Next Article in Journal
Flattening the Curve of COVID-19 Vaccine Rejection—An International Overview
Next Article in Special Issue
Current Advances of Nitric Oxide in Cancer and Anticancer Therapeutics
Previous Article in Journal
High Rates of COVID-19 Vaccine Hesitancy and Its Association with Conspiracy Beliefs: A Study in Jordan and Kuwait among Other Arab Countries
Previous Article in Special Issue
The Potential of Soluble Human Leukocyte Antigen Molecules for Early Cancer Detection and Therapeutic Vaccine Design
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunotherapeutic Strategies for Neuroblastoma: Present, Past and Future

Laboratorio Cellule Staminali Post-Natali e Terapie Cellulari, Istituto Giannina Gaslini (Istituto di Ricerca e Cura a Carattere Scientifico—IRCCS), Via G. Gaslini 5, 16147 Genova, Italy
*
Author to whom correspondence should be addressed.
Vaccines 2021, 9(1), 43; https://doi.org/10.3390/vaccines9010043
Submission received: 30 November 2020 / Revised: 7 January 2021 / Accepted: 10 January 2021 / Published: 13 January 2021
(This article belongs to the Special Issue Cancer Immunotherapy: Advances and Future Prospects)

Abstract

:
Neuroblastoma is the most common extracranial pediatric solid tumor with a heterogeneous clinical course, ranging from spontaneous regression to metastatic disease and death, irrespective of intensive chemotherapeutic regimen. On the basis of several parameters, children affected by neuroblastoma are stratified into low, intermediate and high risk. At present, more than 50% of high-risk patients with metastatic spread display an overall poor long-term outcome also complicated by devastating long-term morbidities. Thus, novel and more effective therapies are desperately needed to improve lifespan of high-risk patients. In this regard, adoptive cell therapy holds great promise and several clinical trials are ongoing, demonstrating safety and tolerability, with no toxicities. Starting from the immunological and clinical features of neuroblastoma, we here discuss the immunotherapeutic approaches currently adopted for high-risk patients and different innovative therapeutic strategies currently under investigation. The latter are based on the infusion of natural killer (NK) cells, as support of consolidation therapy in addition to standard treatments, or chimeric antigen receptor (CAR) T cells directed against neuroblastoma associated antigens (e.g., disialoganglioside GD2). Finally, future perspectives of adoptive cell therapies represented by γδ T lymphocyes and CAR NK cells are envisaged.

Graphical Abstract

1. General Features of Neuroblastoma

Neuroblastoma (NB) is the most widespread extracranial solid tumor in childhood [1,2] and the most common malignancy in the first year of life. The median age at diagnosis is 18 months and 90% of cases are diagnosed before the patient is 10 years of age [3]. It arises from the sympathetic nervous system, mainly from the adrenal medulla [4] and, in most cases, affects the adrenal glands or sympathetic ganglia [5]. At clinical presentation, NB is quite heterogeneous, ranging from asymptomatic tumors to diffuse metastases with systemic manifestations, thus reflecting on differences in outcome of patients that may evolve into spontaneous regression as well as into unfavorable progression, metastasis and death, irrespective of the intensive therapies adopted [6]. Metastases are present at diagnosis in about 50% of patients and mainly involve bone marrow (BM), bone and regional lymph nodes, while involvement of the central nervous system and lungs is rare, being present in less than 5% of metastatic patients at diagnosis [7,8]. In addition, extensive liver involvement may be observed in infants and causes liver disease, renal and lung dysfunction as a consequence of abdominal distention.
Diagnosis is based on a combination of laboratory tests, radiographic imaging and pathology, but many additional biological factors may be helpful to predict the clinical behavior in NB, including histologic and cytogenetic features, as well as molecular changes, in particular the amplification of the MYCN oncogene [9,10].
Remarkable efforts have been done by the International Neuroblastoma Risk Group (INRG) with the help of international groups, i.e., the Children’s Oncology Group and the International Society of Paediatric Oncology European Neuroblastoma, that created a cooperative task force in order to identify homogeneous risk groups before any treatment [11]. The extent of disease was determined by the presence or absence of image defined risk factors and/or metastatic disease at the time of diagnosis, defining disease stages as local (L1 and L2) or metastatic (M and MS). Furthermore, risk stratifications were defined including not only the stage, but different aspects of tumor biology [12] (Table 1).
The INRG collected data from over 8000 patients thanks to cooperation with different groups in North America, Europe and Japan and, when available, explored 35 different potential risk factors including prognostic factors such as age at diagnosis, pathology and genomic characterization (e.g., MYCN amplification and 11q status, cell ploidy and segmental chromosomal abnormalities), comparing these features to event-free and overall survival. Such efforts were of particular relevance since the precise risk stratification of patients were needed to guide therapy, improve the outcome for high-risk patients by intensification or changing treatment, and modify appropriately the chemotherapy for lower risk patients, with the aim of minimizing toxicity and late effects.
Thus, the INRG classified patients as low, intermediate or high risk: for the low and intermediate risk patients high overall survival greater than 90% has been achieved, while minimizing therapy [13,14,15]. By contrast, the high-risk patients show overall poor long-term outcome also complicated by devastating long-term morbidities, indicating that this group is specifically associated with chemo-resistance. The overall survival of high risk patients has improved over the past 20 years, from 29% for patients diagnosed from 1990 to 1994 to 50% for patients diagnosed from 2005 to 2010 [16,17]. Such results were presumably due to the intensification of therapy, myeloablation and immunotherapy, but prognosis of these patients still remains unsatisfactory. Nonetheless, patients with refractory or relapsed NB can rarely be cured and for this reason novel efficacious therapies are urgently needed.

2. Conventional Therapies for High Risk Patients

High-risk patients require intensive and complex therapies that include (i) the induction phase with multiple cycles of chemotherapy before surgery, (ii) a consolidation phase which may include myeloablation and autologous hematopoietic stem cell transplantation, local radiation and anti-disialoganglioside GD2 antibodies (Ab) and (iii) a maintenance phase with immunotherapy and/or differentiation agents [2].
The most widely used conventional cytotoxic chemotherapies are topotecan with either cyclophosphamide or temozolomide [18] or irinotecan and temozolomide [19,20,21] that may offer partial or even complete response with improvement in symptoms and quality of life, especially for low or intermediate risk patients.
At the end or soon after the end of induction chemotherapy, a surgical resection of the tumor mass, when possible, is applied in order to eliminate the remaining primary tumor.
Concerning the consolidation phase, it has been reported that myeloablation may significantly improve the outcome [22,23,24]. Although the autologous hematopoietic stem cell transplantation is commonly used, only marginal effects on event-free survival have been obtained and for this reason the optimal conditioning regimen is still under investigation. In this regard, long-term cures have been achieved by induction and stem-cell transplantation followed by anti-GD2 Ab therapy [25]. Alternatively, radiation therapy can be used locally.
The maintenance phase is generally composed of a combination of anti-GD2 Ab (that will be discussed in the following paragraph) and isotretinoin, known for its ability to induce differentiation and death in tumor cells, finally improving event-free survival in a randomized trial [23]. A phase III clinical trial is still active to test the side effects and efficacy of treating patients with NB (NCT01041638).
Another therapy is represented by the use of metaiodobenzylguanidine (MIBG), based on the finding that 90% of NB tumors express the norepinephrine transporter and therefore take up the sympathomimetic MIBG [26]. Clinical trials conducted in relapsed or refractory high-risk NB patients, using a high dose of 131I-MIBG as monotherapy or in combination with other agents, demonstrated a 30–40% response rate [27,28,29].

3. Immunological Features of NB

Similarly to other pediatric solid tumors, NB shows intrinsic immunological properties that render often ineffective therapies based on recruitment of cellular components of the immune system or the reactivation of them [30]. NB is poorly immunogenic as witnessed by (i) low expression of HLA class I molecules, (ii) defects in antigen-processing machinery, (iii) down-regulation of molecules that activate T and natural killer (NK) cells (i.e., PD-1L, NKG2DL, MIC-A and –B, ULBP-1, ULBP-2, ULBP-3 and DNAM-1 ligand), (iv) membrane expression and release of soluble HLA-G and –E, (v) release of exosomes containing immune-suppressive molecules (i.e., B7-H3) and (vi) secretion of immunomodulatory cytokines (e.g., IL-10 and TGF-β1) [31,32,33,34,35,36].
To complicate this picture, the tumor microenvironment is hypoxic thus contributing to a metabolic challenge for tumor cells themselves [37,38,39] and for infiltrating immune cells, leading to immune-suppression. The latter finding is supported by infiltration of tumor-associated macrophages (TAM) that are effective in paralyzing T cell responses, inducing T cell apoptosis through Fas–Fas ligand interactions and activating myeloid derived suppressor cells and regulatory T cells that, in turn, suppress active immune response [40,41,42]. Furthermore, NB cells express constitutively high levels of gangliosides which further contribute to the immune suppressive microenvironment. Of these, GD2 is one of the most studied surface antigens in NB also used for target therapies [43,44].

4. Immunotherapeutic Approaches for Neuroblastoma

4.1. Antibodies Targeting NB in Clinical Settings: the GD2 Disialoganglioside Prototype and Other Tumor-Associated Antigens

GD2 is an oncofetal antigen expressed during fetal development. It remains expressed after birth in neurons, peripheral nerves and skin melanocytes and it is consistently found in NB and osteosarcoma [43]. In these tumors, specific Ab targeting GD2 have been developed and used in clinical settings with encouraging results [25,45,46].
The effectiveness of GD2 Ab is related to different mechanisms as (i) direct induction of cell death, (ii) Fcγ receptor (FcγR)–mediated antibody-dependent cell-mediated cytotoxicity (ADCC) by immune cells such as NK, macrophages and neutrophils and (iii) complement-dependent cytotoxicity (CDC) [47,48,49,50]. Of note, these mechanisms of cytotoxicity may be highly potentiated by the use of immune stimulating cytokines or adoptive cell therapy. FDA approved two different GD2 Abs for clinical trials that are dinutuximab (Ch14.18, chimeric murine/human antibody to GD2) and naxitamab (hu3F8, a humanized murine antibody to GD2) [45,46,51], currently used in clinical trials in recruiting phase or still active.
Only one of these clinical trials reported data regarding progression-free survival (PFS) and overall survival (OS) comparing patients treated with irinotecan and temozolomide in combination with dinutuximab or temsirolimus [52]. Improved PFS and OS (76.5% and 88.2%, respectively) were observed in patients receiving dinutuximab compared to those treated with temsirolimus (24.7% and 64.7%, respectively)
Figure 1 summarizes the antibodies used for NB immunotherapy with corresponding antigens and mechanisms of action.
These Abs showed similar clinical impact and toxicities mainly manifested as neuropathic pain, fever and allergic reactions. However some differences have been reported probably due to their different half life [30,53].
Cytokines are the most factors used in combination with a GD2 antibody, especially IL-2 and granulocyte-macrophage colony stimulating factor (GM-CSF). IL-2 induces activation T and NK cells [54] and it was approved by FDA for treating adult tumors. In children with high-risk NB several phase I and II trials tested IL-2 as monotherapy, but no significant effective results have been reported [55,56,57]. Recently, Ladestein et al. demonstrated only marginal effects of dinutuximab beta in combination with IL-2. In fact, the 3-year event-free survival was 56% (95% CI 49–63) with dinutuximab beta (83 patients had an event) and 60% (53–66) with dinutuximab beta and subcutaneous IL-2 (80 patients had an event; p = 0.76) [25,48]. By contrast, IL-2 administration with alternating cycles of GM-CSF in combination with dinutuximab resulted in higher rates of event-free (66% versus 46%) and overall survival after 2 years, compared to standard therapy alone (86% versus 75%) [25,48]. GM-CSF is a myeloid growth factor that stimulates differentiation of progenitor cells into granulocytes and monocytes, and boosts immune responses [58]. A human recombinant GM-CSF, named LEUKINE® (sargramostim), has been approved by FDA. Several clinical trials evaluating the role of GM-CSF combined with anti-GD2 in high-risk NB were conducted over the last decade (Table 2). Although this immunotherapy has shown substantial anti-neuroblastoma activity (NCT01757626) [51] even against minimal residual disease (NCT02100930), patients should be closely monitored for the onset of posterior reversible encephalopathy syndrome (NCT01183897) [26] (Table 2).

