumor activities [40,52]. However, CSC heterogeneity often impedes the efficacy of the therapeutic approaches against a single molecular target. The high dependency of normal stem cells on the pathways mentioned above might also explain normal tissue toxicity and side effects in some of these trials [53]. While conventional cancer therapies, such as radio- or chemotherapy, may eliminate the tumor bulk, treatment resistance of CSCs is suspected to be responsible for recurrence. Thus, it is critical to specifically target and destroy these cells to prevent or significantly delay tumor relapse. Immune cells infiltrating the tumor are a powerful natural mechanism to target and eradicate cancer cells. However, CSCs can create an immunosuppressive microenvironment through intrinsic and extrinsic mechanisms [54]. The immunosuppressive TME is produced by CSCs and other tumor and non-cancerous cells, such as CAFs and pro-tumor immune cells (i.e., regulatory T cells (Tregs), tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), and myeloid-derived suppressor cells (MDCSs)) [55,56,57,58]. In addition, other extracellular physical and chemical factors of TME, such as pH and hypoxia, play a role in tumor immune evasion [59,60]. Tumor cells, including CSCs, can escape immune surveillance and immune-mediated cell killing by downregulating tumor-associated antigens (TAAs), increasing the expression of immune checkpoints such as programmed death-ligand 1 (PD-L1) and, therefore, inhibiting CD8+ cytotoxic T cells, and reducing the expression levels of major histocompatibility complex class I (MHC-I) and the transporter associated with antigen processing (TAP) molecules, which play vital roles in antigen processing and presentation processes [61,62,63,64,65,66,67,68,69,70,71,72,73]. Recently, it has been shown that AML stem cells suppress cytokine secretion and impair oxidative phosphorylation in T cells by overexpression of CD200 receptor [74]. It has also been demonstrated that PD-L1 was highly expressed on CD44+ CSCs compared to CD44 non-CSCs in head and neck squamous cell carcinoma (HNSCC) [65] and regulates stemness in breast cancer [71]. CSCs, due to their plasticity, can also evade immunosurveillance by entering a dormant state or converting into quiescent cells, and by selective reduction of their immunogenic properties, whereas circulating tumor cells (CTCs), which share many properties with CSCs [6], are shielded from the cytotoxic activity of natural killer (NK) cells by TANs [75,76]. A high expression of MHC-I molecules on the normal autologous cells inhibits NK cell activation and function. Tumor cells often downregulate MHC-I and therefore reduce their recognition by CD8+ cytotoxic T cells. The MHC-I downregulation in tumors has been associated with unfavorable clinical prognoses [77]. However, some CSCs, e.g., in ovarian and renal cell carcinoma, upregulate MHC-I molecules on their surface that can potentially contribute to the NK cell regulation through MHC-I specific inhibitory receptors and CSC escaping NK cell-mediated cytotoxicity [78,79]. Therefore, activating the immune response targeting of CSCs via cancer vaccines, adoptive T and NK cell therapies, monoclonal antibodies, bispecific antibodies (bsABs), and immune checkpoint inhibitors (ICIs) can be a promising approach for achieving clinical success in the treatment of different cancer types.

2. Bispecific Antibodies and Antibody-Drug Conjugates

The generally accepted view is that CSCs hijack the signaling pathways, attributed to normal stem cells, that regulate the self-renewal and differentiation processes. Therefore, the development of selective targeting strategies for CSCs, although clinically meaningful, is associated with significant challenges because CSCs and normal stem cells share many important signaling mechanisms for their maintenance and survival. Many chemical inhibitors used to target the CSC regulating signaling pathways described earlier, such as Notch, Hh, and Wnt/β-catenin signalings, also have a toxic effect on normal stem cells [80,81,82]. While there have been tremendous preclinical efforts to target CSC populations from inside the cell, considerably less effort has focused on the cell-surface targets. Cancer immunotherapies are based on inducing the anti-tumor immune response by specific activation and targeted redirecting of immune cells toward tumor cells. Since the first characterization of CSC phenotype in AML by Dick and colleagues in the late 1990s, various surface markers of CSC in hematopoietic malignancies and solid tumors have been identified, including CD44, CD133, CD117, CD123, CD47, CD98hc, and others [4,5,6]. These surface proteins play a pivotal role in the CSC interaction with their niche, cell–cell communication, nutrient uptake, and regulation of the immune system, and provide specific targets for CSC-directed immunotherapies (Figure 1).
In 1993, Seiter and colleagues published a seminal study describing the anti-tumor effect of anti-CD44v antibody using preclinical syngeneic rat xenograft models [83]. The first monoclonal antibody (mAb), rituximab, a chimeric IgG1 against the B-cell-specific antigen CD20 highly expressed on non-Hodgkin’s lymphoma (NHL) cells, was approved for treatment of NHL in 1997 [84] paving the way for immunotherapeutic applications in oncological diseases. Shortly after, the clinical studies in patients with solid tumors showed the therapeutic potential and safety of radioimmunotherapy with the (186)Re-labeled humanized mAb bivatuzumab, which as directed against a CSC-related protein, the CD44 isoform variant 6 (CD44v6) (Table 1). The transmembrane glycoproteins of the CD44 family are highly expressed in the different types of malignant cells, including CSCs, and contribute to tumorigenesis by regulating various cell surface receptors [85]. This family includes several variant isoforms, and CD44v6 is one of the best-studied isoforms serving as a co-receptor for the receptor tyrosine kinases (RTKs), such as vascular endothelial growth factor receptor-2 (VEGFR-2), epidermal growth factor receptor (EGFR), and mesenchymal-epithelial transition factor (c-Met) [85,86]. Unfortunately, early clinical testing of bivatuzumab merstansine, a CD44v6-specific antibody conjugated with an antimicrotubule agent, in 2006 led to a fatal outcome related to the binding of the anti-CD44v6 antibody to skin keratinocytes and associated severe skin toxicity [87]. Therefore, further development of the CD44-targeted drug conjugate was discontinued. A newer anti-CD44 recombinant humanized antibody, RG7356, showed an acceptable safety profile in a clinical trial for patients with advanced solid tumors; however the clinical efficacy was modest. The trial was terminated early as no dose–response relationship was reported [88]. However, in patients with AML, RG7356 induced differentiation of CD34+ leukemic stem-like cells and accumulation of CD68+ macrophages [89].
Integrin associated protein (IAP), or CD47, is another promising target overexpressed on cancer cells, including CSCs, in different types of hematological and solid tumors. CD47 plays a critical role in regulating the homeostasis of immune cells (e.g., T cells, macrophages, and dendritic cells (DCs)), including their activation, differentiation, migration, and death. It is also crucial for tumor growth, as CD47 expression on solid tumors results in evasion of the innate immune response [90,91]. CD47 exerts these activities by interacting with integrin receptors, e.g., αvβ3, and activating integrin-dependent signaling molecules such as focal adhesion kinase (FAK) [92]. CD47 also binds to a transmembrane glycoprotein, signal regulatory protein α (SIRPα), therefore activating protein tyrosine phosphatases such as Src homology 2 (SH2) domain-containing phosphatase-1 (SHP-1) and SHP-2 [91]. In addition, CD47 also acts as a receptor for thrombospondin 1 (TSP-1) [93]. Of importance, previous studies using syngeneic prostate tumor models in CD47 deficient mice showed that inhibition of CD47 might have opposite consequences for tumor growth depending on the target cells: CD47/TSP-1 inhibition in tumor stromal ECs induces angiogenesis and tumor progression, and decreased TSP1 production in tumors from CD47 deficient mice reduced macrophage recruitments, whereas blocking the binding of CD47 on tumor cells with SIRPα on macrophages and DCs might, in contrast, induce an antitumor immune response and reduce tumor growth [94]. Breast cancer progression is associated with the development of intratumoral hypoxia and activation of hypoxia-inducible transcriptional factors (HIFs). HIF1 was shown to stimulate the CD47 expression in breast CSCs, enabling them to avoid macrophage-mediated phagocytosis. In addition, CD47 positively regulates breast CSC phenotypes and properties [95]. Furthermore, inhibition of CD47 by anti-CD47 antibodies was shown to effectively target pancreatic CSCs by increasing macrophage-mediated immunity and tumor cell apoptosis [96]. Of importance, CD47 expression and, therefore, immune evasion in ovarian CSCs has been shown to be induced by surrounding bulk tumor cells [97]. Recent studies also demonstrated that CD47 transcription is regulated via HER2–NF-κB pathway, and antibody-mediated blockage of both CD47 and HER2 synergized with radiotherapy for the treatment of syngeneic mouse breast tumors. Notably, radiotherapy increased the rate of macrophage-mediated phagocytosis in the tumors treated with anti-CD47 antibodies in combination with Herceptin or single anti-CD47 antibodies compared to radiotherapy applied alone in the orthotopic breast tumor models. This study showed synergistic tumor inhibition by a combination of radiotherapy plus anti-CD47 and anti-HER2 immunotherapy [98]. IBI188, also known as Letaplimab, is a recombinant human anti-CD47 mAb. IBI188, in combination with azacytidine (AZA), a DNA methyltransferase inhibitor, showed promising efficacy and a manageable toxicity profile in patients with newly diagnosed higher risk myelodysplastic syndrome (MDS) (NCT04485065) [99]. IBI188 is currently being tested in several early-phase clinical trials in combination with AZA in patients with AML (NCT04485052) and as a single treatment in patients with advanced solid cancers and lymphomas (NCT03717103 and NCT03763149).
Another attractive target is the interleukin-3 receptor (IL-3R) alpha chain, or CD123, which is highly expressed in the undifferentiated precursor (blast) cells, but present at a low level on normal hematopoietic stem cells, and is associated with the development of AML, acute lymphoblastic leukemia (ALL), B-lymphoid leukemia (BLL), hairy cell leukemia (HCL), Hodgkin lymphoma, blastic plasmacytoid dendritic neoplasms (BPDCN), and MDS [100,101]. The early clinical trials for the recombinant IL-3 fused with diphtheria toxin (SL-401, or tagraxofusp) targeting CD123 showed clinical effectiveness, especially in patients with BPDCN [102,103]. Based on these clinical studies, tagraxofusp was approved by the Food and Drug Administration (FDA) for the treatment of patients with BPDCN cancer [102].
These clinical investigations fueled the further development of the anti-CD123 treatment using immunological approaches. In particular, a humanized anti-CD123 mAb Talacotuzumab (JNJ-56022473, CSL362) was engineered to have increased affinity for CD16 on NK cells and target CD123 positive cells through NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC) [104]. The preclinical studies demonstrated an efficient depletion of CD123+ blasts in samples from AML patients [104] and in AML xenograft models that was mediated by both allogeneic and autologous NK cells [105,106]. However, in the clinical analysis as a single agent in elderly high-risk MDS or AML patients, Talacotuzumab demonstrated significant toxicities that resulted in early treatment discontinuation as well as in limited clinical efficacy, explained by the alteration in the NK- and T cell repertoire in these patients before treatment start [105]. Furthermore, Talacotuzumab was tested for its safety and efficacy in combination with decitabine, a chemotherapy drug, and in comparison with decitabine alone in patients with AML. These clinical studies showed no improvement of clinical efficacy for combination treatment compared to the single treatment with decitabine [107].
To improve the clinical effectiveness of the CD123-targeted antibody therapy, an antibody-drug conjugate was developed. To this end, a humanized high-affinity anti-CD123 antibody was linked to the DNA-alkylating cytotoxic compound from the class of indolinobenzodiazepine pseudodimer (IGN) [108]. This anti-CD123-targeting antibody-drug conjugate, called IMGN632, demonstrated robust antitumor efficacy in preclinical models for different hematological malignancies, including AML [108] and BPDCN [109] xenograft models, while sparing normal bone marrow cells, which express low levels of CD123. The early clinical study of IMGN632, given as monotherapy or in combination with AZA and venetoclax (VEN), a Bcl-2 inhibitor, in patients with CD123-positive AML, showed a manageable safety profile. The administration of IMGN632 was associated with a high objective response rate (ORR) of 75% and composite complete remission rate (CCR) of 40% in the high-intensity cohort of patients with AML, whereas ORR/CCR rates were even higher in the cohort of VEN-naïve patients (100%/60%, respectively) [110].
In contrast to the conventional monospecific antibody, bsAB are artificially engineered antibodies with dual specificity for different epitopes attributed to the same or different antigens, therefore offering a variety of therapeutic opportunities, including retargeting of immune cells and modulations of the ligand and receptor action with an efficacy that is hard to achieve for single antibodies [111]. The application of bsAB in cancer treatment is a fast growing area of clinical research. The first clinically approved bsAB, Catumaxomab, targeting the T cell antigen CD3 and human epithelial cell adhesion molecule (EpCAM), was designed to recruit T cells to tumors [112]. Since then, bsABs are one of the most promising tools for targeting tumor malignancies through the recruitment of immune cells such as T cells or NK cells (cell-bridging bsAB) or by antigen cross-linking [113]. In the laboratory, a bsAB is generated by genetic engineering, chemical conjugation of two purified monoclonal antibodies, or by quadromas, in which fused hybridoma cells produce bsABs along with non-functional by-products [114]. Recombinant bsABs might have different designs described in detail elsewhere [111,115,116]. The T cell engaging approach with cell-bridging bsAB was used to develop several CSC-related bsABs, which bind to the patient’s T cells through CD3/T cell receptor (TCR), CD28, or other surface molecules mediating T cell activation and proliferation. On the other hand, they bind to target antigens on tumor cells. Thus, T cell engaging bsABs specifically redirect T cells to target-positive tumor cells. The antigen cross-linking with bsAB can be used for simultaneous blocking and inhibition of two targets on the surface of tumor cells, including CSCs, thereby preventing ligand-induced activation and tumor growth [113]. Simultaneous bsAB binding to the checkpoint regulators on the surface of T cells e.g., PD-L1 and CTLA-4, might potentiate anti-tumor immune response. The design of novel bsAB enables binding to multiple targets, therefore making them a multi-specific antibody (MsAb) [115,117]. Table 1 includes several antibody-based CSC-targeted therapies that have already entered clinical trials. These therapies, in particular, include CD123 × CD3 bsAB, whose commercial name is Flotetuzumab (MGD006). Flotetuzumab has been evaluated in phase I/II clinical trials for refractory AML, and has shown encouraging anti-leukemic activity and acceptable safety. In particular, complete remission was observed in 26.7% of patients with refractory AML [118].
Several CD47-targeted bsABs are currently being tested in early-phase clinical trials, including HX009, PD-1 × CD47 bsAB, tested in patients with relapsed or refractory lymphoma (NCT05189093). HX009 binds to PD-1 expressed on T cells and CD47 on tumor cells. By this, HX009 blocks the binding of CD47 on tumor cells with SIRPα on macrophages and DCs and, therefore, activates macrophage-mediated phagocytosis of the CD47-expressing tumor cells. On the other hand, the binding of HX009 to PD-1 prevents the interaction between PD-1 and its ligands, PD-L1 and PD-L2, and inhibits the downstream signaling pathways. This signal inhibition recovers effector T cell functions and activates cytotoxic T cell-mediated antitumor immunity [119]. Preliminary results suggest that HX009 is well-tolerated and shows strong antitumor activity [120,121]. Another CD47 targeting bsAB, IBI322, with a dual specificity for CD47 and PD-L1 [122], is also being tested in several clinical trials in patients with advanced malignant tumors (NCT04328831, NCT04912466, NCT04338659) and hematologic malignancies (NCT04795128), but no clinical data has been reported yet. In addition, bsAB targeting CD47 and the B-lymphocyte antigens CD20 (IMM0306) or CD19 (TG-1801) also entered clinical testing for patients with B-cell lymphoma or chronic lymphocytic leukemia (CLL), (NCT04806035, NCT03804996) and B-cell NHL (NCT04746131).
Another example is Amivantamab, a bsAB targeting EGFR and c-Met driving tumor growth in patients with non-small cell lung cancer (NSCLC). NSCLC progression is frequently associated with activating mutations in the kinase domain of EGFR. Some of these mutations, such as in-frame base pair insertions in exon 20 (ex20-ins), result in tumor resistance to conventional EGFR tyrosine kinase inhibitors [123]. EGFR and another receptor tyrosine-protein kinase, c-Met, cooperatively regulate tumor cell proliferation, migration, and activation of the downstream signaling pathways. Because of the synergy between the EGFR and c-Met pathways, their dual inhibition is critical for the treatment of NSCLC [124]. Amivantamab bsAB inhibits the activation of both receptors by binding to their extracellular domains, preventing ligand-induced activation and triggering receptor degradation. In addition, it activates tumor destruction by effector immune cells through Fc-mediated mechanisms, such as ADCC [125]. Amivantamab was demonstrated to be efficient against NSCLC with a resistance mutation in EGFR and c-Met activation [125]. Clinical trials (e.g., CHRYSALIS; NCT02609776; and others) have shown acceptable toxicity and anti-tumor efficacy of Amivantamab in patients with locally advanced or metastatic NSCLC and EGFR Ex20ins mutations based on the ORR of about 40% and duration of response [126]. In 2021, Amivantamab was approved by the US FDA for the treatment of patients with advanced or metastatic NSCLC with EGFR ex20-ins mutations, whose disease had progressed during or after platinum-based chemotherapy [127]. Of importance, c-Met has been characterized as a regulator of CSC populations in different types of solid tumors, including pancreatic cancer [128,129], prostate cancer [130], and colorectal cancer [131]. A novel bsAB c-Met × CTLA-4 targeting c-Met and CTLA-4, a negative regulator of T cell activation, showed significant anti-tumor activity in lung cancer models in vitro and in vivo. This anti-tumor effect was at least partially mediated by the inhibition of the CD166+ positive lung CSC populations [131].
EpCAM is another marker highly expressed in tumor cells, including CSCs [132]. Catumaxomab (Removab) is a EpCAM × CD3 bsAB approved in the European Union in April 2009 for the treatment of malignant ascites, a condition developing in patients with different types of epithelial cancers [112]. It is called a trifunctional antibody (trAb) due to its ability to bind tumor cells, T cells, and accessory cells (e.g., macrophages, DCs, and NK cells) through its intact Fc region [133]. The results from clinical studies (NCT00836654) demonstrated that the application of catumaxomab is associated with the depletion of CD133+/EpCAM+ CSCs from malignant ascites in patients with the ovarian, pancreatic, and gastric cancer [134].
Antibody-based cancer immunotherapy, particularly monospecific and bsAB therapy, is a promising strategy for cancer treatment [113], and most bsABs are still in the early phase of clinical trials. However, some biological and clinical factors might compromise the efficacy of bsAB and limit their clinical translation. In particular, the application of bsAB, like some other types of immunotherapy, e.g., chimeric antigen receptor (CAR) T cells, might cause potentially fatal adverse effects such as cytokine release syndrome (CRS)—systemic inflammatory response associated with high cytokine levels in peripheral blood [135]. There is a hope that many bsABs, currently being tested in more than 300 clinical trials, will be introduced in clinical practice [136]. However, only one bsAB for treating solid malignancies, Amivantamab, an EGFR/c-Met specific bsAB for the treatment of patients with NSCLC, has been clinically approved [137]. Limitations that might impact the efficacy of the therapeutic antibodies for the treatment of solid tumors include poor tumor penetration, unequal distribution, and endocytic clearance in tumor cells [138]. In addition, some tumor cell populations, such as CSCs, can occupy hypoxic niches where antibody delivery is complicated due to the sparse presence of blood vessels [23,139]. Notably, a robust assay to measure CSC functions in tumor samples, besides surface marker expression, is yet missing. Given the heterogeneity and plasticity of CSC, developing these assays would be essential for the reliable evaluation of CSC-directed immunotherapy in clinical trials [40]. The loss of the tumor antigen, which serves as bsAB's target and consequent tumor resistance and immune escape, is an additional complication for the clinical application of bsAB [140,141]. Furthermore, no single surface marker is currently available to define the entire CSC population in a given tumor entity, or even in one individual tumor [142]. There is evidence of high variability of the CSC phenotype between patients with a given type of cancer and a high heterogeneity of CSC within one individual tumor. Mutation changes, epigenetic reprogramming, and microenvironmental stimuli induce CSC plasticity during the treatment, cancer progression, and upon relapse [142,143,144]. An essential challenge is the scarcity of tumor-specific antigens that are not present in normal tissues: more than 70% of known CSC surface markers appear on normal adult and embryonic stem cells [145]. Even low epitope expression on normal cells can be associated with severe normal tissue toxicities from immune therapy [146]. Therefore, improving treatment specificity by inventing new cancer-specific treatment targets and sequential or combinational targeting of two or more tumor antigens during the course of treatment could reduce normal tissue toxicity and overcome tumor antigen escape. Thus, developing multispecific antibodies [147] and multispecific CAR T cell immunotherapy (as described in the next chapter) could be a promising strategy to improve treatment options for patients with malignant tumors.
Table 1. Selected clinical trials for antibody-based therapies targeting tumor cells, including CSCs.
Table 1. Selected clinical trials for antibody-based therapies targeting tumor cells, including CSCs.
Specificity/Generic NameDescriptionTumor Entity TestedClinical Trials/ApprovalsReferences
CD44v6/Bivatuzumab (BIWA 4)mAb against
CD44v6, (186)Re-labeled
Inoperable recurrent and/or metastatic HNSCC, NSCLC, breast cancerPhase I: NCT02204059,
NCT02204046,
NCT02254018
Outcome: Antitumor effects and effective tumor targeting was observed. Administration is well tolerated.
[148,149]
CD44v6/Bivatuzumab—mertansinemAb against
CD44v6, conjugated mertansine
Incurable HNSCC or esophagus squamous cell carcinoma (ESCC), recurrent or metastatic breast cancerPhase I: NCT02254044, NCT02254031,
NCT02254005,
NCT02254018
Outcome: one fatal drug-related adverse skin event had occurred. Further clinical development was discontinued.
[87,150,151,152]
CD44v6/RG7356mAb against
CD44v6
Advanced CD44-expressing solid malignancies.Phase I study: NCT01358903 Outcome: acceptable safety profile, modest clinical efficacy was observed. The study was terminated due to the absence of a clinical and pharmacodynamic dose-response relationship[88]
CD44v6/RG7356mAb against
CD44v6
AMLPhase I study; NCT01641250 Outcome: the treatment was generally safe and well tolerated. Out of 44 patients, two patients achieved complete or partial response and one patient had stable disease.[89]
CD123/JNJ-56022473
/Talacotuzumab
7G3 mAb against CD123Elderly high-risk MDS or AML failing hypomethylating agentsPhase II: NCT02992860 Talacotuzumab as a single agent;
Outcome: limited clinical efficacy and significant toxicity
[153]
CD123/JNJ-56022473
/Talacotuzumab
7G3 mAb against CD123CD123-positive AMLPhase II/III study: NCT02472145 Talacotuzumab in combination with decitabine versus decitabine alone; Outcome: no improvement in efficacy versus decitabine alone[107]
CD123/IMGN632mAb G4723A against CD123 conjugated with DNA-alkylating payload of the IGN cytotoxic compoundsCD123-positive AMLPhase Ib/II study;
NCT03386513 IMGN632 is given as monotherapy or in combination with AZA and/or VEN; Outcome: manageable toxicity profile; high ORR (of 75%) and CCR (of 40%) in high intensity cohort; ORR/CCR rates were even higher in the cohort of VEN-naïve patients (100%/60%, respectively)
[110,154]
CD47
IBI188/Letaplimab
mAb against
CD47
Newly diagnosed higher risk MDSPhase I study: NCT04485065
The preliminary results suggest that IBI188 in combination with AZA showed a promising efficacy and a manageable toxicity profile
[99]
CD123 and CD3
Flotetuzumab/
MGD006
bsAB (CD3ε × CD123)Relapsed/refractory AMLPhase I/II study: NCT02152956
Outcome: acceptable safety profile, encouraging anti-leukemic activity (the complete remission rate (CRR)/CRR with partial hematological recovery was 26.7%; an overall response rate was 30.0%
[118]
CD47 and PD-1
HX009
bsAB antibody binding CD47 and PD-1Relapsed or refractory lymphomaPhase I/II study: NCT0409776, The preliminary results suggest that HX009 is well-tolerated and showed strong antitumor activity[120,121]
EGFR and c-MET
Amivantamab/Rybrevant/
JNJ-61186372
bsAB antibody binding EGFR with one Fab and c-Met with the other FabAdvanced or metastatic solid tumors including EGFR-mutated NSCLCAmivantamab was approved by the US FDA for the treatment of patients with advanced or metastatic NSCLC with EGFR ex20ins mutations, whose disease has progressed on or after platinum-based chemotherapy.[125,126,127]
EpCAM and CD3
Catumaxomab/Removab
EpCAM × CD3; trAb binding tumor cells, T cells, and accessory cells (e.g., macrophages, DC, and NK cells through its intact Fc regionMalignant ascites derived from epithelial tumorsCatumaxomab was approved in the European Union in April 2009 for the treatment of malignant ascites, but was withdrawn in 2017 for commercial reasons.[112]

