Next Article in Journal
Repurposing of Miltefosine as an Adjuvant for Influenza Vaccine
Next Article in Special Issue
The Potential of Soluble Human Leukocyte Antigen Molecules for Early Cancer Detection and Therapeutic Vaccine Design
Previous Article in Journal
Post-Vaccination Yellow Fever Antiserum Reduces Zika Virus in Embryoid Bodies When Placental Cells are Present
Previous Article in Special Issue
Transcriptional Regulation of Cancer Immune Checkpoints: Emerging Strategies for Immunotherapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Strategies to Improve Chimeric Antigen Receptor Therapies for Neuroblastoma

by
Piamsiri Sawaisorn
1,†,
Korakot Atjanasuppat
1,†,
Usanarat Anurathapan
1,
Somchai Chutipongtanate
2,3,4,* and
Suradej Hongeng
1,*
1
Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
2
Pediatric Translational Research Unit, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
3
Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
4
Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
*
Authors to whom correspondence should be addressed.
Shared equal contribution.
Vaccines 2020, 8(4), 753; https://doi.org/10.3390/vaccines8040753
Submission received: 29 October 2020 / Revised: 4 December 2020 / Accepted: 10 December 2020 / Published: 11 December 2020
(This article belongs to the Special Issue Cancer Immunotherapy: Advances and Future Prospects)

Abstract

:
Chimeric antigen receptors (CARs) are among the curative immunotherapeutic approaches that exploit the antigen specificity and cytotoxicity function of potent immune cells against cancers. Neuroblastomas, the most common extracranial pediatric solid tumors with diverse characteristics, could be a promising candidate for using CAR therapies. Several methods harness CAR-modified cells in neuroblastoma to increase therapeutic efficiency, although the assessment has been less successful. Regarding the improvement of CARs, various trials have been launched to overcome insufficient capacity. However, the reasons behind the inadequate response against neuroblastoma of CAR-modified cells are still not well understood. It is essential to update the present state of comprehension of CARs to improve the efficiency of CAR therapies. This review summarizes the crucial features of CARs and their design for neuroblastoma, discusses challenges that impact the outcomes of the immunotherapeutic competence, and focuses on devising strategies currently being investigated to improve the efficacy of CARs for neuroblastoma immunotherapy.

1. Introduction

Neuroblastoma, an extracranial solid tumor that initiates from the sympathetic nervous system’s neuroendocrine tissue, is one of the most common causes of death in pediatric cancers [1,2]. It is often diagnosed during the perinatal period, which accounts for 8% in patients under 15 years. This childhood neoplasm appears each year in more than 600 cases in the United States and 200 in Japan [3,4,5]. According to clinical presentation, neuroblastoma is an extremely variant characteristic tumor. It ranges from an adrenal mass tumor that regresses without treatment to a metastatic tumor that causes critical illness [6]. At present, while intensive therapies can be beneficial for patients with localized disease, these therapies have frequently not been useful against patients with high-risk disease (approximately 40% of cases associated with the extent of metastases and genetic factors) nor patients with relapse [7,8]. Hence, novel therapies based on immunotherapy were subsequently developed to improve survival for high-risk patients.
One such approach is treatment with anti-GD2 monoclonal antibody, which has already been assessed in a Phase III clinical trial. The use of this antibody-based therapy was compiled into a therapeutic protocol for high-risk neuroblastoma patients and revealed promising results [9,10]. This effectiveness has led to other immunotherapeutic approaches, even though their integration into conventional multimodality therapies requires further investigation.
After discovering that GD2, a disialoganglioside highly expressed in most neuroblastomas, is also targeted by T cells, cellular immunotherapies including genetic engineering of T lymphocytes to express anti-GD2 chimeric antigen receptors (CARs) have emerged and are now being studied. With the combination of antigen specificity and cytolytic capacity, anti-GD2 CAR T cells have demonstrated safety and antitumor efficacy in relapsed neuroblastoma patients [11,12]. Various preclinical studies have improved the antitumor effects, proliferation, and cytokine release of CAR T cells, and some approaches have reached clinical trials (Figure 1). Currently, the anti-GD2 CAR T cell approach might represent a potential therapeutic for pediatric neuroblastoma.
However, unlike the success of using CAR T cells in hematological malignancies [13,14,15], the efficacy of CAR T cells in neuroblastomas has been limited by several factors, including the tumor microenvironment, T cell exhaustion, and T cells’ persistence and potency, which may lead to therapeutic resistance [16,17,18]. Therefore, CAR T cell improvement as a feasible alternative to conventional therapies for patients with neuroblastoma is still a significant challenge to achieving immunotherapy.
In this review, we provide an updated summary of preclinical and clinical experience of CAR-based neuroblastoma therapies and discuss the improvements of CAR in different ways that could overcome clinical problems of applying this approach for treating neuroblastoma. Defining these strategies would suggest an attractive route of improving the potency of CAR immunotherapy.

2. CARs in Neuroblastoma

The knowledge of tumor immunology has been used in translating this comprehension into productive cancer therapies. A number of immunotherapy approaches represent a new borderline in treating cancers. One such strategy to overcome tolerance in cancer is to genetically engineer immune cells to express CAR. This concept of artificial antigen-specific receptors first originated in 1989–1993 [19,20]. By fusing an antibody-derived binding domain to T cell signaling domains, the CAR construct gains the tumor antigen specificity and the capacity to induce multiple signals in the response of immune cells. Over three decades, tremendous progress has been made and CARs were refined into the first, second, third, and currently fourth generations of their structure. Based on the potential of CAR T cells directed against the CD19 protein for treatment of hematologic malignancies shown in clinical trials, cancer immunotherapy was named the “Breakthrough of the Year” in 2013 by Science [21]. In addition, the use of anti-CD19 CAR T cells for relapsed/refractory acute lymphoblastic leukemia and in children and young adults was approved by the US Food and Drug Administration (FDA) in 2017 and two licensed products of CAR T cells including tisagenlecleucel and axicabtagene ciloleucel have been launched [22,23]. This success, therefore, has brought new insights for clinical translation in treating solid cancers.
CARs have been developed to fulfill the applicability of adoptive cellular immunotherapy for neuroblastoma in a major histocompatibility complex (MHC)-unrestricted manner in effector T cells. Effector immune cells, commonly T lymphocytes, have been genetically engineered to express an extracellular antigen-binding domain that is mostly a single-chain variable fragment (scFv) joined with a transmembrane domain and an intracellular signaling domain. The first-generation CARs were designed to have a single CD3-ζ intracellular signaling domain. The second- and third-generation CAR products were improved by adding one or two costimulatory endodomains to the CD3-ζ motif to achieve the optimal activation and survival of CAR cells. Current intracellular endodomains based on the costimulatory receptors include CD27, CD28, 41BB, ICOS, and OX40 [17,24]. Each of the CAR design components reflects the variations of therapeutics achievement, and novel CAR engineering has been developed for decades to broaden CAR therapeutics in solid tumors like neuroblastoma [25].

2.1. Summary of CAR Experience

Several CAR approaches in neuroblastoma have been developed according to discovered putative cancer antigens. There are some novel target antigens for CAR T cell therapy in neuroblastoma that have been investigated in the preclinical phase (Figure 2).
Anaplastic lymphoma kinase (ALK), an oncogene expressed in neuroblastoma cells, is associated with familial neuroblastoma cases [26,27]. Anti-ALK CAR has demonstrated its effectiveness against this neuroblastoma subtype in vitro and in vivo [28,29]. This line of research also suggested that antigen density must be considered to achieve CAR T cell potential. Another tyrosine kinase receptor that may be rendered an ideal target for CAR therapies is glypican 2 (GPC2). The high expression of GPC2 on the neuroblastoma cell surface brought promising clearance of disseminated neuroblastoma in the mouse model by anti-GPC2 CAR T cells [30]. B7H3 (CD276), a checkpoint molecule expressed in neuroblastomas, is another candidate for CAR therapies of neuroblastoma [31,32]. This attractive target brought useful immunotherapeutic strategies, including monoclonal antibodies and CARs targeting B7H3. Recently, the efficacy of anti-B7H3 CAR has been demonstrated in vivo [33,34]. Many target antigens that are specific to neuroblastoma cells have also been more characterized. Such antigens, including neural cell adhesion molecule (NCAM or CD56), New York esophageal squamous cell carcinoma 1 (NY-ESO1), and preferentially expressed antigen in melanoma (PRAME), were investigated both in vitro and in vivo for safety and efficacy, which gained attention for further development as CAR features [35,36,37,38].
To date, only CAR T cells targeting L1-CAM (CD171) and GD2 have reached the early phase of clinical trials (Table 1). L1-CAM, an adhesion molecule in the immunoglobulin superfamily, is another suitable target in neuroblastoma [39]. Because of the specificity of CE7, the monoclonal antibody that can bind to the L1-CAM epitope, the anti-L1-CAM CAR with the scFv from CE7 was generated. The first-generation anti-L1-CAM CARs’ efficacy and safety were investigated in patients with relapsed/refractory neuroblastoma in a Phase 1 clinical trial [12]. To augment the persistence of anti-L1-CAM CAR, second-generation CAR was generated using a 41BB costimulation domain, followed by third-generation CAR, including CD28 costimulation addition, which is currently being investigated in phase 1 clinical trials [40,41]. Until now, the most critical target antigen in neuroblastoma has been GD2, a disialoganglioside highly expressed on neuroblastoma tissue [42]. Owing to the presence of this antigen during chemotherapy and the success of anti-GD2 monoclonal antibody therapy, this antigen has been the most studied targeted for CAR T cell therapy in neuroblastoma [43]. Many approaches of first-generation anti-GD2 CAR have been reported, including anti-GD2 CAR containing a single-chain variable fragment (scFv) derived from 14g2a monoclonal antibody or Epstein–Barr virus-specific cytotoxic T cell transduced CARs (so-called GD2 CAR-CTL), with the knowledge that the prolonged persistence in vivo was associated with the costimulation domain of CAR [44,45,46,47]. Anti-GD2 CAR constructs are now considered on costimulatory endodomains. The second and third generations of CAR were then generated for in vitro and in vivo assessments of CAR T cell survival [48,49]. The third-generation anti-GD2 CAR, containing an inducible caspase 9 (iC9) safety switch, has been tested in clinical trials for its safety (clinicaltrials.gov identifier NCT01953900 and NCT01822652). Various clinical trials based on CAR therapy are underway to augment the reliable therapeutic outcomes. However, improving the efficacy and persistence of CAR is still a significant issue.

2.2. Obstacles to Using CARs in Neuroblastoma

2.2.1. CAR T Cell Persistence and Exhaustion

Restrictive CAR T cell persistence has occurred as a major problem in neuroblastoma. Evidence from the first generation of CAR studies in vivo and clinical trials suggested that the limited persistence of CAR T cells from low activation and proliferation of cells also affected the antitumor efficacy [18,48,50,56]. One clinical study demonstrated that the infused, first-generation, anti-L1-CAM CAR cells were detectable in the peripheral blood up to 1–7 days after adoptive transfer in most patients with bulk disease but significantly longer (42 days) in a patient with limited disease burden [12]. T cell exhaustion might be a significant cause of shortening persistence. This is confirmed by discovering the exhausted CAR T cell phenotype in GD2 CAR T cells with low-level tonic signaling [57]. The persistence of infused CAR T cells might be prolonged if the exhaustion was reduced. Thus, several methods have been proposed to increase the persistence of CAR T cells. One such way is the utilization of second- and third-generation CARs, which improve costimulation after antigen binding (e.g., 4-1BB costimulatory domain) to protect shortened persistence. This development is under investigation for feasibility [13,15,57].

2.2.2. Target Selection and On-Target, Off-Tumor Effect

Ideally, the target antigen for CAR T cells should have a high expression in cancer cells, a low expression in normal cells, and not be associated with oncogenesis [58]. It is known that there are challenges in the path of choosing an optimal CAR T cell target antigen in neuroblastoma, since many target antigens are related to normal peripheral nerves or neural tissue expression. Toxicities caused by particular interactions between the CAR and its target antigen expressed by normal cells termed on-target, off-tumor effects have been reported in previous CAR studies in solid tumors [59,60,61,62]. One clinical trial in metastatic colon cancer reported pulmonary infiltration by CAR T cells that caused a systemic cytokine storm in patients who received HER2-targeted CAR T cell therapy, demonstrating strong evidence of on-target, off-tumor toxicities [61]. On the other hand, there was no such effect in a pediatric sarcoma study using anti-HER2 CAR T cells and anti-GD2 CAR T cells in neuroblastoma studies [47,63,64]. This evidence suggested that the variation of antigen density on the different types of cancer is an additional factor to consider during target selection to avoid on-target, off-tumor effects.

2.2.3. Tumor Microenvironment

Unlike the remarkable success of CAR T cells in the treatment of hematological malignancies, the efficacy of CAR T cells in neuroblastoma can be obstructed by the immunosuppressive tumor microenvironment (TME), which is a manifest barrier to achieve full effective CAR T cell therapy for solid tumors [65]. Significant factors derived from TME in neuroblastomas include immunosuppressive cells like tumor-associated macrophages (TAMs), Type 2 regulatory T cells (Tregs), and myeloid-derived suppressor cells (MDSCs), which contributed to poor results of CAR therapy [16]. Another factor is the inhibitory ligands present in the TME, such as PD-L1, the ligand for an inhibitory receptor expressed on activated T cells, named PD-1 [66,67]. Remarkably, this habitual expression of the inhibitory ligand in neuroblastoma can cause the loss of CAR T cells [68]. In addition to inhibitory signals, the availability of soluble factors in the TME, including galactin 1 and 3, TGF-β, and IL-10, can trigger T cell inhibitory pathways or inhibit T cell function [67,69,70,71,72], while secretory HMGB1 may be responsible for Treg differentiation in the neuroblastoma TME [73]. Furthermore, there are physical barriers that prevent the tumor access of T cells, such as protease fibroblast activation protein (FAP) expressed by tumor-associated stromal fibroblasts, the extracellular matrix (ECM), and immunosuppressive tumor vasculature-like vascular endothelial growth factor (VEGF) [74,75].

2.2.4. CAR Trafficking

Trafficking of CAR cells into solid tumor sites to exert antitumor activity needs to be improved, especially in neuroblastoma. Several chemokines that can mediate immune cell trafficking are generally excreted by tumor or stromal cells like CC-chemokine ligand 17 (CCL17), CCL22, and CCL2 to enhance the localization of immune cells [76]. Moreover, suitable trafficking of immune cells, like T cells, can occur when there is an upregulation of a chemokine receptor that is matched to chemokine-related trafficking on T cells. However, in a previous study using CAR T cells derived from neuroblastoma patients, low expression of CCR2, a chemokine receptor, was detected [77,78,79]. Thus, various approaches to generate CAR T cells with an ability to traffic to neuroblastoma sites are underway.

3. Strategies to Improve CARs in Neuroblastoma

Remarkably the engineering of T lymphocytes to target tumor-associated antigens by forced expression of CARs has been successful against CD19+ leukemia [80,81,82]. Similar results have not yet been achieved against neuroblastoma [50,83,84]. The key factors that challenge the success of CAR immunotherapies are the immune effector cells, the design of the CAR construct, and the intrinsic tumor factors. The proposed strategies for improving CARs’ efficacy in neuroblastoma therapy are illustrated in Figure 3.