4.2. Other NB Associated Antigens as Targets for Antibody-Mediated Immunotherapy

Another target recently considered for development of therapeutic approaches against high risk patients is represented by anaplastic lymphoma kinase (ALK) that was found to be mutated in 9% of NB [59,60]. In 2009 the first clinical trial in pediatric patients with refractory solid tumors was conducted by the Children’s Oncology Group using an ALK inhibitor (i.e., crizotinib) providing discouraging results [61]. Afterwards, better results were obtained using other ALK inhibitor compounds, alone or in combination with chemotherapy, in solid tumors such as lung cancer, but some problems emerged due to secondary mutation and amplification of ALK and off-target mechanisms including activation of ‘bypass’ signaling pathways [62,63,64]. Recently, Sano R. et al. developed an Ab drug conjugate directly targeting ALK receptor, the CDX-0125-TEI, that exhibited cytotoxicity against both wild-type and mutated ALK in patient-derived xenografts [65] (Figure 1).
Finally, an additional interesting target for NB therapy is B7-H3 (CD276), a type I transmembrane glycoprotein molecule homogeneously expressed in both primary and metastatic NB (Figure 1) [62]. Loo D. et al. developed the anti B7-H3 Ab MGA27 that demonstrated to mediate potent cytotoxicity against a broad range of tumor cell types in xenograft models, while avoiding toxicities [66]. This antibody, later named as enoblituzumab, is currently in phase I trials for diverse solid tumors including refractory tumors and pediatric cancers.

5. Adoptive Cell Therapy Based on NK Cells

NK cells represent a subset of lymphocytes belonging to innate immune system, which originate in the BM and exert anti-tumor and anti-viral activities [67,68,69]. NK cell functions are regulated by the integration of multiple signals derived from activating and inhibitory receptors which bind specific ligands expressed on tumors and viral-infected cells. NK cells may be expanded ex vivo and stimulated by different cytokines, including IL-15, IL-12 and IL-18 [67,68,69]. Since NK cells do not cause graft versus host disease (GvHD), they represent an ideal source for allogeneic “off-the-shelf” cellular therapy. The development of therapeutic protocols for NB patients, based on the infusion of NK cells, arose from the success obtained in different clinical trials using monoclonal antibodies targeting NB. In these trials, the observed complete remission or stabilization of the disease was related to an increase of NK cell frequency and activity, including NK cell-mediated ADCC [70,71,72,73,74]. Of note, NK cells are known for their ability to target and lyse tumor cells lacking HLA-class I molecules, which represent the ligands of killer inhibitory receptors [75]. For this reason metastatic NB cells, which show low to absent HLA-class I expression [76], paralleled by the presence of different ligands for NK cell activating receptors [77], represent an ideal target for NK cell mediated lysis.
Different preclinical studies on NB have been described using NK cells. Castriconi et al. [78] analyzed the therapeutic effect of infusion of NK cells activated in vitro with IL-2 in a xenograft model of metastatic NB. SCID/NOD mice, inoculated intravenously with the HTLA-230 NB cell line, showed significant prolonged survival when injected with multiple NK cell doses soon after the inoculation of tumor cells. Such therapeutic effect was further increased by treatment with IL-2 and IL-15 that stimulated NK cells through up-regulation of the activating markers CD69 and CD25 as well as of NKp44 and DNAM-1 receptors [78]. Similarly, Liu and coworkers [79] tested NK cells, either from normal donors or NB patients, in SCID/NOD mice injected with CHLA-255-Fluc cells. At variance with the previous studies, NK cells were expanded in vitro, using a feeder composed of irradiated clinical-grade K562 cells expressing membrane-bound IL-21 (K562 mbIL-21), in the presence of IL-2. The resulting NK cells displayed high expression of DNAM-1, NKG2D, NKp46, CD56 and CD16, and efficiently lysed NB cell lines in vitro, through mechanisms such as (i) granzyme A and B release and (ii) induction of ADCC in the presence of anti-GD2 Ab. Preclinical studies highlighted a limited therapeutic effect of expanded NK cells alone, but a significant inhibition of tumor growth and a prolonged survival of mice when combined with the anti-GD2 ch14.18 Ab. These data suggested that the main therapeutic effect exerted by NK cells against NB in vivo was related to induction of ADCC rather than direct NK cell lysis [79]. The importance of this mechanism was confirmed in the study by Barry et al. that used primary NB xenografted in murine kidney, then analyzed for metastatic disease in the liver and BM. In this model, the administration of NK cells, expanded in vitro using K562 and IL-21, in combination with dinutuximab, not only reduced metastasis in the liver and BM, but also prolonged survival [80].
Starting from the concept that TGFβ is an immunosuppressive cytokine released by NB cells able to down-regulate activating receptors on NK cells and to inhibit the release of granzyme and perforin, Tran and coworkers performed preclinical studies using the TGFβR1 inhibitor galunisertib. They reported that mice, injected in the kidney with NB cell lines and treated with galunisertib plus expanded NK cells and dinutuximab, showed prolonged survival associated with reduction of tumor growth, as compared to mice treated with NK cells and dinutuximab alone. Thus, they paved the way to clinical application of this combined therapy for high-risk NB patients [81].
Clinical trials using NK cell infusions for NB patients are still ongoing (Table 3). A pilot trial was performed by Federico et al. [82] on NB patients that had undergone six different courses of induction chemotherapy, including cyclophosphamide and topotecan, or irinotecan and temozolomide, or ifosfamide, carboplatin and etoposide. Each group was treated with the anti-GD2 Ab hu14.18K322A, GM-CSF and IL2, in the presence or absence of haploidentical parental NK cell infusion. Such GMP-graded NK cells were isolated from leukapheresis by CliniMACS and infused at a median dose of 15.5 × 106/Kg. NK cell infusion was safe and well tolerated, although toxicities related to chemotherapy, anti-GD2 or GM-CSF infusion were observed. The overall response rate of patients was 61.5%, with a complete response/very good partial response rate of 38.5%. More importantly, progression of the disease was never observed. However, the contribution of NK cells to therapeutic success in this limited study was not assessed [82]. A phase II clinical trial was performed in NB patients subjected to multiple cycles of induction chemotherapy with different drugs (including cyclophosphamide, topotecan, cisplatin, etoposide, doxorubicin and vincristine) and infused with the anti-GD2 Ab hu14.18K322A with GM-CSF and IL-2 [83]. Patients were treated, as consolidation therapy, with busulfan/melphalan plus infusion of autologous hematopoietic stem cells (collected in the induction therapy phase). In addition, some patients were treated with an experimental immunotherapy, composed of the hu14.18K322A Ab and GM-CSF in the presence or absence of GMP-graded haploidentical NK cells, which were purified using CliniMACS system and not expanded ex vivo. No difference in toxicities was observed between patients infused with or without NK cells. NK cells were still detected until 18 days post-infusion demonstrating an NK cell engraftment [83]. A longitudinal analysis of this clinical trial revealed that patients receiving haploidentical NK cell infusion had an increased NK cell count at day 7, and a decreased NK cell count at day 21 post-infusion [84]. However, donor NK cells infused at higher doses were still detected after 21 days. Such cells were able to respond to ex vivo stimulation with IL-15 and IL-2 and to lyse K562 cell line in vitro, thus suggesting that NK retained their cytotoxic function. Finally, patients with stable disease displayed a higher number of alloreactive NK cells than those with residual disease [84].
Modak and coworkers [85] performed a phase I clinical study on NB patients receiving an induction therapy with vincristine, cyclophosphamide and topotecan, and then different doses of haploidentical parental NK cells and the anti-GD2 murine Ab 3F8. In this study, NK cells were isolated by CliniMACS and then cultured overnight with IL-2 before being infused. Toxicities were almost related to the infusion of 3F8 Ab, whereas NK cell administration was safe and well tolerated. Moreover, they demonstrated that patients with detectable NK cells in the periphery at day 14 display a complete remission and, in general, patients who received a higher dose of NK cell infusion showed an improved progression-free survival compared to those receiving lower NK cell doses (p = 0.018) [85].
Of note, Heinze and coworkers [86] recently described different methods to expand NK cells in vitro for clinical use analyzing viability, purity and composition of NK cell preparations starting from normal donors’ buffy coat preparations. Either CD56+ enriched cells and CD3+/CD19+ depleted cells were cultured in media supplemented with different combinations of cytokines (i.e., IL-2+IL-15 or IL-15+IL-21). The authors concluded that the highest purity and expansion rate can be obtained starting from CD3+/CD19+ depleted cells using IL-15 and IL-21. These cells showed also increased cytotoxicity and degranulation, when cultured in the presence of NB cell lines, thus suggesting that these NK cell preparations could be useful to test in clinical trials [86].
Collectively, these studies suggested that NK cell infusion, as consolidation therapy, in NB patients is feasible and well tolerated. Therapeutic effects of NK cells against NB hold promise and have to be carefully evaluated in phase III randomized clinical trials. At present, no phase III/IV clinical trials using NK cell infusions are ongoing, whereas 20 phase I/II trials are registered (clinicaltrials.gov), and some of them completed (Table 3).

6. Adoptive Cell Therapy Based on NKT Cells

Another innate cytotoxic cell population, with intermediate features between NK and T cells, is represented by NKT cells which express an invariant TCRα chain, Vα24-Jα18, and are able to recognize self- and microbial-derived glycolipids presented by the monomorphic HLA class I-like molecule CD1d [87]. The contribution of NKT cells to anti-tumor responses has been demonstrated in different tumor models [88,89,90], as underlined by the finding that these cells are decreased in number and/or functionality in cancer patients [91,92,93]. NKT-based clinical trials are ongoing for several malignancies, and are based on the injection of alpha-Galactosylceramide (α-GalCer) or α-GalCer-pulsed APC to expand NKT cells in vivo. Alternatively, autologous NKT cells have been expanded ex vivo using IL-2 and anti-CD3 mAbs [94]. Recent evidence suggested that NKT cells may have anti-tumor activities in NB patients. Liu et al. [95] reported that NKT cell migration towards a co-culture of NB cells and monocytes was increased in hypoxic conditions, a typical feature of NB microenvironments. Indeed, hypoxia down-regulated the secretion of the tumor-associated chemokine CCL2 (which attracts NKT cells) by NB cells, whereas the chemokine CCL20 was increased upon co-culture of NB cells with monocytes. Accordingly, neutralizing monoclonal Abs against CCL20 abrogated NKT cell migration in hypoxic conditions [95]. The authors demonstrated that humanized NOD/SCID Il2rg-/- (NSG) mice inoculated with NB cells in the renal capsule showed a high infiltration of human TAM in the tumor microenvironment, with a high percentage of M2 macrophages. Afterwards, they inoculated expanded NKT cells that highly infiltrated the tumors, but this feature was abrogated by the treatment of mice with anti-CCL2 and anti-CCL20 mAbs. Furthermore, expanded NKT cells, inoculated in preclinical model of metastatic NB, preferentially migrated in the hypoxic areas of metastasis in liver and BM, were inhibited by hypoxia and by contact with tumor cells [95]. To improve NKT cell functions, they transduced NKT cells with a cDNA encoding human IL-15 (NKT/IL-15), demonstrating that the latter cells were able to proliferate in hypoxic conditions in vitro, whereas parental NKT cells were inhibited in the same settings. Finally, they setup a metastatic model of NB in NSG and humanized NSG mice, showing that tumors developed more rapidly in the latter mice, due to the infiltration of human hematopoietic cells in the tumor. Such tumor-promoting effect was totally abrogated by inoculating NKT/IL-15 (but not NKT) and restored in the presence of CD1d blocking mAbs. These findings suggest that adoptive therapy with expanded NKT cells in combination with IL-15 may represent a therapeutic strategy for patients with metastatic NB [95]. So far, no active clinical trials based on NKT cells as therapy are present for NB patients.