3. CSC-Targeted Immune Cells

In contrast to the antibody-based immunotherapy available off-the-shelf, adoptive cell therapy (ACT) has been developed as more individualized anti-cancer immunotherapy when cytotoxic lymphocytes, such as T cells or NK cells, are customized for each patient. The cytotoxic activities of T cells and NK cells are mediated by releasing the pore-forming protein perforin, granzyme serine protease, and pro-inflammatory cytokines, making them attractive candidates for the ACT [155,156,157]. The objective regression of cancer after ACT was first documented in 1988 for patients with metastatic melanoma treated with autologous tumor-infiltrating lymphocytes [158]. The promising results of the ACT applications stimulated the genetic engineering of immune cells to improve tumor-specific response and to broaden ACT application to other types of cancer. For this, T cells were genetically modified using viral vectors to overexpress either conventional TCRs or artificial CAR. T cells equipped with TCRs recognize the antigens presented by MHC molecules on the surface of tumor cells, whereas T cells with CARs recognize tumor-specific cell surface antigens that do not need to be restricted by MHC [159].
The idea for CAR design was coined by Gross and colleagues in 1989 [160]. The CAR structure contains four major modules: (I) the extracellular domain responsible for the antigen binding and made by the variable domains of the heavy (VH) and light (VL) chains of tumor-specific immunoglobulins, (II) the spacer domain connecting the extracellular domain to the transmembrane domain, (III) the transmembrane domain, and (IV) the cytoplasmic domain derived from activating immune receptors, e.g., TCR. The initial design of CARs included a single intracellular CD3ζ motif. These CARs were able to efficiently trigger the signal for the activation and effector function of T cells against target antigen-expressing cells [160]. However, the early phase clinical trials for ovarian cancer patients showed the lack of patient antitumor response associated with a short-term persistence of genetically modified T cells in the blood of patients and poor T cell trafficking to the tumor sites [161]. The next generations of CARs included additional activation motives from costimulatory molecules, such as CD28 [162,163] and 4-1BB/CD137 [164,165], in the intracellular part of CAR molecules. This design is associated with robust T cell proliferation and full activation, better persistence in vivo, and amelioration of T cell exhaustion. The selection of the optimal CAR design is a fast-expanding field of immunology, including specificity, affinity and avidity of antigen binding regions, spacer length, and transmembrane domain interactions, as well as type, number, and order of the costimulatory domains [166]. The design of CAR constructs for NK cells includes similar components. However, the unique characteristics of NK cells motivated researchers to develop CARs containing NKG2D immunoreceptor elements and additional signaling subunits such as DAP10, DAP12 [167,168,169].
The manufacturing of CAR-T cells consists of multiple steps, including the collection of peripheral blood mononuclear cells (PBMCs) from the patient, T cell isolation, activation, genetic manipulation for CAR expression, expansion, and quality checks for further applications [170]. CAR-T cell-based immunotherapy is currently being investigated in more than 800 clinical trials [171]. Due to impressive clinical success rates, several autologous CAR-T based anti-cancer treatments have been clinically approved [172]. Kymriah (Tisagenlecleucel) was the first anti-CD19 CAR-T therapy approved by the FDA in 2017 for relapsed or refractory pediatric and young-adult B-cell ALL, and then for adult relapsed or refractory diffuse large B-cell lymphoma [173]. Since then, four additional anti-CD19 CAR-T-based anti-cancer therapies and one anti-B-cell maturation antigen (BCMA) CAR T product have been approved for the treatment of B-cell lymphomas/leukemias and multiple myeloma [172,174,175], respectively. Analysis of the ongoing clinical trials evaluating CAR-T cells revealed that CD19 and BCMA are the most frequently used antigens for CAR-T therapies against hematological malignancies, whereas, for solid tumors, CAR-T cells are directed against, e.g., mesothelin, carcinoembryonic antigen (CEA), Mucin 1 (MUC1), HER2, EGFR, and glypican-3 (GPC3) [171]. Among these targets, MUC1 was characterized as a stemness driver in colorectal cancer where MUC1 forms a complex with MYC transcriptional factor and activates the expression of leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5) gene, a marker of the intestine stem cells and CSCs [176]. In addition, MUC1 also activates the expression of other CSC markers, such as ALDH1, BMI1, and the pluripotency factors Oct4, Nanog, and Sox2171 [177]. MUC1 induces tumor cell plasticity and epigenetic reprogramming by coupling MYC activation with activation of other transcription factors such as STAT3, NF-κB, and E2F [178]. Furthermore, several CSC-targeted CAR-based therapies entered early-stage clinical trials. These clinical trials include CAR-T cells targeting CD44v6 in stomach cancer lymphosarcoma, AML, and multiple myeloma; CD133 in relapsed and/or chemotherapy refractory advanced malignancies; c-Met in patients with melanoma and breast carcinoma [179], EpCAM in nasopharyngeal carcinoma, breast cancer, gastric cancer and other EpCAM positive solid tumors (Table 2).
Although several CAR-based therapies for blood cancers have been clinically successful, CAR-modified immune cells still face several hurdles in clinical application for solid tumors. Different biological factors might decrease CAR-T cell efficacy, including a loss of tumor antigen (antigen escape) associated with the development of therapy resistance, lack of antigen specificity resulting in the “on-target off-tumor” toxicity against normal tissues, CAR-T exhaustion [180], immunosuppressive tumor microenvironment, and poor CAR-T cell trafficking and tumor infiltration [181]. Furthermore, like other immunotherapy types, such as antibody-based cancer treatment, CAR-T cell-based therapy can cause severe side effects, including the above-described CRS and immune effector cell-associated neurotoxicity syndrome (ICANS) [182]. In addition, the ex vivo manipulation of T cells for the autologous CAR-T cell production is mainly performed as a manual or semi-automated process resulting in considerable variability and high acquisition costs [183].
The growing awareness of these limitations has driven the development of new generations of CAR T cells, and various strategies are being pursued to overcome the existing difficulties. Bispecific CARs, multi-CARs, and logic-gated CAR T cells are being developed to enhance the specificity as well as efficiency, and thereby reduce off-tumor effects of CAR T cell products [184,185,186]. The incorporation of suicide switches [187,188] and “biodegradable” CAR T cells [189,190] are further attempts to particularly address the safety concerns of CAR T cell therapy. Besides these and many other strategies, modular adapter CAR platforms currently represent a rapidly and steadily growing field to create safer and more efficient CAR T cell retargeting strategies [191]. The main concept of this technology is to separate the effector and targeting functions of conventional CARs (Figure 2A). Thus, adaptor CAR T cells are designed to be redirected for tumor cell killing only in combination with a second tumor-specific component. Unlike conventional CAR-T cells, they do not recognize any surface antigen and are, therefore, switched off by default. For cross-linking with target cells, and thus activation, a so-called adapter molecule is required. In principle, it consists of a tumor-specific binding site and an interaction site for CAR-T cell recruitment. The modular concept of adapter CAR approaches allows (I) control of therapy-related side effects by adaptor molecule dosing, (II) highly flexible targeting of different tumor-associated antigens, either simultaneously or sequentially, thereby increasing treatment specificity/efficacy and lowering the risk of tumor escape and off-tumor effects, and (III) co-delivery of payloads to locally enhance anti-tumor effects [191] (Figure 2B). The interaction of adapter CAR T cells and the adapter molecules is based on different connection systems following two major concepts: (I) adapter CARs recognizing diverse tags incorporated into the adapter molecule, e.g., peptide tags [192,193,194], FITC [195], biotin [196], dinitrophenyl [197], and (II) adapter CARs redirected to tumor cells via bispecific antibodies [198,199,200,201,202]. The interaction between human La/SS-B peptide epitopes (E5B9, E7B6) [203,204] and the corresponding anti-La antibody binding domains [205], for example, led to the development of both the peptide-binding adaptor CAR “UniCAR” [206] (Figure 2C) and the corresponding bsAB-binding adaptor CAR “RevCAR” [201] (Figure 2D). Under physiological conditions, naturally occurring human La/SS-B resides in the nucleus and is inaccessible to unwanted interactions with UniCAR/RevCAR components, rendering the corresponding adapter CAR-T cells inactive. The UniCAR is a second-generation CAR constructed by fusing an anti-La single-chain fragment variable (scFv) as an extracellular binding domain to the transmembrane and intracellular domain of CD28, as well as the signaling domains of CD3zeta (Figure 2C). UniCAR T cells can be cross-linked with tumor cells and induce tumor cell lysis only in the presence of a tumor-specific target module (TM). Such TMs are designed by connecting a tumor-specific binding moiety, e.g., peptide ligands [207], nanobodies [208], or antibody-derived fragments [209,210,211,212], to the La-epitopes. Vice versa, in the RevCAR system, La-peptide epitopes are used as the extracellular domain of the RevCARs (Figure 2D) [201,213]. Thus, bsABs (termed RevTMs) simultaneously target the La epitope and a tumor-associated antigen. They are utilized to bridge CAR T cells and tumor cells and, thereby engaging RevCAR T cells for efficient tumor cell lysis. Other recent designs have further focused on developing adaptor CARs that recognize common features of already approved drugs, e.g., the P329G Fc mutation of therapeutic antibodies [214] or a binding pocket within the Fab arm of monoclonal antibodies [215]. In preclinical in vitro and in vivo studies, adapter CAR T cells have been successfully redirected against various CSC-related antigens, such as CD123 [213,216,217], EpCAM [196,200,218] and CD98hc [219]. By modifying T cells with two different RevCARs and fine-tuning the selected adapter molecules, the RevCAR system was successfully applied for dual “AND”-gate targeting of CD33 and CD123 on AML blasts, highlighting the versatility of the platform technology (Figure 2B) [213]. Such logic-gated approaches will allow more specific targeting of tumor cells, including CSCs, thereby reducing unwanted toxicities against healthy tissues. More recently, CD123-directed UniCAR T cells showed the first proof-of-concept for functionality and controllability in a phase I clinical trial with AML patients [220,221]. So far, treatment was completed in 12 patients and proved to be tolerable, with overall mild adverse effects and only one dose-limiting toxicity. The observed treatment-related side effects, e.g., myelosuppression, disappeared rapidly when the infusion of the adapter molecules was interrupted. After the patients had recovered, therapy could be resumed by repeated TM administration. Ten patients treated with UniCAR-T-CD123 therapy have shown a clinical response, including two complete remissions with incomplete count recovery and four partial responses [220,221].
In contrast to CAR-T cells, CAR-NK cells are less likely to induce off-tumor toxicities and adverse side effects, such as CRS and neurotoxicity, that could be partially attributed to their short lifespan in the bloodstream and different types of secreted cytokines [222]. The preclinical and clinical studies demonstrated that allogenic NK cells have a low risk for graft versus host disease (GVHD) [223,224,225], can be prepared from different sources (e.g., cord blood, haploidentical donors, induced pluripotent stem cells, iPSC) [224,225,226,227] and expanded ex vivo for “off-the-shelf” allogeneic applications [228]. Unlike CAR-T cells, CAR-NK cells kill tumor cells through CAR-mediated and CAR-independent mechanisms. Due to these unique properties, several clinical trials are exploiting CAR-NK cells’ cytotoxic activity targeting specific tumor antigens in patients with hematopoietic malignancies and solid tumors [222]. These clinical trials include targeting the CSC-related antigens, such as CAR-NK cells, against CD123 in AML and MUC1 in solid tumors (Table 2). Although CAR-NK therapy emerged as a more cost-efficient and safer immunotherapy than CAR-T cells, it is challenged by the short in vivo lifespan and limited proliferation capacity of NK cells that compromise long-lasting therapeutic responses. Similar to CAR-T cells, CAR-NK anti-cancer efficiency is reduced by tumor heterogeneity, immune suppressive microenvironment, off-tumor cell killing, and poor CAR-NK infiltration into solid tumors [229].
Table 2. Selected clinical trials for immune cell-based therapies targeting CSC-related tumor antigens.
Table 2. Selected clinical trials for immune cell-based therapies targeting CSC-related tumor antigens.
Specificity
/Generic Name
DescriptionTumor Entity TestedClinical TrialsReferences
CD44v6CAR-T cellsCD44v6 positive stomach cancer, lymphosarcomaPhase I/II;
NCT04427449
No results were posted.
CD44v6MLM-CAR44.1 T cells; CD44v6 CAR-T cells were genetically modified to express herpes simplex virus (HSV)-TK Mut2 suicide gene to minimize toxicityAML, Multiple MyelomaPhase I/II; NCT04097301
Outcome: terminated due to the inability to close the study in a clinically relevant time frame.
[230]
CD133CAR-T cellsRelapsed and/or chemotherapy refractory advanced malignancies (liver cancer, pancreatic cancer, brain tumor, breast cancer, ovarian tumor, colorectal cancer, acute myeloid and lymphoid leukemia)Phase I/II; NCT02541370
Outcome: out of 21 enrolled patients, 1 had a partial response, 14 had stable disease during 2–16.3 months, and 6 progressed after treatment start; hyperbilirubinemia was the most common high-grade adverse event
[231]
CD38-CART/
CD33-CART/
CD56-CART/
CD123-CART/
CD117-CART/
CD133-CART/
CD34-CART/
MUCl-CART
single CAR-T or double CAR-T cells with CD33,CD38,
CD56,CD123,
CD117,CD133,
CD34 or MUCl
AMLPhase: n/a; NCT03473457 Outcome: terminated because the therapeutic effect was not as expected.
No results were posted
EpCAMCAR-T cellsNasopharyngeal carcinoma, breast cancer, gastric cancer and other
EpCAM positive solid tumors
Phase I; NCT02915445
No results were posted
EpCAM- and TM4SF1CAR-T cellsRefractory/recurrent advanced pancreatic cancer, colorectal cancer, gastric cancer or lung cancerPhase: n/a; NCT04151186
No results were posted
CD123CAR-NK cellsAMLPhase: I;
NCT05574608
No results were posted
MUC1CAR-NK cellsMUC1 positive relapsed or refractory solid tumorPhase I/II; NCT02839954
No results were posted
CD123Preconditioning (lymphodepletion) with cyclophosphamide and fludarabine followed by treatment with UniCAR-T and CD123 TMRelapsed/refractory AMLPhase I; NCT04230265
Outcome: the initial results suggest that the treatment is well tolerated with mild adverse effects; out of three treated patients, one patient had a partial remission and two patients had complete remission with incomplete hematologic recovery
[220]