3.1. Improving Effector Immune Cells

Solving CAR T cell obstruction or the use of other effector cells might increase CARs’ tumor-killing activity in neuroblastoma. Regarding the killing effect against tumor cells, natural killer T (NKT) [85], gamma-delta T (γδT) [86], and natural killer (NK) cells [87] are valuable candidates for generating CAR-derived cells. These immune cells have different pros and cons. Thus, choosing the proper effector cell for each tumor target is extremely important. This part will assemble information about these CAR-derived effector cells, some of which are summarized in Table 2.

3.1.1. T lymphocytes

To date, the immunophenotype of T cells is well understood as an essential parameter in safety and efficacy features in CAR T products [50,88]. The ratio of subtype composition and an available naïve and central memory T (TCM) cell population are essential to increase the therapeutic efficacy of CAR T cells. Shah et al., reported that enriching CD4+ and CD8+ in the starting T cell population before CAR-lentiviral transfection can enhance the efficacy of CAR T cells on neuroblastoma [89]. The 1:1 ratio of subtype composition between CD4+ and CD8+ possibly promotes high efficiency with more safety in patients [84]. Furthermore, another study exhibited an increased percentage of CD4+ T cells and CD45RO+CD62L+ TCM in CAR treatment of neuroblastoma, shown to prolong the persistence of CAR T cells in clinical trials [50]. Interestingly, CAR T cells derived from naïve T cells and TCM cells strongly proliferated ex vivo, resulting in 89% of the CAR T cell population [90]. Likewise, the procession of CD62L, a standard marker of memory cells, in the CAR population is more effective against both hematological malignancy and solid tumors and promoted longer persistence of peripheral NKT and T cells [91,92,93]. Because of the timing of infiltration into the tumor site, recognizing the tumor antigen, and performing their function, the memory subtype of CAR T cells is an exigency [94]. Selection of the T cell subtype population during CAR production or before administration to patients is recommended for improving the efficacy of CAR T cells against neuroblastomas. Nonetheless, CAR T cells are mostly detected in peripheral blood but have lower tumor infiltration than other effector cells such as NKT and NK cells. Thus, a combination of other infiltrated effector cells or ECM remodeling enzymes may enhance the tumor infiltration of T cells. In contrast, neuroblastoma-targeted CAR T cells may be more appropriate for eradicating the circulating neuroblastoma cells in patients’ blood and lymphatic system.

3.1.2. Natural Killer Cells

Natural killer (NK) cell, lymphoid components of the innate immune system, can prevent pathogen invasion and kill tumor cells without requiring previous stimulation like T lymphocytes [95]. NK cells are mostly found in the peripheral blood, liver, spleen, and bone marrow, and a few are available in the lymphoid tissues [96,97]. NK cells can localize, migrate, and infiltrate into tumor sites to elicit MHC-unrestricted cytotoxicity and secrete proinflammatory cytokines and chemokines against solid tumors [97]. In addition, NK cells showed antitumor responses via the NKG2D and cytotoxic adapter molecule DNAX-activating Protein 10 (DAP10) to eliminate MDSCs, which then reversed the suppressive TME and attracted immune effectors infiltration into the tumor site [98]. According to these antitumor features, NK has been applied in various types of CARs immunotherapy against both hematological malignancies and solid tumors [99,100,101]. Of note, the irradiated NK cell line NK-92 has been adopted as the effector for various designs of CAR to overcome the difficulty of sorting and ex vivo expansion of primary NK cells [102,103].
Several studies reported that CAR-modified NK cells responded to solid tumors and hematological malignancies. CXCR1-expressed NKG2D-targeted CAR NK cells had enhanced tumor trafficking with preserved antitumor effects in ovarian cancer xenograft models [104], while anti-CD147 CAR NK exhibited potent cytotoxicity against hepatocellular carcinoma (HCC) cell lines in vitro and patient-derived HCC xenograft mouse models [100]. As aforementioned, the NK-92 cell line has been used as a CAR effector cell. Li et al. [105] demonstrated that bi-specific PD1-DAP10 and NKG2D CAR NK-92 cells enhanced cytotoxicity to human gastric cells (SGC7901), via triggering apoptosis. These dual-targeting CAR NK-92 cells also displayed significant antitumor activity in the SGC-7901 mouse xenograft. Moreover, the human prostate-specific membrane antigen (PSMA)-targeted modified CAR NK-92 cells recognized and mediated potent antitumor effects against prostate cancer xenograft models [106]. Anti-CD19 CAR NK cells showed potent tumor-specific lethality against NK-resistant lymphoma [107]. In a Phase I/II trial, Liu et al. [108] reported that 8 out of 11 (73%) patients with relapsed or refractory CD19-positive cancers (non-Hodgkin’s lymphoma or chronic lymphocytic leukemia (CLL)) had a response to anti-CD19 CAR NK without the development of major toxic effects. These infused CAR NK cells expanded and persisted in patients at low levels for at least 12 months [108].
For the neuroblastoma treatments, NK cells might requisite proper CAR constructs such as costimulators and/or signaling molecules, which are discussed in the next topic. Furthermore, several researchers indicated that the antitumor properties of CARs NK cells were downregulated by suppressive molecules of the TME, such as TGFβ, thereby limiting the tumor-killing capability of NK cells [98,109]. In this direction, a combination with the inhibitor of immunosuppressive molecules (e.g., anti-TGFβ) might increase the antitumor activities of neuroblastoma-targeted CAR NK cells.

3.1.3. NKT Cells

NKT cells are a subset of innate lymphocytes that share T and NK cells’ properties [110,111]. NKTs have differences in T-cell receptor (TCR) diversity and their cancer immune response, and can be divided into two subtypes; invariant (Type 1, iNKT) and variant (Type 2) NKT cells [102]. Type I NKTs enhance antitumor immunity, while Type II NKTs regulate and inhibit tumor immunity [112,113,114]. iNKT cells have been studied and applied as potent tumor-specific CAR effectors against lymphomas [92,115] melanomas [116], and neuroblastomas [85]. The first use of CAR NKT cells against neuroblastoma was reported in 2014 [85]. Both the second and third generations of anti-GD2 CAR NKT cells exhibited potent antitumor activity and presented significantly prolonged survival without any graft-versus-host disease in a mouse xenograft model [85]. Unlike T cells, NKT effectively traffic and infiltrate into the tumor site [92] to mediate antitumor activities, inhibit tumor-supportive macrophages, and transactivate the localized NK and CD8+ T cells [117,118]. Additionally, a higher percentage of NKT cells in tumor-infiltrating lymphocytes have been detected in neuroblastoma than in patients’ peripheral blood [119]. Likewise, Xu et al. reported that CAR NKT cells enhanced CARs’ treatment efficacy through their persistence and provided high localization onto the tumor site without significant histopathological toxicity [52]. These suggested that NKT cells are more easily infiltrated into neuroblastomas than T lymphocytes. Recently, Heczey et al., [120] reported interim results from three patients with relapsed or resistant neuroblastoma, who enrolled into the Phase I dose escalation trial, receiving dose Level 1 of autologous NKT cells coexpressing GD2 CAR and interleukin-15 (NCT03294954). The results showed that GD2 CAR NKT cells expanded in vivo, localized to tumors, and, in one patient, induced regression of bone metastatic lesions without adverse effects [120].
Neuroblastoma-targeted CAR NKT cells are noticeable as potential effector cells against neuroblastomas and other solid tumors. Of note, an exhausting phenotype of NKTs upon in vitro stimulation is associated with the upregulated expression of PD-1 and TIM-3 [92]. Thus, CAR NKT cells should be used in combination with anti-PD-1 or anti-TIM-3 to generate an intense tumor-killing effect.

3.1.4. γδT Cells

γδT cells are innate T lymphocytes that independently recognize MHC targets, have a natural killing activity against pathogens, and respond to tumor cells [121,122,123]. In contrast to αβT cells, γδT lymphocytes are rare in the peripheral blood and lymphoid organs (<10%), while they mostly reside in a wide variety of tissues including the skin epidermis, gastrointestinal tract mucosa, and the reproductive system [124,125]. Interestingly, γδT cells also found in neuroblastoma tumors [119]. γδT cells can be divided into three subpopulations depending on the differences in TCR-δ chain expression (Vδ1, Vδ2, and Vδ3), paired with the γ9 chain [126]. γδT cells expressing the Vγ9Vδ2 TCR is a common subset that can inhibit proliferation and angiogenesis and encourage tumor cell apoptosis [102]. Moreover, these γδT cells can be activated and proliferated ex vivo for more than 1000-fold expansion [127].
Neuroblastoma mostly escapes from the immune system via downregulation of the MHC Class I molecule [119]. Since γδT TCR does not require MHC-mediated antigen presentation [128,129], γδT cells may be appropriate to address this issue. Furthermore, these cells can differentiate into professional antigen-presenting cells (pAPCs), which present antigenic fragments for CD4+ and CD8+ αβT cells [130]. Although γδT cell exhaustion is associated with PD-1, an immune checkpoint molecule [131], γδT cells have lower expression of PD-1 compared with αβT cells, and are thereby recognized as a promising effector cell for CAR immunotherapy. Like T cells, the naïve and memory phenotype of γδT cells sufficiently survive to promote the persistence of CAR γδT cells in the recipients without decreasing the [132] killing activity against tumor cells [121].
Until now, few studies of CARs γδT cells have been reported. The first anti-CD19 CAR γδT cells showed strong killing efficacy against leukemia [122,132,133] For solid tumors, there have been preclinical studies of CAR γδT cells against melanoma [86] and neuroblastoma [127,134,135]. Therefore, γδT cells are influential candidates for developing CAR immunotherapy against neuroblastoma. At present, two Phase I clinical trials are available in clincaltrial.gov. The first study (NCT04107142) is planning to investigate of the safety and tolerability of NKG2DL-targeting CAR γδT cells in various solid tumors: colorectal, breast, sarcoma, nasopharyngeal carcinoma, prostate, and gastric cancer. The second clinical trial (NCT03885076) is estimating the potential role of anti-CD33-CD28 CAR γδT cells against acute myeloid leukemia (ALL). However, the clinical studies’ results are not yet available. From these properties, the γδT cell is a potential candidate for use as an effector immune cell in adoptive CAR immunotherapy against neuroblastoma.

3.2. Modification of CAR Constructs

The CAR construct design is one of the key features that affect the kinetics of expansion and the duration of persistence. CARs are recombinant receptors responsible for both antigen-binding and activation functions for immune cells [136]. The second-generation CARs offer a clinical benefit via both the TCR stimulatory domain (CD3z) and a single costimulatory domain [89]. Recently, FDA-approved products can contain either CD28 or 4-1BB (CD137) costimulatory domain [80]. Choosing the appropriate combination of costimulatory ligands, chimeric costimulatory receptors, or cytokines in the CARs’ structure for each objective immune cell may differently enhance the effector cells’ function, proliferation, and survival [136]. Good design of CAR structures can overcome the significant barriers of CARs in cancer immunotherapy.

3.2.1. Hinge and Transmembrane Domain

The access of CARs to the target antigen depends on their flexibility and the length of the hinge and transmembrane domain [137]. In the case of anti-CD19 CAR T cells, human CD8α hinge and transmembrane molecules did not promote efficacies differently against tumor xenografts but induced lower cytokine levels compared with CD28 hinge and transmembranes [138,139]. These confirmed that the length and composition of the hinge provided an antigen binding and signaling cascade through the CAR T cells. Accordingly, the CD8α hinge and transmembrane domain may be an excellent choice to generate neuroblastoma-targeted CARs. However, further studies on this topic are required to discover a suitable composition that offers the highest tumor-killing efficacy but bestows the patients with low or no side effect.

3.2.2. Costimulatory Domains

Costimulator molecules play a critical role in the development, activation, and functional response of effector immune cells [92]. A set of costimulatory receptors that are noticeable in human T cells include: CD28, 4-1BB, and OX40 [92], while the costimulators for activating NK cells are CD28, 4-1BB, 2B4, and NKG2D [140,141]. For creating CAR T cells, CD28 has more potent signaling through T cell activation with a short persistence, while 4-1BB provides a less potent signal but longer persistence [142]. To address cytokine releasing syndrome (CRS), the CD28 costimulator is appropriate for the dim target antigen, whereas 4-1BB is suitable for the high density target [139]. Unlike T cells, the CD28 costimulator is suitable for enhancing the proliferation, cancer-killing function, and persistence of NKT cells. Even though CD28 and 4-1BB prevent the loss of CD62L expression that is important to prolong the survival of NKT cells [92], the CD28 costimulator also forces the CAR NKT cell to massive expansion, while 4-1BB induces excessive activation, leading to cell death [52]. For CAR γδT cells, both CD28 and 4-1BB [143] were applied as costimulators [144,145]. However, data on the comparison and optimization of costimulators for CAR γδT cells are scanty. For NK cells, CD28 is mostly used as a costimulator. Oelsner et al. suggested that CD19-CAR NK cells with CD28 costimulators showed better performance against B-cell malignancy than 4-1BB [145]. Fusing DNAM1 and 2B4 costimulatory domains in CAR NK design can enhance cytotoxicity against hepatocellular cancer cells in vitro [146].

3.2.3. Signaling-Transducing Domain

CD3ζ is commonly used as the signaling molecule in CAR constructs to activate T and NK cells functions for both hematologic malignancy and solid tumors [81,89,138,147]. Immunotherapy resistance and CRS are associated with highly active CD3ζ-activated CAR T cells [148,149]. Implementing other activation domains in CAR designs should be considered as an alternative to provide a robust clinical advantage in relieving the risk of CRS and overcoming these barriers.
DNAX activation protein of 12 kDa (DAP12), an immunotyrosine-based activation motif-containing adaptor, is constitutively expressed in NK cells and is expressed in a subset of human T cells [150]. The first use of DAP12 as an alternative cytoplasmic domain in CAR design emerged in 2015 [151]. The chimeric target receptor fusing with the transmembrane and cytoplasmic domains of KIR2DS2, a stimulatory killer immunoglobulin-like receptor, and DAP12 (KIR-CAR DAP12) triggered robust antigen-specific proliferation, effector function, and enhanced antitumor activity in leukemia cell lines [151]. Although only a few studies have used DAP12 as a new activation domain in CAR-modified T cells, the low cytokine markers of CRS could be observed in DAP12-based CAR preclinical studies [148]. Compared with CD3ζ-based second-generation CARs, T cells modified with the natural killer group 2D (NKG2D) ectodomain combined with 4-1BB and the DAP12 signaling domain released lower levels of interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), and interleukin-2 (IL-2) during tumor cell lysis without a significant difference in tumor-killing effects both in vitro and in vivo [148]. DAP12 is also involved in the signal transduction of activating NK cells [152]. DAP12-based CAR NK cells could eradicate neuroblastomas in the mouse xenograft model [152]. This DAP12 might be a brilliant new signaling molecule for high efficacy with more safety in CAR development

3.2.4. Other Generations of CAR Structures

Although the second generation of CARs was mostly used in neuroblastoma clinical trials, the fourth generation of CARs has been developed. With neuroblastoma-targeted antigens incorporating multiple costimulatory molecules and a suicide gene-inducible caspase9 (iCasp9), the success of fourth-CAR T cells against MYCN amplification in neuroblastomas has been reported [53]. This fourth-generation CAR exhibited higher tumor-killing activity than the third-generation CAR with a safety profile, leading to 4 years of survival of a neuroblastoma patient after fourth-generation CAR treatment [53]. Successful fourth-generation CAR T cells are being explored for the new era of treatment for neuroblastoma and other solid tumors.