7. CAR T Cells for Therapy of High Risk NB Patients

The use of T cells genetically modified to express a chimeric antigen receptor (CAR) is a new promising approach of adoptive cell therapy in cancer, combining the antigen specificity of a monoclonal Ab with the effector function and long-term persistence of T cells [96,97,98]. CAR provided success in treating B cell acute lymphoblastic leukemias, with limited clinical benefit in solid tumors, although more than 100 clinical studies have been developed. Indeed, the success of CAR T cell therapy in solid tumors relies on the ability of CAR T cells enter in the tumor site, overcome the immunosuppressive tumor microenvironment, and persist for a long period.
CAR is composed of different domains, including (i) the single-chain variable fragment (scFv) of tumor antigen-specific Ab, (ii) a hinge region, (iii) the trans-membrane domain of CD8α or CD28 molecules and (iv) the intracellular signaling region. Thus, T cells endowed with CAR showed an improved T-cell antigen recognition, T-cell activation and tumor cell lysis. CARs are classified as first, second and third generation on the basis of the presence of one, two or more T cell co-stimulatory molecules [99,100].
First generation CAR included activating/signaling domains of CD3ζ or FcγRIII molecules, whereas second generation CAR included co-stimulatory domains of other molecules, including CD28, ICOS, 4-1BB, OX-40 and CD27). The third generation CAR are endowed with co-stimulatory domains in tandem (i.e., CD28 in combination with 4-1BB), which may increase T cell expansion and anti-tumor functions [101].
FDA approved two different products for CAR T cell therapy in 2017, named Kymriah® (Novartis) and Yescarta® (Kite/Gilead), for the treatment of hematological malignancies [102].
The most common side effects of CAR αβ T cells are cytokine release syndrome (CRS) and GvHD [103]. Of note, to prevent unexpected T-cell–related toxicity such as CRS, an inducible caspase-9 (iC9) gene was introduced in frame with the TCR, thus allowing the prompt elimination of genetically modified T cells by the use of chemical induction of dimerization (CID) drug, AP1903 [104,105].
Two clinical studies have been conducted with first generation CAR T cells in patients with NB [106,107]: in the first GD2 CAR T cells were well tolerated, but only one of six patients had a partial response, in the second five of 11 patients with active disease showed tumor responses and three of them had complete responses. Nonetheless, the response rates in patients with NB remain significantly lower than those observed in patients with acute lymphoblastic leukemia treated with CD19-CAR constructed with the same approach [108]. More recently, Heczey A et al. implemented a third-generation GD2 CAR in which the specific scFv was derived from the murine 14.G2a mAb coupled with the ξ-chain endodomain and two costimulatory endodomains in tandem (CD28 and OX40) [109]. CAR T cells, administered in combination with lymphodepletion to patients with NB, were well tolerated and promoted some objective clinical responses [107,109,110]. Afterwards, Quintarelli et al. [111] reported that insertion of 4-1BB costimulatory domain, in the place of OX40 in a third generation CAR, significantly improved the anti-tumor efficacy of GD2 CAR. The choice of 4-1BB signaling was particularly effective in terms of T cell activation and persistence, control of tumor cell growth and T cell exhaustion [111].
Finally, Chen and coworkers incorporated IL-15 and iC9 within the GD2 CAR construct and demonstrated that transduced T cells enriched in central memory/stem cell–like cells, expressed PD-1 at low level, promoted superior antitumor activity, expansion and survival in vitro and in vivo [112].
Several clinical trials are in progress (Table 4), including those that evaluate CAR T cells that target CD171 [113] in patients with NB and ganglioneuroblastoma (NCT02311621) or B7H3 (NCT04483778) in relapsed/refractory pediatric solid tumors. Many others continue to explore GD2–CAR T cells for NB and include NCT02761915, NCT03373097, NCT03721068, NCT03635632, NCT01822652 and NCT01953900. In addition, several studies have addressed strategies to implement the efficacy of CAR T cell therapies in NB patients [114].
Figure 2 summarizes current approaches for adoptive cell therapy in the clinical setting of NB.

8. Future Prospects: CAR NK Cells and γδ T Cells

γδ T cells and NK cells may represent an ideal source for adoptive cell therapy due to some shared features. Both cell types recognize and kill tumor cells irrespective of the expression of a single tumor-associated antigen, thus avoiding tumor immune escape related to single antigen loss [67,68,69,94,114,115]. In addition, γδ T and NK cells act through MHC-independent recognition of target cells, thus reducing the risk of alloreactivity and GvHD [116] and are able to provide immediate release of effector cytokines in different tissues. Of note γδ T cells show a natural tissue tropism which provide a migratory advantage over αβ T cells, leading to a superior ability to infiltrate in hypoxic tumors [117,118]. Finally, γδ T cells can interact with APC and other immune cells, and also act as APCs by priming the antigens for αβ T cells, thus orchestrating the anti-tumor immune responses [119,120].
Taken together, these features render NK and γδ T cells an optimal source for immunotherapy, and different genetic engineering strategies have been developed to enhance and redirect their cytotoxicity toward tumor antigens as well as to improve their in vivo or ex vivo expansion that is crucial for their broad clinical application [121].
In this regard, although γδ T cells may be difficult to expand efficiently ex vivo, sufficient numbers for adoptive transfer immunotherapy especially for pediatric patients are easily obtained. For example, the Vγ9Vδ2 population may be efficiently expanded using zoledronic acid and IL-2 both ex vivo and in vivo starting from peripheral blood mononuclear cells [99]. γδ T cell infusion proved to be safe and showed promising results in recent clinical trials. Indeed, ongoing studies are addressing the increase of γδ T cell survival and potency for clinical purposes [122].
Although CAR γδ T cells have shown preclinical efficacy, their optimization and the choice of the best co-stimulatory domains are still open [123]. γδ T cells express a wide panel of co-stimulatory molecules, including CD28, CD27 and 4-1BB that enhance γδ T cell survival and proliferation. Thus, the interaction between signals derived from γδ TCR and these molecules should be studied in detail to design more suitable CAR constructs [124].
The clinical efficacy of CAR T cells in solid tumors may be inhibited by immune suppressive conditions in the tumor microenvironment [123]. In this regard, γδ T cells may be resistant to immune-suppression due to the expression of peculiar receptors. In addition, the addition of treatments targeting the stroma and checkpoint inhibitors may improve the treatment efficacy in solid tumors.
Polito et al. [125] recently described an efficient method to produce allogeneic third-party and ready-to-use γδ CAR T cells. They expanded memory Vδ1 subpopulation using engineered APCs expressing CD86, 4-1BBL, CD40L and the CMV-antigen-pp65, obtaining polyclonal γδ T cells expressing activation and memory markers, with potent anti-tumor activity in vitro and in vivo with no sign of alloreactivity. Finally, the authors engineered these cells with a third generation anti-GD2 CAR (GD2.CD28.4-1BBζ), further enhancing anti-NB cytotoxicity.
A handful of clinical trials using γδ T cells for cancer treatment have been performed, several are in progress and include hematological malignancies (NCT02656147, NCT03533816, NCT04008381 and NCT03885076), glioblastoma (NCT04165941), hepatocellular carcinoma (NCT00562666, NCT02425735 and NCT04518774), renal carcinoma (NCT00582790), breast cancer (NCT02418481 and NCT03183206), prostate cancer (NCT00582556), lung cancer (NCT0318323, NCT02425748, NCT00588913) and pancreatic cancer (NCT03180437). These clinical studies revealed that adoptive transfer of γδ T cells is safe and feasible [126] and, in most cases, objective responses were observed.
NK cells represent another ideal source for allogeneic “off-the-shelf” cellular therapy, since such cells (i) do not cause GvHD, (ii) lack in vivo clonal expansion and (iii) display a limited persistence. These features reduce the risk of CRS, a life-threatening toxicity observed in several CAR αβ T cell trials [127]. However, a limitation of this therapeutic approach may be related to a different persistence between CAR T and CAR NK cells according to disease, the tumor burden, lymphodepletion settings and other factors (i.e., the potency of the administered T cells and CAR construct). Thus, some strategies have been developed to overcome this issue, including the co-transfection of stimulatory cytokines (e.g., IL-15) and multiple administration of CAR cells at different time points.
First, second and third generation CAR NK cells have been generated, using lentiviral vectors, to target tumor antigens such as CD19 [128,129], CD20 [128], CD33 [130], CD7 [131], CD22 [132], ErbB2 [133] and EGFR [134,135]. Such CARs contained the scFv region of a tumor antigen-specific Ab combined with (i) enhancers like Kozak sequence (HU 2020), (ii) signaling domains including 4-1BB [129,130,136,137] and/or CD28 [130,133,134,135,136,138], (iii) CD8α hinge region [137,138], (iv) myc tag sequence [139] and (v) CD3ζ chain of TCR complex [128,129,133,134,135,138,140].
Esser [141] and Romanski [142] used amphotropic retroviral vectors to generate CAR NK cells. The construct was composed of scFv of ch14.18 anti-GD2 chimeric Ab or of anti-CD19 Ab, respectively, combined with IgVH signal peptide, myc tag, the hinge region of CD8α and trans-membrane/intracellular domains of CD3ζ chain. Such CAR NK cells recognized and eliminated GD2 expressing cells which were resistant to parental NK-92 cells. In addition, the authors observed increased killing of NK-sensitive tumor cell lines using retargeted NK-92 cells. Such lysis was strictly dependent on specific recognition of the target antigen.
The potency of this therapeutic approach was further implemented by Kailayangiri et al. [143] by the integration of co-stimulatory molecules (i.e., trans-membrane domain of CD28 and signaling domains of 4-1BB and CD3ζ) and by extended cytokine support during NK cell expansion in vitro. However, the in vivo response was low, due to the immunosuppressive activities of HLA-G molecule.
To date, only a few phase I/II clinical trials are in recruiting status for NB therapy (NCT03940820, NCT02839954 and NCT03941457), but many others are in progress for different diseases including Covid-19 (NCT04324996), solid tumors (NCT03940820, NCT02839954, NCT03941457), glioblastoma (NCT03383978) and hematological malignancies (NCT04008394, NCT02274584, NCT04526834, NCT04538599, NCT03940833, NCT04623944, NCT03056339, NCT04004637, NCT03049449, NCT04033302, NCT04288726, NCT04538599 and NCT04008394).

9. Conclusions

In the last 20 years several biological parameters that impact on the course of NB and on patients’ survival have been identified. These are of particular relevance for the best risk stratification, thus identifying high risk patients who need more aggressive treatments to improve their clinical outcome. In this line, a plethora of therapeutic strategies have been setup and applied in preclinical and clinical settings for patients with refractory or relapsed tumors. Nonetheless, these patients can rarely be cured, and for this reason innovative and more effective protocols are urgently needed.
Anti-GD2 Abs represent the first immunotherapy applied in NB which greatly increase overall and progression-free survival of children, especially when used in combination with cytokines, such as IL-2 and GM-CSF.
Recent advances have been achieved using NK cells as adoptive cell therapy in support of consolidation phase, and infusion of CAR T cells. In this regard, many phase I/II clinical trials have been completed, some are still ongoing, demonstrating that adoptive cell therapy is safe, well tolerated and without relevant toxicities. However, efficacy of both adoptive cellular therapeutic strategies has to be confirmed in future phase III/IV studies. Of note, NK cells will be used as adjuvant therapy after at least one cycle of standard therapy plus anti-GD2 mAbs whereas CAR T cells will be infused in patients with relapse or refractory to conventional treatments.
Future perspectives of adoptive cell therapies for high-risk NB patients are represented by the use of γδ T cells and by the generation of novel CAR cells obtained by the transfection of NK and γδ T cells. Undoubtedly, the peculiar advantage of these cells is the generation of “off the shelf” therapies avoiding a strict selection of the donors irrespective of the recipients, with no risk of GvHD. To date, few phase I/II on NB patients are ongoing, however promising results have been obtained in other malignancies, demonstrating the safety of these therapeutic approaches.