4. Cancer Stem Cell Vaccines

An additional promising approach to boost immune responses against CSCs is to use CSCs as a source of antigens to pulse antigen-presenting DCs and develop anti-CSC DC vaccines. DCs were first discovered in 1973 by Ralph Steinmann and Zanvil A. Cohn as a phagocytic cell population in the murine spleen [232]. DCs are an immune cell population bridging innate and adaptive immune responses. They present the processed epitopes to CD4+ T cells and CD8+ T cells through MHC II and MHC I, respectively, and secrete cytokines critical for the survival and proliferation of T cells, NK cells, and T cell tumor infiltration [233]. DC vaccination is a promising form of immunotherapy, and many DC vaccines have been developed in the past years and tested in clinical trials. The DC vaccines are most commonly prepared by ex vivo differentiation of the autologous precursor cells into immature DCs, the maturation of DCs by addition of a cytokine cocktail, and then pulsing them with cancer antigens in the form of antigen peptides, tumor cell lysates, exosomes, or mRNAs. Afterwards, mature DCs are administered back into the patients where they activate antigen-specific T cells [231] (Figure 3). Numerous clinical studies have shown that DC vaccinations are both safe and efficient anti-cancer therapies capable of inducing an immunological response, increasing tumor-infiltrating lymphocytes, and improving overall survival (OS) [231,234].
Dendritic CSC vaccination (CSC-DC) is a promising form of DC-mediated immunotherapy. The administration of DCs loaded with MUC1-derived peptide, alone or in combination with other tumor-specific antigens, has been tested in several clinical trials for patients with refractory NSCLC [235], pancreatic cancer [236,237,238], biliary cancer [238], and castration-resistant prostate cancer (CRPC) [239]. These studies showed that the vaccine was well tolerated and associated with clinical responses.
Another approach employing the peptide-loaded DCs is the ALDH peptide-based DC vaccine [240,241]. ALDH is a family of metabolic enzymes responsible for the detoxification of intracellular aldehydes through their oxidation to the carboxylic acids [242]. A high level of ALDH activity measured by the ALDEFLUOR analysis is used as a marker to isolate CSC populations in a wide variety of solid malignancies, such as breast cancer, prostate cancer, lung cancer, colon cancer, sarcoma, and HNSCC [240,242,243]. The ALDH family includes 19 genes. Out of them, several ALDH isoforms are highly expressed in CSCs, including ALDH1A1 and ALDH1A3 [242]. In addition to being markers for CSC, both genes play critical functional roles in the regulation of the activation of the retinoic acid signaling, PI3K/AKT pathway, ethanol and amino acid metabolism, and cell defense against ROS [242,243,244]. The critical role of ALDH proteins in tumor development and therapy resistance makes them promising therapeutic targets. In the first adoptive therapy experiments for targeting ALDH-positive CSCs, ALDH1A1-specific CD8+ T cells were induced in vitro by the DCs pulsed with ALDH1 [88,89,90,91,92,93,94,95,96] peptide and injected intravenously into the xenograft-bearing immunodeficient mice [241]. This study demonstrated that ALDH1 peptide-specific CD8+ T cells inhibited primary tumors growing subcutaneously and lung metastases [241].
Similar to other immunotherapy directed against a single antigen, the efficacy of the peptide-based DC vaccines could be compromised by heterogeneity and plasticity of antigen expression in tumor cells, including CSCs. A pulsing of DCs with the entire tumor cell lysate, potentially including the whole repertoire of the tumor antigens, is another promising strategy for developing CSC-specific DC vaccines. The tumor cells with high ALDH activity (ALDHhigh) can be identified and isolated quickly by fluorescence-activated cell sorting (FACS), providing a source of antigens for developing CSC-targeted therapeutic approaches. Ning and coauthors first confirmed high tumorigenicity of ALDHhigh cell populations compared to ALDHnegative cells using murine melanoma D5 and squamous cell carcinoma SCC7 syngeneic xenograft tumor models in the immunocompetent C3H and C57BL/6 mice. Next, they evaluated the antitumor immunity induced by vaccination with murine bone-marrow-derived DCs pulsed with the lysate of ALDHhigh cells (CSC-tumor pulsed DC, CSC-TPDC) compared with the lysate of whole unsorted heterogeneous tumor cells (H-TPDC). They demonstrated that the vaccination of DCs pulsed with the lysate of ALDHhigh cells induced significantly higher protective immunity against tumors than H-TPDCs, as well as DCs pulsed with the lysate of ALDHnegative cells [245]. An additional approach to increase anti-tumor immunogenicity by targeting multiple epitopes is a pulsing of DCs with mRNA derived from CSCs. A recent in vitro study by Sumransub et al. demonstrated a preclinical efficacy of an mRNA-based DC vaccine using patient-derived breast cancer cells. In this study, DCs were differentiated from PBMCs of a healthy donor and pulsed with mRNA isolated from CD44+/CD24 CSC population. This finding revealed that CSC mRNA induces a more potent cytotoxic T cell response as compared to mRNA isolated from the entire tumor cell population [72].
The DC vaccine based on the CSC lysates was also recently tested in a clinical trial for newly diagnosed or recurrent glioblastoma (GBM). In particular, the phase I clinical trial assessed the safety and tolerability of the autologous DC vaccine pulsed with lysate derived from allogeneic GBM stem-like cells. The glioblastoma cells used for the vaccine preparation were isolated from a single patient and propagated as neurospheres in serum-free conditions for CSC enrichment. For the autologous vaccine preparation, PBMCs were collected by leukapheresis and used to isolate monocytes, which were differentiated to DCs and pulsed with GBM CSC lysates. DC vaccines were administered intradermally. The therapy was safe and well tolerated. A subset of the patients (9 of the assessed 25 patients) developed a cytotoxic T cell immune response. The study was not powered to evaluate treatment efficacy, although a comparison of the progression-free survival (PFS) and OS with historical control suggested that CSC-targeted immunotherapy can be a treatment of patients with GBM [246].
Results of the clinical studies for DC-based vaccines demonstrated that their combination with therapies that stimulate immune responses and inhibit immune suppression might be more effective for cancer treatment than vaccine administration alone [238]. ICI represents one of the most used treatments in the last decade. It targets immune checkpoint molecules, including CTLA-4, PD-1, and PD-L1, and thereby enhances the immune response to cancer [234]. As discussed in chapter 1.2, CSCs can contribute to tumor immune evasion. Recent studies revealed that PD-L1 promotes the expression of stemness markers [65,70,71,72] and CSC populations with a high level of PD-L1 expression might be associated with tumor immune evasion. To overcome these therapeutic challenges, several studies combined CSC-DC vaccines and ICIs. For example, Hassani Najafabadi et al. developed a nanoparticle vaccine system to deliver ALDH1A1 and ALDH1A3 epitope peptides to antigen-presenting cells (APCs) in vivo and induce T cell responses against ALDH-high CSCs [247]. According to the study, vaccination with high-density lipoprotein nanodisks (ND) loaded with ALDH epitopes reduced the frequency of ALDHhigh CSCs in tumor tissue when combined with anti-PD-L1 therapy, and it exerted strong inhibitory effects on tumor growth in the syngeneic D5 melanoma and 4T1 breast cancer models [247]. Liao et al. used the synthetic ALDH1A1, and ALDHA1A3 peptides and DCs derived from murine bone marrow for the preparation of the CSC-targeted DC vaccines. DCs were administered subcutaneously into a C57BL/6 mice tumor model bearing syngeneic D5 murine melanoma. This study demonstrated that the DC vaccine induced T cell proliferation, humoral immune response, and T cell cytotoxicity against ALDHhigh tumor cells, and inhibited D5 tumor growth in vivo. The dual ALDH1A1 and ALDH1A3 peptide DC vaccine possessed significantly higher antitumor activity as compared to the single ALDH1A1 or ALDH1A3 peptide vaccines [240]. Of importance, this study also proved that anti-PD-L1 treatment significantly enhanced the number of CD3+ tumor-infiltrating lymphocytes and antitumor effect of this vaccine [240]. Zheng et al. investigated the CSC targeting effect of the CSC-DC vaccine combined with a dual blockade of immune checkpoints, such as PD-L1 and CTLA-4. This study confirmed the efficacy of the ALDHhigh-DC vaccines for the treatment of the syngeneic melanoma B16-F10 tumors growing in the immunocompetent C57BL/6 mice. Importantly, this study revealed that animals treated with the dual blockade of PD-L1 and CTLA-4 and CSC-DC vaccine conferred significantly more tumor regression than the CSC-DC vaccine alone. They also showed that the combination of the CSC-DC vaccine and immune checkpoint blockade significantly induced CD8+ T cell proliferation and CSC-specific cytotoxic T cell activity compared with the CSC-DC vaccine alone. This study provided the scientific basis for a clinical trial involving the combination of the CSC-DC vaccine and simultaneous PD-L1 and CTLA-4 blockades for improved tumor control in patients with cancer [248].
A combination of CSC-targeted DC vaccines with conventional therapy is a promising approach to target both CSC and non-CSC cell subsets and to prevent the interconversion between the cell populations. Lu et al. evaluated the therapeutic efficiency of ALDHhigh-CSC lysate-pulsed DCs in combination with local tumor radiation therapy (RT) given in 6 doses of 8.5 Gy. They employed syngeneic D5 melanoma and squamous sarcoma SCC7 tumor model in immunocompetent C57BL/6 and C3H mice. This study confirmed the therapeutic efficacy of the CSC-DC vaccine and demonstrated that it has a higher anti-cancer efficiency in the adjuvant setting when administered after RT. The studies conducted by Hu et al. also assessed the therapeutic potential of CSC-DC vaccination in adjuvant setting [249]. They revealed that ALDHhigh-DC treatment after surgical tumor resection significantly reduced local recurrence, prevented lung metastases, and reduced tumor ALDHhigh CSC populations in the immunocompetent C3H mice bearing syngeneic squamous carcinoma SCC7 tumors. The study of El-Ashmawy et al. suggested that the efficacy of the cisplatin-based chemotherapy for the treatment of murine Ehrlich carcinoma can be improved by its combination with the DC vaccine developed against the CD44+/CD24 CSC-like cell population [250].
Taken together, the immune targeting of CSCs represents a promising approach to cancer treatment. Furthermore, its combination with ICIs and conventional therapy such as surgery and chemo- and radiotherapy could be a strategy to optimize its therapeutic effectiveness.

5. Preclinical and Clinical Trials of Combination Therapies with Immunotherapy and Conventional Therapies Targeting CSC Markers

Conventional treatment strategies for patients with malignant diseases include surgery, chemotherapy, and radiotherapy, referred to as the traditional “three pillars” of cancer treatment. Combining immunotherapies with conventional treatments has recently become a cornerstone of cancer therapy (Table 3). It has been reported that many conventional cancer treatments, such as radiotherapy and chemotherapy, have additional immune activation mechanisms of action, including the depletion of immunosuppressive Tregs and MDSCs. The therapy-induced cell death releases tumor antigens recognized, processed, and presented to T lymphocytes by APCs [251,252,253,254,255]. Many studies have depicted the significance of combination therapy for the treatment of CSCs. In preclinical studies using an AML model, a combination of a DNA methylation inhibitor AZA and CD47 blockade via 5F9 mAb have resulted in increased macrophage-mediated phagocytosis in vitro compared to single treatments, inhibited AML growth, and prolonged survival in xenograft mice models [251]. Following these findings, a phase 1b study has demonstrated that the combination of magrolimab, a well-tolerated humanized anti-CD47 antibody, with AZA showed a greater ORR compared to AZA treatment alone, with a more rapid response time in AML patients ([252], NCT03248479). Bone marrow analysis has depicted significantly lower levels of leukemic stem cells in responding patients treated with the combination treatment. In another preclinical study, the combination of targeting receptor tyrosine kinase-like orphan receptor 1 (ROR1)-dependent signaling, which is associated with CSC maintenance and self-renewal, via cirmtuzumab and ibrutinib, which blocks B-cell receptor signaling, was more effective than single-agent treatments in reducing the number of CLL cells in the spleens of immunodeficient mice [253]. These preclinical findings have provided a rationale for clinical studies. A phase 1/2 trial is currently testing this combination in CLL patients, showing encouraging complete response results compared to ibrutinib treatment alone, suggesting a synergistic effect of the therapy ([254], NCT03088878). There are also preclinical studies that test the combination of immunotherapy with radiotherapies. Sequential treatment with fractionated photon irradiation followed by CD98hc-directed UniCAR treatment has exhibited a synergistic cytotoxic effect on radioresistant HNSCC spheroids compared to single treatments, underlining an improved antitumor effect [246]. A phase 2 clinical trial composed of induction treatment with FOLFOXIRI (folinic acid, 5-fluorouracil, oxaliplatin, and irinotecan) drug combination and bevacizumab, a mAb targeting angiogenic factor VEGF, followed by chemoradiotherapy and bevacizumab treatment, has been conducted in patients with advanced and resectable rectal adenocarcinoma. Still, no results have been posted on the ClinicalTrials.gov website yet (NCT03085992). Of importance, small-molecule inhibitors of fat mass and obesity-associated protein (FTO) demonstrated to suppress AML stem cell self-renewal as well as inhibit immune checkpoint expression, and immune evasion, hinting at the broad potential of anti-CSC therapy [255]. The CSC model predicts that therapies targeting the tumor bulk may induce tumor shrinkage; however, the responses will not be durable. Accumulating evidence suggests that stemness is rather a transient feature, and the de-differentiation of the non-CSC populations can replenish the pool of CSCs. Therefore, combining conventional therapies and CSC-targeting treatment could be a more efficient therapeutic strategy to improve therapeutic efficacy.

6. Conclusions

The therapeutic success of anti-cancer immunotherapy depends on the ability of the immune system to detect and destroy tumors as foreign tissue. However, the efficiency of cancer immunotherapy can be limited by immune evasion. CSCs possess multiple mechanisms to escape immune surveillance and create an immunosuppressive microenvironment. Results of the preclinical studies demonstrated that a combination of CSC-targeted immunotherapies with an immune checkpoint blockade stimulates anti-tumor immune responses and might be a more promising cancer therapy for further clinical studies. Tumor cell plasticity and heterogeneity also reduce the efficacy of anti-CSC therapy. Developing immunotherapy against more than one CSC antigen might lower the risk of tumor escape and non-specific toxicity. A combination of CSC-specific immunotherapy with conventional treatments, such as radio- and chemotherapy targeting the bulk tumor cells and ICI enhancing the anti-tumor immune response, could be a strategy to prevent CSC replenishment by non-CSC de-differentiation. Furthermore, conventional therapy such as radiotherapy and chemotherapy can activate multiple immune-stimulating mechanisms, highlighting the consideration for their combination with anti-CSC immunotherapy [266,267,268,269,270]. The development of preclinical models that can recapitulate human immunity and heterogeneous CSC populations in human tumors is another challenge for developing efficient and clinically relevant CSC-targeted treatment. These demands for improving the translational potential of preclinical immunology models are currently addressed using humanized [271], naturalized [272], syngeneic [273], and genetically engineered mice models [274]. These models better recapitulate the human immune system compared to conventional human tumor xenografts and can be used for precise monitoring of immune–tumor interaction [275] and for hypothesis-driven experimentation [274]. In addition, different alternative strategies to modeling immunity are being further refined, including ex vivo cultures preserving the human tissue structures, microfluidic devices, and 3D engineered tissues providing physiologically relevant microenvironments [276]. The development of robust CSC analysis in the patient-derived specimens during treatment is essential to assess the efficacy of the CSC-targeted immunotherapy [40]. It is important to notice that most of the above-described clinical trials are developed to target the bulk tumor cells and do not specifically aim to eliminate CSCs. Although several above-described immunotherapies are showing promising clinical efficacy, including considerable CRR, for example, for the patients with AML treated with IMGN632 and Flotetuzumab, the analyses of bulk tumor response for evaluating the treatment efficacy might not be suitable indicators for its specificity against CSCs. Therefore, CSC-related assays are crucial to assessing the clinical response in these studies, including analyses of CSC frequency and characterization of their self-renewal and tumor-initiating properties. The preclinical efforts to improve the effectiveness of CSC-targeting approaches, including bsAB, CARs, and vaccines, and the data obtained from ongoing clinical trials, might pave the road for CSC-directed treatments to become a clinical reality.

Author Contributions

Conceptualization, A.D. and A.S.K.; writing—review and editing, A.D., C.A., M.B., A.S.K. and S.D.G.; visualization, A.D., A.S.K., S.D.G. and C.A.; supervision, A.D. All authors have read and agreed to the published version of the manuscript.

Funding

The research in A.D. lab was funded by German Cancer Aid (Deutsche Krebshilfe), project ID: 70114659, and Deutsche Forschungsgemeinschaft (DFG) project ID: 491692296 and 416001651. The project was funded by the German Federal Ministry of Education and Research (BMBF; TurbiCAR, project ID: 01EK1513A) granted to M.B. and by the Helmholtz Initiative and Networking Fund (Radio-Immunotheranostics (MHELTHERA), project ID: InterLabs-0031) granted to M.B. The project was further supported by the Mildred Scheel Early Career Center Dresden P2/German Cancer Aid (Deutsche Krebshilfe) (project ID: 74MSNZNGAr) granted to C.A.

Acknowledgments

C.A. is a fellow of the Mildred Scheel Early Career Center Dresden P2 funded by the German Cancer Aid (Deutsche Krebshilfe).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ABCATP-binding cassette
ACTAdoptive cell therapy
ADCCAntibody-dependent cellular cytotoxicity
ALDHAldehyde dehydrogenase
ALDHhighHigh ALDH activity
ALLAcute lymphoblastic leukemia
AMLAcute myeloid leukemia
APCAntigen-presenting cell
AZAAzacitidine
BCMAB-cell maturation antigen
BLLB-lymphoid leukemia
BPDCNBlastic plasmacytoid dendritic neoplasm
bsABBispecific antibody
BTKBruton’s tyrosine kinase
CAFCancer-associated fibroblast
CARChimeric antigen receptor
CCRComposite complete remission rate
CD44v6CD44 isoform variant 6
CEACarcinoembryonic antigen
CLLChronic lymphocytic leukemia
CRPCCastration-resistant prostate cancer
CRRComplete remission rate
CRSCytokine release syndrome
CSCCancer stem cell
CSC-DCDendritic CSC vaccination
CSC-TPDCCSC-tumor pulsed DC
CTLA-4Cytotoxic T-lymphocyte-associated protein 4
CTCCirculating tumor cell
DCDendritic cell
ECEndothelial cell
ECMExtracellular matrix
EGFREpidermal growth factor receptor
EpCAMEpithelial cell adhesion molecule
ESCCEsophagus squamous cell carcinoma
ex20-insInsertions in exon 20
FACSFluorescence-activated cell sorting
FAKFocal adhesion kinase
FDAFood and Drug Administration
FOLFOXIRIFolinic acid, 5-fluorouracil, oxaliplatin and irinotecan
FTOFat mass and obesity-associated protein
GPC3Glypican-3
GVHDGraft versus host disease
H-TPDCHeterogenous-tumor pulsed DC
HCLHairy cell leukemia
HhHedgehog
HIFHypoxia-inducible transcriptional factor
HNSCCHead and neck squamous cell carcinoma
ICANSImmune effector cell-associated neurotoxicity syndrome
ICIImmune checkpoint inhibitor
IGNIndolinobenzodiazepine pseudodimer
IL-3RInterleukin-3 receptor
iPSCInduced pluripotent stem cell
LGR5Leucine-rich repeat-containing G protein-coupled receptor 5
mAbMonoclonal antibody
MDCSMyeloid-derived suppressor cell
MDSMyelodysplastic syndrome
METMesenchymal-epithelial transition
MHC-IMajor histocompatibility complex class I
MMP-2Matrix metalloproteinase-2
MsAbMulti-specific antibody
MSCMesenchymal cells
MUC1Mucin 1
NDNanodisk
NF-κBNuclear factor kappa B
NHLNon-Hodgkin’s lymphoma
NKNatural killer cell
NSCLCNon-small cell lung cancer
ORRObjective response rate
OSOverall survival
PBMCPeripheral blood mononuclear cell
PD-1Programmed cell death protein 1
PD-L1Programmed death-ligand 1
PFSProgression-free survival
ROR1Receptor tyrosine kinase-like orphan receptor 1
ROSReactive oxygen species
RTRadiation therapy
RTKReceptor tyrosine kinase
scFvSingle-chain variable region
SCIDSevere combined immunodeficient
SH2Src homology 2
SHP-1Src homology 2 domain-containing phosphatase-1
SHP-2Src homology 2 domain-containing phosphatase-2
SIRPαSignal regulatory protein α
STAT3Signal transducer and activator of transcription 3
TAATumor-associated antigen
TAMTumor-associated macrophage
TAPThe transporter associated with antigen processing
TANTumor-associated neutrophil
TCRT cell receptor
TMTargeting module
TMETumor microenvironment
TregRegulatory T cell
trAbTrifunctional antibody
TSP-1Thrombospondin 1
UniCARUniversal CAR
VEGFVascular endothelial growth factor
VEGFR-2Vascular endothelial growth factor receptor-2
VENVenetoclax
VHHeavy chain
VLLight chain