3.3. Overcoming the TME to Improve the Efficacy of CAR Immunotherapy

The intrinsic tumor factors, including the extracellular matrix (ECM) and the TME, are critical challenges for CAR immunotherapy against neuroblastoma [153]. The counterstrategies for these tumor escape mechanisms are shown in this topic.

3.3.1. ECM: Trafficking and Infiltration of Effector Cells

Low T cell infiltration has been observed in so-called immunologically “cold” neuroblastoma tumors [154]. Various studies demonstrated that poor trafficking to and limited CAR T cell persistence in solid tumors are significant burdens in CAR immunotherapy [155]. To increase the trafficking ability of CAR T cells via presenting matched chemokine receptors or adding therapeutic agents against tumor-enriched chemokines may be a practical strategy for neuroblastoma and other solid tumor treatments [156]. For example, in hepatocellular carcinoma in which a high expression level of CXCR2 ligand was observed, Liu et al. confirmed that using CXCR2 CAR T cells could enhance in vivo trafficking and tumor cytotoxicity [157]. Additionally, the fusion of IL-7 and CCL19 on the CAR structure showed increased infiltration and exhibited high antitumor activity of CAR T cells [158]. Moreover, given the prevalence of IL-8 production in human cancer [159], this approach may find broad applicability in the potentiation of CAR T cell immunotherapy for solid tumors. Accordingly, the tumor chemokine–chemokine receptor network in neuroblastoma should be identified. Studies showed that VEGF, IL-6, and IL-10 receptor (IL-10R) in neuroblastoma were associated with poor outcomes [160,161,162]. Thus, the fusion of anti-VEGF, anti-IL-6, or anti-IL-10R into the CAR structure may also be effective against neuroblastoma.

3.3.2. Myeloid-Derived Suppressor Cells

MDSCs are immunosuppressive immune cells found in the TME [163,164]. These MDSCs enhance tumor growth and suppress the infiltration, proliferation, and tumor-killing activity of CAR T cells [165]. These cells also express PD-L1 themselves, which contributes to immune escape in various solid tumors [166]. Researchers found that the NKG2D ligand, a cytotoxicity receptor activated by non-classical MHC molecules, is overexpressed in these tumor-infiltrating MDSCs [167,168]. A combination with NKG2D-CAR NK cells enhanced the tumor infiltration and expansion of CAR T cells at tumor sites and prolonged CAR T cell survival compared with single treatment [98,169]. The mixed NKG2D-CAR NK and neuroblastoma-targeted CAR T cell treatment may safely enhance antitumor activity against neuroblastomas that are supported and protected by MDSCs.

3.3.3. Tumor Extracellular Vesicles

Extracellular vesicles (EVs) are lipid bilayer vesicles released from the cells [170], which can be classified into three major groups (exosomes, microvesicles, and apoptotic bodies), based on their size [171]. Like cargo, EVs contain soluble proteins such as growth factors, cytokines, chemokines [172], lipids, metabolites, and nucleic acids, including regulatory microRNAs (miRs) [171,173]. Several studies reported that EVs play a crucial role in tumor–tumor [174,175] and tumor–immune cell communications [176] and affect tumor cells’ phenotype and metastatic potential [177,178,179]. Neuroblastoma cells release EVs in their extracellular space [172]. Previous studies showed that neuroblastoma EVs induced the production of pro-tumorigenic cytokines and chemokines such as IL-6, IL-8, VEGF, and CCL2 by MSCs [180]. Additionally, neuroblastoma EVs also trigger a proinflammatory response in monocytes and promote neuroblastoma chemoresistance [181]. Future studies aiming to reveal the exact roles of EVs in the communication between the neuroblastoma and the TME is required to decipher a novel treatment strategy against neuroblastoma TMEs.

3.4. Combination of CARs

3.4.1. PD-1/PDL-1

Programmed cell death 1 (PD-1 or PDCD1) is a coinhibitory receptor expressed by all CAR effector cells, including αβT [182], γδT [143], NKT [52], and NK cells [131]. PD-1 binds to its ligand, PD-L1 [or CD274], leading to a decrease in effector cell receptors’ downstream signaling and diminishing T cell activation [183]. By using immunohistochemical analysis, Zuo et al. [184] reported that up to one-third (11/31) of neuroblastoma patients showed positive PD-L1 expression in tumor tissues. It was not surprising that PD-L1 positivity was associated with decreased overall survival [184]. PD-L1 expression in metastatic neuroblastomas also plays a key role in immune resistance mechanisms [185]. Several reports suggested that inhibition of PD-1/PD-L1 interaction by anti-PD-1 or anti-PD-L1 antibody enhanced T cells’ killing efficacy against tumor cells [67,186,187]. Li et al. reported that combining anti-PD-1 antibody with CAR T cells engineered to secrete PD-1 inhibitors can enhance expansion and inhibitory effects in a human lung carcinoma xenograft mouse model [188]. The modified human CAR T cell secreting PD-1 blocking scFvs (E27-secreting CAR T) had also been established and investigated for therapeutic efficacy in hematopoietic malignancies as compared with their prototype CAR T cells. The results showed that E27-secreting CAR T cells significantly eliminated tumor cells and increased the survival of tumor-bearing mice [189]. Rupp et al. successfully established PD-1 deficient anti-CD19 CAR T cells combined with Cas9 ribonucleoprotein (Cas9 RNP)-mediated gene editing to disturb PD-L1-positive tumor xenografts. The results showed that Cas9-edited CAR T cells can improve the therapeutic efficacy for cancer immunotherapy in vivo [190]. Since PD-L1 is highly upregulated in multiple solid tumors, PD-L1-targeting CAR T cells have been designed. A vector bearing the extracellular and transmembrane regions of human PD-1 (dPD1z) and a CAR vector against PD-L1 (CARPD-L1z) have been established. Both dPD1z and CARPD-L1z effectively suppress the growth of multiple types of tumors in patient-derived xenograft models [191].
The combination of anti-PD1 and CAR T therapy has been investigated in Phase I clinical trials for hematological (NCT04134325, NCT03287817, NCT04337606) and malignant pleural mesothelioma (NCT04577326). Likewise, the safety and efficacy of anti-PD-L1 combined with CAR T cells have been investigated for B-cell malignancies (NCT03310619) and various types of solid tumor including cervical cancer, sarcoma, and non-small cell lung cancer (NCT04556669). In this direction, checkpoint inhibition may be turning the neuroblastoma microenvironment to respond to immunological treatment and enhance the efficacy of CAR therapy against neuroblastomas as well [192,193].

3.4.2. Cytokines

Another approach to improve the potency of CARs is to genetically modify the effector cells secreting pro-inflammatory or pro-proliferative cytokines, aiming to strengthen effector function, proliferation, and persistence, and to alter the TME [136,194]. The cytokines IFN-γ, IL-2, IL-7, IL-12, IL-15, IL-18, IL-21, and IL-27, promoted neuroblastoma regression via enhancing the efficacy of effector cells, whereas VEGF, IL-6, and IL-10 induced neuroblastoma progression through their immunosuppressive activities. Several studies reported that increasing the proinflammatory chemokine IL-8 levels in patients was related to poor prognosis in neuroblastomas and other solid tumors [195,196]. This information is useful for the new design of CAR constructs or combination therapy to enhance the efficacy of CARs against neuroblastoma.
To date, IL-15 cytokine has been studied for its ability to enhance CARs’ antitumor effects. IL-15 plays a crucial role in T and NKT cells [52,197] via enhanced CAR antitumor activity and survival in peripheral blood and tumor tissue [197]. This study suggested that the IL-15-conjugated CD28-CAR structure reduced the exhaustion marker and improved the persistence and the tumor-killing activity of NKT-established CARs. Recently, Xin et al. utilized mice bearing neuroblastoma xenografts to demonstrate that GD2-CAR IL-15 NKT cells enhanced in vivo persistence, increased localization to tumor sites, and improved tumor control as compared with GD2-CAR NKT cells [52]. Interestingly, IL-15-conjugated GD2 CAR T cells reduced the expression of the PD-1 receptor [197]. These characteristics have made IL-15 a promising candidate for CAR T (or NKT) design for neuroblastoma immunotherapy.

4. Conclusions and Future Perspectives

The strategies to improve CARs for use in neuroblastomas are mostly concerned with increasing the antitumor activity and persistence of the infused CAR cells. Therefore, discovering new tumor antigens that are more restricted to neuroblastomas with less neuronal toxicity is very necessary. The evaluation of new target antigens such as B7H3 or o-acetyl-GD2 (oaGD2) is ongoing [198,199]. Researchers can also modify the construct of CARs to be more effective by adding new costimulatory endodomains such as 4-1BB or inducible T-cell costimulator (ICOS), which can be seen in the fourth generation of CAR therapy [200,201]. Seeking a way to overcome the immunosuppressive TME in neuroblastomas is a critical challenge in CAR studies. Various trials of CAR cell therapy in neuroblastomas, which relied on the strategies mentioned above, are underway to validate improved outcomes. Alternatively, administration of antitumor-related cytokines might enhance the immune response and amplify the direct neuroblastoma cell-killing effect of CAR cells in a complex TME. For example, IL-15 has been used to improve in vivo persistence and antitumor activity against neuroblastoma. Hence, another area of interest in CAR structure design is the induction of cytokine expression.
Given all these findings, understanding the mechanism of how the TME affects the resistance of CAR cells and inhibits the in vivo antitumor function is vastly relevant. The exploration of exosomal miRs or exosome decoys released within the neuroblastoma TME may be essential alternatives to overcome the barriers posed by the TME. Furthermore, by understanding the molecular mechanisms within the TME, researchers can efficiently engineer molecular targeting CAR cells to restore TME resistance sensitivity.
Lastly, a more practical approach for CAR-modified cell manufacturing is still needed. The ability to manufacture prompt quantities of an efficacious cell product is required for neuroblastoma patients with various conditions. In the end, good manufacturing might help to create highly active CAR cells for patients with relapsed disease in time.
In summary, immunotherapy using CARs in neuroblastoma treatment has shown promising efficacy and safety. To utilize the therapeutic benefits of CARs as the first line of immunotherapy in patients with a high risk of relapsed neuroblastoma, several approaches have been explored further to augment the antitumor function of CAR-modified cells in vivo. However, unlike the remarkable success in hematological malignancies, various barriers restrict the effective use of this CAR treatment in clinical trials, as indicated earlier. The strategies provided in this review may offer suitable approaches to address the benefits of CAR therapy in neuroblastoma.

Author Contributions

Conceptualization, S.C.; writing—original draft preparation, P.S. and K.A.; writing—review and editing, U.A., S.C., and S.H.; visualization, P.S., K.A., and S.C.; supervision, U.A. and S.H.; funding acquisition, S.H. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the Research Chair Grant from the National Science and Technology Development Agency of Thailand, grant number FDA-CO-2559-3325-TH.