Author Contributions

F.M., F.S. and I.A. wrote and revised the manuscript, M.P. revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Italian Ministry of Health, Progetti di Ricerca Corrente 2020.

Institutional Review Board Statement

Not Applicable.

Informed Consent Statement

Not Applicable.

Data Availability Statement

Data sharing not applicable.

Acknowledgments

We thank BioRender.com for sharing the tools for building cartoons and Chiara Brignole for help in the choice of such graphic tool.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Maris, J.M. Recent advances in neuroblastoma. N. Engl. J. Med. 2010, 362, 2202–2211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Tolbert, V.P.; Matthay, K.K. Neuroblastoma: Clinical and biological approach to risk stratification and treatment. Cell Tissue Res. 2018, 372, 195–209. [Google Scholar] [CrossRef]
  3. London, W.B.; Castleberry, R.P.; Matthay, K.K.; Look, A.T.; Seeger, R.C.; Shimada, H.; Thorner, P.; Brodeur, G.; Maris, J.M.; Reynolds, C.P.; et al. Evidence for an age cutoff greater than 365 days for neuroblastoma risk group stratification in the Children’s Oncology Group. J. Clin. Oncol. 2005, 23, 6459–6465. [Google Scholar] [CrossRef] [PubMed]
  4. Vo, K.T.; Matthay, K.K.; Neuhaus, J.; London, W.B.; Hero, B.; Ambros, P.F.; Nakagawara, A.; Miniati, D.; Wheeler, K.; Pearson, A.D.; et al. Clinical, biologic, and prognostic differences on the basis of primary tumor site in neuroblastoma: A report from the international neuroblastoma risk group project. J. Clin. Oncol. 2014, 32, 3169–3176. [Google Scholar] [CrossRef] [PubMed]
  5. Irwin, M.S.; Park, J.R. Neuroblastoma: Paradigm for precision medicine. Pediatr. Clin. N. Am. 2015, 62, 225–256. [Google Scholar] [CrossRef]
  6. Matthay, K.K.; Maris, J.M.; Schleiermacher, G.; Nakagawara, A.; Mackall, C.L.; Diller, L.; Weiss, W.A. Neuroblastoma. Nat. Rev. Dis. Prim. 2016, 2, 16078. [Google Scholar] [CrossRef]
  7. DuBois, S.G.; Kalika, Y.; Lukens, J.N.; Brodeur, G.M.; Seeger, R.C.; Atkinson, J.B.; Haase, G.M.; Black, C.T.; Perez, C.; Shimada, H.; et al. Metastatic sites in stage IV and IVS neuroblastoma correlate with age, tumor biology, and survival. J. Pediatr. Hematol. Oncol. 1999, 21, 181–189. [Google Scholar] [CrossRef]
  8. Morgenstern, D.A.; London, W.B.; Stephens, D.; Volchenboum, S.L.; Simon, T.; Nakagawara, A.; Shimada, H.; Schleiermacher, G.; Matthay, K.K.; Cohn, S.L.; et al. Prognostic significance of pattern and burden of metastatic disease in patients with stage 4 neuroblastoma: A study from the International Neuroblastoma Risk Group database. Eur. J. Cancer 2016, 65, 1–10. [Google Scholar] [CrossRef]
  9. Brodeur, G.; Seeger, R.; Schwab, M.; Varmus, H.; Bishop, J. Amplification of N-myc in untreated human neuroblastomas correlates with advanced disease stage. Science 1984, 224, 1121–1124. [Google Scholar] [CrossRef]
  10. Seeger, R.C.; Brodeur, G.M.; Sather, H.; Dalton, A.; Siegel, S.E.; Wong, K.Y.; Hammond, D. Association of multiple copies of the N-myc oncogene with rapid progression of neuroblastomas. N. Engl. J. Med. 1985, 313, 1111–1116. [Google Scholar] [CrossRef]
  11. Matthay, K.K.; Shulkin, B.; Ladenstein, R.; Michon, J.; Giammarile, F.; Lewington, V.; Pearson, A.D.; Cohn, S.L. Criteria for evaluation of disease extent by (123)I-metaiodobenzylguanidine scans in neuroblastoma: A report for the International Neuroblastoma Risk Group (INRG) Task Force. Br. J. Cancer 2010, 102, 1319–1326. [Google Scholar] [CrossRef]
  12. Cohn, S.L.; Pearson, A.D.J.; London, W.B.; Monclair, T.; Ambros, P.F.; Brodeur, G.M.; Faldum, A.; Hero, B.; Iehara, T.; Machin, D.; et al. The International Neuroblastoma Risk Group (INRG) classification system: An INRG Task Force report. J. Clin. Oncol. 2009, 27, 289–297. [Google Scholar] [CrossRef] [PubMed]
  13. Baker, D.L.; Schmidt, M.L.; Cohn, S.L.; Maris, J.M.; London, W.B.; Buxton, A.; Stram, D.; Castleberry, R.P.; Shimada, H.; Sandler, A.; et al. Outcome after Reduced Chemotherapy for Intermediate-Risk Neuroblastoma. N. Engl. J. Med. 2010, 363, 1313–1323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Rubie, H.; De Bernardi, B.; Gerrard, M.; Canete, A.; Ladenstein, R.; Couturier, J.; Ambros, P.; Munzer, C.; Pearson, A.D.; Garaventa, A.; et al. Excellent outcome with reduced treatment in infants with nonmetastatic and unresectable neuroblastoma without MYCN amplification: Results of the prospective INES 99.1. J. Clin. Oncol. 2011, 29, 449–455. [Google Scholar] [CrossRef] [PubMed]
  15. Strother, D.R.; London, W.B.; Schmidt, M.L.; Brodeur, G.M.; Shimada, H.; Thorner, P.; Collins, M.H.; Tagge, E.; Adkins, S.; Reynolds, C.P.; et al. Outcome after surgery alone or with restricted use of chemotherapy for patients with low-risk neuroblastoma: Results of Children’s Oncology Group study P9641. J. Clin. Oncol. 2012, 30, 1842–1848. [Google Scholar] [CrossRef] [PubMed]
  16. Morgenstern, D.A.; Pötschger, U.; Moreno, L.; Papadakis, V.; Owens, C.; Ash, S.; Pasqualini, C.; Luksch, R.; Garaventa, A.; Canete, A.; et al. Risk stratification of high-risk metastatic neuroblastoma: A report from the HR-NBL-1/SIOPEN study. Pediatr. Blood Cancer 2018, 65, 17. [Google Scholar] [CrossRef]
  17. 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]
  18. Di Giannatale, A.; Dias-Gastellier, N.; Devos, A.; Mc Hugh, K.; Boubaker, A.; Courbon, F.; Verschuur, A.; Ducassoul, S.; Malekzadeh, K.; Casanova, M.; et al. Phase II study of temozolomide in combination with topotecan (TOTEM) in relapsed or refractory neuroblastoma: A European Innovative Therapies for Children with Cancer-SIOP-European Neuroblastoma study. Eur. J. Cancer 2014, 50, 170–177. [Google Scholar] [CrossRef]
  19. Bagatell, R.; London, W.B.; Wagner, L.M.; Voss, S.D.; Stewart, C.F.; Maris, J.M.; Kretschmar, C.; Cohn, S.L. Phase II study of irinotecan and temozolomide in children with relapsed or refractory neuroblastoma: A Children’s Oncology Group study. J. Clin. Oncol. 2011, 29, 208–213. [Google Scholar] [CrossRef] [Green Version]
  20. Kushner, B.H.; Kramer, K.; Modak, S.; Cheung, N.-K.V. Irinotecan Plus Temozolomide for Relapsed or Refractory Neuroblastoma. J. Clin. Oncol. 2006, 24, 5271–5276. [Google Scholar] [CrossRef]
  21. London, W.B.; Frantz, C.N.; Campbell, L.A.; Seeger, R.C.; Brumback, B.A.; Cohn, S.L.; Matthay, K.K.; Castleberry, R.P.; Diller, L. Phase II randomized comparison of topotecan plus cyclophosphamide versus topotecan alone in children with recurrent or refractory neuroblastoma: A Children’s Oncology Group study. J. Clin. Oncol. 2010, 28, 3808–3815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Berthold, F.; Boos, J.; Burdach, S.; Erttmann, R.; Henze, G.; Hermann, J.; Klingebiel, T.; Kremens, B.; Schilling, F.H.; Schrappe, M.; et al. Myeloablative megatherapy with autologous stem-cell rescue versus oral maintenance chemotherapy as consolidation treatment in patients with high-risk neuroblastoma: A randomised controlled trial. Lancet Oncol. 2005, 6, 649–658. [Google Scholar] [CrossRef]
  23. Matthay, K.K.; Villablanca, J.G.; Seeger, R.C.; Stram, D.O.; Harris, R.E.; Ramsay, N.K.; Swift, P.; Shimada, H.; Black, C.T.; Brodeur, G.M.; et al. Treatment of high-risk neuroblastoma with intensive chemotherapy, radiotherapy, autologous bone marrow transplantation, and 13-cis-retinoic acid. Children’s Cancer Group. N. Engl. J. Med. 1999, 341, 1165–1173. [Google Scholar] [CrossRef] [PubMed]
  24. Pritchard, J.; Cotterill, S.J.; Germond, S.M.; Imeson, J.; de Kraker, J.; Jones, D.R. High dose melphalan in the treatment of advanced neuroblastoma: Results of a randomised trial (ENSG-1) by the European Neuroblastoma Study Group. Pediatr. Blood Cancer 2005, 44, 348–357. [Google Scholar] [CrossRef] [PubMed]
  25. 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] [PubMed] [Green Version]
  26. Kayano, D.; Kinuya, S. Current Consensus on I-131 MIBG Therapy. Nucl. Med. Mol. Imaging 2018, 52, 254–265. [Google Scholar] [CrossRef] [PubMed]
  27. Matthay, K.K.; Yanik, G.; Messina, J.; Quach, A.; Huberty, J.; Cheng, S.C.; Veatch, J.; Goldsby, R.; Brophy, P.; Kersun, L.S.; et al. Phase II study on the effect of disease sites, age, and prior therapy on response to iodine-131-metaiodobenzylguanidine therapy in refractory neuroblastoma. J. Clin. Oncol. 2007, 25, 1054–1060. [Google Scholar] [CrossRef] [PubMed]
  28. Wilson, J.S.; Gains, J.E.; Moroz, V.; Wheatley, K.; Gaze, M.N. A systematic review of 131I-meta iodobenzylguanidine molecular radiotherapy for neuroblastoma. Eur. J. Cancer 2014, 50, 801–815. [Google Scholar] [CrossRef]
  29. Zhou, M.J.; Doral, M.Y.; DuBois, S.G.; Villablanca, J.G.; Yanik, G.A.; Matthay, K.K. Different outcomes for relapsed versus refractory neuroblastoma after therapy with (131)I-metaiodobenzylguanidine ((131)I-MIBG). Eur. J. Cancer 2015, 51, 2465–2472. [Google Scholar] [CrossRef] [Green Version]
  30. Park, J.A.; Cheung, N.-K.V. Targets and Antibody Formats for Immunotherapy of Neuroblastoma. J. Clin. Oncol. 2020, 38, 1836–1848. [Google Scholar] [CrossRef]
  31. Corrias, M.V.; Occhino, M.; Croce, M.; De Ambrosis, A.; Pistillo, M.P.; Bocca, P.; Pistoia, V.; Ferrini, S. Lack of HLA-class I antigens in human neuroblastoma cells: Analysis of its relationship to TAP and tapasin expression. Tissue Antigens 2001, 57, 110–117. [Google Scholar] [CrossRef] [PubMed]
  32. Marimpietri, D.; Petretto, A.; Raffaghello, L.; Pezzolo, A.; Gagliani, C.; Tacchetti, C.; Mauri, P.; Melioli, G.; Pistoia, V. Proteome profiling of neuroblastoma-derived exosomes reveal the expression of proteins potentially involved in tumor progression. PLoS ONE 2013, 8, e75054. [Google Scholar] [CrossRef] [PubMed]
  33. Morandi, F.; Levreri, I.; Bocca, P.; Galleni, B.; Raffaghello, L.; Ferrone, S.; Prigione, I.; Pistoia, V. Human Neuroblastoma Cells Trigger an Immunosuppressive Program in Monocytes by Stimulating Soluble HLA-G Release. Cancer Res. 2007, 67, 6433–6441. [Google Scholar] [CrossRef] [Green Version]