References

  1. Ferlay, J.; Colombet, M.; Soerjomataram, I.; Parkin, D.M.; Piñeros, M.; Znaor, A.; Bray, F. Cancer statistics for the year 2020: An overview. Int. J. Cancer 2021, 149, 778–789. [Google Scholar] [CrossRef] [PubMed]
  2. Nowell, P.C. The clonal evolution of tumor cell populations. Science 1976, 194, 23–28. [Google Scholar] [CrossRef] [PubMed]
  3. Lapidot, T.; Sirard, C.; Vormoor, J.; Murdoch, B.; Hoang, T.; Caceres-Cortes, J.; Minden, M.; Paterson, B.; Michael, A.; Caligiuri, J.; et al. A cell initiating human acute myeloid leukemia after transplantation into SCID mice. Nature 1994, 367, 645–648. [Google Scholar] [CrossRef]
  4. Visvader, J.E.; Lindeman, G.J. Cancer Stem Cells: Current Status and Evolving Complexities. Cell Stem Cell 2012, 10, 717–728. [Google Scholar] [CrossRef] [Green Version]
  5. Bonnet, D.; Dick, J.E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 1997, 3, 730–737. [Google Scholar] [CrossRef]
  6. Peitzsch, C.; Tyutyunnykova, A.; Pantel, K.; Dubrovska, A. Cancer stem cells: The root of tumor recurrence and metastases. Semin. Cancer Biol. 2017, 44, 10–24. [Google Scholar] [CrossRef]
  7. Al-Hajj, M.; Wicha, M.S.; Benito-Hernandez, A.; Morrison, S.J.; Clarke, M.F. Prospective identification of tumorigenic breast cancer cells. Proc. Natl. Acad. Sci. USA 2003, 100, 3983–3988. [Google Scholar] [CrossRef] [Green Version]
  8. Singh, S.K.; Hawkins, C.; Clarke, I.D.; Squire, J.A.; Bayani, J.; Hide, T.; Henkelman, R.M.; Cusimano, M.D.; Dirks, P.B. Identification of human brain tumour initiating cells. Nature 2004, 432, 396–401. [Google Scholar] [CrossRef] [PubMed]
  9. Zhou, B.-B.S.; Zhang, H.; Damelin, M.; Geles, K.G.; Grindley, J.C.; Dirks, P.B. Tumour-initiating cells: Challenges and opportunities for anticancer drug discovery. Nat. Rev. Drug Discov. 2009, 8, 806–823. [Google Scholar] [CrossRef] [PubMed]
  10. Chen, L.; Peng, M.; Li, N.; Song, Q.; Yao, Y.; Xu, B.; Liu, H.; Ruan, P. Combined use of EpCAM and FRalpha enables the high-efficiency capture of circulating tumor cells in non-small cell lung cancer. Sci. Rep. 2018, 8, 1188. [Google Scholar] [CrossRef] [Green Version]
  11. Huang, X.; Wan, J.; Leng, D.; Zhang, Y.; Yang, S. Dual-targeting nanomicelles with CD133 and CD44 aptamers for enhanced delivery of gefitinib to two populations of lung cancer-initiating cells. Exp. Ther. Med. 2019, 19, 192–204. [Google Scholar] [CrossRef]
  12. Hadjimichael, C.; Chanoumidou, K.; Papadopoulou, N.; Arampatzi, P.; Papamatheakis, J.; Kretsovali, A. Common stemness regu-lators of embryonic and cancer stem cells. World J. Stem Cells 2015, 7, 1150–1184. [Google Scholar]
  13. Ginestier, C.; Hur, M.H.; Charafe-Jauffret, E.; Monville, F.; Dutcher, J.; Brown, M.; Jacquemier, J.; Viens, P.; Kleer, C.G.; Liu, S.; et al. ALDH1 Is a Marker of Normal and Malignant Human Mammary Stem Cells and a Predictor of Poor Clinical Outcome. Cell Stem Cell 2007, 1, 555–567. [Google Scholar] [CrossRef] [Green Version]
  14. Ju, F.; Atyah, M.M.; Horstmann, N.; Gul, S.; Vago, R.; Bruns, C.J.; Zhao, Y.; Dong, Q.-Z.; Ren, N. Characteristics of the cancer stem cell niche and therapeutic strategies. Stem Cell Res. Ther. 2022, 13, 1–17. [Google Scholar] [CrossRef] [PubMed]
  15. Han, M.-E.; Kim, H.-J.; Shin, D.H.; Hwang, S.-H.; Kang, C.-D.; Oh, S.-O. Overexpression of NRG1 promotes progression of gastric cancer by regulating the self-renewal of cancer stem cells. J. Gastroenterol. 2014, 50, 645–656. [Google Scholar] [CrossRef]
  16. Hu, Y.; Yan, C.; Mu, L.; Huang, K.; Li, X.; Tao, D.; Wu, Y.; Qin, J. Fibroblast-derived exosomes contribute to chemoresistance through priming cancer stem cells in colorectal cancer. PLoS ONE 2015, 10, e0125625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Prieto-Vila, M.; Takahashi, R.-U.; Usuba, W.; Kohama, I.; Ochiya, T. Drug Resistance Driven by Cancer Stem Cells and Their Niche. Int. J. Mol. Sci. 2017, 18, 2574. [Google Scholar] [CrossRef] [Green Version]
  18. Meurette, O.; Mehlen, P. Notch Signaling in the Tumor Microenvironment. Cancer Cell 2018, 34, 536–548. [Google Scholar] [CrossRef] [Green Version]
  19. Lau, E.Y.-T.; Ho, N.P.-Y.; Lee, T.K.-W. Cancer Stem Cells and Their Microenvironment: Biology and Therapeutic Implications. Stem Cells Int. 2017, 2017, 1–11. [Google Scholar] [CrossRef] [Green Version]
  20. Grange, C.; Tapparo, M.; Collino, F.; Vitillo, L.; Damasco, C.; Deregibus, M.C.; Tetta, C.; Bussolati, B.; Camussi, G. Microvesicles Released from Human Renal Cancer Stem Cells Stimulate Angiogenesis and Formation of Lung Premetastatic Niche. Cancer Res. 2011, 71, 5346–5356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Ajani, J.A.; Song, S.; Hochster, H.S.; Steinberg, I.B. Cancer Stem Cells: The Promise and the Potential. Semin. Oncol. 2015, 42, S3–S17. [Google Scholar] [CrossRef] [PubMed]
  22. Krause, M.; Dubrovska, A.; Linge, A.; Baumann, M. Cancer stem cells: Radioresistance, prediction of radiotherapy outcome and specific targets for combined treatments. Adv. Drug Deliv. Rev. 2016, 109, 63–73. [Google Scholar] [CrossRef] [Green Version]
  23. Schulz, A.; Meyer, F.; Dubrovska, A.; Borgmann, K. Cancer Stem Cells and Radioresistance: DNA Repair and Beyond. Cancers 2019, 11, 862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Cojoc, M.; Mäbert, K.; Muders, M.H.; Dubrovska, A. A role for cancer stem cells in therapy resistance: Cellular and molecular mechanisms. Semin. Cancer Biol. 2015, 31, 16–27. [Google Scholar] [CrossRef]
  25. Ayob, A.Z.; Ramasamy, T.S. Cancer stem cells as key drivers of tumour progression. J. Biomed. Sci. 2018, 25, 20. [Google Scholar] [CrossRef] [Green Version]
  26. Batlle, E.; Clevers, H. Cancer stem cells revisited. Nat. Med. 2017, 23, 1124–1134. [Google Scholar] [CrossRef] [PubMed]
  27. Pasello, M.; Giudice, A.M.; Scotlandi, K. The ABC subfamily A transporters: Multifaceted players with incipient potentialities in cancer. Semin. Cancer Biol. 2019, 60, 57–71. [Google Scholar] [CrossRef]
  28. Prager, B.C.; Xie, Q.; Bao, S.; Rich, J.N. Cancer Stem Cells: The Architects of the Tumor Ecosystem. Cell Stem Cell 2019, 24, 41–53. [Google Scholar] [CrossRef] [Green Version]
  29. Liao, W.-T.; Ye, Y.-P.; Deng, Y.-J.; Bian, X.-W.; Ding, Y.-Q. Metastatic cancer stem cells: From the concept to therapeutics. Am. J. Stem Cells 2014, 3, 46–62. [Google Scholar]
  30. Garcia-Mayea, Y.; Mir, C.; Masson, F.; Paciucci, R.; Lleonart, M. Insights into new mechanisms and models of cancer stem cell multidrug resistance. Semin. Cancer Biol. 2020, 60, 166–180. [Google Scholar] [CrossRef]
  31. Ramos, E.K.; Hoffmann, A.D.; Gerson, S.L.; Liu, H. New Opportunities and Challenges to Defeat Cancer Stem Cells. Trends Cancer 2017, 3, 780–796. [Google Scholar] [CrossRef]
  32. Kolev, V.N.; Wright, Q.G.; Vidal, C.M.; Ring, J.E.; Shapiro, I.M.; Ricono, J.; Weaver, D.T.; Padval, M.V.; Pachter, J.A.; Xu, Q. PI3K/mTOR Dual Inhibitor VS-5584 Preferentially Targets Cancer Stem Cells. Cancer Res 2015, 75, 446–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Mossahebi-Mohammadi, M.; Quan, M.; Zhang, J.-S.; Li, X. FGF Signaling Pathway: A Key Regulator of Stem Cell Pluripotency. Front. Cell Dev. Biol. 2020, 8, 79. [Google Scholar] [CrossRef] [Green Version]
  34. Kim, B.N.; Ahn, D.H.; Kang, N.; Yeo, C.D.; Kim, Y.K.; Lee, K.Y.; Kim, T.J.; Lee, S.H.; Park, M.S.; Yim, H.W.; et al. TGF-beta induced EMT and stemness characteristics are associated with epigenetic reg-ulation in lung cancer. Sci. Rep. 2020, 10, 10597. [Google Scholar] [CrossRef]
  35. Kumar, V.; Vashishta, M.; Kong, L.; Wu, X.; Lu, J.J.; Guha, C.; Dwarakanath, B.S. The Role of Notch, Hedgehog, and Wnt Signaling Pathways in the Resistance of Tumors to Anticancer Therapies. Front. Cell Dev. Biol. 2021, 9, 650772. [Google Scholar] [CrossRef]
  36. Song, J.I.; Grandis, J.R. STAT signaling in head and neck cancer. Oncogene 2000, 19, 2489–2495. [Google Scholar] [CrossRef] [Green Version]
  37. Kaltschmidt, C.; Banz-Jansen, C.; Benhidjeb, T.; Beshay, M.; Förster, C.; Greiner, J.; Hamelmann, E.; Jorch, N.; Mertzlufft, F.; Pfitzenmaier, J.; et al. A Role for NF-kappaB in Organ Specific Cancer and Cancer Stem Cells. Cancers 2019, 11, 655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Peitzsch, C.; Cojoc, M.; Hein, L.; Kurth, I.; Mäbert, K.; Trautmann, F.; Klink, B.; Schröck, E.; Wirth, M.P.; Krause, M.; et al. An Epigenetic Reprogramming Strategy to Resensitize Radioresistant Prostate Cancer Cells. Cancer Res. 2016, 76, 2637–2651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Kroon, P.; Berry, P.A.; Stower, M.J.; Rodrigues, G.; Mann, V.M.; Simms, M.; Bhasin, D.; Chettiar, S.; Li, C.; Li, P.-K.; et al. JAK-STAT Blockade Inhibits Tumor Initiation and Clonogenic Recovery of Prostate Cancer Stem-like Cells. Cancer Res. 2013, 73, 5288–5298. [Google Scholar] [CrossRef] [Green Version]
  40. Saygin, C.; Matei, D.; Majeti, R.; Reizes, O.; Lathia, J.D. Targeting Cancer Stemness in the Clinic: From Hype to Hope. Cell Stem. Cell 2018, 24, 25–40. [Google Scholar] [CrossRef] [Green Version]
  41. Kim, S.; Dubrovska, A.; Salamone, R.J.; Walker, J.R.; Grandinetti, K.B.; Bonamy, G.M.C.; Orth, A.P.; Elliott, J.; Graus Porta, D.; Garcia-Echeverria, C.; et al. FGFR2 promotes breast tumorigenicity through maintenance of breast tumor-initiating cells. PLoS ONE 2013, 8, e51671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Yuan, Z.; Goetz, J.A.; Singh, S.; Ogden, S.K.; Petty, W.J.; Black, C.C.; Memoli, V.A.; Dmitrovsky, E.; Robbins, D.J. Frequent requirement of hedgehog signaling in non-small cell lung carcinoma. Oncogene 2007, 26, 1046–1055. [Google Scholar] [CrossRef] [Green Version]
  43. Fu, L.; Zhang, C.; Zhang, L.Y.; Dong, S.S.; Lu, L.H.; Chen, J.; Dai, Y.; Li, Y.; Kong, K.L.; Kwong, D.L.; et al. Wnt2 secreted by tumour fibroblasts promotes tumour progression in oesophageal cancer by activation of the Wnt/beta-catenin signalling pathway. Gut 2011, 60, 1635–1643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Rajakulendran, N.; Rowland, K.J.; Selvadurai, H.J.; Ahmadi, M.; Park, N.I.; Naumenko, S.; Dolma, S.; Ward, R.J.; So, M.; Lee, L.; et al. Wnt and Notch signaling govern self-renewal and differentiation in a subset of human glioblastoma stem cells. Genes Dev. 2019, 33, 498–510. [Google Scholar] [CrossRef] [PubMed]
  45. van Groningen, T.; Akogul, N.; Westerhout, E.M.; Chan, A.; Hasselt, N.E.; Zwijnenburg, D.A.; Broekmans, M.; Stroeken, P.; Haneveld, F.; Hooijer, G.K.J.; et al. A NOTCH feed-forward loop drives reprogramming from adrenergic to mesenchymal state in neuroblastoma. Nat. Commun. 2019, 10, 1530. [Google Scholar] [CrossRef] [Green Version]
  46. Yang, J.; Xing, H.; Lu, D.; Wang, J.; Li, B.; Tang, J.; Gu, F.; Hong, L. Role of Jagged1/STAT3 signalling in platinum-resistant ovarian cancer. J. Cell Mol. Med. 2019, 23, 4005–4018. [Google Scholar] [CrossRef] [Green Version]
  47. Qi, X.-T.; Li, Y.-L.; Zhang, Y.-Q.; Xu, T.; Lu, B.; Fang, L.; Gao, J.-Q.; Yu, L.-S.; Zhu, D.-F.; Yang, B.; et al. KLF4 functions as an oncogene in promoting cancer stem cell-like characteristics in osteosarcoma cells. Acta Pharmacol. Sin. 2018, 40, 546–555. [Google Scholar] [CrossRef] [PubMed]
  48. Rosner, M.H.; Vigano, M.A.; Ozato, K.; Timmons, P.M.; Poirie, F.; Rigby, P.W.J.; Staudt, L.M. A POU-domain transcription factor in early stem cells and germ cells of the mammalian embryo. Nature 1990, 345, 686–692. [Google Scholar] [CrossRef]
  49. Lundberg, I.V.; Edin, S.; Eklof, V.; Oberg, A.; Palmqvist, R.; Wikberg, M.L. SOX2 expression is associated with a cancer stem cell state and down-regulation of CDX2 in colorectal cancer. BMC Cancer 2016, 16, 471. [Google Scholar] [CrossRef] [Green Version]
  50. Nathansen, J.; Lukiyanchuk, V.; Hein, L.; Stolte, M.I.; Borgmann, K.; Löck, S.; Kurth, I.; Baumann, M.; Krause, M.; Linge, A.; et al. Oct4 confers stemness and radioresistance to head and neck squamous cell car-cinoma by regulating the homologous recombination factors PSMC3IP and RAD54L. Oncogene 2021, 40, 4214–4228. [Google Scholar] [CrossRef]
  51. Jeter, C.R.; Liu, B.; Liu, X.; Chen, X.; Liu, C.; Calhoun-Davis, T.; Repass, J.; Zaehres, H.; Shen, J.J.; Tang, D.G. NANOG promotes cancer stem cell characteristics and prostate cancer resistance to androgen deprivation. Oncogene 2011, 30, 3833–3845. [Google Scholar] [CrossRef] [Green Version]
  52. Yang, L.; Shi, P.; Zhao, G.; Xu, J.; Peng, W.; Zhang, J.; Zhang, G.; Wang, X.; Dong, Z.; Chen, F.; et al. Targeting Cancer Stem Cell Pathways for Cancer Therapy. Signal Transduct. Target. Ther. 2020, 5, 8. [Google Scholar] [CrossRef] [Green Version]
  53. Allen, F.; Maillard, I. Therapeutic Targeting of Notch Signaling: From Cancer to Inflammatory Disorders. Front. Cell Dev. Biol. 2021, 9, 649205. [Google Scholar] [CrossRef]
  54. Ruiu, R.; Tarone, L.; Rolih, V.; Barutello, G.; Bolli, E.; Riccardo, F.; Cavallo, F.; Conti, L. Cancer stem cell immunology and immunotherapy: Harnessing the immune system against cancer's source. Prog. Mol. Biol. Transl. Sci. 2019, 164, 119–188. [Google Scholar]
  55. Xiang, T.; Long, H.; He, L.; Han, X.; Lin, K.; Liang, Z.; Zhuo, W.; Xie, R.; Zhu, B. Interleukin-17 produced by tumor microenvironment promotes self-renewal of CD133+ cancer stem-like cells in ovarian cancer. Oncogene 2013, 34, 165–176. [Google Scholar] [CrossRef]
  56. Casbon, A.-J.; Reynaud, D.; Park, C.; Khuc, E.; Gan, D.D.; Schepers, K.; Passegué, E.; Werb, Z. Invasive breast cancer reprograms early myeloid differentiation in the bone marrow to generate immunosuppressive neutrophils. Proc. Natl. Acad. Sci. USA 2015, 112, E566–E575. [Google Scholar] [CrossRef] [Green Version]
  57. Jenkins, L.; Jungwirth, U.; Avgustinova, A.; Iravani, M.; Mills, A.P.; Haider, S.; Harper, J.; Isacke, C.M. Cancer-Associated Fibroblasts Suppress CD8+ T-cell Infiltration and Confer Resistance to Immune-Checkpoint Blockade. Cancer Res. 2022, 82, 2904–2917. [Google Scholar] [CrossRef]
  58. Labani-Motlagh, A.; Ashja-Mahdavi, M.; Loskog, A. The Tumor Microenvironment: A Milieu Hindering and Obstructing An-titumor Immune Responses. Front Immunol. 2020, 11, 940. [Google Scholar] [CrossRef]
  59. Vito, A.; El-Sayes, N.; Mossman, K. Hypoxia-Driven Immune Escape in the Tumor Microenvironment. Cells 2020, 9, 992. [Google Scholar] [CrossRef]
  60. Calcinotto, A.; Filipazzi, P.; Grioni, M.; Iero, M.; De Milito, A.; Ricupito, A.; Cova, A.; Canese, R.; Jachetti, E.; Rossetti, M.; et al. Modulation of Microenvironment Acidity Reverses Anergy in Human and Murine Tumor-Infiltrating T Lymphocytes. Cancer Res. 2012, 72, 2746–2756. [Google Scholar] [CrossRef] [Green Version]
  61. Schatton, T.; Schütte, U.; Frank, N.Y.; Zhan, Q.; Hoerning, A.; Robles, S.C.; Zhou, J.; Hodi, F.S.; Spagnoli, G.C.; Murphy, G.F.; et al. Modulation of T-Cell Activation by Malignant Melanoma Initiating Cells. Cancer Res. 2010, 70, 697–708. [Google Scholar] [CrossRef] [Green Version]
  62. Reim, F.; Dombrowski, Y.; Ritter, C.; Buttmann, M.; Haüsler, S.; Ossadnik, M.; Krockenberger, M.; Beier, D.; Beier, C.P.; Dietl, J.; et al. Immunoselection of Breast and Ovarian Cancer Cells with Trastuzumab and Natural Killer Cells: Selective Escape of CD44high/CD24low/HER2low Breast Cancer Stem Cells. Cancer Res. 2009, 69, 8058–8066. [Google Scholar] [CrossRef] [Green Version]
  63. Golan, H.; Shukrun, R.; Caspi, R.; Vax, E.; Pode-Shakked, N.; Goldberg, S.; Pleniceanu, O.; Bar-Lev, D.D.; Mark-Danieli, M.; Pri-Chen, S.; et al. In Vivo Expansion of Cancer Stemness Affords Novel Cancer Stem Cell Targets: Malignant Rhabdoid Tumor as an Example. Stem Cell Rep. 2018, 11, 795–810. [Google Scholar] [CrossRef] [Green Version]
  64. Bruttel, V.S.; Wischhusen, J. Cancer stem cell immunology: Key to understanding tumorigenesis and tumor immune escape? Front. Immunol. 2014, 5, 360. [Google Scholar] [CrossRef] [Green Version]
  65. Lee, Y.; Shin, J.H.; Longmire, M.; Wang, H.; Kohrt, H.E.; Chang, H.Y.; Sunwoo, J.B. CD44+ Cells in Head and Neck Squamous Cell Carcinoma Suppress T-Cell–Mediated Immunity by Selective Constitutive and Inducible Expression of PD-LClin. Cancer Res. 2016, 22, 3571–3581. [Google Scholar] [CrossRef] [Green Version]
  66. Miao, Y.; Yang, H.; Levorse, J.; Yuan, S.; Polak, L.; Sribour, M.; Singh, B.; Rosenblum, M.D.; Fuchs, E. Adaptive Immune Resistance Emerges from Tumor-Initiating Stem Cells. Cell 2019, 177, 1172–1186.e14. [Google Scholar] [CrossRef]
  67. Dong, P.; Xiong, Y.; Yue, J.; Hanley, S.J.B.; Watari, H. Tumor-Intrinsic PD-L1 Signaling in Cancer Initiation, Development and Treatment: Beyond Immune Evasion. Front. Oncol. 2018, 8, 386. [Google Scholar] [CrossRef] [Green Version]