Acknowledgments

This study was supported by the Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand. S.C. was financially supported by the Faculty Staff Development Program of the Faculty of Medicine Ramathibodi Hospital, Mahidol University, for his research activities. All figures were created with BioRender.com.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analysis, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Ahmed, A.A.; Zhang, L.; Reddivalla, N.; Hetherington, M. Neuroblastoma in children: Update on clinicopathologic and genetic prognostic factors. Pediatr. Hematol. Oncol. 2017, 34, 165–185. [Google Scholar] [CrossRef]
  2. Moreno, L.; Rubie, H.; Varo, A.; Le Deley, M.C.; Amoroso, L.; Chevance, A.; Garaventa, A.; Gambart, M.; Bautista, F.; Valteau-Couanet, D.; et al. Outcome of children with relapsed or refractory neuroblastoma: A meta-analysis of ITCC/SIOPEN European phase II clinical trials. Pediatr. Blood Cancer 2017, 64, 25–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Smith, M.A.; Altekruse, S.F.; Adamson, P.C.; Reaman, G.H.; Seibel, N.L. Declining childhood and adolescent cancer mortality. Cancer 2014, 120, 2497–2506. [Google Scholar] [CrossRef] [PubMed]
  4. Ries, L.A.G.; Smith, M.A.; Gurney, J.G.; Linet, M.; Tamra, T.; Young, J.L.; Bunin, G.R.; Bernstein, L.; Key, C.R.; Lynch, C.F.; et al. (Eds.) Cancer Incidence and Survival among Children and Adolescents: United States SEER Program 1975–1995; National Cancer Institute, SEER Program: Bethesda, MD, USA, 1999; NIH Pub. No. 99-4649. [Google Scholar]
  5. Matthay, K.K.; Maris, J.M.; Schleiermacher, G.; Nakagawara, A.; Mackall, C.L.; Diller, L.; Weiss, W.A. Neuroblastoma. Nat. Rev. Dis. Primers 2016, 2, 16078. [Google Scholar] [CrossRef] [PubMed]
  6. Brodeur, G.M. Neuroblastoma: Biological insights into a clinical enigma. Nat. Rev. Cancer 2003, 3, 203–216. [Google Scholar] [CrossRef]
  7. Huibregtse, K.E.; Vo, K.T.; DuBois, S.G.; Fetzko, S.; Neuhaus, J.; Batra, V.; Maris, J.M.; Weiss, B.; Marachelian, A.; Yanik, G.A.; et al. Incidence and risk factors for secondary malignancy in patients with neuroblastoma after treatment with (131)I-metaiodobenzylguanidine. Eur. J. Cancer 2016, 66, 144–152. [Google Scholar] [CrossRef]
  8. Li, R.; Polishchuk, A.; DuBois, S.; Hawkins, R.; Lee, S.W.; Bagatell, R.; Shusterman, S.; Hill-Kayser, C.; Al-Sayegh, H.; Diller, L.; et al. Patterns of Relapse in High-Risk Neuroblastoma Patients Treated With and Without Total Body Irradiation. Int. J. Radiat. Oncol. Biol. Phys. 2017, 97, 270–277. [Google Scholar] [CrossRef]
  9. Cheung, I.Y.; Kushner, B.H.; Modak, S.; Basu, E.M.; Roberts, S.S.; Cheung, N.V. Phase I trial of anti-GD2 monoclonal antibody hu3F8 plus GM-CSF: Impact of body weight, immunogenicity and anti-GD2 response on pharmacokinetics and survival. Oncoimmunology 2017, 6, e1358331. [Google Scholar] [CrossRef] [Green Version]
  10. Yu, A.L.; Gilman, A.L.; Ozkaynak, M.F.; London, W.B.; Kreissman, S.G.; Chen, H.X.; Smith, M.; Anderson, B.; Villablanca, J.G.; Matthay, K.K.; et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N. Engl. J. Med. 2010, 363, 1324–1334. [Google Scholar] [CrossRef] [Green Version]
  11. Kushner, B.H.; Ostrovnaya, I.; Cheung, I.Y.; Kuk, D.; Kramer, K.; Modak, S.; Yataghene, K.; Cheung, N.K. Prolonged progression-free survival after consolidating second or later remissions of neuroblastoma with Anti-GD2 immunotherapy and isotretinoin: A prospective Phase II study. Oncoimmunology 2015, 4, e1016704. [Google Scholar] [CrossRef] [Green Version]
  12. Park, J.R.; Digiusto, D.L.; Slovak, M.; Wright, C.; Naranjo, A.; Wagner, J.; Meechoovet, H.B.; Bautista, C.; Chang, W.C.; Ostberg, J.R.; et al. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol. Ther. 2007, 15, 825–833. [Google Scholar] [CrossRef]
  13. Gardner, R.A.; Finney, O.; Annesley, C.; Brakke, H.; Summers, C.; Leger, K.; Bleakley, M.; Brown, C.; Mgebroff, S.; Kelly-Spratt, K.S.; et al. Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. Blood 2017, 129, 3322–3331. [Google Scholar] [CrossRef] [PubMed]
  14. Lee, D.W.; Kochenderfer, J.N.; Stetler-Stevenson, M.; Cui, Y.K.; Delbrook, C.; Feldman, S.A.; Fry, T.J.; Orentas, R.; Sabatino, M.; Shah, N.N.; et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: A phase 1 dose-escalation trial. Lancet 2015, 385, 517–528. [Google Scholar] [CrossRef]
  15. Maude, S.L.; Frey, N.; Shaw, P.A.; Aplenc, R.; Barrett, D.M.; Bunin, N.J.; Chew, A.; Gonzalez, V.E.; Zheng, Z.; Lacey, S.F.; et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 2014, 371, 1507–1517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Asgharzadeh, S.; Salo, J.A.; Ji, L.; Oberthuer, A.; Fischer, M.; Berthold, F.; Hadjidaniel, M.; Liu, C.W.; Metelitsa, L.S.; Pique-Regi, R.; et al. Clinical significance of tumor-associated inflammatory cells in metastatic neuroblastoma. J. Clin. Oncol. 2012, 30, 3525–3532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Heczey, A.; Louis, C.U. Advances in chimeric antigen receptor immunotherapy for neuroblastoma. Discov. Med. 2013, 16, 287–294. [Google Scholar] [PubMed]
  18. Pule, M.A.; Savoldo, B.; Myers, G.D.; Rossig, C.; Russell, H.V.; Dotti, G.; Huls, M.H.; Liu, E.; Gee, A.P.; Mei, Z.; et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: Persistence and antitumor activity in individuals with neuroblastoma. Nat. Med. 2008, 14, 1264–1270. [Google Scholar] [CrossRef] [PubMed]
  19. Gross, G.; Gorochov, G.; Waks, T.; Eshhar, Z. Generation of effector T cells expressing chimeric T cell receptor with antibody type-specificity. Transplant. Proc. 1989, 21, 127–130. [Google Scholar]
  20. 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]
  21. Couzin-Frankel, J. Breakthrough of the year 2013. Cancer immunotherapy. Science 2013, 342, 1432–1433. [Google Scholar] [CrossRef] [Green Version]
  22. Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.; Myers, G.D.; et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 439–448. [Google Scholar] [CrossRef] [PubMed]
  23. Neelapu, S.S.; Locke, F.L.; Bartlett, N.L.; Lekakis, L.J.; Miklos, D.B.; Jacobson, C.A.; Braunschweig, I.; Oluwole, O.O.; Siddiqi, T.; Lin, Y.; et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 2531–2544. [Google Scholar] [CrossRef] [PubMed]
  24. Savoldo, B.; Dotti, G. Chimeric antigen receptors (CARs) from bench-to-bedside. Immunol. Lett. 2013, 155, 40–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Richards, R.M.; Sotillo, E.; Majzner, R.G. CAR T Cell Therapy for Neuroblastoma. Front. Immunol. 2018, 9, 2380. [Google Scholar] [CrossRef] [Green Version]
  26. Mosse, Y.P.; Laudenslager, M.; Longo, L.; Cole, K.A.; Wood, A.; Attiyeh, E.F.; Laquaglia, M.J.; Sennett, R.; Lynch, J.E.; Perri, P.; et al. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature 2008, 455, 930–935. [Google Scholar] [CrossRef] [Green Version]
  27. De Brouwer, S.; De Preter, K.; Kumps, C.; Zabrocki, P.; Porcu, M.; Westerhout, E.M.; Lakeman, A.; Vandesompele, J.; Hoebeeck, J.; Van Maerken, T.; et al. Meta-analysis of neuroblastomas reveals a skewed ALK mutation spectrum in tumors with MYCN amplification. Clin. Cancer Res. 2010, 16, 4353–4362. [Google Scholar] [CrossRef] [Green Version]
  28. Moog-Lutz, C.; Degoutin, J.; Gouzi, J.Y.; Frobert, Y.; Brunet-de Carvalho, N.; Bureau, J.; Creminon, C.; Vigny, M. Activation and inhibition of anaplastic lymphoma kinase receptor tyrosine kinase by monoclonal antibodies and absence of agonist activity of pleiotrophin. J. Biol. Chem. 2005, 280, 26039–26048. [Google Scholar] [CrossRef] [Green Version]
  29. Walker, A.J.; Majzner, R.G.; Zhang, L.; Wanhainen, K.; Long, A.H.; Nguyen, S.M.; Lopomo, P.; Vigny, M.; Fry, T.J.; Orentas, R.J.; et al. Tumor Antigen and Receptor Densities Regulate Efficacy of a Chimeric Antigen Receptor Targeting Anaplastic Lymphoma Kinase. Mol. Ther. 2017, 25, 2189–2201. [Google Scholar] [CrossRef] [Green Version]
  30. Li, N.; Fu, H.; Hewitt, S.M.; Dimitrov, D.S.; Ho, M. Therapeutically targeting glypican-2 via single-domain antibody-based chimeric antigen receptors and immunotoxins in neuroblastoma. Proc. Natl. Acad. Sci. USA 2017, 114, E6623–E6631. [Google Scholar] [CrossRef] [Green Version]
  31. Baral, A.; Ye, H.X.; Jiang, P.C.; Yao, Y.; Mao, Y. B7-H3 and B7-H1 expression in cerebral spinal fluid and tumor tissue correlates with the malignancy grade of glioma patients. Oncol. Lett. 2014, 8, 1195–1201. [Google Scholar] [CrossRef] [Green Version]
  32. Zhou, Z.; Luther, N.; Ibrahim, G.M.; Hawkins, C.; Vibhakar, R.; Handler, M.H.; Souweidane, M.M. B7-H3, a potential therapeutic target, is expressed in diffuse intrinsic pontine glioma. J. Neurooncol. 2013, 111, 257–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Du, H.; Hirabayashi, K.; Ahn, S.; Kren, N.P.; Montgomery, S.A.; Wang, X.; Tiruthani, K.; Mirlekar, B.; Michaud, D.; Greene, K.; et al. Antitumor Responses in the Absence of Toxicity in Solid Tumors by Targeting B7-H3 via Chimeric Antigen Receptor T Cells. Cancer Cell 2019, 35, 221–237 e228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Majzner, R.G.; Theruvath, J.L.; Nellan, A.; Heitzeneder, S.; Cui, Y.; Mount, C.W.; Rietberg, S.P.; Linde, M.H.; Xu, P.; Rota, C.; et al. CAR T Cells Targeting B7-H3, a Pan-Cancer Antigen, Demonstrate Potent Preclinical Activity Against Pediatric Solid Tumors and Brain Tumors. Clin. Cancer Res. 2019, 25, 2560–2574. [Google Scholar] [CrossRef] [PubMed]
  35. Crossland, D.L.; Denning, W.L.; Ang, S.; Olivares, S.; Mi, T.; Switzer, K.; Singh, H.; Huls, H.; Gold, K.S.; Glisson, B.S.; et al. Antitumor activity of CD56-chimeric antigen receptor T cells in neuroblastoma and SCLC models. Oncogene 2018, 37, 3686–3697. [Google Scholar] [CrossRef]
  36. Zeromski, J.; Nyczak, E.; Dyszkiewicz, W. Significance of cell adhesion molecules, CD56/NCAM in particular, in human tumor growth and spreading. Folia Histochem. Cytobiol. 2001, 39 (Suppl. 2), 36–37. [Google Scholar]
  37. Singh, N.; Kulikovskaya, I.; Barrett, D.M.; Binder-Scholl, G.; Jakobsen, B.; Martinez, D.; Pawel, B.; June, C.H.; Kalos, M.D.; Grupp, S.A. T cells targeting NY-ESO-1 demonstrate efficacy against disseminated neuroblastoma. Oncoimmunology 2016, 5, e1040216. [Google Scholar] [CrossRef] [Green Version]
  38. Spel, L.; Boelens, J.J.; van der Steen, D.M.; Blokland, N.J.; van Noesel, M.M.; Molenaar, J.J.; Heemskerk, M.H.; Boes, M.; Nierkens, S. Natural killer cells facilitate PRAME-specific T-cell reactivity against neuroblastoma. Oncotarget 2015, 6, 35770–35781. [Google Scholar] [CrossRef] [Green Version]
  39. Arlt, M.J.; Novak-Hofer, I.; Gast, D.; Gschwend, V.; Moldenhauer, G.; Grunberg, J.; Honer, M.; Schubiger, P.A.; Altevogt, P.; Kruger, A. Efficient inhibition of intra-peritoneal tumor growth and dissemination of human ovarian carcinoma cells in nude mice by anti-L1-cell adhesion molecule monoclonal antibody treatment. Cancer Res. 2006, 66, 936–943. [Google Scholar] [CrossRef] [Green Version]
  40. Wang, X.; Chang, W.C.; Wong, C.W.; Colcher, D.; Sherman, M.; Ostberg, J.R.; Forman, S.J.; Riddell, S.R.; Jensen, M.C. A transgene-encoded cell surface polypeptide for selection, in vivo tracking, and ablation of engineered cells. Blood 2011, 118, 1255–1263. [Google Scholar] [CrossRef]
  41. Kunkele, A.; Taraseviciute, A.; Finn, L.S.; Johnson, A.J.; Berger, C.; Finney, O.; Chang, C.A.; Rolczynski, L.S.; Brown, C.; Mgebroff, S.; et al. Preclinical Assessment of CD171-Directed CAR T-cell Adoptive Therapy for Childhood Neuroblastoma: CE7 Epitope Target Safety and Product Manufacturing Feasibility. Clin. Cancer Res. 2017, 23, 466–477. [Google Scholar] [CrossRef] [Green Version]
  42. Rossig, C.; Bollard, C.M.; Nuchtern, J.G.; Merchant, D.A.; Brenner, M.K. Targeting of G(D2)-positive tumor cells by human T lymphocytes engineered to express chimeric T-cell receptor genes. Int. J. Cancer 2001, 94, 228–236. [Google Scholar] [CrossRef] [PubMed]
  43. Suzuki, M.; Cheung, N.K.V. Disialoganglioside GD2 as a therapeutic target for human diseases. Expert Opin. Ther. Tar. 2015, 19, 349–362. [Google Scholar] [CrossRef] [PubMed]
  44. Cheung, N.K.; Cheung, I.Y.; Kramer, K.; Modak, S.; Kuk, D.; Pandit-Taskar, N.; Chamberlain, E.; Ostrovnaya, I.; Kushner, B.H. Key role for myeloid cells: Phase II results of anti-G(D2) antibody 3F8 plus granulocyte-macrophage colony-stimulating factor for chemoresistant osteomedullary neuroblastoma. Int. J. Cancer 2014, 135, 2199–2205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Gilman, A.L.; Ozkaynak, M.F.; Matthay, K.K.; Krailo, M.; Yu, A.L.; Gan, J.; Sternberg, A.; Hank, J.A.; Seeger, R.; Reaman, G.H.; et al. Phase I study of ch14.18 with granulocyte-macrophage colony-stimulating factor and interleukin-2 in children with neuroblastoma after autologous bone marrow transplantation or stem-cell rescue: A report from the Children’s Oncology Group. J. Clin. Oncol. 2009, 27, 85–91. [Google Scholar] [CrossRef] [PubMed]