  34. Morandi, F.; Rizzo, R.; Fainardi, E.; Rouas-Freiss, N.; Pistoia, V. Recent Advances in Our Understanding of HLA-G Biology: Lessons from a Wide Spectrum of Human Diseases. J. Immunol. Res. 2016, 2016, 4326495. [Google Scholar] [CrossRef] [Green Version]
  35. Raffaghello, L.; Prigione, I.; Airoldi, I.; Camoriano, M.; Morandi, F.; Bocca, P.; Gambini, C.; Ferrone, S.; Pistoia, V. Mechanisms of immune evasion of human neuroblastoma. Cancer Lett. 2005, 228, 155–161. [Google Scholar] [CrossRef] [PubMed]
  36. Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 2011, 331, 1565–1570. [Google Scholar] [CrossRef] [Green Version]
  37. Feichtinger, R.G.; Neureiter, D.; Mayr, J.A.; Zimmermann, F.A.; Berthold, F.; Jones, N.; Sperl, W.; Kofler, B. Loss of mitochondria in ganglioneuromas. Front. Biosci. 2011, 3, 179–186. [Google Scholar] [CrossRef] [Green Version]
  38. Feichtinger, R.G.; Zimmermann, F.; Mayr, J.A.; Neureiter, D.; Hauser-Kronberger, C.; Schilling, F.H.; Jones, N.; Sperl, W.; Kofler, B. Low aerobic mitochondrial energy metabolism in poorly- or undifferentiated neuroblastoma. BMC Cancer 2010, 10, 149. [Google Scholar] [CrossRef] [Green Version]
  39. Morscher, R.J.; Aminzadeh-Gohari, S.; Feichtinger, R.G.; Mayr, J.A.; Lang, R.; Neureiter, D.; Sperl, W.; Kofler, B. Inhibition of Neuroblastoma Tumor Growth by Ketogenic Diet and/or Calorie Restriction in a CD1-Nu Mouse Model. PLoS ONE 2015, 10, e0129802. [Google Scholar] [CrossRef] [Green Version]
  40. 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] [Green Version]
  41. Pelizzo, G.; Veschi, V.; Mantelli, M.; Croce, S.; Di Benedetto, V.; D’Angelo, P.; Maltese, A.; Catenacci, L.; Apuzzo, T.; Scavo, E.; et al. Microenvironment in neuroblastoma: Isolation and characterization of tumor-derived mesenchymal stromal cells. BMC Cancer 2018, 18, 1176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. 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]
  43. Lammie, G.; Cheung, N.; Gerald, W.; Rosenblum, M.; Cordoncardo, C. Ganglioside gd(2) expression in the human nervous-system and in neuroblastomas—An immunohistochemical study. Int. J. Oncol. 1993, 3, 909–915. [Google Scholar] [CrossRef] [PubMed]
  44. Suzuki, M.; Cheung, N.K. Disialoganglioside GD2 as a therapeutic target for human diseases. Expert Opin. Ther. Targets 2015, 19, 349–362. [Google Scholar] [CrossRef] [PubMed]
  45. Heiner, J.P.; Miraldi, F.; Kallick, S.; Makley, J.; Neely, J.; Smith-Mensah, W.H.; Cheung, N.K. Localization of GD2-specific monoclonal antibody 3F8 in human osteosarcoma. Cancer Res. 1987, 47, 5377–5381. [Google Scholar] [PubMed]
  46. Ploessl, C.; Pan, A.; Maples, K.T.; Lowe, D.K. Dinutuximab: An Anti-GD2 Monoclonal Antibody for High-Risk Neuroblastoma. Ann. Pharmacother. 2016, 50, 416–422. [Google Scholar] [CrossRef]
  47. Kushner, B.H.; Cheung, N.K. GM-CSF enhances 3F8 monoclonal antibody-dependent cellular cytotoxicity against human melanoma and neuroblastoma. Blood 1989, 73, 1936–1941. [Google Scholar] [CrossRef] [Green Version]
  48. Ladenstein, R.; Pötschger, U.; Valteau-Couanet, D.; Luksch, R.; Castel, V.; Yaniv, I.; Laureys, G.; Brock, P.; Michon, J.M.; Owens, C.; et al. Interleukin 2 with anti-GD2 antibody ch14.18/CHO (dinutuximab beta) in patients with high-risk neuroblastoma (HR-NBL1/SIOPEN): A multicentre, randomised, phase 3 trial. Lancet Oncol. 2018, 19, 1617–1629. [Google Scholar] [CrossRef]
  49. Munn, D.H.; Cheung, N.K. Antibody-dependent antitumor cytotoxicity by human monocytes cultured with recombinant macrophage colony-stimulating factor. Induction of efficient antibody-mediated antitumor cytotoxicity not detected by isotope release assays. J. Exp. Med. 1989, 170, 511–526. [Google Scholar] [CrossRef] [Green Version]
  50. Saarinen, U.M.; Coccia, P.F.; Gerson, S.L.; Pelley, R.; Cheung, N.K. Eradication of neuroblastoma cells in vitro by monoclonal antibody and human complement: Method for purging autologous bone marrow. Cancer Res. 1985, 45, 5969–5975. [Google Scholar]
  51. Kushner, B.H.; Cheung, I.Y.; Modak, S.; Basu, E.M.; Roberts, S.S.; Cheung, N.-K. Humanized 3F8 Anti-GD2 Monoclonal Antibody Dosing With Granulocyte-Macrophage Colony-Stimulating Factor in Patients With Resistant Neuroblastoma: A Phase 1 Clinical Trial. JAMA Oncol. 2018, 4, 1729–1735. [Google Scholar] [CrossRef] [Green Version]
  52. Mody, R.; Naranjo, A.; Van Ryn, C.; Yu, A.L.; London, W.B.; Shulkin, B.L.; Parisi, M.T.; Servaes, S.-E.N.; Diccianni, M.B.; Sondel, P.M.; et al. Irinotecan-temozolomide with temsirolimus or dinutuximab in children with refractory or relapsed neuroblastoma (COG ANBL1221): An open-label, randomised, phase 2 trial. Lancet. Oncol. 2017, 18, 946–957. [Google Scholar] [CrossRef] [Green Version]
  53. Voeller, J.; Sondel, P.M. Advances in Anti-GD2 Immunotherapy for Treatment of High-risk Neuroblastoma. J. Pediatr. Hematol. Oncol. 2019, 41, 163–169. [Google Scholar] [CrossRef] [PubMed]
  54. Liao, W.; Lin, J.-X.; Leonard, W.J. Interleukin-2 at the crossroads of effector responses, tolerance, and immunotherapy. Immunity 2013, 38, 13–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Bauer, M.; Reaman, G.H.; Hank, J.A.; Cairo, M.S.; Anderson, P.; Blazar, B.R.; Frierdich, S.; Sondel, P.M. A phase II trial of human recombinant lnterleukin-2 administered as a 4-day continuous infusion for children with refractory neuroblastoma, non-Hodgkin’s lymphoma, sarcoma, renal cell carcinoma, and malignant melanoma. A childrens cancer group study. Cancer 1995, 75, 2959–2965. [Google Scholar] [CrossRef]
  56. Kalwak, K.; Ussowicz, M.; Gorczyńska, E.; Turkiewicz, D.; Toporski, J.; Dobaczewski, G.; Latos-Grazyńska, E.; Ryczan, R.; Noworolska-Sauren, D.; Chybicka, A. Immunologic effects of intermediate-dose IL-2 i.v. after autologous hematopoietic cell transplantation in pediatric solid tumors. J. Interf. Cytokine Res. 2003, 23, 173–181. [Google Scholar] [CrossRef]
  57. Roper, M.; Smith, M.A.; Sondel, P.M.; Gillespie, A.; Reaman, G.H.; Hammond, G.D.; Levitt, D.; Rosolen, A.; Colamonici, O.R.; Neckers, L.M.; et al. A phase I study of interleukin-2 in children with cancer. Am. J. Pediatr. Hematol. Oncol. 1992, 14, 305–311. [Google Scholar] [CrossRef]
  58. Becher, B.; Tugues, S.; Greter, M. GM-CSF: From Growth Factor to Central Mediator of Tissue Inflammation. Immunity 2016, 45, 963–973. [Google Scholar] [CrossRef] [Green Version]
  59. Bresler, S.C.; Weiser, D.A.; Huwe, P.J.; Park, J.H.; Krytska, K.; Ryles, H.; Laudenslager, M.; Rappaport, E.F.; Wood, A.C.; McGrady, P.W.; et al. ALK mutations confer differential oncogenic activation and sensitivity to ALK inhibition therapy in neuroblastoma. Cancer Cell 2014, 26, 682–694. [Google Scholar] [CrossRef] [Green Version]
  60. Schulte, J.H.; Schulte, S.; Heukamp, L.C.; Astrahantseff, K.; Stephan, H.; Fischer, M.; Schramm, A.; Eggert, A. Targeted Therapy for Neuroblastoma: ALK Inhibitors. Klin. Padiatr. 2013, 225, 303–308. [Google Scholar] [CrossRef]
  61. Mossé, Y.P.; Lim, M.S.; Voss, S.D.; Wilner, K.; Ruffner, K.; Laliberte, J.; Rolland, D.; Balis, F.M.; Maris, J.M.; Weigel, B.J.; et al. Safety and activity of crizotinib for paediatric patients with refractory solid tumours or anaplastic large-cell lymphoma: A Children’s Oncology Group phase 1 consortium study. Lancet Oncol. 2013, 14, 472–480. [Google Scholar] [CrossRef] [Green Version]
  62. Camidge, D.R.; Doebele, R.C. Treating ALK-positive lung cancer--early successes and future challenges. Nat. Rev. Clin. Oncol. 2012, 9, 268–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Doebele, R.C.; Pilling, A.B.; Aisner, D.L.; Kutateladze, T.G.; Le, A.T.; Weickhardt, A.J.; Kondo, K.L.; Linderman, D.J.; Heasley, L.E.; Franklin, W.A.; et al. Mechanisms of Resistance to Crizotinib in Patients with ALK Gene Rearranged Non–Small Cell Lung Cancer. Clin. Cancer Res. 2012, 18, 1472–1482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Katayama, R.; Shaw, A.T.; Khan, T.M.; Mino-Kenudson, M.; Solomon, B.J.; Halmos, B.; Jessop, N.A.; Wain, J.C.; Yeo, A.T.; Benes, C.; et al. Mechanisms of acquired crizotinib resistance in ALK-rearranged lung Cancers. Sci. Transl. Med. 2012, 4, 120ra117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Sano, R.; Krytska, K.; Larmour, C.E.; Raman, P.; Martinez, D.; Ligon, G.F.; Lillquist, J.S.; Cucchi, U.; Orsini, P.; Rizzi, S.; et al. An antibody-drug conjugate directed to the ALK receptor demonstrates efficacy in preclinical models of neuroblastoma. Sci. Transl. Med. 2019, 11. [Google Scholar] [CrossRef] [PubMed]
  66. Loo, D.; Alderson, R.F.; Chen, F.Z.; Huang, L.; Zhang, W.; Gorlatov, S.; Burke, S.; Ciccarone, V.; Li, H.; Yang, Y.; et al. Development of an Fc-Enhanced Anti–B7-H3 Monoclonal Antibody with Potent Antitumor Activity. Clin. Cancer Res. 2012, 18, 3834–3845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Caligiuri, M.A. Human natural killer cells. Blood 2008, 112, 461–469. [Google Scholar] [CrossRef]
  68. Vivier, E.; Raulet, D.H.; Moretta, A.; Caligiuri, M.A.; Zitvogel, L.; Lanier, L.L.; Yokoyama, W.M.; Ugolini, S. Innate or Adaptive Immunity? The Example of Natural Killer Cells. Science 2011, 331, 44–49. [Google Scholar] [CrossRef] [Green Version]
  69. Vivier, E.; Tomasello, E.; Baratin, M.; Walzer, T.; Ugolini, S. Functions of natural killer cells. Nat. Immunol. 2008, 9, 503–510. [Google Scholar] [CrossRef]
  70. Brenner, M.K.; Heslop, H.; Krance, R.; Horowitz, M.; Strother, D.; Nuchtern, J.; Grilley, B.; Martingano, E.; Cooper, K. Phase I study of chemokine and cytokine gene-modified autologous neuroblastoma cells for treatment of relapsed/refractory neuroblastoma using an adenoviral vector. Hum. Gene Ther. 2000, 11, 1477–1488. [Google Scholar] [CrossRef]