  68. Shaim, H.; Shanley, M.; Basar, R.; Daher, M.; Gumin, J.; Zamler, D.B.; Uprety, N.; Wang, F.; Huang, Y.; Gabrusiewicz, K.; et al. Targeting the αv integrin/TGF-β axis improves natural killer cell function against glio-blastoma stem cells. J. Clin. Investig. 2021, 131, e142116. [Google Scholar] [CrossRef]
  69. Hsu, J.-M.; Xia, W.; Hsu, Y.-H.; Chan, L.-C.; Yu, W.-H.; Cha, J.-H.; Chen, C.-T.; Liao, H.-W.; Kuo, C.-W.; Khoo, K.-H.; et al. STT3-dependent PD-L1 accumulation on cancer stem cells promotes immune evasion. Nat. Commun. 2018, 9, 1–17. [Google Scholar] [CrossRef] [Green Version]
  70. Zavros, Y. Initiation and Maintenance of Gastric Cancer: A Focus on CD44 Variant Isoforms and Cancer Stem Cells. Cell. Mol. Gastroenterol. Hepatol. 2017, 4, 55–63. [Google Scholar] [CrossRef] [Green Version]
  71. Gao, L.; Guo, Q.; Li, X.; Yang, X.; Ni, H.; Wang, T.; Zhao, Q.; Liu, H.; Xing, Y.; Xi, T.; et al. MiR-873/PD-L1 axis regulates the stemness of breast cancer cells. eBioMedicine 2019, 41, 395–407. [Google Scholar] [CrossRef] [Green Version]
  72. Sumransub, N.; Jirapongwattana, N.; Jamjuntra, P.; Thongchot, S.; Chieochansin, T.; Yenchitsomanus, P.; Thuwajit, P.; Warnnissorn, M.; O-Charoenrat, P.; Thuwajit, C. Breast cancer stem cell RNA-pulsed dendritic cells enhance tumor cell killing by effector T cells. Oncol. Lett. 2020, 19, 2422–2430. [Google Scholar] [CrossRef] [Green Version]
  73. Lu, L.; Jiang, J.; Zhan, M.; Zhang, H.; Wang, Q.T.; Sun, S.N.; Guo, X.K.; Yin, H.; Wei, Y.; Liu, J.O.; et al. Targeting Neoantigens in Hepatocellular Carcinoma for Immunotherapy: A Futile Strategy? Hepatology 2021, 73, 414–421. [Google Scholar] [CrossRef] [Green Version]
  74. Herbrich, S.; Baran, N.; Cai, T.; Weng, C.; Aitken, M.J.L.; Post, S.M.; Henderson, J.; Shi, C.; Havranek, O.; Richard-Carpentier, G.; et al. Overexpression of CD200 is a stem cell-specific mechanism of immune evasion in AML. J. Immunother. Cancer 2021, 9, e002968. [Google Scholar] [CrossRef]
  75. Dianat-Moghadam, H.; Heidarifard, M.; Jahanban-Esfahlan, R.; Panahi, Y.; Hamishehkar, H.; Pouremamali, F.; Rahbarghazi, R.; Nouri, M. Cancer stem cells-emanated therapy resistance: Implications for liposomal drug delivery systems. J. Control. Release 2018, 288, 62–83. [Google Scholar] [CrossRef]
  76. Chernosky, N.M.; Tamagno, I. The Role of the Innate Immune System in Cancer Dormancy and Relapse. Cancers 2021, 13, 5621. [Google Scholar] [CrossRef]
  77. Cornel, A.M.; Mimpen, I.L.; Nierkens, S. MHC Class I Downregulation in Cancer: Underlying Mechanisms and Potential Targets for Cancer Immunotherapy. Cancers 2020, 12, 1760. [Google Scholar] [CrossRef]
  78. Akhter, Z.; Sharawat, S.K.; Kumar, V.; Kochat, V.; Equbal, Z.; Ramakrishnan, M.; Kumar, U.; Mathur, S.; Kumar, L.; Mukhopadhyay, A. Aggressive serous epithelial ovarian cancer is potentially propagated by EpCAM+CD45+ phenotype. Oncogene 2018, 37, 2089–2103. [Google Scholar] [CrossRef]
  79. Zhong, Y.; Guan, K.; Guo, S.; Zhou, C.; Wang, D.; Ma, W.; Zhang, Y.; Li, C.; Zhang, S. Spheres derived from the human SK-RC-42 renal cell carcinoma cell line are enriched in cancer stem cells. Cancer Lett. 2010, 299, 150–160. [Google Scholar] [CrossRef]
  80. Kushwah, R.; Guezguez, B.; Lee, J.B.; Hopkins, C.I.; Bhatia, M. Pleiotropic roles of Notch signaling in normal, malignant, and developmental hematopoiesis in the human. EMBO Rep. 2014, 15, 1128–1138. [Google Scholar] [CrossRef] [Green Version]
  81. Liu, S.; Dontu, G.; Mantle, I.D.; Patel, S.; Ahn, N.-S.; Jackson, K.W.; Suri, P.; Wicha, M.S. Hedgehog Signaling and Bmi-1 Regulate Self-renewal of Normal and Malignant Human Mammary Stem Cells. Cancer Res 2006, 66, 6063–6071. [Google Scholar] [CrossRef] [Green Version]
  82. Kahn, M. Can we safely target the WNT pathway? Nat. Rev. Drug. Discov. 2014, 13, 513–532. [Google Scholar] [CrossRef] [Green Version]
  83. Tilley, L.P.; Liu, S.K.; Gilbertson, S.R.; Wagner, B.M.; Lord, P.F. Primary myocardial disease in the cat. A model for human car-dio-myopathy. Am. J. Pathol. 1977, 86, 493–522. [Google Scholar]
  84. Maloney, D.G.; Grillo-Lopez, A.J.; White, C.A.; Bodkin, D.; Schilder, R.J.; Neidhart, J.A.; Janakiraman, N.; Foon, K.A.; Liles, T.M.; Dallaire, B.K.; et al. IDEC-C2B8 (Rituximab) anti-CD20 monoclonal antibody therapy in patients with relapsed low-grade non-Hodgkin's lymphoma. Blood 1997, 90, 2188–2195. [Google Scholar] [CrossRef] [Green Version]
  85. Chen, C.; Zhao, S.; Karnad, A.; Freeman, J.W. The biology and role of CD44 in cancer progression: Therapeutic implications. J. Hematol. Oncol. 2018, 11, 1–23. [Google Scholar] [CrossRef] [Green Version]
  86. Morath, I.; Jung, C.; Lévêque, R.; Linfeng, C.; Toillon, R.-A.; Warth, A.; Orian-Rousseau, V. Differential recruitment of CD44 isoforms by ErbB ligands reveals an involvement of CD44 in breast cancer. Oncogene 2018, 37, 1472–1484. [Google Scholar] [CrossRef] [PubMed]
  87. Tijink, B.M.; Buter, J.; de Bree, R.; Giaccone, G.; Lang, M.S.; Staab, A.; Leemans, C.R.; van Dongen, G.A. A Phase I Dose Escalation Study with Anti-CD44v6 Bivatuzumab Mertansine in Patients with Incurable Squamous Cell Carcinoma of the Head and Neck or Esophagus. Clin. Cancer Res. 2006, 12, 6064–6072. [Google Scholar] [CrossRef] [Green Version]
  88. Menke-van der Houven van Oordt, C.W.; Gomez-Roca, C.; van Herpen, C.; Coveler, A.L.; Mahalingam, D.; Verheul, H.M.W.; van der Graaf, W.T.A.; Christen, R.; Rüttinger, D.; Weigand, S.; et al. First-in-human phase I clinical trial of RG7356, an anti-CD44 humanized antibody, in patients with advanced, CD44-expressing solid tumors. Oncotarget. 2016, 7, 80046–80058. [Google Scholar] [CrossRef] [Green Version]
  89. Vey, N.; Delaunay, J.; Martinelli, G.; Fiedler, W.; Raffoux, E.; Prebet, T.; Gomez-Roca, C.; Papayannidis, C.; Kebenko, M.; Paschka, P.; et al. Phase I clinical study of RG7356, an anti-CD44 humanized antibody, in patients with acute myeloid leukemia. Oncotarget 2016, 7, 32532–32542. [Google Scholar] [CrossRef] [Green Version]
  90. Matlung, H.L.; Szilagyi, K.; Barclay, N.A.; van den Berg, T.K. The CD47-SIRPalpha signaling axis as an innate immune checkpoint in cancer. Immunol. Rev. 2017, 276, 145–164. [Google Scholar] [CrossRef]
  91. Barclay, A.N.; Van den Berg, T.K. The interaction between signal regulatory protein alpha (SIRPalpha) and CD47: Structure, function, and therapeutic target. Annu. Rev. Immunol. 2014, 32, 25–50. [Google Scholar] [CrossRef] [PubMed]
  92. Gao, A.G.; Lindberg, F.P.; Dimitry, J.M.; Brown, E.J.; Frazier, W.A. Thrombospondin modulates alpha v beta 3 function through integrin-associated protein. J. Cell Biol. 1996, 135, 533–544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Hayat, S.M.G.; Bianconi, V.; Pirro, M.; Jaafari, M.R.; Hatamipour, M.; Sahebkar, A. CD47: Role in the immune system and application to cancer therapy. Cell. Oncol. 2019, 43, 19–30. [Google Scholar] [CrossRef]
  94. Gao, L.; Chen, K.; Gao, Q.; Wang, X.; Sun, J.; Yang, Y.-G. CD47 deficiency in tumor stroma promotes tumor progression by enhancing angiogenesis. Oncotarget 2016, 8, 22406–22413. [Google Scholar] [CrossRef] [Green Version]
  95. Zhang, H.; Lu, H.; Xiang, L.; Bullen, J.W.; Zhang, C.; Samanta, D.; Gilkes, D.M.; He, J.; Semenza, G.L. HIF-1 regulates CD47 expression in breast cancer cells to promote evasion of phagocytosis and maintenance of cancer stem cells. Proc. Natl. Acad. Sci. USA 2015, 112, E6215–E6223. [Google Scholar] [CrossRef] [Green Version]
  96. Cioffi, M.; Trabulo, S.; Hidalgo, M.; Costello, E.; Greenhalf, W.; Erkan, M.; Kleeff, J.; Sainz, B.; Heeschen, C. Inhibition of CD47 Effectively Targets Pancreatic Cancer Stem Cells via Dual Mechanisms. Clin. Cancer Res. 2015, 21, 2325–2337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Chang, C.L.; Wu, C.C.; Hsu, Y.T.; Hsu, Y.C. Immune vulnerability of ovarian cancer stem-like cells due to low CD47 expression is protected by surrounding bulk tumor cells. Oncoimmunology 2020, 9, 1803530. [Google Scholar] [CrossRef]
  98. Candas-Green, D.; Xie, B.; Huang, J.; Fan, M.; Wang, A.; Menaa, C.; Zhang, Y.; Zhang, L.; Jing, D.; Azghadi, S.; et al. Dual blockade of CD47 and HER2 eliminates radioresistant breast cancer cells. Nat. Commun. 2020, 11, 1–15. [Google Scholar] [CrossRef]
  99. Miao, M.; Wu, D.; Xie, S.; Hong, M.; Chang, C.; Yu, L.; Gao, S.; Li, Y.; Li, Y.; Zhu, Z.; et al. A Phase 1b Study to Evaluate Safety and Efficacy of IBI188 in Combination with Azacitidine (AZA) As a First-Line Treatment in Subjects with Newly Diagnosed Higher Risk Myelodysplastic Syndrome. Blood 2022, 140, 4045–4046. [Google Scholar] [CrossRef]
  100. Testa, U.; Pelosi, E.; Castelli, G. CD123 as a Therapeutic Target in the Treatment of Hematological Malignancies. Cancers 2019, 11, 1358. [Google Scholar] [CrossRef] [Green Version]
  101. El Achi, H.; Dupont, E.; Paul, S.; Khoury, J.D. CD123 as a Biomarker in Hematolymphoid Malignancies: Principles of Detection and Targeted Therapies. Cancers 2020, 12, 3087. [Google Scholar] [CrossRef] [PubMed]
  102. Pemmaraju, N.; Lane, A.A.; Sweet, K.L.; Stein, A.S.; Vasu, S.; Blum, W.; Rizzieri, D.A.; Wang, E.S.; Duvic, M.; Sloan, J.M.; et al. Tagraxofusp in Blastic Plasmacytoid Dendritic-Cell Neoplasm. N. Engl. J. Med. 2019, 380, 1628–1637. [Google Scholar] [CrossRef] [PubMed]
  103. Alkharabsheh, O.; Frankel, A.E. Clinical Activity and Tolerability of SL-401 (Tagraxofusp): Recombinant Diphtheria Toxin and Interleukin-3 in Hematologic Malignancies. Biomedicines 2019, 7, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Xie, L.H.; Biondo, M.; Busfield, S.J.; Arruda, A.; Yang, X.; Vairo, G.; Minden, M.D. CD123 target validation and preclinical evaluation of ADCC activity of anti-CD123 antibody CSL362 in combination with NKs from AML patients in remission. Blood Cancer J. 2017, 7, e567. [Google Scholar] [CrossRef] [PubMed]
  105. Lee, E.M.; Yee, D.; Busfield, S.J.; McManus, J.F.; Cummings, N.; Vairo, G.; Wei, A.; Ramshaw, H.; Powell, J.; Lopez, A.F.; et al. Efficacy of an Fc-modified anti-CD123 antibody (CSL362) combined with chemotherapy in xenograft models of acute myelogenous leukemia in immunodeficient mice. Haematologica 2015, 100, 914–926. [Google Scholar] [CrossRef]
  106. Nievergall, E.; Ramshaw, H.S.; Yong, A.S.M.; Biondo, M.; Busfield, S.J.; Vairo, G.; Lopez, A.F.; Hughes, T.P.; White, D.L.; Hiwase, D.K. Monoclonal antibody targeting of IL-3 receptor α with CSL362 effectively depletes CML progenitor and stem cells. Blood 2014, 123, 1218–1228. [Google Scholar] [CrossRef] [Green Version]
  107. Montesinos, P.; Roboz, G.J.; Bulabois, C.-E.; Subklewe, M.; Platzbecker, U.; Ofran, Y.; Papayannidis, C.; Wierzbowska, A.; Shin, H.J.; Doronin, V.; et al. Safety and efficacy of talacotuzumab plus decitabine or decitabine alone in patients with acute myeloid leukemia not eligible for chemotherapy: Results from a multicenter, randomized, phase 2/3 study. Leukemia 2020, 35, 62–74. [Google Scholar] [CrossRef] [Green Version]
  108. Kovtun, Y.; Jones, G.E.; Adams, S.; Harvey, L.; Audette, C.A.; Wilhelm, A.; Bai, C.; Rui, L.; Laleau, R.; Liu, F.; et al. A CD123-targeting antibody-drug conjugate, IMGN632, designed to eradicate AML while sparing normal bone marrow cells. Blood Adv. 2018, 2, 848–858. [Google Scholar] [CrossRef] [Green Version]
  109. Zhang, Q.C.T.; Han, L.; Mary Kuruvilla, V.; Adams, S.; Callum, S.M.; Harutyunyan, K.; Lane, A.A.; Kovtun, Y.; Daver, N.G.; Pemmaraju, N.; et al. Pre-Clinical Efficacy of CD123-Targeting Antibody-Drug Conjugate IMGN632 in Blastic Plasmacytoid Dentritic Cell Neoplasm (BPDCN) Models. Blood 2018, 132 (Suppl. S1), 3956. [Google Scholar] [CrossRef]
  110. Daver, N.; Aribi, A.; Montesinos, P.; Roboz, G.J.; Wang, E.S.; Walter, R.B.; Jeyakumar, D.; DeAngelo, D.J.; Erba, H.P.; Advani, A.; et al. Safety and Efficacy from a Phase 1b/2 Study of IMGN632 in Combination with Aza-citidine and Venetoclax for Patients with CD123-Positive Acute Myeloid Leukemia. Blood 2021, 138 (Suppl. S1), 372. [Google Scholar] [CrossRef]
  111. Brinkmann, U.; Kontermann, R.E. Bispecific antibodies. Science 2021, 372, 916–917. [Google Scholar] [CrossRef] [PubMed]
  112. Linke, R.; Klein, A.; Seimetz, D. Catumaxomab: Clinical development and future directions. MABS 2010, 2, 129–136. [Google Scholar] [CrossRef] [Green Version]
  113. Wang, S.; Chen, K.; Lei, Q.; Ma, P.; Yuan, A.Q.; Zhao, Y.; Jiang, Y.; Fang, H.; Xing, S.; Fang, Y.; et al. The state of the art of bispecific antibodies for treating human malignancies. EMBO Mol. Med. 2021, 13, e14291. [Google Scholar] [CrossRef] [PubMed]
  114. Zhang, X.; Yang, Y.; Fan, D.; Xiong, D. The development of bispecific antibodies and their applications in tumor immune escape. Exp. Hematol. Oncol. 2017, 6, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Brinkmann, U.; Kontermann, R.E. The making of bispecific antibodies. mAbs 2016, 9, 182–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Ma, J.; Mo, Y.; Tang, M.; Shen, J.; Qi, Y.; Zhao, W.; Huang, Y.; Xu, Y.; Qian, C. Bispecific Antibodies: From Research to Clinical Application. Front. Immunol. 2021, 12, 626616. [Google Scholar] [CrossRef]
  117. Sweet-Jones, J.; Ahmad, M.; Martin, A.C. Antibody markup language (AbML)—A notation language for antibody-based drug formats and software for creating and rendering AbML (abYdraw). mAbs 2022, 14, 2101183. [Google Scholar] [CrossRef]
  118. Uy, G.L.; Aldoss, I.; Foster, M.C.; Sayre, P.H.; Wieduwilt, M.J.; Advani, A.S.; Godwin, J.E.; Arellano, M.L.; Sweet, K.L.; Emadi, A.; et al. Flotetuzumab as salvage immunotherapy for refractory acute myeloid leukemia. Blood 2021, 137, 751–762. [Google Scholar] [CrossRef]
  119. Simon, S.; Labarriere, N. PD-1 expression on tumor-specific T cells: Friend or foe for immunotherapy? Oncoimmunology 2017, 7, e1364828. [Google Scholar] [CrossRef]
  120. Roohullah, A.; Ganju, V.; Zhang, F.; Zhang, L.; Yu, T.; Wilkinson, K.; Cooper, A.; de Souza, P. First-in-human phase 1 dose escalation study of HX009, a novel recombinant humanized anti-PD-1 and CD47 bispecific antibody, in patients with advanced malignancies. J. Clin. Oncol. 2021, 39, 2517. [Google Scholar] [CrossRef]
  121. An, X.; Li, H.; Ke, H.; Xiong, L.; Zhang, F.; Zang, M.; Tu, X.; Wang, J.; Liu, D.; Chen, C.; et al. 491 Preclinical pharmacology modeling of HX009, a clinical stage first-in-class PD-1xCD47 BsAb, for anti-lymphoma applications. J. Immuno. Ther. Cancer 2022, 10, 491. [Google Scholar]
  122. Wang, Y.; Ni, H.; Zhou, S.; He, K.; Gao, Y.; Wu, W.; Wu, M.; Wu, Z.; Qiu, X.; Zhou, Y.; et al. Tumor-selective blockade of CD47 signaling with a CD47/PD-L1 bispecific antibody for enhanced anti-tumor activity and limited toxicity. Cancer Immunol. Immunother. 2021, 70, 365–376. [Google Scholar] [CrossRef]
  123. Arcila, M.E.; Nafa, K.; Chaft, J.E.; Rekhtman, N.; Lau, C.; Reva, B.A.; Zakowski, M.F.; Kris, M.G.; Ladanyi, M. EGFR exon 20 insertion mutations in lung adenocarcinomas: Prevalence, molecular het-erogeneity, and clinicopathologic characteristics. Mol Cancer Ther. 2013, 12, 220–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Puri, N.; Salgia, R. Synergism of EGFR and c-Met pathways, cross-talk and inhibition, in non-small cell lung cancer. J. Carcinog. 2008, 7, 9. [Google Scholar] [CrossRef] [PubMed]
  125. Moores, S.L.; Chiu, M.L.; Bushey, B.S.; Chevalier, K.; Luistro, L.; Dorn, K.; Brezski, R.J.; Haytko, P.; Kelly, T.; Wu, S.-J.; et al. A Novel Bispecific Antibody Targeting EGFR and cMet Is Effective against EGFR Inhibitor–Resistant Lung Tumors. Cancer Res. 2016, 76, 3942–3953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Syed, Y.Y. Amivantamab: First Approval. Drugs 2021, 81, 1349–1353. [Google Scholar] [CrossRef]
  127. Parums, D.V. Editorial: Current Status of Two Adjuvanted Malaria Vaccines and the World Health Organization (WHO) Strategy to Eradicate Malaria by 2030. Med. Sci. Monit. 2023, 29, e939357. [Google Scholar] [CrossRef] [PubMed]
  128. Li, C.; Wu, J.; Hynes, M.; Dosch, J.; Sarkar, B.; Welling, T.H.; di Magliano, M.P.; Simeone, D.M. c-Met Is a Marker of Pancreatic Cancer Stem Cells and Therapeutic Target. Gastroenterology 2011, 141, 2218–2227.e5. [Google Scholar] [CrossRef]
  129. Yan, B.; Jiang, Z.; Cheng, L.; Chen, K.; Zhou, C.; Sun, L.; Qian, W.; Li, J.; Cao, J.; Xu, Q.; et al. Paracrine HGF/c-MET enhances the stem cell-like potential and glycolysis of pancreatic cancer cells via activation of YAP/HIF-1α. Exp. Cell Res. 2018, 371, 63–71. [Google Scholar] [CrossRef]