  46. Ozkaynak, M.F.; Sondel, P.M.; Krailo, M.D.; Gan, J.; Javorsky, B.; Reisfeld, R.A.; Matthay, K.K.; Reaman, G.H.; Seeger, R.C. Phase I study of chimeric human/murine anti-ganglioside G(D2) monoclonal antibody (ch14.18) with granulocyte-macrophage colony-stimulating factor in children with neuroblastoma immediately after hematopoietic stem-cell transplantation: A Children’s Cancer Group Study. J. Clin. Oncol. 2000, 18, 4077–4085. [Google Scholar] [CrossRef] [PubMed]
  47. Straathof, K.; Flutter, B.; Wallace, R.; Thomas, S.; Cheung, G.; Collura, A.; Gileadi, T.; Barton, J.; Wright, G.; Inglott, S.; et al. A Cancer Research UK phase I trial of anti-GD2 chimeric antigen receptor (CAR) transduced T-cells (1RG-CART) in patients with relapsed or refractory neuroblastoma. Cancer Res. 2018, 78. [Google Scholar] [CrossRef]
  48. Pule, M.A.; Straathof, K.C.; Dotti, G.; Heslop, H.E.; Rooney, C.M.; Brenner, M.K. A chimeric T cell antigen receptor that augments cytokine release and supports clonal expansion of primary human T cells. Mol. Ther. 2005, 12, 933–941. [Google Scholar] [CrossRef]
  49. Heczey, A.; Louis, C.U.; Savoldo, B.; Dakhova, O.; Durett, A.; Grilley, B.; Liu, H.; Wu, M.F.F.; Mei, Z.Y.; Gee, A.; et al. CAR T Cells Administered in Combination with Lymphodepletion and PD-1 Inhibition to Patients with Neuroblastoma. Mol. Ther. 2017, 25, 2214–2224. [Google Scholar] [CrossRef] [Green Version]
  50. Louis, C.U.; Savoldo, B.; Dotti, G.; Pule, M.; Yvon, E.; Myers, G.D.; Rossig, C.; Russell, H.V.; Diouf, O.; Liu, E.L.; et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood 2011, 118, 6050–6056. [Google Scholar] [CrossRef]
  51. Sun, J.; Huye, L.E.; Lapteva, N.; Mamonkin, M.; Hiregange, M.; Ballard, B.; Dakhova, O.; Raghavan, D.; Durett, A.G.; Perna, S.K.; et al. Early transduction produces highly functional chimeric antigen receptor-modified virus-specific T-cells with central memory markers: A Production Assistant for Cell Therapy (PACT) translational application. J. Immunother. Cancer 2015, 3, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Xu, X.; Huang, W.; Heczey, A.; Liu, D.; Guo, L.; Wood, M.; Jin, J.; Courtney, A.N.; Liu, B.; Di Pierro, E.J.; et al. NKT Cells Coexpressing a GD2-Specific Chimeric Antigen Receptor and IL15 Show Enhanced In Vivo Persistence and Antitumor Activity against Neuroblastoma. Clin. Cancer Res. 2019, 25, 7126–7138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Xu, X.; Zhao, W.; Yue, Z.; Qin, M.; Jin, M.; Chang, L.J.; Ma, X. 4SCAR-GD2-modified T-cell therapy in neuroblastoma with MYCN amplification: A case report with over 4-year follow-up data. Pediatr. Investig. 2020, 4, 55–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Hattinger, C.M.; Patrizio, M.P.; Magagnoli, F.; Luppi, S.; Serra, M. An update on emerging drugs in osteosarcoma: Towards tailored therapies? Expert Opin. Emerg. Drugs 2019, 24, 153–171. [Google Scholar] [CrossRef] [PubMed]
  55. Tanaka, M.; Tashiro, H.; Omer, B.; Lapteva, N.; Ando, J.; Ngo, M.; Mehta, B.; Dotti, G.; Kinchington, P.R.; Leen, A.M.; et al. Vaccination Targeting Native Receptors to Enhance the Function and Proliferation of Chimeric Antigen Receptor (CAR)-Modified T Cells. Clin. Cancer Res. 2017, 23, 3499–3509. [Google Scholar] [CrossRef] [Green Version]
  56. Robbins, P.F.; Dudley, M.E.; Wunderlich, J.; El-Gamil, M.; Li, Y.F.; Zhou, J.; Huang, J.; Powell, D.J., Jr.; Rosenberg, S.A. Cutting edge: Persistence of transferred lymphocyte clonotypes correlates with cancer regression in patients receiving cell transfer therapy. J. Immunol. 2004, 173, 7125–7130. [Google Scholar] [CrossRef] [Green Version]
  57. 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]
  58. Himoudi, N.; Yan, M.Y.; Papanastasiou, A.; Anderson, J. MYCN as a target for cancer immunotherapy. Cancer Immunol. Immun. 2008, 57, 693–700. [Google Scholar] [CrossRef]
  59. Ahmed, N.; Brawley, V.S.; Hegde, M.; Robertson, C.; Ghazi, A.; Gerken, C.; Liu, E.; Dakhova, O.; Ashoori, A.; Corder, A.; et al. Human Epidermal Growth Factor Receptor 2 (HER2) -Specific Chimeric Antigen Receptor-Modified T Cells for the Immunotherapy of HER2-Positive Sarcoma. J. Clin. Oncol. 2015, 33, 1688–1696. [Google Scholar] [CrossRef]
  60. Goff, S.L.; Morgan, R.A.; Yang, J.C.; Sherry, R.M.; Robbins, P.F.; Restifo, N.P.; Feldman, S.A.; Lu, Y.C.; Lu, L.; Zheng, Z.; et al. Pilot Trial of Adoptive Transfer of Chimeric Antigen Receptor-transduced T Cells Targeting EGFRvIII in Patients With Glioblastoma. J. Immunother. 2019, 42, 126–135. [Google Scholar] [CrossRef]
  61. Morgan, R.A.; Yang, J.C.; Kitano, M.; Dudley, M.E.; Laurencot, C.M.; Rosenberg, S.A. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. 2010, 18, 843–851. [Google Scholar] [CrossRef]
  62. Thistlethwaite, F.C.; Gilham, D.E.; Guest, R.D.; Rothwell, D.G.; Pillai, M.; Burt, D.J.; Byatte, A.J.; Kirillova, N.; Valle, J.W.; Sharma, S.K.; et al. The clinical efficacy of first-generation carcinoembryonic antigen (CEACAM5)-specific CAR T cells is limited by poor persistence and transient pre-conditioning-dependent respiratory toxicity. Cancer Immunol. Immunother. 2017, 66, 1425–1436. [Google Scholar] [CrossRef] [PubMed]
  63. Hegde, M.; DeRenzo, C.C.; Zhang, H.M.; Mata, M.; Gerken, C.; Shree, A.; Yi, Z.Z.; Brawley, V.; Dakhova, O.; Wu, M.F.; et al. Expansion of HER2-CAR T cells after lymphodepletion and clinical responses in patients with advanced sarcoma. J. Clin. Oncol. 2017, 35. [Google Scholar] [CrossRef]
  64. Navai, S.A.; Derenzo, C.; Joseph, S.; Sanber, K.; Byrd, T.; Zhang, H.M.; Mata, M.; Gerken, C.; Shree, A.; Mathew, P.R.; et al. Administration of HER2-CAR T cells after lymphodepletion safely improves T cell expansion and induces clinical responses in patients with advanced sarcomas. Cancer Res. 2019, 79. [Google Scholar] [CrossRef]
  65. Mina, M.; Boldrini, R.; Citti, A.; Romania, P.; D’Alicandro, V.; De Ioris, M.; Castellano, A.; Furlanello, C.; Locatelli, F.; Fruci, D. Tumor-infiltrating T lymphocytes improve clinical outcome of therapy-resistant neuroblastoma. Oncoimmunology 2015, 4, e1019981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Majzner, R.G.; Simon, J.S.; Grosso, J.F.; Martinez, D.; Pawel, B.R.; Santi, M.; Merchant, M.S.; Geoerger, B.; Hezam, I.; Marty, V.; et al. Assessment of programmed death-ligand 1 expression and tumor-associated immune cells in pediatric cancer tissues. Cancer 2017, 123, 3807–3815. [Google Scholar] [CrossRef] [Green Version]
  67. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [Green Version]
  68. Moon, E.K.; Wang, L.C.; Dolfi, D.V.; Wilson, C.B.; Ranganathan, R.; Sun, J.; Kapoor, V.; Scholler, J.; Pure, E.; Milone, M.C.; et al. Multifactorial T-cell hypofunction that is reversible can limit the efficacy of chimeric antigen receptor-transduced human T cells in solid tumors. Clin. Cancer Res. 2014, 20, 4262–4273. [Google Scholar] [CrossRef] [Green Version]
  69. Dwivedi, V.P.; Tousif, S.; Bhattacharya, D.; Prasad, D.V.; Van Kaer, L.; Das, J.; Das, G. Transforming growth factor-beta protein inversely regulates in vivo differentiation of interleukin-17 (IL-17)-producing CD4+ and CD8+ T cells. J. Biol. Chem. 2012, 287, 2943–2947. [Google Scholar] [CrossRef] [Green Version]
  70. Seo, N.; Hayakawa, S.; Takigawa, M.; Tokura, Y. Interleukin-10 expressed at early tumour sites induces subsequent generation of CD4(+) T-regulatory cells and systemic collapse of antitumour immunity. Immunology 2001, 103, 449–457. [Google Scholar] [CrossRef]
  71. Soldati, R.; Berger, E.; Zenclussen, A.C.; Jorch, G.; Lode, H.N.; Salatino, M.; Rabinovich, G.A.; Fest, S. Neuroblastoma triggers an immunoevasive program involving galectin-1-dependent modulation of T cell and dendritic cell compartments. Int. J. Cancer 2012, 131, 1131–1141. [Google Scholar] [CrossRef]
  72. Kloss, C.C.; Lee, J.; Zhang, A.; Chen, F.; Melenhorst, J.J.; Lacey, S.F.; Maus, M.V.; Fraietta, J.A.; Zhao, Y.; June, C.H. Dominant-Negative TGF-beta Receptor Enhances PSMA-Targeted Human CAR T Cell Proliferation And Augments Prostate Cancer Eradication. Mol. Ther. 2018, 26, 1855–1866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Vanichapol, T.; Chiangjong, W.; Panachan, J.; Anurathapan, U.; Chutipongtanate, S.; Hongeng, S. Secretory High-Mobility Group Box 1 Protein Affects Regulatory T Cell Differentiation in Neuroblastoma Microenvironment In Vitro. J. Oncol. 2018, 2018, 7946021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Hainaut, P.; Plymoth, A. Targeting the hallmarks of cancer: Towards a rational approach to next-generation cancer therapy. Curr. Opin. Oncol. 2013, 25, 50–51. [Google Scholar] [CrossRef]
  75. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Nagarsheth, N.; Wicha, M.S.; Zou, W.P. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat. Rev. Immunol. 2017, 17, 559–572. [Google Scholar] [CrossRef] [Green Version]
  77. Craddock, J.A.; Lu, A.; Bear, A.; Pule, M.; Brenner, M.K.; Rooney, C.M.; Foster, A.E. Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by expression of the chemokine receptor CCR2b. J. Immunother. 2010, 33, 780–788. [Google Scholar] [CrossRef] [Green Version]
  78. Di Stasi, A.; De Angelis, B.; Rooney, C.M.; Zhang, L.; Mahendravada, A.; Foster, A.E.; Heslop, H.E.; Brenner, M.K.; Dotti, G.; Savoldo, B. T lymphocytes coexpressing CCR4 and a chimeric antigen receptor targeting CD30 have improved homing and antitumor activity in a Hodgkin tumor model. Blood 2009, 113, 6392–6402. [Google Scholar] [CrossRef] [Green Version]
  79. Mohammed, S.; Sukumaran, S.; Bajgain, P.; Watanabe, N.; Heslop, H.E.; Rooney, C.M.; Brenner, M.K.; Fisher, W.E.; Leen, A.M.; Vera, J.F. Improving Chimeric Antigen Receptor-Modified T Cell Function by Reversing the Immunosuppressive Tumor Microenvironment of Pancreatic Cancer. Mol. Ther. 2017, 25, 249–258. [Google Scholar] [CrossRef] [Green Version]
  80. Davila, M.L.; Papapetrou, E.P. CARs Move To the Fast Lane. Mol. Ther. 2014, 22, 477–478. [Google Scholar] [CrossRef]
  81. Prasongtanakij, S.; Anurathapan, U.; Vanichapol, T.; Jittorntrum, B.; Atjanasuppat, K.; Pongpitcha, P.; Pakakasama, S.; Songdej, D.; Sirachainan, N.; Paisooksantivatana, K.; et al. Production and characterization of haploidentical CD19 CAR T cells: Validated to induce a continuous complete remission in a patient with relapsed refractory B-cell ALL. Asia Pac. J. Clin. Oncol. 2020. [Google Scholar] [CrossRef]
  82. Feins, S.; Kong, W.; Williams, E.F.; Milone, M.C.; Fraietta, J.A. An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. Am. J. Hematol. 2019, 94, S3–S9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Shusterman, S.; London, W.B.; Gillies, S.D.; Hank, J.A.; Voss, S.D.; Seeger, R.C.; Reynolds, C.P.; Kimball, J.; Albertini, M.R.; Wagner, B.; et al. Antitumor activity of hu14.18-IL2 in patients with relapsed/refractory neuroblastoma: A Children’s Oncology Group (COG) phase II study. J. Clin. Oncol. 2010, 28, 4969–4975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Abramson, J.S.; McGree, B.; Noyes, S.; Plummer, S.; Wong, C.; Chen, Y.B.; Palmer, E.; Albertson, T.; Ferry, J.A.; Arrillaga-Romany, I.C. Anti-CD19 CAR T Cells in CNS Diffuse Large-B-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 783–784. [Google Scholar] [CrossRef] [PubMed]
  85. Heczey, A.; Liu, D.; Tian, G.; Courtney, A.N.; Wei, J.; Marinova, E.; Gao, X.; Guo, L.; Yvon, E.; Hicks, J.; et al. Invariant NKT cells with chimeric antigen receptor provide a novel platform for safe and effective cancer immunotherapy. Blood 2014, 124, 2824–2833. [Google Scholar] [CrossRef] [Green Version]
  86. Harrer, D.C.; Simon, B.; Fujii, S.I.; Shimizu, K.; Uslu, U.; Schuler, G.; Gerer, K.F.; Hoyer, S.; Dorrie, J.; Schaft, N. RNA-transfection of gamma/delta T cells with a chimeric antigen receptor or an alpha/beta T-cell receptor: A safer alternative to genetically engineered alpha/beta T cells for the immunotherapy of melanoma. BMC Cancer 2017, 17, 551. [Google Scholar] [CrossRef]
  87. Daher, M.; Rezvani, K. Next generation natural killer cells for cancer immunotherapy: The promise of genetic engineering. Curr. Opin. Immunol. 2018, 51, 146–153. [Google Scholar] [CrossRef] [PubMed]
  88. Turtle, C.J.; Hanafi, L.A.; Berger, C.; Hudecek, M.; Pender, B.; Robinson, E.; Hawkins, R.; Chaney, C.; Cherian, S.; Chen, X.; et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci. Transl. Med. 2016, 8, 355ra116. [Google Scholar] [CrossRef] [Green Version]
  89. Shah, N.N.; Fry, T.J. Mechanisms of resistance to CAR T cell therapy. Nat. Rev. Clin. Oncol. 2019, 16, 372–385. [Google Scholar] [CrossRef] [PubMed]
  90. Schmueck-Henneresse, M.; Omer, B.; Shum, T.; Tashiro, H.; Mamonkin, M.; Lapteva, N.; Sharma, S.; Rollins, L.; Dotti, G.; Reinke, P.; et al. Comprehensive Approach for Identifying the T Cell Subset Origin of CD3 and CD28 Antibody-Activated Chimeric Antigen Receptor-Modified T Cells. J. Immunol. 2017, 199, 348–362. [Google Scholar] [CrossRef] [Green Version]
  91. Graef, P.; Buchholz, V.R.; Stemberger, C.; Flossdorf, M.; Henkel, L.; Schiemann, M.; Drexler, I.; Hofer, T.; Riddell, S.R.; Busch, D.H. Serial transfer of single-cell-derived immunocompetence reveals stemness of CD8(+) central memory T cells. Immunity 2014, 41, 116–126. [Google Scholar] [CrossRef] [Green Version]