  71. Handgretinger, R.; Baader, P.; Dopfer, R.; Klingebiel, T.; Reuland, P.; Treuner, J.; Reisfeld, R.A.; Niethammer, D. A phase I study of neuroblastoma with the anti-ganglioside GD2 antibody 14.G2a. Cancer Immunol. Immunother. 1992, 35, 199–204. [Google Scholar] [CrossRef]
  72. Kushner, B.H.; Kramer, K.; Cheung, N.K. Phase II trial of the anti-G(D2) monoclonal antibody 3F8 and granulocyte-macrophage colony-stimulating factor for neuroblastoma. J. Clin. Oncol. 2001, 19, 4189–4194. [Google Scholar] [CrossRef]
  73. 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]
  74. Rousseau, R.F.; Haight, A.E.; Hirschmann-Jax, C.; Yvon, E.S.; Rill, D.R.; Mei, Z.; Smith, S.C.; Inman, S.; Cooper, K.; Alcoser, P.; et al. Local and systemic effects of an allogeneic tumor cell vaccine combining transgenic human lymphotactin with interleukin-2 in patients with advanced or refractory neuroblastoma. Blood 2003, 101, 1718–1726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Moretta, L.; Moretta, A. Killer immunoglobulin-like receptors. Curr. Opin. Immunol. 2004, 16, 626–633. [Google Scholar] [CrossRef] [PubMed]
  76. Castriconi, R.; Dondero, A.; Corrias, M.V.; Lanino, E.; Pende, D.; Moretta, L.; Bottino, C.; Moretta, A. Natural killer cell-mediated killing of freshly isolated neuroblastoma cells: Critical role of DNAX accessory molecule-1-poliovirus receptor interaction. Cancer Res. 2004, 64, 9180–9184. [Google Scholar] [CrossRef] [Green Version]
  77. Castriconi, R.; Dondero, A.; Augugliaro, R.; Cantoni, C.; Carnemolla, B.; Sementa, A.R.; Negri, F.; Conte, R.; Corrias, M.V.; Moretta, L.; et al. Identification of 4Ig-B7-H3 as a neuroblastoma-associated molecule that exerts a protective role from an NK cell-mediated lysis. Proc. Natl. Acad. Sci. USA 2004, 101, 12640–12645. [Google Scholar] [CrossRef] [Green Version]
  78. Castriconi, R.; Dondero, A.; Cilli, M.; Ognio, E.; Pezzolo, A.; De Giovanni, B.; Gambini, C.; Pistoia, V.; Moretta, L.; Moretta, A.; et al. Human NK cell infusions prolong survival of metastatic human neuroblastoma-bearing NOD/scid mice. Cancer Immunol. Immunother. 2007, 56, 1733–1742. [Google Scholar] [CrossRef]
  79. Liu, Y.; Wu, H.W.; Sheard, M.A.; Sposto, R.; Somanchi, S.S.; Cooper, L.J.; Lee, D.A.; Seeger, R.C. Growth and activation of natural killer cells ex vivo from children with neuroblastoma for adoptive cell therapy. Clin. Cancer Res. 2013, 19, 2132–2143. [Google Scholar] [CrossRef] [Green Version]
  80. Barry, W.E.; Jackson, J.R.; Asuelime, G.E.; Wu, H.W.; Sun, J.; Wan, Z.; Malvar, J.; Sheard, M.A.; Wang, L.; Seeger, R.C.; et al. Activated Natural Killer Cells in Combination with Anti-GD2 Antibody Dinutuximab Improve Survival of Mice after Surgical Resection of Primary Neuroblastoma. Clin. Cancer Res. 2019, 25, 325–333. [Google Scholar] [CrossRef] [Green Version]
  81. Tran, H.C.; Wan, Z.; Sheard, M.A.; Sun, J.; Jackson, J.R.; Malvar, J.; Xu, Y.; Wang, L.; Sposto, R.; Kim, E.S.; et al. TGFβR1 Blockade with Galunisertib (LY2157299) Enhances Anti-Neuroblastoma Activity of the Anti-GD2 Antibody Dinutuximab (ch14.18) with Natural Killer Cells. Clin. Cancer Res. 2017, 23, 804–813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Federico, S.M.; McCarville, M.B.; Shulkin, B.L.; Sondel, P.M.; Hank, J.A.; Hutson, P.; Meagher, M.; Shafer, A.; Ng, C.Y.; Leung, W.; et al. A Pilot Trial of Humanized Anti-GD2 Monoclonal Antibody (hu14.18K322A) with Chemotherapy and Natural Killer Cells in Children with Recurrent/Refractory Neuroblastoma. Clin. Cancer Res. 2017, 23, 6441–6449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Talleur, A.C.; Triplett, B.M.; Federico, S.; Mamcarz, E.; Janssen, W.; Wu, J.; Shook, D.; Leung, W.; Furman, W.L. Consolidation Therapy for Newly Diagnosed Pediatric Patients with High-Risk Neuroblastoma Using Busulfan/Melphalan, Autologous Hematopoietic Cell Transplantation, Anti-GD2 Antibody, Granulocyte-Macrophage Colony–Stimulating Factor, Interleukin-2, and Haploidentical Natural Killer Cells. Biol. Blood Marrow Transplant. 2017, 23, 1910–1917. [Google Scholar] [CrossRef]
  84. Nguyen, R.; Sahr, N.; Sykes, A.; McCarville, M.B.; Federico, S.M.; Sooter, A.; Cullins, D.; Rooney, B.; Janssen, W.E.; Talleur, A.C.; et al. Longitudinal NK cell kinetics and cytotoxicity in children with neuroblastoma enrolled in a clinical phase II trial. J. Immunother. Cancer 2020, 8, e000176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Modak, S.; Le Luduec, J.B.; Cheung, I.Y.; Goldman, D.A.; Ostrovnaya, I.; Doubrovina, E.; Basu, E.; Kushner, B.H.; Kramer, K.; Roberts, S.S.; et al. Adoptive immunotherapy with haploidentical natural killer cells and Anti-GD2 monoclonal antibody m3F8 for resistant neuroblastoma: Results of a phase I study. Oncoimmunology 2018, 7, e1461305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Heinze, A.; Grebe, B.; Bremm, M.; Huenecke, S.; Munir, T.A.; Graafen, L.; Frueh, J.T.; Merker, M.; Rettinger, E.; Soerensen, J.; et al. The Synergistic Use of IL-15 and IL-21 for the Generation of NK Cells From CD3/CD19-Depleted Grafts Improves Their ex vivo Expansion and Cytotoxic Potential Against Neuroblastoma: Perspective for Optimized Immunotherapy Post Haploidentical Stem Cell Transplantation. Front. Immunol. 2019, 10, 2816. [Google Scholar] [CrossRef] [Green Version]
  87. Kronenberg, M.; Gapin, L. The unconventional lifestyle of NKT cells. Nat. Rev. Immunol. 2002, 2, 557–568. [Google Scholar] [CrossRef]
  88. Berzofsky, J.A.; Terabe, M. The contrasting roles of NKT cells in tumor immunity. Curr. Mol. Med. 2009, 9, 667–672. [Google Scholar] [CrossRef] [Green Version]
  89. Swann, J.; Crowe, N.Y.; Hayakawa, Y.; Godfrey, D.I.; Smyth, M.J. Regulation of antitumour immunity by CD1d-restricted NKT cells. Immunol. Cell Biol. 2004, 82, 323–331. [Google Scholar] [CrossRef]
  90. Swann, J.B.; Coquet, J.M.; Smyth, M.J.; Godfrey, D.I. CD1-restricted T cells and tumor immunity. Curr. Top. Microbiol. Immunol. 2007, 314, 293–323. [Google Scholar] [CrossRef]
  91. Dhodapkar, M.V.; Geller, M.D.; Chang, D.H.; Shimizu, K.; Fujii, S.-I.; 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]
  92. Muhammad Ali Tahir, S.; Cheng, O.; Shaulov, A.; Koezuka, Y.; Bubley, G.J.; Wilson, S.B.; Balk, S.P.; Exley, M.A. Loss of IFN-γ Production by Invariant NK T Cells in Advanced Cancer. J. Immunol. 2001, 167, 4046–4050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Yanagisawa, K.; Seino, K.-i.; Ishikawa, Y.; Nozue, M.; Todoroki, T.; Fukao, K. Impaired Proliferative Response of Vα24 NKT Cells from Cancer Patients Against α-Galactosylceramide. J. Immunol. 2002, 168, 6494–6499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Sabry, M.; Lowdell, M.W. Killers at the crossroads: The use of innate immune cells in adoptive cellular therapy of cancer. STEM CELLS Transl. Med. 2020, 9, 974–984. [Google Scholar] [CrossRef] [PubMed]
  95. Liu, D.; Song, L.; Wei, J.; Courtney, A.N.; Gao, X.; Marinova, E.; Guo, L.; Heczey, A.; Asgharzadeh, S.; Kim, E.; et al. IL-15 protects NKT cells from inhibition by tumor-associated macrophages and enhances antimetastatic activity. J. Clin. Investig. 2012, 122, 2221–2233. [Google Scholar] [CrossRef] [Green Version]
  96. 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]
  97. Hartmann, J.; Schüßler-Lenz, M.; Bondanza, A.; Buchholz, C.J. Clinical development of CAR T cells—challenges and opportunities in translating innovative treatment concepts. EMBO Mol. Med. 2017, 9, 1183–1197. [Google Scholar] [CrossRef]
  98. Majzner, R.G.; Mackall, C.L. Clinical lessons learned from the first leg of the CAR T cell journey. Nat. Med. 2019, 25, 1341–1355. [Google Scholar] [CrossRef]
  99. Yazdanifar, M.; Barbarito, G.; Bertaina, A.; Airoldi, I. γδ T Cells: The Ideal Tool for Cancer Immunotherapy. Cells 2020, 9, 1305. [Google Scholar] [CrossRef]
  100. Yazdanifar, M.; Zhou, R.; Mukherjee, P. Emerging immunotherapeutics in adenocarcinomas: A focus on CAR-T cells. Curr. Trends Immunol. 2016, 17, 95–115. [Google Scholar] [PubMed]
  101. Allegra, A.; Innao, V.; Gerace, D.; Vaddinelli, D.; Musolino, C. Adoptive immunotherapy for hematological malignancies: Current status and new insights in chimeric antigen receptor T cells. Blood Cells Mol. Dis. 2016, 62, 49–63. [Google Scholar] [CrossRef] [PubMed]
  102. Chavez, J.C.; Bachmeier, C.; Kharfan-Dabaja, M.A. CAR T-cell therapy for B-cell lymphomas: Clinical trial results of available products. Ther. Adv. Hematol. 2019, 10, 2040620719841581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Li, J.; Li, W.; Huang, K.; Zhang, Y.; Kupfer, G.; Zhao, Q. Chimeric antigen receptor T cell (CAR-T) immunotherapy for solid tumors: Lessons learned and strategies for moving forward. J. Hematol. Oncol. 2018, 11, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Gargett, T.; Brown, M.P. The inducible caspase-9 suicide gene system as a “safety switch” to limit on-target, off-tumor toxicities of chimeric antigen receptor T cells. Front. Pharmacol. 2014, 5, 235. [Google Scholar] [CrossRef] [PubMed]
  105. Iuliucci, J.D.; Oliver, S.D.; Morley, S.; Ward, C.; Ward, J.; Dalgarno, D.; Clackson, T.; Berger, H.J. Intravenous safety and pharmacokinetics of a novel dimerizer drug, AP1903, in healthy volunteers. J. Clin. Pharmacol. 2001, 41, 870–879. [Google Scholar] [CrossRef]
  106. 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]
  107. 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]
  108. 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] [Green Version]
  109. Heczey, A.; Louis, C.U.; Savoldo, B.; Dakhova, O.; Durett, A.; Grilley, B.; Liu, H.; Wu, M.F.; Mei, Z.; 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]
  110. Louis, C.U.; Savoldo, B.; Dotti, G.; Pule, M.; Yvon, E.; Myers, G.D.; Rossig, C.; Russell, H.V.; Diouf, O.; Liu, E.; 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]