  130. van Leenders, G.J.; Sookhlall, R.; Teubel, W.J.; Reneman, S.; Sacchetti, A.; Vissers, K.J.; van Weerden, W.; Jenster, G. Activation of c-MET induces a stem-like phenotype in human prostate cancer. PLoS ONE 2011, 6, e26753. [Google Scholar] [CrossRef] [Green Version]
  131. Lin, Y.; Lu, C.; Hsiang, Y.; Pi, S.; Chen, C.; Cheng, K.; Pan, H.; Chien, P.; Chen, Y. c-Met inhibition is required for the celecoxib-attenuated stemness property of human colorectal cancer cells. J. Cell. Physiol. 2018, 234, 10336–10344. [Google Scholar] [CrossRef]
  132. Imrich, S.; Hachmeister, M.; Gires, O. EpCAM and its potential role in tumor-initiating cells. Cell Adhes. Migr. 2012, 6, 30–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Seimetz, D.; Lindhofer, H.; Bokemeyer, C. Development and approval of the trifunctional antibody catumaxomab (an-ti-EpCAM x anti-CD3) as a targeted cancer immunotherapy. Cancer Treat Rev. 2010, 36, 458–467. [Google Scholar] [CrossRef]
  134. Jager, M.; Schoberth, A.; Ruf, P.; Hess, J.; Hennig, M.; Schmalfeldt, B.; Wimberger, P.; Strohlein, M.; Theissen, B.; Heiss, M.M.; et al. Immunomonitoring results of a phase II/III study of malignant ascites patients treated with the trifunctional antibody catumaxomab (anti-EpCAM x anti-CD3). Cancer Res. 2012, 72, 24–32. [Google Scholar] [CrossRef] [Green Version]
  135. Morris, E.C.; Neelapu, S.S.; Giavridis, T.; Sadelain, M. Cytokine release syndrome and associated neurotoxicity in cancer im-mu-notherapy. Nat. Rev. Immunol. 2022, 22, 85–96. [Google Scholar] [CrossRef]
  136. Esfandiari, A.; Cassidy, S.; Webster, R.M. Bispecific antibodies in oncology. Nat. Rev. Drug Discov. 2022, 21, 411–412. [Google Scholar] [CrossRef]
  137. Gera, N. The evolution of bispecific antibodies. Expert Opin. Biol. Ther. 2022, 22, 945–949. [Google Scholar] [CrossRef]
  138. Krief, P.; Boucheix, C.; Billard, C.; Mishal, Z.; van Agthoven, A.; Fiers, W.; Azzarone, B. Modulation of expression of class II histocompatibility antigens by secretion of a cellular inhibitor in K562 leukemic cells. Eur. J. Immunol. 1987, 17, 1021–1025. [Google Scholar] [CrossRef]
  139. Peitzsch, C.; Perrin, R.; Hill, R.P.; Dubrovska, A.; Kurth, I. Hypoxia as a biomarker for radioresistant cancer stem cells. Int. J. Radiat. Biol. 2014, 90, 636–652. [Google Scholar] [CrossRef] [PubMed]
  140. Cruz, E.; Kayser, V. Monoclonal antibody therapy of solid tumors: Clinical limitations and novel strategies to enhance treatment efficacy. Biol. Targets Ther. 2019, 13, 33–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Hegde, P.S.; Chen, D.S. Top 10 Challenges in Cancer Immunotherapy. Immunity 2020, 52, 17–35. [Google Scholar] [CrossRef]
  142. Prasetyanti, P.R.; Medema, J.P. Intra-tumor heterogeneity from a cancer stem cell perspective. Mol. Cancer 2017, 16, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Schwarz, F.M.; Schniewind, I.; Besso, M.J.; Lange, S.; Linge, A.; Patil, S.G.; Loeck, S.; Klusa, D.; Dietrich, A.; Voss-Boehme, A.; et al. Plasticity within Aldehyde Dehydrogenase–Positive Cells Determines Prostate Cancer Radiosensitivity. Mol. Cancer Res. 2022, 20, 794–809. [Google Scholar] [CrossRef]
  144. Schniewind, I.; Hadiwikarta, W.W.; Grajek, J.; Poleszczuk, J.; Richter, S.; Peitzsch, M.; Muller, J.; Klusa, D.; Beyreuther, E.; Lock, S.; et al. Cellular plasticity upon proton irradiation determines tumor cell radio-sen-sitivity. Cell Rep. 2022, 38, 110422. [Google Scholar] [CrossRef] [PubMed]
  145. Kim, W.-T.; Ryu, C.J. Cancer stem cell surface markers on normal stem cells. BMB Rep. 2017, 50, 285–298. [Google Scholar] [CrossRef] [Green Version]
  146. Betts, A.; van der Graaf, P.H. Mechanistic Quantitative Pharmacology Strategies for the Early Clinical Development of Bispecific Antibodies in Oncology. Clin. Pharm. Ther. 2020, 108, 528–541. [Google Scholar] [CrossRef] [PubMed]
  147. Elshiaty, M.; Schindler, H.; Christopoulos, P. Principles and Current Clinical Landscape of Multispecific Antibodies against Cancer. Int. J. Mol. Sci. 2021, 22, 5632. [Google Scholar] [CrossRef]
  148. Börjesson, P.K.E.; Postema, E.J.; Roos, J.C.; Colnot, D.R.; Marres, H.A.M.; Van Schie, M.H.; Stehle, G.; De Bree, R.; Snow, G.B.; Oyen, W.J.G.; et al. Phase I therapy study with (186)Re-labeled humanized monoclonal antibody BIWA 4 (bivatuzumab) in patients with head and neck squamous cell carcinoma. Clin. Cancer Res. 2003, 9, 3961. [Google Scholar]
  149. Colnot, D.R.; Quak, J.J.; Roos, J.C.; Van Lingen, A.; Wilhelm, A.J.; Van Kamp, G.J.; Huijgens, P.C.; Snow, G.B.; Van Dongen, G.A. Phase I therapy study of 186Re-labeled chimeric monoclonal antibody U36 in patients with squamous cell carcinoma of the head and neck. J. Nucl. Med. 2000, 41, 1999–2010. [Google Scholar] [PubMed]
  150. Riechelmann, H.; Sauter, A.; Golze, W.; Hanft, G.; Schroen, C.; Hoermann, K.; Erhardt, T.; Gronau, S. Phase I trial with the CD44v6-targeting immunoconjugate bivatuzumab mertansine in head and neck squamous cell carcinoma. Oral. Oncol. 2008, 44, 823–829. [Google Scholar] [CrossRef]
  151. Rupp, U.; Schoendorf-Holland, E.; Eichbaum, M.; Schuetz, F.; Lauschner, I.; Schmidt, P.; Staab, A.; Hanft, G.; Huober, J.; Sinn, H.-P.; et al. Safety and pharmacokinetics of bivatuzumab mertansine in patients with CD44v6-positive metastatic breast cancer: Final results of a phase I study. Anti-Cancer Drugs 2007, 18, 477–485. [Google Scholar] [CrossRef] [PubMed]
  152. Sauter, A.; Kloft, C.; Gronau, S.; Bogeschdorfer, F.; Erhardt, T.; Golze, W.; Schroen, C.; Staab, A.; Riechelmann, H.; Hoermann, K. Pharmacokinetics, immunogenicity and safety of bivatuzumab mertansine, a novel CD44v6-targeting immunoconjugate, in patients with squamous cell carcinoma of the head and neck. Int. J. Oncol. 2007, 30, 927–935. [Google Scholar] [CrossRef] [PubMed]
  153. Kubasch, A.S.; Schulze, F.; Giagounidis, A.; Götze, K.S.; Krönke, J.; Sockel, K.; Middeke, J.M.; Chermat, F.; Gloaguen, S.; Puttrich, M.; et al. Single agent talacotuzumab demonstrates limited efficacy but considerable toxicity in elderly high-risk MDS or AML patients failing hypomethylating agents. Leukemia 2019, 34, 1182–1186. [Google Scholar] [CrossRef] [Green Version]
  154. Daver, N.G.; Montesinos, P.; DeAngelo, D.J.; Roboz, G.J.; Wang, E.S.; Walter, R.B.; Jeyakumar, D.; DeAngelo, D.J.; Erba, H.P.; Advani, A.; et al. A phase I/II study of IMGN632, a novel CD123-targeting antibody-drug conjugate, in patients with relapsed/refractory acute myeloid leukemia, blastic plasmacytoid dendritic cell neoplasm, and other CD123-positive hematologic malignancies. J. Clin. Oncol. 2020, 38 (Suppl. S15), TPS7563. [Google Scholar] [CrossRef]
  155. Voskoboinik, I.; Whisstock, J.C.; Trapani, J.A. Perforin and granzymes: Function, dysfunction and human pathology. Nat. Rev. Immunol. 2015, 15, 388–400. [Google Scholar] [CrossRef] [PubMed]
  156. Wolf, N.K.; Kissiov, D.U.; Raulet, D.H. Roles of natural killer cells in immunity to cancer, and applications to immunotherapy. Nat. Rev. Immunol. 2022, 23, 90–105. [Google Scholar] [CrossRef]
  157. Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef]
  158. Rosenberg, S.A.; Packard, B.S.; Aebersold, P.M.; Solomon, D.; Topalian, S.L.; Toy, S.T.; Simon, P.; Lotze, M.T.; Yang, J.C.; Seipp, C.A.; et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immu-no-therapy of patients with metastatic melanoma. A preliminary report. N. Engl. J. Med. 1988, 319, 1676–1680. [Google Scholar] [CrossRef]
  159. Rosenberg, S.A.; Restifo, N.P. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 2015, 348, 62–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Gross, G.; Waks, T.; Eshhar, Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl. Acad. Sci. USA 1989, 86, 10024–10028. [Google Scholar] [CrossRef] [Green Version]
  161. Kershaw, M.H.; Westwood, J.A.; Parker, L.L.; Wang, G.; Eshhar, Z.; Mavroukakis, S.A.; White, D.E.; Wunderlich, J.R.; Canevari, S.; Rogers-Freezer, L.; et al. A Phase I Study on Adoptive Immunotherapy Using Gene-Modified T Cells for Ovarian Cancer. Clin. Cancer Res. 2006, 12, 6106–6115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Maher, J.; Brentjens, R.J.; Gunset, G.; Riviere, I.; Sadelain, M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta /CD28 receptor. Nat. Biotechnol. 2002, 20, 70–75. [Google Scholar] [CrossRef] [PubMed]
  163. Krause, A.; Guo, H.-F.; Latouche, J.-B.; Tan, C.; Cheung, N.-K.V.; Sadelain, M. Antigen-dependent CD28 Signaling Selectively Enhances Survival and Proliferation in Genetically Modified Activated Human Primary T Lymphocytes. J. Exp. Med. 1998, 188, 619–626. [Google Scholar] [CrossRef] [Green Version]
  164. Long, A.H.; Haso, W.M.; Shern, J.F.; Wanhainen, K.M.; Murgai, M.; Ingaramo, M.; Smith, J.P.; Walker, A.J.; Kohler, M.E.; Venkateshwara, V.R.; et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat. Med. 2015, 21, 581–590. [Google Scholar] [CrossRef] [Green Version]
  165. Zhong, X.-S.; Matsushita, M.; Plotkin, J.; Riviere, I.; Sadelain, M. Chimeric Antigen Receptors Combining 4-1BB and CD28 Signaling Domains Augment PI3kinase/AKT/Bcl-XL Activation and CD8+ T Cell–mediated Tumor Eradication. Mol. Ther. 2010, 18, 413–420. [Google Scholar] [CrossRef]
  166. Jayaraman, J.; Mellody, M.P.; Hou, A.J.; Desai, R.P.; Fung, A.W.; Pham, A.H.T.; Chen, Y.Y.; Zhao, W. CAR-T design: Elements and their synergistic function. eBiomedicine 2020, 58, 102931. [Google Scholar] [CrossRef] [PubMed]
  167. Billadeau, D.D.; Upshaw, J.L.; Schoon, R.A.; Dick, C.J.; Leibson, P.J. NKG2D-DAP10 triggers human NK cell–mediated killing via a Syk-independent regulatory pathway. Nat. Immunol. 2003, 4, 557–564. [Google Scholar] [CrossRef]
  168. Töpfer, K.; Cartellieri, M.; Michen, S.; Wiedemuth, R.; Müller, N.; Lindemann, D.; Bachmann, M.; Füssel, M.; Schackert, G.; Temme, A. DAP12-Based Activating Chimeric Antigen Receptor for NK Cell Tumor Immunotherapy. J. Immunol. 2015, 194, 3201–3212. [Google Scholar] [CrossRef] [Green Version]
  169. Curio, S.; Jonsson, G.; Marinović, S. A summary of current NKG2D-based CAR clinical trials. Immunother. Adv. 2021, 1, ltab018. [Google Scholar] [CrossRef]
  170. Wang, X.; Rivière, I. Clinical manufacturing of CAR T cells: Foundation of a promising therapy. Mol. Ther. Oncolytics 2016, 3, 16015. [Google Scholar] [CrossRef] [Green Version]
  171. Barros, L.R.C.; Couto, S.C.F.; Santurio, D.D.S.; Paixão, E.A.; Cardoso, F.; da Silva, V.J.; Klinger, P.; Ribeiro, P.D.A.C.; Rós, F.A.; Oliveira, T.G.M.; et al. Systematic Review of Available CAR-T Cell Trials around the World. Cancers 2022, 14, 2667. [Google Scholar] [CrossRef] [PubMed]
  172. Lin, X.; Lee, S.; Sharma, P.; George, B.; Scott, J. Summary of US Food and Drug Administration Chimeric Antigen Receptor (CAR) T-Cell Biologics License Application Approvals from a Statistical Perspective. J. Clin. Oncol. 2022, 40, 3501–3509. [Google Scholar] [CrossRef]
  173. Braendstrup, P.; Levine, B.L.; Ruella, M. The long road to the first FDA-approved gene therapy: Chimeric antigen receptor T cells targeting CD. Cytotherapy 2020, 22, 57–69. [Google Scholar] [CrossRef] [PubMed]
  174. Yi, D.; Gergis, M.; Hsu, J.; Yang, Y.; Bi, X.; Aljurf, M.; Gergis, U. Next-Generation Chimeric Antigen Receptor T Cells. Hematol. Stem Cell Ther. 2022, 15, 11. [Google Scholar] [CrossRef]
  175. Sengsayadeth, S.; Savani, B.N.; Oluwole, O.; Dholaria, B. Overview of approved CAR-T therapies, ongoing clinical trials, and its impact on clinical practice. eJHaem 2022, 3 (Suppl. S1), 6–10. [Google Scholar] [CrossRef] [PubMed]
  176. Morgan, R.G.; Mortensson, E.; Williams, A.C. Targeting LGR5 in Colorectal Cancer: Therapeutic gold or too plastic? Br. J. Cancer 2018, 118, 1410–1418. [Google Scholar] [CrossRef]
  177. Li, W.; Zhang, N.; Jin, C.; Long, M.D.; Rajabi, H.; Yasumizu, Y.; Fushimi, A.; Yamashita, N.; Hagiwara, M.; Zheng, R.; et al. MUC1-C drives stemness in progression of colitis to colorectal cancer. J. Clin. Investig. 2020, 5, e137112. [Google Scholar] [CrossRef]
  178. Kufe, D.W. Chronic activation of MUC1-C in wound repair promotes progression to cancer stem cells. J. Cancer Metastasis Treat. 2022, 8, 12. [Google Scholar] [CrossRef]
  179. Shah, P.D.; Huang, A.C.C.; Xu, X.; Zhang, P.Z.; Orlowski, R.; Matlawski, T.; Shea, J.; Cervini, A.; Amaravadi, R.K.; Tchou, J.C.; et al. Phase I trial of autologous cMET-directed CAR-T cells administered intravenously in patients with melanoma & breast carcinoma. J. Clin. Oncol. 2020, 38 (Suppl. S15), 10035. [Google Scholar]
  180. Blank, C.U.; Haining, W.N.; Held, W.; Hogan, P.G.; Kallies, A.; Lugli, E.; Lynn, R.C.; Philip, M.; Rao, A.; Restifo, N.P.; et al. Defining ‘T cell exhaustion’. Nat. Rev. Immunol. 2019, 19, 665–674. [Google Scholar] [CrossRef]
  181. Rodriguez-Garcia, A.; Lynn, R.C.; Poussin, M.; Eiva, M.A.; Shaw, L.C.; O’Connor, R.S.; Minutolo, N.G.; Casado-Medrano, V.; Lopez, G.; Matsuyama, T.; et al. CAR-T cell-mediated depletion of immunosuppressive tumor-associated macrophages promotes endogenous antitumor immunity and augments adoptive immunotherapy. Nat. Commun. 2021, 12, 1–17. [Google Scholar]
  182. Baur, K.; Heim, D.; Beerlage, A.; Poerings, A.S.; Kopp, B.; Medinger, M.; Dirks, J.C.; Passweg, J.R.; Holbro, A. Dasatinib for treatment of CAR T-cell therapy-related complications. J. Immunother. Cancer 2022, 10, e005956. [Google Scholar] [CrossRef]
  183. Ran, T.; Eichmüller, S.B.; Schmidt, P.; Schlander, M. Cost of decentralized CAR T-cell production in an academic nonprofit setting. Int. J. Cancer 2020, 147, 3438–3445. [Google Scholar] [CrossRef] [PubMed]
  184. Zah, E.; Nam, E.; Bhuvan, V.; Tran, U.; Ji, B.Y.; Gosliner, S.B.; Wang, X.; Brown, C.E.; Chen, Y.Y. Systematically optimized BCMA/CS1 bispecific CAR-T cells robustly control heterogeneous multiple myeloma. Nat. Commun. 2020, 11, 1–13. [Google Scholar] [CrossRef]
  185. Kloss, C.C.; Condomines, M.; Cartellieri, M.; Bachmann, M.; Sadelain, M. Combinatorial antigen recognition with balanced sig-naling promotes selective tumor eradication by engineered T cells. Nat. Biotechnol. 2013, 31, 71–75. [Google Scholar] [CrossRef] [PubMed]
  186. Bielamowicz, K.; Fousek, K.; Byrd, T.T.; Samaha, H.; Mukherjee, M.; Aware, N.; Wu, M.-F.; Orange, J.S.; Sumazin, P.; Man, T.-K.; et al. Trivalent CAR T cells overcome interpatient antigenic variability in glioblastoma. Neuro-Oncol. 2018, 20, 506–518. [Google Scholar] [CrossRef] [Green Version]
  187. Philip, B.; Kokalaki, E.; Mekkaoui, L.; Thomas, S.; Straathof, K.; Flutter, B.; Marin, V.; Marafioti, T.; Chakraverty, R.; Linch, D.; et al. A highly compact epitope-based marker/suicide gene for easier and safer T-cell therapy. Blood 2014, 124, 1277–1287. [Google Scholar] [CrossRef] [PubMed]
  188. Jones, B.S.; Lamb, L.S.; Egoldman, F.; Stasi, A.E. Improving the safety of cell therapy products by suicide gene transfer. Front. Pharmacol. 2014, 5, 254. [Google Scholar] [CrossRef]
  189. Raes, L.; Stremersch, S.; Fraire, J.C.; Brans, T.; Goetgeluk, G.; De Munter, S.; Van Hoecke, L.; Verbeke, R.; Van Hoeck, J.; Xiong, R.; et al. Intracellular Delivery of mRNA in Adherent and Suspension Cells by Vapor Nanobubble Photoporation. Nano-Micro Lett. 2020, 12, 1–17. [Google Scholar] [CrossRef]
  190. Foster, J.B.; Barrett, D.M.; Karikó, K. The Emerging Role of In Vitro-Transcribed mRNA in Adoptive T Cell Immunotherapy. Mol. Ther. 2019, 27, 747–756. [Google Scholar] [CrossRef] [Green Version]
  191. Arndt, C.; Fasslrinner, F.; Loureiro, L.R.; Koristka, S.; Feldmann, A.; Bachmann, M. Adaptor CAR Platforms—Next Generation of T Cell-Based Cancer Immunotherapy. Cancers 2020, 12, 1302. [Google Scholar] [CrossRef] [PubMed]
  192. Koristka, M.S.; Cartellieri, M.; Feldmann, A.; Arndt, M.C.; Loff, M.S.; Michalk, M.I.; Aliperta, M.R.; Von Bonin, M.; Bornhäuser, M.; Ehninger, A.; et al. Flexible Antigen-Specific Redirection of Human Regulatory T Cells Via a Novel Universal Chimeric Antigen Receptor System. Blood 2014, 124, 3494. [Google Scholar] [CrossRef]
  193. Cartellieri, M.; Loff, S.; von Bonin, M.; Arndt, C.; Loff, S.; Ehninger, A.; von Bonin, M.; Bejestani, E.P.; Ehninger, G.; Bachmann, M.P. Unicar: A Novel Modular Retargeting Platform Technology for CAR T Cells. Blood 2015, 125, 5549. [Google Scholar] [CrossRef]
  194. Cao, Y.; Rodgers, D.T.; Du, J.; Ahmad, I.; Hampton, E.N.; Ma, J.S.Y.; Mazagova, M.; Choi, S.-H.; Yun, H.Y.; Xiao, H.; et al. Design of Switchable Chimeric Antigen Receptor T Cells Targeting Breast Cancer. Angew. Chem. Int. Ed. 2016, 55, 7520–7524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Tamada, K.; Geng, D.; Sakoda, Y.; Bansal, N.; Srivastava, R.; Li, Z.; Davila, E. Redirecting Gene-Modified T Cells toward Various Cancer Types Using Tagged Antibodies. Clin. Cancer Res. 2012, 18, 6436–6445. [Google Scholar] [CrossRef] [Green Version]