  92. Tian, G.; Courtney, A.N.; Jena, B.; Heczey, A.; Liu, D.; Marinova, E.; Guo, L.; Xu, X.; Torikai, H.; Mo, Q.; et al. CD62L+ NKT cells have prolonged persistence and antitumor activity in vivo. J. Clin. Investig. 2016, 126, 2341–2355. [Google Scholar] [CrossRef]
  93. Sommermeyer, D.; Hudecek, M.; Kosasih, P.L.; Gogishvili, T.; Maloney, D.G.; Turtle, C.J.; Riddell, S.R. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia 2016, 30, 492–500. [Google Scholar] [CrossRef] [Green Version]
  94. Rodriguez-Garcia, A.; Palazon, A.; Noguera-Ortega, E.; Powell, D.J., Jr.; Guedan, S. CAR-T Cells Hit the Tumor Microenvironment: Strategies to Overcome Tumor Escape. Front. Immunol. 2020, 11, 1109. [Google Scholar] [CrossRef]
  95. Pietra, G.; Vitale, C.; Pende, D.; Bertaina, A.; Moretta, F.; Falco, M.; Vacca, P.; Montaldo, E.; Cantoni, C.; Mingari, M.C.; et al. Human natural killer cells: News in the therapy of solid tumors and high-risk leukemias. Cancer Immunol. Immunother. 2016, 65, 465–476. [Google Scholar] [CrossRef]
  96. Rezvani, K.; Rouce, R.; Liu, E.; Shpall, E. Engineering Natural Killer Cells for Cancer Immunotherapy. Mol. Ther. 2017, 25, 1769–1781. [Google Scholar] [CrossRef]
  97. Caligiuri, M.A. Human natural killer cells. Blood 2008, 112, 461–469. [Google Scholar] [CrossRef]
  98. Parihar, R.; Rivas, C.; Huynh, M.; Omer, B.; Lapteva, N.; Metelitsa, L.S.; Gottschalk, S.M.; Rooney, C.M. NK Cells Expressing a Chimeric Activating Receptor Eliminate MDSCs and Rescue Impaired CAR-T Cell Activity against Solid Tumors. Cancer Immunol. Res. 2019, 7, 363–375. [Google Scholar] [CrossRef]
  99. Jamali, A.; Hadjati, J.; Madjd, Z.; Mirzaei, H.R.; Thalheimer, F.B.; Agarwal, S.; Bonig, H.; Ullrich, E.; Hartmann, J. Highly Efficient Generation of Transgenically Augmented CAR NK Cells Overexpressing CXCR4. Front. Immunol. 2020, 11, 2028. [Google Scholar] [CrossRef]
  100. Tseng, H.C.; Xiong, W.; Badeti, S.; Yang, Y.; Ma, M.; Liu, T.; Ramos, C.A.; Dotti, G.; Fritzky, L.; Jiang, J.G.; et al. Efficacy of anti-CD147 chimeric antigen receptors targeting hepatocellular carcinoma. Nat. Commun. 2020, 11, 4810. [Google Scholar] [CrossRef]
  101. Morgan, M.A.; Buning, H.; Sauer, M.; Schambach, A. Use of Cell and Genome Modification Technologies to Generate Improved “Off-the-Shelf” CAR T and CAR NK Cells. Front. Immunol. 2020, 11, 1965. [Google Scholar] [CrossRef]
  102. Rotolo, R.; Leuci, V.; Donini, C.; Cykowska, A.; Gammaitoni, L.; Medico, G.; Valabrega, G.; Aglietta, M.; Sangiolo, D. CAR-Based Strategies beyond T Lymphocytes: Integrative Opportunities for Cancer Adoptive Immunotherapy. Int. J. Mol. Sci. 2019, 20, 2839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Klingemann, H.; Boissel, L.; Toneguzzo, F. Natural Killer Cells for Immunotherapy-Advantages of the NK-92 Cell Line over Blood NK Cells. Front. Immunol. 2016, 7, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Ng, Y.Y.; Tay, J.C.K.; Wang, S. CXCR1 Expression to Improve Anti-Cancer Efficacy of Intravenously Injected CAR-NK Cells in Mice with Peritoneal Xenografts. Mol. Ther. Oncolytics 2020, 16, 75–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Li, M.; Zhi, L.; Yin, M.; Guo, C.; Zhang, H.; Lu, C.; Zhu, W. A novel bispecific chimeric PD1-DAP10/NKG2D receptor augments NK92-cell therapy efficacy for human gastric cancer SGC-7901 cell. Biochem. Biophys. Res. Commun. 2020, 523, 745–752. [Google Scholar] [CrossRef] [PubMed]
  106. Montagner, I.M.; Penna, A.; Fracasso, G.; Carpanese, D.; Dalla Pieta, A.; Barbieri, V.; Zuccolotto, G.; Rosato, A. Anti-PSMA CAR-engineered NK-92 Cells: An Off-the-shelf Cell Therapy for Prostate Cancer. Cells 2020, 9, 1382. [Google Scholar] [CrossRef] [PubMed]
  107. Gang, M.; Marin, N.D.; Wong, P.; Neal, C.C.; Marsala, L.; Foster, M.; Schappe, T.; Meng, W.; Tran, J.; Schaettler, M.; et al. CAR-modified memory-like NK cells exhibit potent responses to NK-resistant lymphomas. Blood 2020, 136, 2308–2318. [Google Scholar] [CrossRef] [PubMed]
  108. Liu, E.; Marin, D.; Banerjee, P.; Macapinlac, H.A.; Thompson, P.; Basar, R.; Nassif Kerbauy, L.; Overman, B.; Thall, P.; Kaplan, M.; et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N. Engl. J. Med. 2020, 382, 545–553. [Google Scholar] [CrossRef]
  109. Dasgupta, S.; Bhattacharya-Chatterjee, M.; O’Malley, B.W., Jr.; Chatterjee, S.K. Inhibition of NK cell activity through TGF-beta 1 by down-regulation of NKG2D in a murine model of head and neck cancer. J. Immunol. 2005, 175, 5541–5550. [Google Scholar] [CrossRef]
  110. Porcelli, S.; Yockey, C.E.; Brenner, M.B.; Balk, S.P. Analysis of T cell antigen receptor (TCR) expression by human peripheral blood CD4-8- alpha/beta T cells demonstrates preferential use of several V beta genes and an invariant TCR alpha chain. J. Exp. Med. 1993, 178, 1–16. [Google Scholar] [CrossRef] [Green Version]
  111. Bendelac, A. CD1: Presenting unusual antigens to unusual T lymphocytes. Science 1995, 269, 185–186. [Google Scholar] [CrossRef]
  112. Halder, R.C.; Aguilera, C.; Maricic, I.; Kumar, V. Type II NKT cell-mediated anergy induction in type I NKT cells prevents inflammatory liver disease. J. Clin. Investig. 2007, 117, 2302–2312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Chang, D.H.; Deng, H.; Matthews, P.; Krasovsky, J.; Ragupathi, G.; Spisek, R.; Mazumder, A.; Vesole, D.H.; Jagannath, S.; Dhodapkar, M.V. Inflammation-associated lysophospholipids as ligands for CD1d-restricted T cells in human cancer. Blood 2008, 112, 1308–1316. [Google Scholar] [CrossRef] [PubMed]
  114. Dhodapkar, M.V.; Geller, M.D.; Chang, D.H.; Shimizu, K.; Fujii, S.; Dhodapkar, K.M.; Krasovsky, J. A reversible defect in natural killer T cell function characterizes the progression of premalignant to malignant multiple myeloma. J. Exp. Med. 2003, 197, 1667–1676. [Google Scholar] [CrossRef] [PubMed]
  115. Rotolo, A.; Caputo, V.S.; Holubova, M.; Baxan, N.; Dubois, O.; Chaudhry, M.S.; Xiao, X.; Goudevenou, K.; Pitcher, D.S.; Petevi, K.; et al. Enhanced Anti-lymphoma Activity of CAR19-iNKT Cells Underpinned by Dual CD19 and CD1d Targeting. Cancer Cell 2018, 34, 596–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Simon, B.; Wiesinger, M.; Marz, J.; Wistuba-Hamprecht, K.; Weide, B.; Schuler-Thurner, B.; Schuler, G.; Dorrie, J.; Uslu, U. The Generation of CAR-Transfected Natural Killer T Cells for the Immunotherapy of Melanoma. Int. J. Mol. Sci. 2018, 19, 2365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Metelitsa, L.S. Anti-tumor potential of type-I NKT cells against CD1d-positive and CD1d-negative tumors in humans. Clin. Immunol. 2011, 140, 119–129. [Google Scholar] [CrossRef] [Green Version]
  118. Vivier, E.; Ugolini, S.; Blaise, D.; Chabannon, C.; Brossay, L. Targeting natural killer cells and natural killer T cells in cancer. Nat. Rev. Immunol. 2012, 12, 239–252. [Google Scholar] [CrossRef]
  119. Olle Hurtado, M.; Wolbert, J.; Fisher, J.; Flutter, B.; Stafford, S.; Barton, J.; Jain, N.; Barone, G.; Majani, Y.; Anderson, J. Tumor infiltrating lymphocytes expanded from pediatric neuroblastoma display heterogeneity of phenotype and function. PLoS ONE 2019, 14, e0216373. [Google Scholar] [CrossRef] [Green Version]
  120. Heczey, A.; Courtney, A.N.; Montalbano, A.; Robinson, S.; Liu, K.; Li, M.; Ghatwai, N.; Dakhova, O.; Liu, B.; Raveh-Sadka, T.; et al. Anti-GD2 CAR-NKT cells in patients with relapsed or refractory neuroblastoma: An interim analysis. Nat. Med. 2020, 26, 1686–1690. [Google Scholar] [CrossRef]
  121. Fisher, J.; Kramer, A.M.; Gustafsson, K.; Anderson, J. Non-V delta 2 gamma delta T lymphocytes as effectors of cancer immunotherapy. Oncoimmunology 2015, 4, e973808. [Google Scholar] [CrossRef] [Green Version]
  122. Fisher, J.P.; Heuijerjans, J.; Yan, M.; Gustafsson, K.; Anderson, J. gammadelta T cells for cancer immunotherapy: A systematic review of clinical trials. Oncoimmunology 2014, 3, e27572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Himoudi, N.; Morgenstern, D.A.; Yan, M.; Vernay, B.; Saraiva, L.; Wu, Y.; Cohen, C.J.; Gustafsson, K.; Anderson, J. Human gammadelta T lymphocytes are licensed for professional antigen presentation by interaction with opsonized target cells. J. Immunol. 2012, 188, 1708–1716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Chien, Y.H.; Meyer, C.; Bonneville, M. gammadelta T cells: First line of defense and beyond. Annu. Rev. Immunol. 2014, 32, 121–155. [Google Scholar] [CrossRef] [PubMed]
  125. Di Lorenzo, B.; Ravens, S.; Silva-Santos, B. High-throughput analysis of the human thymic Vdelta1(+) T cell receptor repertoire. Sci. Data 2019, 6, 115. [Google Scholar] [CrossRef] [Green Version]
  126. Vantourout, P.; Hayday, A. Six-of-the-best: Unique contributions of gammadelta T cells to immunology. Nat. Rev. Immunol. 2013, 13, 88–100. [Google Scholar] [CrossRef] [Green Version]
  127. Fisher, J.P.; Yan, M.; Heuijerjans, J.; Carter, L.; Abolhassani, A.; Frosch, J.; Wallace, R.; Flutter, B.; Capsomidis, A.; Hubank, M.; et al. Neuroblastoma killing properties of Vdelta2 and Vdelta2-negative gammadeltaT cells following expansion by artificial antigen-presenting cells. Clin. Cancer Res. 2014, 20, 5720–5732. [Google Scholar] [CrossRef] [Green Version]
  128. Miyagawa, F.; Tanaka, Y.; Yamashita, S.; Minato, N. Essential requirement of antigen presentation by monocyte lineage cells for the activation of primary human gamma delta T cells by aminobisphosphonate antigen. J. Immunol. 2001, 166, 5508–5514. [Google Scholar] [CrossRef]
  129. Deseke, M.; Prinz, I. Ligand recognition by the gammadelta TCR and discrimination between homeostasis and stress conditions. Cell. Mol. Immunol. 2020, 17, 914–924. [Google Scholar] [CrossRef]
  130. Brandes, M.; Willimann, K.; Moser, B. Professional antigen-presentation function by human gammadelta T Cells. Science 2005, 309, 264–268. [Google Scholar] [CrossRef]
  131. Hamid, O.; Carvajal, R.D. Anti-programmed death-1 and anti-programmed death-ligand 1 antibodies in cancer therapy. Expert Opin. Biol. Ther. 2013, 13, 847–861. [Google Scholar] [CrossRef]
  132. Deniger, D.C.; Switzer, K.; Mi, T.; Maiti, S.; Hurton, L.; Singh, H.; Huls, H.; Olivares, S.; Lee, D.A.; Champlin, R.E.; et al. Bispecific T-cells expressing polyclonal repertoire of endogenous gammadelta T-cell receptors and introduced CD19-specific chimeric antigen receptor. Mol. Ther. 2013, 21, 638–647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Rozenbaum, M.; Meir, A.; Aharony, Y.; Itzhaki, O.; Schachter, J.; Bank, I.; Jacoby, E.; Besser, M.J. Gamma-Delta CAR-T Cells Show CAR-Directed and Independent Activity Against Leukemia. Front. Immunol. 2020, 11, 1347. [Google Scholar] [CrossRef] [PubMed]
  134. Capsomidis, A.; Benthall, G.; Van Acker, H.H.; Fisher, J.; Kramer, A.M.; Abeln, Z.; Majani, Y.; Gileadi, T.; Wallace, R.; Gustafsson, K.; et al. Chimeric Antigen Receptor-Engineered Human Gamma Delta T Cells: Enhanced Cytotoxicity with Retention of Cross Presentation. Mol. Ther. 2018, 26, 354–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Fisher, J.; Abramowski, P.; Wisidagamage Don, N.D.; Flutter, B.; Capsomidis, A.; Cheung, G.W.; Gustafsson, K.; Anderson, J. Avoidance of On-Target Off-Tumor Activation Using a Co-stimulation-Only Chimeric Antigen Receptor. Mol. Ther. 2017, 25, 1234–1247. [Google Scholar] [CrossRef] [Green Version]
  136. Sadelain, M.; Brentjens, R.; Riviere, I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013, 3, 388–398. [Google Scholar] [CrossRef] [Green Version]
  137. Jensen, M.C.; Riddell, S.R. Designing chimeric antigen receptors to effectively and safely target tumors. Curr. Opin. Immunol. 2015, 33, 9–15. [Google Scholar] [CrossRef] [Green Version]
  138. Alabanza, L.; Pegues, M.; Geldres, C.; Shi, V.; Wiltzius, J.J.W.; Sievers, S.A.; Yang, S.; Kochenderfer, J.N. Function of Novel Anti-CD19 Chimeric Antigen Receptors with Human Variable Regions Is Affected by Hinge and Transmembrane Domains. Mol. Ther. 2017, 25, 2452–2465. [Google Scholar] [CrossRef] [Green Version]
  139. Rafiq, S.; Hackett, C.S.; Brentjens, R.J. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat. Rev. Clin. Oncol. 2020, 17, 147–167. [Google Scholar] [CrossRef]
  140. Suck, G.; Odendahl, M.; Nowakowska, P.; Seidl, C.; Wels, W.S.; Klingemann, H.G.; Tonn, T. NK-92: An ‘off-the-shelf therapeutic’ for adoptive natural killer cell-based cancer immunotherapy. Cancer Immunol. Immunother. 2016, 65, 485–492. [Google Scholar] [CrossRef]
  141. Bryceson, Y.T.; Chiang, S.C.; Darmanin, S.; Fauriat, C.; Schlums, H.; Theorell, J.; Wood, S.M. Molecular mechanisms of natural killer cell activation. J. Innate Immun. 2011, 3, 216–226. [Google Scholar] [CrossRef]
  142. Salter, A.I.; Pont, M.J.; Riddell, S.R. Chimeric antigen receptor-modified T cells: CD19 and the road beyond. Blood 2018, 131, 2621–2629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Lo Presti, E.; Pizzolato, G.; Corsale, A.M.; Caccamo, N.; Sireci, G.; Dieli, F.; Meraviglia, S. gammadelta T Cells and Tumor Microenvironment: From Immunosurveillance to Tumor Evasion. Front. Immunol. 2018, 9, 1395. [Google Scholar] [CrossRef] [PubMed]