  111. Quintarelli, C.; Orlando, D.; Boffa, I.; Guercio, M.; Polito, V.A.; Petretto, A.; Lavarello, C.; Sinibaldi, M.; Weber, G.; Del Bufalo, F.; et al. Choice of costimulatory domains and of cytokines determines CAR T-cell activity in neuroblastoma. Oncoimmunology 2018, 7, e1433518. [Google Scholar] [CrossRef] [PubMed]
  112. 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] [PubMed] [Green Version]
  113. Künkele, 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] [PubMed] [Green Version]
  114. Sawaisorn, P.; Atjanasuppat, K.; Anurathapan, U.; Chutipongtanate, S.; Hongeng, S. Strategies to Improve Chimeric Antigen Receptor Therapies for Neuroblastoma. Vaccines 2020, 8, 753. [Google Scholar] [CrossRef] [PubMed]
  115. Vermijlen, D.; Gatti, D.; Kouzeli, A.; Rus, T.; Eberl, M. γδ T cell responses: How many ligands will it take till we know? Semin. Cell Dev. Biol. 2018, 84, 75–86. [Google Scholar] [CrossRef] [PubMed]
  116. Minculescu, L.; Sengeløv, H. The role of gamma delta T cells in haematopoietic stem cell transplantation. Scand. J. Immunol. 2015, 81, 459–468. [Google Scholar] [CrossRef]
  117. Davey, M.S.; Willcox, C.R.; Joyce, S.P.; Ladell, K.; Kasatskaya, S.A.; McLaren, J.E.; Hunter, S.; Salim, M.; Mohammed, F.; Price, D.A.; et al. Clonal selection in the human Vδ1 T cell repertoire indicates γδ TCR-dependent adaptive immune surveillance. Nat. Commun. 2017, 8, 14760. [Google Scholar] [CrossRef]
  118. Nussbaumer, O.; Koslowski, M. The emerging role of γδ T cells in cancer immunotherapy. Immuno-Oncol. Technol. 2019, 1, 3–10. [Google Scholar] [CrossRef] [Green Version]
  119. Brandes, M.; Willimann, K.; Moser, B. Professional antigen-presentation function by human gammadelta T Cells. Science 2005, 309, 264–268. [Google Scholar] [CrossRef]
  120. 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] [Green Version]
  121. Silva-Santos, B.; Serre, K.; Norell, H. γδ T cells in cancer. Nat. Rev. Immunol. 2015, 15, 683–691. [Google Scholar] [CrossRef] [PubMed]
  122. Morandi, F.; Yazdanifar, M. Engineering the Bridge between Innate and Adaptive Immunity for Cancer Immunotherapy: Focus on γδ T and NK Cells. Cells 2020, 9, 1757. [Google Scholar] [CrossRef] [PubMed]
  123. Lo Presti, E.; Pizzolato, G.; Corsale, A.M.; Caccamo, N.; Sireci, G.; Dieli, F.; Meraviglia, S. γδ T Cells and Tumor Microenvironment: From Immunosurveillance to Tumor Evasion. Front. Immunol. 2018, 9, 1395. [Google Scholar] [CrossRef] [PubMed]
  124. Mirzaei, H.R.; Mirzaei, H.; Lee, S.Y.; Hadjati, J.; Till, B.G. Prospects for chimeric antigen receptor (CAR) γδ T cells: A potential game changer for adoptive T cell cancer immunotherapy. Cancer Lett. 2016, 380, 413–423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Polito, V.A.; Cristantielli, R.; Weber, G.; Del Bufalo, F.; Belardinilli, T.; Arnone, C.M.; Petretto, A.; Antonucci, L.; Giorda, E.; Tumino, N.; et al. Universal Ready-to-Use Immunotherapeutic Approach for the Treatment of Cancer: Expanded and Activated Polyclonal γδ Memory T Cells. Front. Immunol. 2019, 10, 2717. [Google Scholar] [CrossRef] [Green Version]
  126. Buccheri, S.; Guggino, G.; Caccamo, N.; Li Donni, P.; Dieli, F. Efficacy and safety of γδT cell-based tumor immunotherapy: A meta-analysis. J. Biol. Regul. Homeost. Agents 2014, 28, 81–90. [Google Scholar]
  127. Rezvani, K.; Rouce, R.; Liu, E.; Shpall, E. Engineering Natural Killer Cells for Cancer Immunotherapy. Mol. Ther. 2017, 25, 1769–1781. [Google Scholar] [CrossRef]
  128. Boissel, L.; Betancur-Boissel, M.; Lu, W.; Krause, D.S.; Van Etten, R.A.; Wels, W.S.; Klingemann, H. Retargeting NK-92 cells by means of CD19- and CD20-specific chimeric antigen receptors compares favorably with antibody-dependent cellular cytotoxicity. Oncoimmunology 2013, 2, e26527. [Google Scholar] [CrossRef] [Green Version]
  129. Herrera, L.; Santos, S.; Vesga, M.A.; Anguita, J. Adult peripheral blood and umbilical cord blood NK cells are good sources for effective CAR therapy against CD19 positive leukemic cells. Sci. Rep. 2019, 9, 18729. [Google Scholar] [CrossRef] [Green Version]
  130. Tang, X.; Yang, L.; Li, Z.; Nalin, A.P.; Dai, H.; Xu, T.; Yin, J.; You, F.; Zhu, M.; Shen, W.; et al. First-in-man clinical trial of CAR NK-92 cells: Safety test of CD33-CAR NK-92 cells in patients with relapsed and refractory acute myeloid leukemia. Am. J. Cancer Res. 2018, 8, 1083–1089. [Google Scholar]
  131. You, F.; Wang, Y.; Jiang, L.; Zhu, X.; Chen, D.; Yuan, L.; An, G.; Meng, H.; Yang, L. A novel CD7 chimeric antigen receptor-modified NK-92MI cell line targeting T-cell acute lymphoblastic leukemia. Am. J. Cancer Res. 2019, 9, 64–78. [Google Scholar] [PubMed]
  132. Colamartino, A.B.L.; Lemieux, W.; Bifsha, P.; Nicoletti, S.; Chakravarti, N.; Sanz, J.; Roméro, H.; Selleri, S.; Béland, K.; Guiot, M.; et al. Efficient and Robust NK-Cell Transduction With Baboon Envelope Pseudotyped Lentivector. Front. Immunol. 2019, 10, 2873. [Google Scholar] [CrossRef] [PubMed]
  133. Zhang, C.; Burger, M.C.; Jennewein, L.; Genßler, S.; Schönfeld, K.; Zeiner, P.; Hattingen, E.; Harter, P.N.; Mittelbronn, M.; Tonn, T.; et al. ErbB2/HER2-Specific NK Cells for Targeted Therapy of Glioblastoma. J. Natl. Cancer Inst. 2016, 108. [Google Scholar] [CrossRef] [PubMed]
  134. Chen, X.; Han, J.; Chu, J.; Zhang, L.; Zhang, J.; Chen, C.; Chen, L.; Wang, Y.; Wang, H.; Yi, L.; et al. A combinational therapy of EGFR-CAR NK cells and oncolytic herpes simplex virus 1 for breast cancer brain metastases. Oncotarget 2016, 7, 27764–27777. [Google Scholar] [CrossRef] [Green Version]
  135. Han, J.; Chu, J.; Keung Chan, W.; Zhang, J.; Wang, Y.; Cohen, J.B.; Victor, A.; Meisen, W.H.; Kim, S.H.; Grandi, P.; et al. CAR-Engineered NK Cells Targeting Wild-Type EGFR and EGFRvIII Enhance Killing of Glioblastoma and Patient-Derived Glioblastoma Stem Cells. Sci. Rep. 2015, 5, 11483. [Google Scholar] [CrossRef]
  136. Pinz, K.G.; Yakaboski, E.; Jares, A.; Liu, H.; Firor, A.E.; Chen, K.H.; Wada, M.; Salman, H.; Tse, W.; Hagag, N.; et al. Targeting T-cell malignancies using anti-CD4 CAR NK-92 cells. Oncotarget 2017, 8, 112783–112796. [Google Scholar] [CrossRef] [Green Version]
  137. Zhang, E.; Yang, P.; Gu, J.; Wu, H.; Chi, X.; Liu, C.; Wang, Y.; Xue, J.; Qi, W.; Sun, Q.; et al. Recombination of a dual-CAR-modified T lymphocyte to accurately eliminate pancreatic malignancy. J. Hematol. Oncol. 2018, 11, 102. [Google Scholar] [CrossRef] [Green Version]
  138. Yu, M.; Luo, H.; Fan, M.; Wu, X.; Shi, B.; Di, S.; Liu, Y.; Pan, Z.; Jiang, H.; Li, Z. Development of GPC3-Specific Chimeric Antigen Receptor-Engineered Natural Killer Cells for the Treatment of Hepatocellular Carcinoma. Mol. Ther. 2018, 26, 366–378. [Google Scholar] [CrossRef] [Green Version]
  139. Chu, J.; Deng, Y.; Benson, D.M.; He, S.; Hughes, T.; Zhang, J.; Peng, Y.; Mao, H.; Yi, L.; Ghoshal, K.; et al. CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma. Leukemia 2014, 28, 917–927. [Google Scholar] [CrossRef] [Green Version]
  140. Zhang, Q.; Zhang, H.; Ding, J.; Liu, H.; Li, H. Combination Therapy with EpCAM-CAR-NK-92 Cells and Regorafenib against Human Colorectal Cancer Models. J. Immunol. Res. 2018, 2018, 4263520. [Google Scholar] [CrossRef] [Green Version]
  141. Esser, R.; Müller, T.; Stefes, D.; Kloess, S.; Seidel, D.; Gillies, S.D.; Aperlo-Iffland, C.; Huston, J.S.; Uherek, C.; Schönfeld, K.; et al. NK cells engineered to express a GD2-specific antigen receptor display built-in ADCC-like activity against tumour cells of neuroectodermal origin. J. Cell. Mol. Med. 2012, 16, 569–581. [Google Scholar] [CrossRef] [PubMed]
  142. Romanski, A.; Uherek, C.; Bug, G.; Seifried, E.; Klingemann, H.; Wels, W.S.; Ottmann, O.G.; Tonn, T. CD19-CAR engineered NK-92 cells are sufficient to overcome NK cell resistance in B-cell malignancies. J. Cell. Mol. Med. 2016, 20, 1287–1294. [Google Scholar] [CrossRef] [PubMed]
  143. Kailayangiri, S.; Altvater, B.; Spurny, C.; Jamitzky, S.; Schelhaas, S.; Jacobs, A.H.; Wiek, C.; Roellecke, K.; Hanenberg, H.; Hartmann, W.; et al. Targeting Ewing sarcoma with activated and GD2-specific chimeric antigen receptor-engineered human NK cells induces upregulation of immune-inhibitory HLA-G. Oncoimmunology 2017, 6, e1250050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. NB immunotherapy based on antibodies and mechanisms of action. The figure describes mechanisms of action of antibodies currently used in clinical settings. These antibodies specific for NB-associated antigens may induce direct cell death, complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC). In addition, cytokines such as IL-2 and GM-CSF may increase the anti-tumor activity exerted by these antibodies.
Figure 1. NB immunotherapy based on antibodies and mechanisms of action. The figure describes mechanisms of action of antibodies currently used in clinical settings. These antibodies specific for NB-associated antigens may induce direct cell death, complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC). In addition, cytokines such as IL-2 and GM-CSF may increase the anti-tumor activity exerted by these antibodies.
Vaccines 09 00043 g001
Figure 2. Current adoptive cell therapies in the clinical setting of neuroblastoma (NB) treatment. This figure describes different adoptive cell therapy approaches currently investigated for immunotherapy of high-risk NB patients. This includes CAR T cells and NK cells as adjuvant therapy, used alone or in combination with cytokines. Cytokines may also be used to expand NK cells before infusion.
Figure 2. Current adoptive cell therapies in the clinical setting of neuroblastoma (NB) treatment. This figure describes different adoptive cell therapy approaches currently investigated for immunotherapy of high-risk NB patients. This includes CAR T cells and NK cells as adjuvant therapy, used alone or in combination with cytokines. Cytokines may also be used to expand NK cells before infusion.