  196. Urbanska, K.; Lanitis, E.; Poussin, M.; Lynn, R.C.; Gavin, B.P.; Kelderman, S.; Yu, J.; Scholler, N.; Powell, D.J., Jr. A Universal Strategy for Adoptive Immunotherapy of Cancer through Use of a Novel T-cell Antigen Receptor. Cancer Res. 2012, 72, 1844–1852. [Google Scholar] [CrossRef] [Green Version]
  197. Rong, L.; Lim, R.M.; Yin, X.; Tan, L.; Yang, J.H.; Xie, J. Site-Specific Dinitrophenylation of Single-Chain Antibody Fragments for Redirecting a Universal CAR-T Cell against Cancer Antigens. J. Mol. Biol. 2022, 434, 167513. [Google Scholar] [CrossRef]
  198. Urbanska, K.; Lynn, R.C.; Stashwick, C.; Thakur, A.; Lum, L.G.; Powell, D.J. Targeted cancer immunotherapy via combination of designer bispecific antibody and novel gene-engineered T cells. J. Transl. Med. 2014, 12, 1–12. [Google Scholar] [CrossRef] [Green Version]
  199. Klesmith, J.R.; Su, L.; Wu, L.; Klesmith, J.R.; Su, L.; Wu, L.; Schrack, I.A.; Dufort, F.J.; Birt, A.; Ambrose, C.; et al. Retargeting CD19 Chimeric Antigen Receptor T Cells via Engineered CD19-Fusion Proteins. Mol. Pharm. 2019, 16, 3544–3558. [Google Scholar] [CrossRef] [PubMed]
  200. Karches, C.H.; Benmebarek, M.-R.; Schmidbauer, M.L.; Kurzay, M.; Klaus, R.; Geiger, M.; Rataj, F.; Cadilha, B.L.; Lesch, S.; Heise, C.; et al. Bispecific Antibodies Enable Synthetic Agonistic Receptor-Transduced T Cells for Tumor Immunotherapy. Clin. Cancer Res. 2019, 25, 5890–5900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Feldmann, A.; Hoffmann, A.; Bergmann, R.; Koristka, S.; Berndt, N.; Arndt, C.; Loureiro, L.R.; Kittel-Boselli, E.; Mitwasi, N.; Kegler, A.; et al. Versatile chimeric antigen receptor platform for controllable and combinatorial T cell therapy. Oncoimmunology 2020, 9, 1785608. [Google Scholar] [CrossRef]
  202. Borrok, M.J.; Li, Y.; Harvilla, P.B.; Maruthachalam, B.V.; Tamot, N.; Prokopowitz, C.; Chen, J.; Venkataramani, S.; Grewal, I.S.; Ganesan, R.; et al. Conduit CAR: Redirecting CAR T-Cell Specificity with A Universal and Adaptable Bispecific Antibody Platform. Cancer Res. Commun. 2022, 2, 146–157. [Google Scholar] [CrossRef]
  203. Carmo-Fonseca, M.; Pfeifer, K.; Schröder, H.C.; Vaz, M.; Fonseca, J.; Müller, W.E.; Bachmann, M. Identification of La ribonucleoproteins as a component of interchromatin granules. Exp. Cell Res. 1989, 185, 73–85. [Google Scholar] [CrossRef] [PubMed]
  204. Bartsch, T.; Arndt, C.; Loureiro, L.R.; Kegler, A.; Puentes-Cala, E.; Soto, J.A.; Kurien, B.T.; Feldmann, A.; Berndt, N.; Bachmann, M.P. A Small Step, a Giant Leap: Somatic Hypermutation of a Single Amino Acid Leads to Anti-La Autoreactivity. Int. J. Mol. Sci. 2021, 22, 12046. [Google Scholar] [CrossRef]
  205. Bachmann, M.; Bartsch, T.; Bippes, C.; Bachmann, D.; Puentes-Cala, E.; Bachmann, J.; Bartsch, H.; Arndt, C.; Koristka, S.; Loureiro, L.; et al. T Cell Mediated Conversion of a Non-Anti-La Reactive B Cell to an Autoreactive Anti-La B Cell by Somatic Hypermutation. Int. J. Mol. Sci. 2021, 22, 1198. [Google Scholar] [CrossRef] [PubMed]
  206. Bachmann, M. The UniCAR system: A modular CAR T cell approach to improve the safety of CAR T cells. Immunol. Lett. 2019, 211, 13–22. [Google Scholar] [CrossRef]
  207. Arndt, C.; Feldmann, A.; Koristka, S.; Schäfer, M.; Bergmann, R.; Mitwasi, N.; Berndt, N.; Bachmann, D.; Kegler, A.; Schmitz, M.; et al. A theranostic PSMA ligand for PET imaging and retargeting of T cells expressing the universal chimeric antigen receptor UniCAR. Oncoimmunology 2019, 8, 1659095. [Google Scholar] [CrossRef] [Green Version]
  208. Albert, S.; Arndt, C.; Koristka, S.; Berndt, N.; Bergmann, R.; Feldmann, A.; Schmitz, M.; Pietzsch, J.; Steinbach, J.; Bachmann, M. From mono- to bivalent: Improving theranostic properties of target modules for redi-rection of UniCAR T cells against EGFR-expressing tumor cells in vitro and in vivo. Oncotarget 2018, 9, 25597–25616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Feldmann, A.; Arndt, C.; Bergmann, R.; Loff, S.; Cartellieri, M.; Bachmann, D.; Aliperta, R.; Hetzenecker, M.; Ludwig, F.; Albert, S.; et al. Retargeting of T lymphocytes to PSCA- or PSMA positive prostate cancer cells using the novel modular chimeric antigen receptor platform technology “UniCAR”. Oncotarget 2017, 8, 31368–31385. [Google Scholar] [CrossRef] [Green Version]
  210. Koristka, S.; Ziller-Walter, P.; Bergmann, R.; Arndt, C.; Feldmann, A.; Kegler, A.; Cartellieri, M.; Ehninger, A.; Ehninger, G.; Bornhäuser, M.; et al. Anti-CAR-engineered T cells for epitope-based elimination of autologous CAR T cells. Cancer Immunol. Immunother. 2019, 68, 1401–1415. [Google Scholar] [CrossRef] [Green Version]
  211. Loureiro, L.R.; Feldmann, A.; Bergmann, R.; Koristka, S.; Berndt, N.; Máthé, D.; Hegedüs, N.; Szigeti, K.; Videira, P.A.; Bachmann, M.; et al. Extended half-life target module for sustainable UniCAR T-cell treatment of STn-expressing cancers. J. Exp. Clin. Cancer Res. 2020, 39, 77. [Google Scholar] [CrossRef]
  212. Mitwasi, N.; Feldmann, A.; Bergmann, R.; Berndt, N.; Arndt, C.; Koristka, S.; Kegler, A.; Jureczek, J.; Hoffmann, A.; Ehninger, A.; et al. Development of novel target modules for retargeting of UniCAR T cells to GD2 positive tumor cells. Oncotarget 2017, 8, 108584–108603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Kittel-Boselli, E.; Soto, K.E.G.; Loureiro, L.R.; Hoffmann, A.; Bergmann, R.; Arndt, C.; Koristka, S.; Mitwasi, N.; Kegler, A.; Bartsch, T.; et al. Targeting Acute Myeloid Leukemia Using the RevCAR Platform: A Pro-grammable, Switchable and Combinatorial Strategy. Cancers 2021, 13, 4785. [Google Scholar] [CrossRef]
  214. Stock, S.; Benmebarek, M.-R.; Kluever, A.-K.; Darowski, D.; Jost, C.; Stubenrauch, K.-G.; Benz, J.; Freimoser-Grundschober, A.; Moessner, E.; Umana, P.; et al. Chimeric antigen receptor T cells engineered to recognize the P329G-mutated Fc part of effector-silenced tumor antigen-targeting human IgG1 antibodies enable modular targeting of solid tumors. J. Immunother. Cancer 2022, 10, e005054. [Google Scholar] [CrossRef]
  215. Kuo, Y.-C.; Kuo, C.-F.; Jenkins, K.; Hung, A.F.-H.; Chang, W.-C.; Park, M.; Aguilar, B.; Starr, R.; Hibbard, J.; Brown, C.; et al. Antibody-based redirection of universal Fabrack-CAR T cells selectively kill antigen bearing tumor cells. J. Immunother. Cancer 2022, 10, e003752. [Google Scholar] [CrossRef]
  216. Cartellieri, M.; Feldmann, A.; Koristka, S.; Arndt, C.; Loff, S.; Ehninger, A.V.; Von Bonin, M.; Bejestani, E.P.; Ehninger, G.; Bachmann, M.P. Switching CAR T cells on and off: A novel modular platform for retargeting of T cells to AML blasts. Blood Cancer J. 2016, 6, e458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Loff, S.; Dietrich, J.; Meyer, J.-E.; Riewaldt, J.; Spehr, J.; von Bonin, M.; Gründer, C.; Swayampakula, M.; Franke, K.; Feldmann, A.; et al. Rapidly Switchable Universal CAR-T Cells for Treatment of CD123-Positive Leukemia. Mol. Ther. Oncolytics 2020, 17, 408–420. [Google Scholar] [CrossRef] [PubMed]
  218. Minutolo, N.G.; Sharma, P.; Poussin, M.; Shaw, L.C.; Brown, D.P.; Hollander, E.E.; Smole, A.; Rodriguez-Garcia, A.; Hui, J.Z.; Zappala, F.; et al. Quantitative Control of Gene-Engineered T-Cell Activity through the Covalent Attachment of Targeting Ligands to a Universal Immune Receptor. J. Am. Chem. Soc. 2020, 142, 6554–6568. [Google Scholar] [CrossRef]
  219. Arndt, C.; Loureiro, L.R.; Feldmann, A.; Jureczek, J.; Bergmann, R.; Máthé, D.; Hegedüs, N.; Berndt, N.; Koristka, S.; Mitwasi, N.; et al. UniCAR T cell immunotherapy enables efficient elimination of radioresistant cancer cells. Oncoimmunology 2020, 9, 1743036. [Google Scholar] [CrossRef] [Green Version]
  220. Wermke, M.; Kraus, S.; Ehninger, A.; Bargou, R.C.; Goebeler, M.E.; Middeke, J.M.; Kreissig, C.; von Bonin, M.; Koedam, J.; Pehl, M.; et al. Proof of concept for a rapidly switchable universal CAR-T platform with UniCAR-T-CD123 in relapsed/refractory AML. Blood 2021, 137, 3145–3148. [Google Scholar] [CrossRef]
  221. Ehninger, G.; Kraus, S.; Sala, E.; Metzelder, S.K.; Vucinic, V.; Fiedler, W.; Goebeler, M.E.; Middeke, J.M.; von Bonin, M.; Kreissig, C.; et al. Phase 1 Dose Escalation Study of the Rapidly Switchable Universal CAR-T Therapy Unicar-T-CD123 in Relapsed/Refractory AML. Blood 2022, 140, 2367–2368. [Google Scholar] [CrossRef]
  222. Xie, G.; Dong, H.; Liang, Y.; Ham, J.D.; Rizwan, R.; Chen, J. CAR-NK cells: A promising cellular immunotherapy for cancer. eBiomedicine 2020, 59, 102975. [Google Scholar] [CrossRef]
  223. Simonetta, F.; Alvarez, M.; Negrin, R.S. Natural Killer Cells in Graft-versus-Host-Disease after Allogeneic Hematopoietic Cell Transplantation. Front. Immunol. 2017, 8, 465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Miller, J.S.; Soignier, Y.; Panoskaltsis-Mortari, A.; McNearney, S.A.; Yun, G.H.; Fautsch, S.K.; McKenna, D.; Le, C.; Defor, T.E.; Burns, L.J.; et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 2005, 105, 3051–3057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Bjorklund, A.T.; Carlsten, M.; Sohlberg, E.; Liu, L.L.; Clancy, T.; Karimi, M.; Cooley, S.; Miller, J.S.; Klimkowska, M.; Schaffer, M.; et al. Complete Remission with Reduction of High-Risk Clones following Hap-loidentical NK-Cell Therapy against MDS and AML. Clin. Cancer Res. 2018, 24, 1834–1844. [Google Scholar] [CrossRef] [Green Version]
  226. Du, W.; Cui, L.; Zhang, J.; Zhang, H.; Liu, R.; Yang, W.; Zhang, Y. Generation of universal natural killer cells from a cryopreserved cord blood mononuclear cell-derived induced pluripotent stem cell library. FEBS Open Bio 2022, 12, 1771–1781. [Google Scholar] [CrossRef] [PubMed]
  227. Kerbauy, L.N.; Marin, N.D.; Kaplan, M.; Banerjee, P.P.; Berrien-Elliott, M.M.; Becker-Hapak, M.; Basar, R.; Foster, M.; Garcia Melo, L.; Neal, C.C.; et al. Combining AFM13, a Bispecific CD30/CD16 Antibody, with Cytokine-Activated Blood and Cord Blood-Derived NK Cells Facilitates CAR-like Responses Against CD30(+) Malignancies. Clin. Cancer Res. 2021, 27, 3744–3756. [Google Scholar] [CrossRef]
  228. Heipertz, E.L.; Zynda, E.R.; Stav-Noraas, T.E.; Hungler, A.D.; Boucher, S.E.; Kaur, N.; Vemuri, M.C. Current Perspectives on “Off-The-Shelf” Allogeneic NK and CAR-NK Cell Therapies. Front. Immunol. 2021, 12, 732135. [Google Scholar] [CrossRef] [PubMed]
  229. Valeri, A.; García-Ortiz, A.; Castellano, E.; Córdoba, L.; Maroto-Martín, E.; Encinas, J.; Leivas, A.; Río, P.; Martínez-López, J. Overcoming tumor resistance mechanisms in CAR-NK cell therapy. Front. Immunol. 2022, 13, 953849. [Google Scholar] [CrossRef]
  230. Carrabba, M.G.C.; Hudecek, M.; Quintarelli, C.; Briones, J.; Hajek, R.; Sierra, J.; Locatelli, F.; Einsele, H.; Bordignon, C.; Ciceri, F.; et al. Phase I-IIa Clinical Trial to Assess Safety and Efficacy of MLM-CAR44.1, a CD44v6 Directed CAR-T in Relapsed/Refractory Acute Myeloid Leukemia (AML) and Multiple Myeloma (MM). Blood 2018, 132 (Suppl. S1), 5790. [Google Scholar] [CrossRef]
  231. Dai, H.; Tong, C.; Shi, D.; Chen, M.; Guo, Y.; Chen, D.; Han, X.; Wang, H.; Wang, Y.; Shen, P. Efficacy and biomarker analysis of CD133-directed CAR T cells in advanced hepatocellular car-cinoma: A single-arm, open-label, phase II trial. Oncoimmunology 2020, 9, 1846926. [Google Scholar] [CrossRef]
  232. Rowley, D.A.; Fitch, F.W. The road to the discovery of dendritic cells, a tribute to Ralph Steinman. Cell. Immunol. 2012, 273, 95–98. [Google Scholar] [CrossRef] [PubMed]
  233. Laureano, R.S.; Sprooten, J.; Vanmeerbeerk, I.; Borras, D.M.; Govaerts, J.; Naulaerts, S.; Berneman, Z.N.; Beuselinck, B.; Bol, K.F.; Borst, J.; et al. Trial watch: Dendritic cell (DC)-based immunotherapy for cancer. Oncoimmunology 2022, 11, 2096363. [Google Scholar] [CrossRef] [PubMed]
  234. van Willigen, W.W.; Bloemendal, M.; Gerritsen, W.R.; Schreibelt, G.; de Vries, I.J.M.; Bol, K.F. Dendritic Cell Cancer Therapy: Vac-cinating the Right Patient at the Right Time. Front Immunol. 2018, 9, 2265. [Google Scholar] [CrossRef] [PubMed]
  235. Teramoto, K.; Ozaki, Y.; Hanaoka, J.; Sawai, S.; Tezuka, N.; Fujino, S.; Daigo, Y.; Kontani, K. Predictive biomarkers and effectiveness of MUC1-targeted dendritic-cell-based vaccine in patients with refractory non-small cell lung cancer. Ther. Adv. Med Oncol. 2016, 9, 147–157. [Google Scholar] [CrossRef] [Green Version]
  236. Ogasawara, M.; Miyashita, M.; Yamagishi, Y.; Ota, S. Dendritic cell vaccination combined with a conventional chemotherapy for patients with relapsed or advanced pancreatic ductal adenocarcinoma: A single-center phase I/II trial. Ther. Apher. Dial. 2021, 25, 415–424. [Google Scholar] [CrossRef]
  237. Nagai, K.; Adachi, T.; Harada, H.; Eguchi, S.; Sugiyama, H.; Miyazaki, Y. Dendritic Cell-based Immunotherapy Pulsed with Wilms Tumor 1 Peptide and Mucin 1 as an Adjuvant Therapy for Pancreatic Ductal Adenocarcinoma After Curative Resection: A Phase I/IIa Clinical Trial. Anticancer. Res. 2020, 40, 5765–5776. [Google Scholar] [CrossRef]
  238. Lepisto, A.J.; Moser, A.J.; Zeh, H.; Lee, K.; Bartlett, D.; McKolanis, J.R.; Geller, B.A.; Schmotzer, A.; Potter, D.P.; Whiteside, T.; et al. A phase I/II study of a MUC1 peptide pulsed autologous dendritic cell vaccine as adjuvant therapy in patients with resected pancreatic and biliary tumors. Cancer Ther. 2008, 6, 955–964. [Google Scholar]
  239. Westdorp, H.; Creemers, J.H.A.; van Oort, I.M.; Schreibelt, G.; Gorris, M.A.J.; Mehra, N.; Simons, M.; de Goede, A.L.; van Rossum, M.M.; Croockewit, A.J.; et al. Blood-derived dendritic cell vaccinations induce immune responses that correlate with clinical outcome in patients with chemo-naive castration-resistant prostate cancer. J. Immunother. Cancer 2019, 7, 302. [Google Scholar] [CrossRef]
  240. Liao, F.; Zhang, J.; Hu, Y.; Najafabadi, A.H.; Moon, J.J.; Wicha, M.S.; Kaspo, B.; Whitfield, J.; Chang, A.E.; Li, Q. Efficacy of an ALDH peptide-based dendritic cell vaccine targeting cancer stem cells. Cancer Immunol. Immunother. 2022, 71, 1959–1973. [Google Scholar] [CrossRef]
  241. Visus, C.; Wang, Y.; Lozano-Leon, A.; Ferris, R.L.; Silver, S.; Szczepanski, M.J.; Brand, R.E.; Ferrone, C.R.; Whiteside, T.L.; Ferrone, S.; et al. Targeting ALDH (bright) human carcinoma-initiating cells with ALDH1A1-specific CD8(+) T cells. Clin Cancer Res. 2011, 17, 6174–6184. [Google Scholar] [CrossRef] [Green Version]
  242. Püschel, J.; Dubrovska, A.; Gorodetska, I. The Multifaceted Role of Aldehyde Dehydrogenases in Prostate Cancer Stem Cells. Cancers 2021, 13, 4703. [Google Scholar] [CrossRef] [PubMed]
  243. Cojoc, M.; Peitzsch, C.; Kurth, I.; Trautmann, F.; Kunz-Schughart, L.A.; Telegeev, G.D.; Stakhovsky, E.A.; Walker, J.R.; Simin, K.; Lyle, S.; et al. Aldehyde Dehydrogenase Is Regulated by beta-Catenin/TCF and Promotes Radioresistance in Prostate Cancer Progenitor Cells. Cancer Res. 2015, 75, 1482–1494. [Google Scholar] [CrossRef] [Green Version]
  244. Mukha, A.; Kahya, U.; Linge, A.; Chen, O.; Lock, S.; Lukiyanchuk, V.; Richter, S.; Alves, T.C.; Peitzsch, M.; Telychko, V.; et al. GLS-driven glutamine catabolism contributes to prostate cancer radiosensitivity by reg-ulating the redox state, stemness and ATG5-mediated autophagy. Theranostics 2021, 11, 7844–7868. [Google Scholar] [CrossRef]
  245. Ning, N.; Pan, Q.; Zheng, F.; Teitz-Tennenbaum, S.; Egenti, M.; Yet, J.; Li, M.; Ginestier, C.; Wicha, M.S.; Moyer, J.S.; et al. Cancer Stem Cell Vaccination Confers Significant Antitumor Immunity. Cancer Res 2012, 72, 1853–1864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Köseer, A.S.; Loureiro, L.R.; Jureczek, J.; Mitwasi, N.; Soto, K.E.G.; Aepler, J.; Bartsch, T.; Feldmann, A.; Kunz-Schughart, L.A.; Linge, A.; et al. Validation of CD98hc as a Therapeutic Target for a Combination of Radiation and Immunotherapies in Head and Neck Squamous Cell Carcinoma. Cancers 2022, 14, 1677. [Google Scholar] [CrossRef] [PubMed]
  247. Hassani Najafabadi, A.; Zhang, J.; Aikins, M.E.; Najaf Abadi, Z.I.; Liao, F.; Qin, Y.; Okeke, E.B.; Scheetz, L.M.; Nam, J.; Xu, Y.; et al. Cancer Immunotherapy via Targeting Cancer Stem Cells Using Vaccine Nanodiscs. Nano Lett. 2020, 20, 7783–7792. [Google Scholar] [CrossRef]
  248. Zheng, F.; Dang, J.; Zhang, H.; Xu, F.; Ba, D.; Zhang, B.; Cheng, F.; Chang, A.E.; Wicha, M.S.; Li, Q. Cancer Stem Cell Vaccination with PD-L1 and CTLA-4 Blockades Enhances the Eradication of Melanoma Stem Cells in a Mouse Tumor Model. J. Immunother. 2018, 41, 361–368. [Google Scholar] [CrossRef]