  144. Mirzaei, H.R.; Mirzaei, H.; Lee, S.Y.; Hadjati, J.; Till, B.G. Prospects for chimeric antigen receptor (CAR) gammadelta T cells: A potential game changer for adoptive T cell cancer immunotherapy. Cancer Lett. 2016, 380, 413–423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Oelsner, S.; Friede, M.E.; Zhang, C.; Wagner, J.; Badura, S.; Bader, P.; Ullrich, E.; Ottmann, O.G.; Klingemann, H.; Tonn, T.; et al. Continuously expanding CAR NK-92 cells display selective cytotoxicity against B-cell leukemia and lymphoma. Cytotherapy 2017, 19, 235–249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Huang, Y.; Zeng, J.; Liu, T.; Xu, Q.; Song, X.; Zeng, J. DNAM1 and 2B4 Costimulatory Domains Enhance the Cytotoxicity of Anti-GPC3 Chimeric Antigen Receptor-Modified Natural Killer Cells Against Hepatocellular Cancer Cells in vitro. Cancer Manag. Res. 2020, 12, 3247–3255. [Google Scholar] [CrossRef]
  147. Fernandez, L.; Metais, J.Y.; Escudero, A.; Vela, M.; Valentin, J.; Vallcorba, I.; Leivas, A.; Torres, J.; Valeri, A.; Patino-Garcia, A.; et al. Memory T Cells Expressing an NKG2D-CAR Efficiently Target Osteosarcoma Cells. Clin. Cancer Res. 2017, 23, 5824–5835. [Google Scholar] [CrossRef] [PubMed]
  148. Ng, Y.Y.; Tay, J.C.K.; Li, Z.; Wang, J.; Zhu, J.; Wang, S. T Cells Expressing NKG2D CAR with a DAP12 Signaling Domain Stimulate Lower Cytokine Production While Effective in Tumor Eradication. Mol. Ther. 2020. [Google Scholar] [CrossRef]
  149. Tan, A.H.J.; Vinanica, N.; Campana, D. Chimeric antigen receptor-T cells with cytokine neutralizing capacity. Blood Adv. 2020, 4, 1419–1431. [Google Scholar] [CrossRef] [Green Version]
  150. Moretta, A.; Biassoni, R.; Bottino, C.; Pende, D.; Vitale, M.; Poggi, A.; Mingari, M.C.; Moretta, L. Major histocompatibility complex class I-specific receptors on human natural killer and T lymphocytes. Immunol. Rev. 1997, 155, 105–117. [Google Scholar] [CrossRef]
  151. Wang, E.; Wang, L.C.; Tsai, C.Y.; Bhoj, V.; Gershenson, Z.; Moon, E.; Newick, K.; Sun, J.; Lo, A.; Baradet, T.; et al. Generation of Potent T-cell Immunotherapy for Cancer Using DAP12-Based, Multichain, Chimeric Immunoreceptors. Cancer Immunol. Res. 2015, 3, 815–826. [Google Scholar] [CrossRef] [Green Version]
  152. Topfer, K.; Cartellieri, M.; Michen, S.; Wiedemuth, R.; Muller, N.; Lindemann, D.; Bachmann, M.; Fussel, M.; Schackert, G.; Temme, A. DAP12-based activating chimeric antigen receptor for NK cell tumor immunotherapy. J. Immunol. 2015, 194, 3201–3212. [Google Scholar] [CrossRef] [PubMed]
  153. Vanichapol, T.; Chutipongtanate, S.; Anurathapan, U.; Hongeng, S. Immune Escape Mechanisms and Future Prospects for Immunotherapy in Neuroblastoma. Biomed. Res. Int. 2018, 2018, 1812535. [Google Scholar] [CrossRef] [PubMed]
  154. Trujillo, J.A.; Sweis, R.F.; Bao, R.; Luke, J.J. T Cell-Inflamed versus Non-T Cell-Inflamed Tumors: A Conceptual Framework for Cancer Immunotherapy Drug Development and Combination Therapy Selection. Cancer Immunol. Res. 2018, 6, 990–1000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. 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] [Green Version]
  156. Whilding, L.M.; Halim, L.; Draper, B.; Parente-Pereira, A.C.; Zabinski, T.; Davies, D.M.; Maher, J. CAR T-Cells Targeting the Integrin alphavbeta6 and Co-Expressing the Chemokine Receptor CXCR2 Demonstrate Enhanced Homing and Efficacy against Several Solid Malignancies. Cancers 2019, 11, 674. [Google Scholar] [CrossRef] [Green Version]
  157. Liu, G.; Rui, W.; Zheng, H.; Huang, D.; Yu, F.; Zhang, Y.; Dong, J.; Zhao, X.; Lin, X. CXCR2-modified CAR-T cells have enhanced trafficking ability that improves treatment of hepatocellular carcinoma. Eur. J. Immunol. 2020, 50, 712–724. [Google Scholar] [CrossRef]
  158. Adachi, K.; Kano, Y.; Nagai, T.; Okuyama, N.; Sakoda, Y.; Tamada, K. IL-7 and CCL19 expression in CAR-T cells improves immune cell infiltration and CAR-T cell survival in the tumor. Nat. Biotechnol. 2018, 36, 346–351. [Google Scholar] [CrossRef]
  159. Liu, L.B.; Xie, F.; Chang, K.K.; Li, M.Q.; Meng, Y.H.; Wang, X.H.; Li, H.; Li, D.J.; Yu, J.J. Hypoxia promotes the proliferation of cervical carcinoma cells through stimulating the secretion of IL-8. Int. J. Clin. Exp. Pathol. 2014, 7, 575–583. [Google Scholar]
  160. Kim, E.S.; Serur, A.; Huang, J.; Manley, C.A.; McCrudden, K.W.; Frischer, J.S.; Soffer, S.Z.; Ring, L.; New, T.; Zabski, S.; et al. Potent VEGF blockade causes regression of coopted vessels in a model of neuroblastoma. Proc. Natl. Acad. Sci. USA 2002, 99, 11399–11404. [Google Scholar] [CrossRef] [Green Version]
  161. Ara, T.; Song, L.; Shimada, H.; Keshelava, N.; Russell, H.V.; Metelitsa, L.S.; Groshen, S.G.; Seeger, R.C.; DeClerck, Y.A. Interleukin-6 in the bone marrow microenvironment promotes the growth and survival of neuroblastoma cells. Cancer Res. 2009, 69, 329–337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Brignole, C.; Marimpietri, D.; Pastorino, F.; Di Paolo, D.; Pagnan, G.; Loi, M.; Piccardi, F.; Cilli, M.; Tradori-Cappai, A.; Arrigoni, G.; et al. Anti-IL-10R antibody improves the therapeutic efficacy of targeted liposomal oligonucleotides. J. Control. Release 2009, 138, 122–127. [Google Scholar] [CrossRef]
  163. Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef]
  164. Marvel, D.; Gabrilovich, D.I. Myeloid-derived suppressor cells in the tumor microenvironment: Expect the unexpected. J. Clin. Investig. 2015, 125, 3356–3364. [Google Scholar] [CrossRef] [PubMed]
  165. Diaz-Montero, C.M.; Salem, M.L.; Nishimura, M.I.; Garrett-Mayer, E.; Cole, D.J.; Montero, A.J. Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin-cyclophosphamide chemotherapy. Cancer Immunol. Immunother. 2009, 58, 49–59. [Google Scholar] [CrossRef] [Green Version]
  166. Noguchi, T.; Ward, J.P.; Gubin, M.M.; Arthur, C.D.; Lee, S.H.; Hundal, J.; Selby, M.J.; Graziano, R.F.; Mardis, E.R.; Korman, A.J.; et al. Temporally Distinct PD-L1 Expression by Tumor and Host Cells Contributes to Immune Escape. Cancer Immunol. Res. 2017, 5, 106–117. [Google Scholar] [CrossRef] [Green Version]
  167. Whittle, S.B.; Smith, V.; Doherty, E.; Zhao, S.; McCarty, S.; Zage, P.E. Overview and recent advances in the treatment of neuroblastoma. Expert. Rev. Anticancer Ther. 2017, 17, 369–386. [Google Scholar] [CrossRef] [Green Version]
  168. Pinto, N.R.; Applebaum, M.A.; Volchenboum, S.L.; Matthay, K.K.; London, W.B.; Ambros, P.F.; Nakagawara, A.; Berthold, F.; Schleiermacher, G.; Park, J.R.; et al. Advances in Risk Classification and Treatment Strategies for Neuroblastoma. J. Clin. Oncol. 2015, 33, 3008–3017. [Google Scholar] [CrossRef]
  169. Hui, E.; Cheung, J.; Zhu, J.; Su, X.; Taylor, M.J.; Wallweber, H.A.; Sasmal, D.K.; Huang, J.; Kim, J.M.; Mellman, I.; et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 2017, 355, 1428–1433. [Google Scholar] [CrossRef]
  170. Thery, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [Green Version]
  171. Antonyak, M.A.; Cerione, R.A. Microvesicles as mediators of intercellular communication in cancer. Methods Mol. Biol. 2014, 1165, 147–173. [Google Scholar] [CrossRef] [PubMed]
  172. Blavier, L.; Yang, R.M.; DeClerck, Y.A. The Tumor Microenvironment in Neuroblastoma: New Players, New Mechanisms of Interaction and New Perspectives. Cancers 2020, 12, 2912. [Google Scholar] [CrossRef] [PubMed]
  173. Becker, A.; Thakur, B.K.; Weiss, J.M.; Kim, H.S.; Peinado, H.; Lyden, D. Extracellular Vesicles in Cancer: Cell-to-Cell Mediators of Metastasis. Cancer Cell 2016, 30, 836–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Fonseka, P.; Liem, M.; Ozcitti, C.; Adda, C.G.; Ang, C.S.; Mathivanan, S. Exosomes from N-Myc amplified neuroblastoma cells induce migration and confer chemoresistance to non-N-Myc amplified cells: Implications of intra-tumour heterogeneity. J. Extracell Vesicles 2019, 8, 1597614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Wortzel, I.; Dror, S.; Kenific, C.M.; Lyden, D. Exosome-Mediated Metastasis: Communication from a Distance. Dev. Cell 2019, 49, 347–360. [Google Scholar] [CrossRef]
  176. Ali, S.; Toews, K.; Schwiebert, S.; Klaus, A.; Winkler, A.; Grunewald, L.; Oevermann, L.; Deubzer, H.E.; Tuns, A.; Jensen, M.C.; et al. Tumor-Derived Extracellular Vesicles Impair CD171-Specific CD4(+) CAR T Cell Efficacy. Front. Immunol. 2020, 11, 531. [Google Scholar] [CrossRef]
  177. Peinado, H.; Aleckovic, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; Garcia-Santos, G.; Ghajar, C.; et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 2012, 18, 883–891. [Google Scholar] [CrossRef] [Green Version]
  178. Costa-Silva, B.; Aiello, N.M.; Ocean, A.J.; Singh, S.; Zhang, H.; Thakur, B.K.; Becker, A.; Hoshino, A.; Mark, M.T.; Molina, H.; et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 2015, 17, 816–826. [Google Scholar] [CrossRef]
  179. Ji, Q.; Zhou, L.; Sui, H.; Yang, L.; Wu, X.; Song, Q.; Jia, R.; Li, R.; Sun, J.; Wang, Z.; et al. Primary tumors release ITGBL1-rich extracellular vesicles to promote distal metastatic tumor growth through fibroblast-niche formation. Nat. Commun. 2020, 11, 1211. [Google Scholar] [CrossRef] [Green Version]
  180. Nakata, R.; Shimada, H.; Fernandez, G.E.; Fanter, R.; Fabbri, M.; Malvar, J.; Zimmermann, P.; DeClerck, Y.A. Contribution of neuroblastoma-derived exosomes to the production of pro-tumorigenic signals by bone marrow mesenchymal stromal cells. J. Extracell Vesicles 2017, 6, 1332941. [Google Scholar] [CrossRef]
  181. Challagundla, K.B.; Wise, P.M.; Neviani, P.; Chava, H.; Murtadha, M.; Xu, T.; Kennedy, R.; Ivan, C.; Zhang, X.; Vannini, I.; et al. Exosome-mediated transfer of microRNAs within the tumor microenvironment and neuroblastoma resistance to chemotherapy. J. Natl. Cancer Inst. 2015, 107. [Google Scholar] [CrossRef] [Green Version]
  182. Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Yokosuka, T.; Takamatsu, M.; Kobayashi-Imanishi, W.; Hashimoto-Tane, A.; Azuma, M.; Saito, T. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J. Exp. Med. 2012, 209, 1201–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Zuo, S.; Sho, M.; Sawai, T.; Kanehiro, H.; Maeda, K.; Yoshida, M.; Tsukada, R.; Nomura, M.; Okuyama, H. Potential role of the PD-L1 expression and tumor-infiltrating lymphocytes on neuroblastoma. Pediatr. Surg. Int. 2020, 36, 137–143. [Google Scholar] [CrossRef] [PubMed]
  185. Dondero, A.; Pastorino, F.; Della Chiesa, M.; Corrias, M.V.; Morandi, F.; Pistoia, V.; Olive, D.; Bellora, F.; Locatelli, F.; Castellano, A.; et al. PD-L1 expression in metastatic neuroblastoma as an additional mechanism for limiting immune surveillance. Oncoimmunology 2016, 5, e1064578. [Google Scholar] [CrossRef] [Green Version]
  186. Sharma, P.; Allison, J.P. The future of immune checkpoint therapy. Science 2015, 348, 56–61. [Google Scholar] [CrossRef]
  187. Taube, J.M.; Klein, A.; Brahmer, J.R.; Xu, H.; Pan, X.; Kim, J.H.; Chen, L.; Pardoll, D.M.; Topalian, S.L.; Anders, R.A. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin. Cancer Res. 2014, 20, 5064–5074. [Google Scholar] [CrossRef] [Green Version]
  188. Li, S.; Siriwon, N.; Zhang, X.; Yang, S.; Jin, T.; He, F.; Kim, Y.J.; Mac, J.; Lu, Z.; Wang, S.; et al. Enhanced Cancer Immunotherapy by Chimeric Antigen Receptor-Modified T Cells Engineered to Secrete Checkpoint Inhibitors. Clin. Cancer Res. 2017, 23, 6982–6992. [Google Scholar] [CrossRef] [Green Version]
  189. Rafiq, S.; Yeku, O.O.; Jackson, H.J.; Purdon, T.J.; van Leeuwen, D.G.; Drakes, D.J.; Song, M.; Miele, M.M.; Li, Z.; Wang, P.; et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat. Biotechnol. 2018, 36, 847–856. [Google Scholar] [CrossRef]
  190. Rupp, L.J.; Schumann, K.; Roybal, K.T.; Gate, R.E.; Ye, C.J.; Lim, W.A.; Marson, A. CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells. Sci. Rep. 2017, 7, 737. [Google Scholar] [CrossRef]
  191. Qin, L.; Zhao, R.; Chen, D.; Wei, X.; Wu, Q.; Long, Y.; Jiang, Z.; Li, Y.; Wu, H.; Zhang, X.; et al. Chimeric antigen receptor T cells targeting PD-L1 suppress tumor growth. Biomark. Res. 2020, 8, 19. [Google Scholar] [CrossRef]
  192. Gargett, T.; Yu, W.; Dotti, G.; Yvon, E.S.; Christo, S.N.; Hayball, J.D.; Lewis, I.D.; Brenner, M.K.; Brown, M.P. GD2-specific CAR T Cells Undergo Potent Activation and Deletion Following Antigen Encounter but can be Protected From Activation-induced Cell Death by PD-1 Blockade. Mol. Ther. 2016, 24, 1135–1149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Toews, K.; Grunewald, L.; Schwiebert, S.; Klaus, A.; Winkler, A.; Ali, S.; Zirngibl, F.; Astrahantseff, K.; Wagner, D.L.; Henssen, A.G.; et al. Central memory phenotype drives success of checkpoint inhibition in combination with CAR T cells. Mol. Carcinog. 2020, 59, 724–735. [Google Scholar] [CrossRef] [PubMed]
  194. Jabbari, P.; Hanaei, S.; Rezaei, N. State of the art in immunotherapy of neuroblastoma. Immunotherapy 2019, 11, 831–850. [Google Scholar] [CrossRef] [PubMed]