  249. Hu, Y.; Lu, L.; Xia, Y.; Chen, X.; Chang, A.E.; Hollingsworth, R.E.; Hurt, E.; Owen, J.; Moyer, J.S.; Prince, M.E.; et al. Therapeutic Efficacy of Cancer Stem Cell Vaccines in the Adjuvant Setting. Cancer Res. 2016, 76, 4661–4672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  250. El-Ashmawy, N.E.; Salem, M.L.; Khedr, E.G.; El-Zamarany, E.A.; Ibrahim, A.O. Dual-targeted therapeutic strategy combining CSC–DC-based vaccine and cisplatin overcomes chemo-resistance in experimental mice model. Clin. Transl. Oncol. 2019, 22, 1155–1165. [Google Scholar] [CrossRef]
  251. Feng, D.; Gip, P.; McKenna, B.K.M.; Zhao, F.; Mata, O.; Choi, B.T.S.; Duan, M.J.; Sompalli, M.K.; Majeti, R.; Weissman, I.L.; et al. Combination Treatment with 5F9 and Azacitidine Enhances Phagocytic Elimination of Acute Myeloid Leukemia. Blood 2018, 132, 2729. [Google Scholar] [CrossRef]
  252. Sallman, D.A.; Asch, A.S.; Al Malki, M.M.; Lee, D.J.; Donnellan, W.B.; Marcucci, G.; Kambhampati, S.; Daver, N.G.; Garcia-Manero, G.; Komrokji, R.S.; et al. The First-in-Class Anti-CD47 Antibody Magrolimab (5F9) in Combination with Azacitidine Is Effective in MDS and AML Patients: Ongoing Phase 1b Results. Blood 2019, 134, 569. [Google Scholar] [CrossRef]
  253. Yu, J.; Chen, L.; Cui, B.; Wu, C.; Choi, M.Y.; Chen, Y.; Zhang, L.; Rassenti, L.Z.; Ii, G.F.W.; Kipps, T.J. Cirmtuzumab inhibits Wnt5a-induced Rac1 activation in chronic lymphocytic leukemia treated with ibrutinib. Leukemia 2016, 31, 1333–1339. [Google Scholar] [CrossRef] [PubMed]
  254. Lee, H.J.; Choi, M.Y.; Siddiqi, T.; Barrientos, J.C.; Wierda, W.G.; Isufi, I.; Tuscano, J.M.; Lamanna, N.; Subbiah, S.; Koff, J.L.; et al. Phase 1/2 study of cirmtuzumab and ibrutinib in mantle cell lymphoma (MCL) or chronic lymphocytic leukemia (CLL). J. Clin. Oncol. 2021, 39 (Suppl. S15), 7556. [Google Scholar] [CrossRef]
  255. Su, R.; Dong, L.; Li, Y.; Gao, M.; Han, L.; Wunderlich, M.; Deng, X.; Li, H.; Huang, Y.; Gao, L.; et al. Targeting FTO Suppresses Cancer Stem Cell Maintenance and Immune Evasion. Cancer Cell 2020, 38, 79–96.e11. [Google Scholar] [CrossRef]
  256. Wu, X.; Huang, S. HER2-specific chimeric antigen receptor-engineered natural killer cells combined with apatinib for the treatment of gastric cancer. Bull. Du Cancer 2019, 106, 946–958. [Google Scholar] [CrossRef] [PubMed]
  257. Wang, D.; Xu, J.; Liu, B.; He, X.; Zhou, L.; Hu, X.; Qiao, F.; Zhang, A.; Xu, X.; Zhang, H.; et al. IL6 blockade potentiates the anti-tumor effects of γ-secretase inhibitors in Notch3-expressing breast cancer. Cell Death Differ. 2017, 25, 330–339. [Google Scholar] [CrossRef]
  258. Zhang, Q.; Zhang, H.; Ding, J.; Liu, H.; Li, H.; Li, H.; Lu, M.; Miao, Y.; Li, L.; Zheng, J. Combination Therapy with EpCAM-CAR-NK-92 Cells and Regorafenib against Human Colorectal Cancer Models. J. Immunol. Res. 2018, 2018, 4263520. [Google Scholar] [CrossRef] [Green Version]
  259. Han, Y.; Sun, B.; Cai, H.; Xuan, Y. Simultaneously target of normal and stem cells-like gastric cancer cells via cisplatin and an-ti-CD133 CAR-T combination therapy. Cancer Immunol. Immunother. 2021, 70, 2795–2803. [Google Scholar] [CrossRef]
  260. Klapdor, R.; Wang, S.; Hacker, U.; Büning, H.; Morgan, M.; Dörk, T.; Hillemanns, P.; Schambach, A. Improved Killing of Ovarian Cancer Stem Cells by Combining a Novel Chimeric Antigen Receptor–Based Immunotherapy and Chemotherapy. Hum. Gene Ther. 2017, 28, 886–896. [Google Scholar] [CrossRef]
  261. Fleurence, J.; Bahri, M.; Fougeray, S.; Faraj, S.; Vermeulen, S.; Pinault, E.; Geraldo, F.; Oliver, L.; Véziers, J.; Marquet, P.; et al. Impairing temozolomide resistance driven by glioma stem-like cells with adjuvant immunotherapy targeting O-acetyl GD2 ganglioside. Int. J. Cancer 2019, 146, 424–438. [Google Scholar] [CrossRef]
  262. Madhav, A.; Andres, A.; Duong, F.; Mishra, R.; Haldar, S.; Liu, Z.; Angara, B.; Gottlieb, R.; Zumsteg, Z.S.; Bhowmick, N.A. Antagonizing CD105 enhances radiation sensitivity in prostate cancer. Oncogene 2018, 37, 4385–4397. [Google Scholar] [CrossRef]
  263. Nishiga, Y.; Drainas, A.P.; Baron, M.; Bhattacharya, D.; Barkal, A.A.; Ahrari, Y.; Mancusi, R.; Ross, J.B.; Takahashi, N.; Thomas, A.; et al. Radiotherapy in combination with CD47 blockade elicits a macrophage-mediated abscopal effect. Nat. Cancer 2022, 3, 1351–1366. [Google Scholar] [CrossRef]
  264. Darragh, L.B.; Gadwa, J.; Pham, T.T.; Van Court, B.; Neupert, B.; Olimpo, N.A.; Nguyen, K.; Nguyen, D.; Knitz, M.W.; Hoen, M.; et al. Elective nodal irradiation mitigates local and systemic immunity generated by com-bination radiation and immunotherapy in head and neck tumors. Nat. Commun. 2022, 13, 7015. [Google Scholar] [CrossRef] [PubMed]
  265. Smith, D.C.; Eisenberg, P.D.; Manikhas, G.; Chugh, R.; Gubens, M.A.; Stagg, R.J.; Kapoun, A.M.; Xu, L.; Dupont, J.; Sikic, B. A Phase I Dose Escalation and Expansion Study of the Anticancer Stem Cell Agent Demcizumab (Anti-DLL4) in Patients with Previously Treated Solid Tumors. Clin. Cancer Res. 2014, 20, 6295–6303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Zitvogel, L.; Apetoh, L.; Ghiringhelli, F.; Andre, F.; Tesniere, A.; Kroemer, G. The anticancer immune response: Indispensable for therapeutic success? J. Clin. Investig. 2008, 118, 1991–2001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  267. Ménard, C.; Martin, F.; Apetoh, L.; Bouyer, F.; Ghiringhelli, F. Cancer chemotherapy: Not only a direct cytotoxic effect, but also an adjuvant for antitumor immunity. Cancer Immunol. Immunother 2008, 57, 1579–1587. [Google Scholar] [CrossRef]
  268. Formenti, S.C.; Demaria, S. Systemic effects of local radiotherapy. Lancet Oncol. 2009, 10, 718–726. [Google Scholar] [CrossRef] [Green Version]
  269. Opzoomer, J.W.; Sosnowska, D.; Anstee, J.E.; Spicer, J.F.; Arnold, J.N. Cytotoxic Chemotherapy as an Immune Stimulus: A Molecular Perspective on Turning Up the Immunological Heat on Cancer. Front. Immunol. 2019, 10, 1654. [Google Scholar] [CrossRef] [Green Version]
  270. Zhang, Z.; Liu, X.; Chen, D.; Yu, J. Radiotherapy combined with immunotherapy: The dawn of cancer treatment. Signal Transduct. Target. Ther. 2022, 7, 1–34. [Google Scholar] [CrossRef] [PubMed]
  271. Allen, T.; Brehm, M.; Bridges, S.; Ferguson, S.; Kumar, P.; Mirochnitchenko, O.; Palucka, K.; Pelanda, R.; Sanders-Beer, B.; Shultz, L.D.; et al. Humanized immune system mouse models: Progress, challenges and opportunities. Nat. Immunol. 2019, 20, 770–774. [Google Scholar] [CrossRef] [PubMed]
  272. Graham, A.L. Naturalizing mouse models for immunology. Nat. Immunol. 2021, 22, 111–117. [Google Scholar] [CrossRef] [PubMed]
  273. Taylor, M.A.; Hughes, A.M.; Walton, J.; Coenen-Stass, A.M.L.; Magiera, L.; Mooney, L.; Bell, S.; Staniszewska, A.; Sandin, L.C.; Barry, S.T.; et al. Longitudinal immune characterization of syngeneic tumor models to enable model selection for immune oncology drug discovery. J. Immunother. Cancer 2019, 7, 1–16. [Google Scholar] [CrossRef] [PubMed]
  274. DuPage, M.; Jacks, T. Genetically engineered mouse models of cancer reveal new insights about the antitumor immune response. Curr. Opin. Immunol. 2013, 25, 192–199. [Google Scholar] [CrossRef] [Green Version]
  275. Chawda, C.; McMorrow, R.; Gaspar, N.; Zambito, G.; Mezzanotte, L. Monitoring Immune Cell Function Through Optical Imaging: A Review Highlighting Transgenic Mouse Models. Mol. Imaging Biol. 2021, 24, 250–263. [Google Scholar] [CrossRef]
  276. Hammel, J.H.; Cook, S.R.; Belanger, M.C.; Munson, J.M.; Pompano, R.R. Modeling Immunity In Vitro: Slices, Chips, and Engineered Tissues. Annu. Rev. Biomed. Eng. 2021, 23, 461–491. [Google Scholar] [CrossRef] [PubMed]
Figure 1. CSC-directed immunotherapeutic approaches. (A) Therapeutic antibody induces immune-mediated tumor cell killing by the induction of antibody-dependent cellular cytotoxicity (ADCC) (1). Antibody-mediated antigen delivery to the antigen-presenting cells (APC), such as dendritic cells (DCs), leads to effective T cell activation (2). T cells are also activated by inhibition of the immune checkpoint proteins: cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), and its ligand, PD-L1. Antibody-drug conjugates deliver potent chemotherapeutic agents to the target cells positive for a tumor-associated antigen (TAA) (3). Bispecific antibodies (bsAB) retarget immune cells to the TAA-expressing tumor cells (4). (B) CAR-T cells/CAR-NK cells are T or natural killer (NK) cells that are genetically modified with artificial chimeric antigen receptor (CAR) recruiting them to tumor cells. (C) Universal CAR (UniCAR) T cells recognize tumor cells via a short-lived target module (TM) which could be designed against more than one TAA. CAR—chimeric antigen receptor; CSC—cancer stem cells; MHC—major histocompatibility complex; TCR—T cell receptor. Created with BioRender.
Figure 1. CSC-directed immunotherapeutic approaches. (A) Therapeutic antibody induces immune-mediated tumor cell killing by the induction of antibody-dependent cellular cytotoxicity (ADCC) (1). Antibody-mediated antigen delivery to the antigen-presenting cells (APC), such as dendritic cells (DCs), leads to effective T cell activation (2). T cells are also activated by inhibition of the immune checkpoint proteins: cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), and its ligand, PD-L1. Antibody-drug conjugates deliver potent chemotherapeutic agents to the target cells positive for a tumor-associated antigen (TAA) (3). Bispecific antibodies (bsAB) retarget immune cells to the TAA-expressing tumor cells (4). (B) CAR-T cells/CAR-NK cells are T or natural killer (NK) cells that are genetically modified with artificial chimeric antigen receptor (CAR) recruiting them to tumor cells. (C) Universal CAR (UniCAR) T cells recognize tumor cells via a short-lived target module (TM) which could be designed against more than one TAA. CAR—chimeric antigen receptor; CSC—cancer stem cells; MHC—major histocompatibility complex; TCR—T cell receptor. Created with BioRender.
Cancers 15 01608 g001
Figure 2. Adapter CAR approaches. (A) Adapter CAR T cells do not recognize a surface antigen. Thus, they are inactive (“OFF”). CAR T cells are activated for tumor cell killing (“ON”) only in the presence of a crosslinking adapter molecule. (B) Due to the modular design, adapter CAR T cell activity can be controlled via the adapter molecule (dosing, timing). Simultaneous or sequential application of different adapter molecules enables multiple and logic-gated tumor targeting. In the adapter CAR system, additional payloads can be co-delivered via adapter molecules to enhance anti-tumor responses. (C) In the UniCAR system, as an example of peptide-binding adapter CARs, CAR T cells are redirected for tumor cell killing via a so-called target module (TM) that is composed of a tumor-specific binding arm and a short La-peptide epitope (E5B9 or E7B6) recognized by the UniCAR. (D) In the RevCAR system, bispecific antibodies (bsAB), termed reversed target modules (RevTMs), mediate RevCAR T cell/tumor cell interactions and, thus, tumor cell killing. RevTMs simultaneously recognize a tumor antigen and the La-peptide epitope (E5B9 or E7B6) used to construct the RevCAR.
Figure 2. Adapter CAR approaches. (A) Adapter CAR T cells do not recognize a surface antigen. Thus, they are inactive (“OFF”). CAR T cells are activated for tumor cell killing (“ON”) only in the presence of a crosslinking adapter molecule. (B) Due to the modular design, adapter CAR T cell activity can be controlled via the adapter molecule (dosing, timing). Simultaneous or sequential application of different adapter molecules enables multiple and logic-gated tumor targeting. In the adapter CAR system, additional payloads can be co-delivered via adapter molecules to enhance anti-tumor responses. (C) In the UniCAR system, as an example of peptide-binding adapter CARs, CAR T cells are redirected for tumor cell killing via a so-called target module (TM) that is composed of a tumor-specific binding arm and a short La-peptide epitope (E5B9 or E7B6) recognized by the UniCAR. (D) In the RevCAR system, bispecific antibodies (bsAB), termed reversed target modules (RevTMs), mediate RevCAR T cell/tumor cell interactions and, thus, tumor cell killing. RevTMs simultaneously recognize a tumor antigen and the La-peptide epitope (E5B9 or E7B6) used to construct the RevCAR.
Cancers 15 01608 g002
Figure 3. Development of the DC autologous vaccines. The DC vaccines are most commonly prepared by ex vivo differentiation of the autologous monocytes into immature DCs. After maturation, DCs are pulsed with cancer antigens and administered back to the patient, where they activate antigen-specific T cells. CSC—cancer stem cells; MHC I and MHC II—major histocompatibility complex class I and class II; PBMC—peripheral blood mononuclear cells; TAA—tumor-associated antigen. Created with BioRender.
Figure 3. Development of the DC autologous vaccines. The DC vaccines are most commonly prepared by ex vivo differentiation of the autologous monocytes into immature DCs. After maturation, DCs are pulsed with cancer antigens and administered back to the patient, where they activate antigen-specific T cells. CSC—cancer stem cells; MHC I and MHC II—major histocompatibility complex class I and class II; PBMC—peripheral blood mononuclear cells; TAA—tumor-associated antigen. Created with BioRender.
Cancers 15 01608 g003
Table 3. Preclinical/clinical trials of combination therapies targeting CSC markers.
Table 3. Preclinical/clinical trials of combination therapies targeting CSC markers.
Preclinical Studies
Experimental ModelTargetImmunotherapyCombined TherapyCancer TypeRefs
In vitro (cell line)CD475F9 mAbAZA (cytotoxic analogue of the nucleoside cytidine, inhibitor of DNA methyltransferase)AML[251]
In vivo (cell-line derived xenograft)
In vivo (patient-derived xenograft)ROR1CirmtuzumabIbrutinib
(Bruton’s tyrosine kinase (BTK) inhibitor)
CLL[253]
In vitro (cell line)EGFRCAR NK-92Cabozantinib
(VEGFR-2 inhibitor)
Renal cell carcinoma[114]
In vivo (cell-line derived xenograft)
In vitro (cell line)Carbonic Anhydrase IX (CAIX)CAR TSunitinib
(multi-targeted receptor kinase inhibitor)
[131]
In vivo (cell-line derived xenograft)
In vitro (cell line)HER2HER2 CAR NK-92Apatinib
(VEGFR-2 inhibitor)
Gastric cancer[256]
In vivo (cell-line derived xenograft)
In vitro (cell line)IL-6TocilizumabMK-0752
(γ-secretase inhibitor)
Breast cancer[257]
In vivo (cell-line derived xenograft)
In vivo (patient-derived xenograft)
In vivo (cell-line derived xenograft)EpCAMCAR-NK-92Regorafenib
(multitargeted kinase inhibitor)
Colorectal cancer[258]
In vivo (syngeneic models)ALDHHigh CSCsALDHHigh-DC vaccineAnti PD-L1 antibodyD5 murine melanoma and 4T1 murine breast cancer[247]
In vivo (syngeneic models)
ALDHHigh CSCsALDHHigh-DC vaccineAnti PD-L1 antibody;
Anti CTLA-4 antibody
B16-F10 murine melanoma tumors[248]
In vivo (cell-line derived xenograft)CD133CAR-TCisplatin
(DNA-binding cytotoxic drug)
Gastric cancer[259]
In vitro (Cell line)CAR NK-92Ovarian cancer[260]
In vivo (cell-line derived xenograft)CD44+/CD24 CSCsCD44+/CD24− CSC-pulsed DC vaccineEhrlich carcinoma[250]
In vitro (patient-derived cell line)OAcGD28B6 mAbTemozolomide
(alkylating agent)
GBM[261]
In vivo (Patient-derived xenograft)
In vivo (cell-line derived xenograft)CD105TRC105Conventional fractionated RT (5 × 2 Gy)Prostate cancer[262]
In vivo (Cell-line derived xenograft)CD47Anti-CD47 mAbConventional fractionated RT
(1 × 5 Gy; 4 × 5 Gy; 1 × 10 Gy)
Small cell lung cancer, colon cancer[263]
In vitro (3D model)CD98UniCAR T + CD98 TMConventional fractionated RT (2 × 2 Gy)HNSCC[246]
In vivo (Cell-line derived xenograft)CD25Anti-CD25 mAbConventional fractionated RT[264]
Clinical trials
NCT numberTargetImmunotherapyCombined therapyCancer typeRefs
NCT03248479CD47Magrolimab (Hu5F9-G4)AZAAML/MDS[252]
NCT03088878ROR1CirmtuzumabIbrutinibCLL[254]
NCT02259582DLL4DemcizumabCarboplatin + pemetrexedNon-squamous NSCLC (DENALI)[265]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Köseer, A.S.; Di Gaetano, S.; Arndt, C.; Bachmann, M.; Dubrovska, A. Immunotargeting of Cancer Stem Cells. Cancers 2023, 15, 1608. https://doi.org/10.3390/cancers15051608

AMA Style

Köseer AS, Di Gaetano S, Arndt C, Bachmann M, Dubrovska A. Immunotargeting of Cancer Stem Cells. Cancers. 2023; 15(5):1608. https://doi.org/10.3390/cancers15051608

Chicago/Turabian Style

Köseer, Ayse Sedef, Simona Di Gaetano, Claudia Arndt, Michael Bachmann, and Anna Dubrovska. 2023. "Immunotargeting of Cancer Stem Cells" Cancers 15, no. 5: 1608. https://doi.org/10.3390/cancers15051608

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

Article Metrics

Back to TopTop