  195. Sanmamed, M.F.; Carranza-Rua, O.; Alfaro, C.; Onate, C.; Martin-Algarra, S.; Perez, G.; Landazuri, S.F.; Gonzalez, A.; Gross, S.; Rodriguez, I.; et al. Serum interleukin-8 reflects tumor burden and treatment response across malignancies of multiple tissue origins. Clin. Cancer Res. 2014, 20, 5697–5707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Kuwada, Y.; Sasaki, T.; Morinaka, K.; Kitadai, Y.; Mukaida, N.; Chayama, K. Potential involvement of IL-8 and its receptors in the invasiveness of pancreatic cancer cells. Int. J. Oncol. 2003, 22, 765–771. [Google Scholar] [CrossRef]
  197. Chen, Y.; Sun, C.; Landoni, E.; Metelitsa, L.; Dotti, G.; Savoldo, B. Eradication of Neuroblastoma by T Cells Redirected with an Optimized GD2-Specific Chimeric Antigen Receptor and Interleukin-15. Clin. Cancer Res. 2019, 25, 2915–2924. [Google Scholar] [CrossRef] [Green Version]
  198. Alvarez-Rueda, N.; Desselle, A.; Cochonneau, D.; Chaumette, T.; Clemenceau, B.; Leprieur, S.; Bougras, G.; Supiot, S.; Mussini, J.M.; Barbet, J.; et al. A monoclonal antibody to O-acetyl-GD2 ganglioside and not to GD2 shows potent anti-tumor activity without peripheral nervous system cross-reactivity. PLoS ONE 2011, 6, e25220. [Google Scholar] [CrossRef] [Green Version]
  199. Kramer, K.; Khan, O.; Haque, S. Central Nervous System Neuroblastoma Metastases Pseudoprogression Following Intraventricular Anti-B7h3 Radioimmunotherapy. Neuro-Oncology 2019, 21, 95. [Google Scholar] [CrossRef]
  200. Yang, L.H.; Ma, X.L.; Liu, Y.C.; Zhao, W.; Yu, L.H.; Qin, M.Q.; Zhu, G.H.; Wang, K.; Shi, X.D.; Zhang, Z.X.; et al. Chimeric Antigen Receptor 4SCAR-GD2-Modified T Cells Targeting High-Risk and Recurrent Neuroblastoma: A Phase II Multi-Center Trial in China. Blood 2017, 130. [Google Scholar] [CrossRef]
  201. Zhao, W.; Dong, L.; Wang, H.; Zheng, X.; Zhang, C.; Wang, K.; Zhu, G.; Wang, B.; Qin, M.; Liu, Y.; et al. Chimeric Antigen Receptor 4SCAR-GD2/CD56-Modified T Cells in the Refractory and Recurrent Paediatric Solid Tumors. Pediatric Blood Cancer 2016, 63, S38. [Google Scholar]
Figure 1. Cumulative publications of chimeric antigen receptor (CAR) immunotherapy in clinical trials.
Figure 1. Cumulative publications of chimeric antigen receptor (CAR) immunotherapy in clinical trials.
Vaccines 08 00753 g001
Figure 2. Target antigens conducted on the safety and efficacy of CAR therapy for neuroblastoma. Six surface antigens of neuroblastoma, including L1-CAM, GPC2, NCAM, GD2, ALK, and B7H3, are under development and investigation. L1-CAM and GD2 are the only two target antigens currently in completed clinical trials for neuroblastoma (labeled with star).
Figure 2. Target antigens conducted on the safety and efficacy of CAR therapy for neuroblastoma. Six surface antigens of neuroblastoma, including L1-CAM, GPC2, NCAM, GD2, ALK, and B7H3, are under development and investigation. L1-CAM and GD2 are the only two target antigens currently in completed clinical trials for neuroblastoma (labeled with star).
Vaccines 08 00753 g002
Figure 3. Proposed strategies to improve CARs’ efficacy against neuroblastoma. To enhance the antitumor toxicity of chimeric antigen receptor (CAR) T cells, several strategies can be applied: (a) The use of extracellular vesicle (EV) inhibitors. EVs are cargo ships containing functional molecules that enhance tumor growth and metastasis and may decoy CAR immunotherapy. Inhibiting the secretion of EVs from tumor cells may improve the killing efficiency CAR T treatment. (b) Engineering CAR T cells with a suicide gene. To improve CARs, and any side effects of CARs, fusing the iCaspase-9 gene in CARs’ structure can break T cell activation via induction of apoptosis in CAR T cells; (c) Engineering CAR T cells secreting cytokine to support function. IL-15 cytokines showed an ability to increase CARs’ antitumor toxicity in both T and natural killer T (NKT) cells. (d) Use of T cell subsets with greater proliferative ability. Naïve and central memory effector cells have the potential to enhance both CARs’ efficacy and the persistency of immune effector cells. (e) Use of different effector cells with greater function. For various solid tumors, CARs derived from natural killer (NK), NKT, or γδT cells showed more penetration into tumor sites and more potent toxicity than CAR T cells. (f) Engineering CAR T cells to overcome the tumor microenvironment (TME) and increase penetration. Supplementing with tumor-enriched chemokine inhibitor or using extracellular matrix remodeling enzyme during treatment might improve the trafficking and infiltration of CAR T cells; (g) Finally, engineering CAR NK cells to support the function of CAR T cells. Inhibition of immunosuppressive cell function in the TME via CAR NK cells can enhance the homing and efficacy of neuroblastoma-targeted CAR T cells.
Figure 3. Proposed strategies to improve CARs’ efficacy against neuroblastoma. To enhance the antitumor toxicity of chimeric antigen receptor (CAR) T cells, several strategies can be applied: (a) The use of extracellular vesicle (EV) inhibitors. EVs are cargo ships containing functional molecules that enhance tumor growth and metastasis and may decoy CAR immunotherapy. Inhibiting the secretion of EVs from tumor cells may improve the killing efficiency CAR T treatment. (b) Engineering CAR T cells with a suicide gene. To improve CARs, and any side effects of CARs, fusing the iCaspase-9 gene in CARs’ structure can break T cell activation via induction of apoptosis in CAR T cells; (c) Engineering CAR T cells secreting cytokine to support function. IL-15 cytokines showed an ability to increase CARs’ antitumor toxicity in both T and natural killer T (NKT) cells. (d) Use of T cell subsets with greater proliferative ability. Naïve and central memory effector cells have the potential to enhance both CARs’ efficacy and the persistency of immune effector cells. (e) Use of different effector cells with greater function. For various solid tumors, CARs derived from natural killer (NK), NKT, or γδT cells showed more penetration into tumor sites and more potent toxicity than CAR T cells. (f) Engineering CAR T cells to overcome the tumor microenvironment (TME) and increase penetration. Supplementing with tumor-enriched chemokine inhibitor or using extracellular matrix remodeling enzyme during treatment might improve the trafficking and infiltration of CAR T cells; (g) Finally, engineering CAR NK cells to support the function of CAR T cells. Inhibition of immunosuppressive cell function in the TME via CAR NK cells can enhance the homing and efficacy of neuroblastoma-targeted CAR T cells.
Vaccines 08 00753 g003
Table 1. An outline of clinical trials for CAR immunotherapy in neuroblastoma.
Table 1. An outline of clinical trials for CAR immunotherapy in neuroblastoma.
Clinicaltrials.Gov
Identifier
Type of StudyStatusTitleInterventionsTarget/ScFv/Signaling DomainDateKey ResultReference
N/APhase I
N = 6
CompletedN/AAnti- L1-CAM CAR T cellsL1-CAM/CE7R/
CD3ζ
N/APartial response in one patient[12]
NCT02761915Phase I
N = 27
RecruitingA UK cancer research trial of anti-GD2 T cells (1RG-CART)Anti-GD2 CAR
T cells
GD2/KM8138/
CD28.CD3ζ
Study start:
February 2016
Primary completion:
February 2023
On target activity in bone and bone marrow were detected[47]
NCT01822652Phase I
N = 11
Active, not
recruiting
Third generation GD-2 chimeric antigen receptor and icaspase suicide safety switch,
neuroblastoma, GRAIN
iC9-GD2 CAR
T cells
GD2/14g2a/
CD28.OX40.CD3ζ
Study start:
August 2013
Primary completion:
December 2015
Modest early antitumor responses were detected[49]
NCT00085930Phase I
N = 19
Active, not
recruiting
Blood T-cells and EBV specific CTLs expressing GD-2 specific chimeric T Cell receptors to
neuroblastoma patients
EBV-specific CTLsGD2/14g2a/
CD3ζ
Study start:
April 2003
Primary completion:
January 2010
Complete response in one patient[18,50]
NCT01460901Phase I
N = 5
CompletedStudy of donor-derived, multi-virus-specific, cytotoxic T-lymphocytes for relapsed/refractory neuroblastoma (STALLONe)GD2 CAR modified Tri-virus-specific cytotoxic t-cellsGD2/14g2a/CD3ζStudy start:
October 2012
Primary completion:
January 2015
Partial response in 30% of patients[51]
NCT03294954Phase I
N = 24
RecruitingGD2-specific CAR and interleukin-15 expressing autologous NKT cells to treat children with neuroblastoma (GINAKIT2)GINAKIT CellsGD2/14g2a/
CD28.CD3ζ
Study start:
18 January 2018
Primary completion:
1 September 2021
N/A[52]
NCT02765243Phase II
N = 34
RecruitingAnti-GD2 fourth generation CART cells targeting refractory and/
or recurrent neuroblastoma
Anti-GD2 CAR T cellsGD2/N/A/CD28.4-1BB.CD27.CD3ζStudy start:
23 March 2016
Primary completion:
12 May 2019
Partial response in 15% of patients[53]
NCT03618381Phase I
N = 36
RecruitingEGFR806 CAR T cell immunotherapy for recurrent/refractory solid tumors in children and young adultsAnti-EGFR806-EGFRt
CAR T cells
EGFR/N/A/4-1BB.CD3ζStudy start:
18 June 2019
Primary completion:
June 2021
N/A[54]
NCT01953900Phase I
N = 26
Active, not
recruiting
iC9-GD2-CAR-VZV-CTLs/refractory or metastatic GD2-positive sarcoma and neuroblastomaAnti-GD2 CAR T cellsGD2/14g2a/
CD28.OX40.CD3ζ
Study start:
April 2014
Primary completion:
April 2021
N/A[54,55]
NCT03635632Phase I
N = 94
RecruitingC7R-GD2.CAR T cells for patients with relapsed or refractory neuroblastoma and other GD2 Positive cancers (GAIL-N)C7R-GD2 CAR T cellsGD2/14g2a/
CD28.OX40.CD3ζ
Study start:
23 April 2019
Primary completion:
June 2022
N/AUnpublished
NCT03373097Phase I/II
N = 42
RecruitingAnti-GD2 CAR T cells in pediatric patients affected by high risk and/or relapsed/refractory neuroblastoma or other GD2-positive solid tumorsAnti-GD2 CAR T cellsGD2/14g2a/
CD28.4-1BB.CD3ζ
Study start:
January 2018
Primary completion:
December 2024
N/AUnpublished
NCT03721068Phase I
N = 18
RecruitingStudy of CAR T cells targeting the GD2 with IL-15+icaspase9 for relapsed/refractory neuroblastomaiC9-GD2 CAR T cellsGD2/N/AStudy start:
19 February 2019
Primary completion:
19 June 2024
N/AUnpublished
NCT04637503Phase I/II
N = 100
Recruiting4SCAR-T therapy targeting GD2, PSMA and CD276 for treating neuroblastoma4SCAR T cellGD2, PSMA, CD276/N/AStudy start:
18 November 2020
Primary completion:
19 November 2020
N/AUnpublished
NCT04539366Phase I
N = 67
Not yet recruitingTesting a new immune cell therapy, GD2-targeted modified T-cells (GD2CART), in children, adolescents, and young adults with relapsed/refractory osteosarcoma and neuroblastoma, the GD2-CAR persistence trialAnti-GD2 CAR
T cells
GD2/N/A/
CD28.OX40.CD3ζ
Study start:
1 January 2021
Primary completion:
1 August 2024
N/AUnpublished
NCT04483778Phase I
N = 68
RecruitingB7H3 CAR T cell immunotherapy for recurrent/refractory solid tumors in children and young adultsAnti B7H3-EGFRt-DHFR CAR T cellsB7H3/N/A/4-1BB.CD3ζStudy start:
13 July 2020
Primary completion:
December 2025
N/AUnpublished
NCT02107963Phase I
N = 15
CompletedA Phase I trial of T cells expressing an anti-GD2 chimeric antigen receptor in children and young adults with GD2+ solid tumorsAnti-GD2 CAR
T cells
GD2/14g2a/OX40.CD28.CD3ζStudy start:
28 February 2014
Primary completion:
15 August 2016
N/AUnpublished
Table 2. Comparison of CAR-derived effector cells for neuroblastoma.
Table 2. Comparison of CAR-derived effector cells for neuroblastoma.
Effector CellEx vivo ExpansionType of Target CellsCAR DesignCombination
LocalizedCTCCostimulatingSignalingCytokineEffector CellSignaling Inhibitor
T*******Dim expression target: CD28
High expression target: 4-1BB
Normally: CD3ζ Decrease CRS: DAP12IL-2, IL-7, IL-15NKG2D.CAR NKanti-PD-1/PD-L1, anti-IL-10, IL-6 inhibitor
NK******CD28, 4-1BB, DNAM1 and 2B4CD3ζ and DAP12IL-2, IL-7, IL-21NB-targeted CAR Tanti-PD-1/PD-L1, anti-TGFβ, anti-IL-10, IL-6 inhibitor
NKT******CD28CD3ζ and DAP12IL-2, IL-7, IL-15N/Aanti-PD-1/PD-L1, anti-IL-10, IL-6 inhibitor
γδT*******Both CD28 and 4-1BBCD3ζ [no data for DAP12]IL-2, IL-7, IL-15N/Aanti-PD-1/PD-L1, anti-IL-10, IL-6 inhibitor
Score: *** = high, ** = moderate, * = low. CAR, chimeric antigen receptor; CTC, circulating tumor cells; DAP12, DNAX-activating protein of 12 kDa; DNAM1, DNAX accessory Molecule-1; DNAX-activating protein 12; ECM, extracellular matrix; IL, interleukin; N/A, not applicable; NB, neuroblastoma; NK, natural killer cell; NKG2D, natural killer cell receptor D; NKT, natural killer T lymphocyte cell; PD-1, programmed cell death-1; PD-L1, programmed cell death-ligand 1; T, T lymphocyte; γδT, gamma delta T lymphocyte.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sawaisorn, P.; Atjanasuppat, K.; Anurathapan, U.; Chutipongtanate, S.; Hongeng, S. Strategies to Improve Chimeric Antigen Receptor Therapies for Neuroblastoma. Vaccines 2020, 8, 753. https://doi.org/10.3390/vaccines8040753

AMA Style

Sawaisorn P, Atjanasuppat K, Anurathapan U, Chutipongtanate S, Hongeng S. Strategies to Improve Chimeric Antigen Receptor Therapies for Neuroblastoma. Vaccines. 2020; 8(4):753. https://doi.org/10.3390/vaccines8040753

Chicago/Turabian Style

Sawaisorn, Piamsiri, Korakot Atjanasuppat, Usanarat Anurathapan, Somchai Chutipongtanate, and Suradej Hongeng. 2020. "Strategies to Improve Chimeric Antigen Receptor Therapies for Neuroblastoma" Vaccines 8, no. 4: 753. https://doi.org/10.3390/vaccines8040753

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