Vaccines 09 00043 g002
Table 1. International Neuroblastoma Risk Group Pretreatment Classification Scheme.
Table 1. International Neuroblastoma Risk Group Pretreatment Classification Scheme.
NRG StageAge (Months)Histologic CategoryGrade of Tumor DifferentiationMYCN11q AberrationPloidyPretreatment Risk Group
L1/L2 GN maturing, GNB intermixed A (very low)
L1 Any, except GN maturing or GNB intermixed NA B (very low)
Amplified K (high)
L2<18Any, except GN maturing or GNB intermixed NANo D (low)
Yes G (intermediate)
≥18GNB nodular, neuroblastomaDifferentiatingNANo E (low)
Yes H (intermediate)
Poorly differentiated or undifferentiatedNA H (intermediate)
Amplified N (high)
M<18 NA HyperdiploidF (low)
<12 NA DiploidI (intermediate)
12 < 18 NA DiploidJ (intermediate)
<18 Amplified O (high)
≥18 P (high)
MS<18 NANo C (very low)
Yes Q (high)
Amplified R (high)
Abbreviations: GN = ganglioneuroma; GNB = ganglioneuroblastoma; INRG = International Neuroblastoma Risk Group; NA = not amplified.
Table 2. Clinical trials using granulocyte-macrophage colony stimulating factor (GM-CSF) and anti-GD2 in the last ten years.
Table 2. Clinical trials using granulocyte-macrophage colony stimulating factor (GM-CSF) and anti-GD2 in the last ten years.
IdentifierStudy TitlePhaseStart DatePatients enrolledStatusPrimary Aims and References
NCT011838973F8/GM-CSF immunotherapy plus 13-cis-retinoic acid for primary refractory Neuroblastoma in bone marrowII201031CompletedTo find out what effects, good and/or bad, the combination of 3F8 and GM-CSF had on the patient and the cancer
Another purpose was to see if high-dose 3F8 combined with GM-CSF is better than standard dose 3F8 in treating neuroblastoma
Kushner BH et al. Cancer. 2013.
NCT011834293F8/GM-CSF immunotherapy plus 13-cis-retinoic acid for consolidation of first remission after non-myeloablative therapy in patients with high-risk neuroblastomaII201039Completed
NCT01183416High-Dose 3F8/GM-CSF immunotherapy plus 13-Cis-Retinoic acid for consolidation of first remission after myeloablative therapy and autologous stem-cell transplantationII20104Completed
NCT01041638Monoclonal antibody Ch14.18, sargramostim, aldesleukin and isotretinoin after autologous stem cell transplant in treating patients with neuroblastomaIII2010105Active, not recruitingTo study the side effects of giving monoclonal antibody Ch14.18 together with sargramostim, aldesleukin and isotretinoin after autologous stem cell transplant in treating patients with neuroblastoma
NCT01334515Biological therapy, sargramostim and isotretinoinin treating patients with relapsed or refractory neuroblastomaII201152CompletedTo analyse how well hu14.18-interleukin-2 (IL2) fusion protein works when given together with sargramostim and isotretinoin in treating patients with relapsed or refractory neuroblastoma (Shusteman S et al. Clin Cancer Res. 2019)
NCT01757626Combination therapy of antibody Hu3F8 with GM-CSF in patients with relapsed/refractory high-risk neuroblastomaI/II2012224RecruitingTo find out if an antibody called Humanized 3F8 (Hu3F8) combined with granulocyte-macrophage colony stimulating factor is safe for treating neuroblastoma (Kushner BH et al. JAMA Oncol. 2018).
NCT01592045ch14.18 Pharmacokinetic study in high-risk neuroblastomaI/II201228CompletedTo compare the pharmacokinetics (blood levels) and safety of chimeric 14.18 manufactured by two independent drug makers
NCT01767194Irinotecan hydrochloride and temozolomide with temsirolimus or dinutuximab in treating younger patients with refractory or relapsed neuroblastomaII201373CompletedTo investigate how well irinotecan hydrochloride and temozolomide with temsirolimus or dinutuximab work in treating younger patients with neuroblastoma that has returned or does not respond to treatment.
Mody R et al. Lancet Oncol. 2017.
NCT02100930Anti-GD2 3F8 monoclonal antibody and GM-CSF for high-risk neuroblastomaNA201469CompletedTo supply an experimental combination of drugs called 3F8 and sargramostim to patients with high-risk neuroblastoma who may benefit from treatment.
NCT02173093Activated T cells armed with GD2 bispecific antibody in children and young adults with neuroblastoma and osteosarcomaI/II201440RecruitingTo study the side effects and best dose of activated T cells armed with GD2 bispecific antibody and how well they work in treating patients with neuroblastoma, osteosarcoma and other GD2-positive solid tumors.
NCT02130869A pilot study of immunotherapy including haploidentical NK cell infusion following CD133+ positively-selected autologous hematopoietic stem cells in children with high risk solid tumors or lymphomasI20148CompletedIn NB, to establish NK cell engraftment in patients receiving high-dose chemotherapy, stem cell infusion, hu14.18K322A, IL-2, haploidentical natural killer cell infusion, G-CSF and GM-CSF.
NCT02786719High-risk neuroblastoma chemotherapy without G-CSFNA201613CompletedWhittle SB et al. Blood Cancer. 2020
NCT03363373Naxitamab for high-risk neuroblastoma patients with primary refractory disease or incomplete response to salvage treatment in bone and/or bone marrowII201795RecruitingTo define the effects of naxitamab and GM-CSF in children diagnosed with high-risk neuroblastoma with primary refractory disease or incomplete response to salvage treatment
NCT03033303A study of the effect of Hu3F8/GM-CSF immunotherapy plus isotretinoin in patients in first remission of high-risk neuroblastomaII201759RecruitingTo test the combined effects of Humanized 3F8 in combination with granulocyte-macrophage colony stimulating factor
NCT03189706Study of chemoimmunotherapy for high-risk neuroblastomaI201762RecruitingTo test the efficacy and safety of Hu3F8 combined with the chemotherapy drugs irinotecan and temozolomide or GM-CSF
NCT04211675NK cells infusions with irinotecan, temozolomide, and dinutuximabI/II201931Not yet recruitingPhase 1: to assess the safety and tolerability of autologous expanded NK cells in combination with irinotecan, temozolomide and dinituximab.
Phase 2: to estimate the response to treatment
NCT03794349Irinotecan hydrochloride, temozolomide, and dinutuximab with or without eflornithine in treating patients with relapsed or refractory neuroblastomaII201995RecruitingTo study how well irinotecan hydrochloride, temozolomide and dinutuximab work with or without eflornithine in patients with relapsed or refractory neuroblastoma
NCT04560166IT with or without naxitamab and GM CSF in patients with high-risk neuroblastomaIII2020117Not yet recruitingThis is an open label, randomized, controlled, multicenter phase 3 trial, in patients ≥ 12 months of age with high-risk NB with primary refractory disease or in first relapse
NCT04385277Treatment with dinutuximab, sargramostim (GM-CSF), and isotretinoin in combination with irinotecan and temozolomide after intensive therapy for people with high-risk neuroblastoma (NBL)II202045Not yet recruitingSafety of dinutuximab, GM-CSF and isotretinoin in combination with irinotecan and temozolomide, in high-risk neuroblastoma patients after consolidation therapy
Abbreviations: 3F8, a murine immunoglobulin 3 monoclonal antibody specific for disialoganglioside; Hu3F8, Humanized murine IgG3 anti-GD2 antibody m3F8; GM-CSF, subcutaneous granulocyte-macrophage-colony-stimulating factor.
Table 3. Phase I/II Clinical trials of NK Immunotherapy.
Table 3. Phase I/II Clinical trials of NK Immunotherapy.
IdentifierStudy TitlePhaseStart DatePatients EnrolledStatusPrimary Aims and References
NCT01386619NK DLI in patients after human leukocyte antigen (HLA)-haploidentical hematopoietic stem cell transplantation (HSCT)I/II200415CompletedTo analyse the feasibility of expanded NK-cell DLI production and their safety, by evaluating regards transfusion associated adverse events and the absence of acute graft-versus-host disease 30 days after the last NK DLI infusion. The efficacy of NK DLI infusions has been also assessed by evaluating the rates of overall and disease free survival and the rate of disease relapse compared to patients treated with haploidentical HSCT without NK DLI infusions.
NCT00569283Donor natural killer cell infusion in preventing relapse or graft failure in patients who have undergone donor bone marrow transplantI200718CompletedTo evaluate the safety of donor natural killer (NK) cells, given as a single intravenous infusion in patients that underwent HLA-haploidentical familial donor bone marrow transplantation and to determine the maximum tolerated dose of donor NK cells infusion. The effectiveness of donor NK cell infusion was evaluated as ability to prevent tumor relapse and graft failure.
NCT00877110Anti-GD2 3F8 antibody and allogeneic natural killer cells for high-risk neuroblastomaI200971CompletedTo assess the feasibility and safety of administering allogeneic haploidentical NK infusions with mAb 3F8 in patients with high-risk NB. Moreover, the efficacy of allogeneic NK infusions plus 3F8 was evaluated as anti-tumor activity. The impact of KIR/HLA immunogenetics and CD16 polymorphism on disease response to NK/3F8 was also evaluated.
NCT01462396Allogeneic stem cell transplantation for advanced neuroblastoma using MHC mismatched related donorsI20114CompletedTo evaluate the safety of a fludarabine based reduced intensity conditioning regimen and CD34+ stem cell selected mis-matched, related, allogeneic transplant in patients with relapsed/refractory neuroblastoma, by monitoring mortality, toxicity, acute and chronic graft versus host disease and engraftment rate. Anti-tumor effect has been also evaluated.
NCT01287104A phase I study of NK cell infusion following allogeneic peripheral blood stem cell transplantation from related or matched unrelated donors in pediatric patients with solid tumors and leukemiasI201134CompletedTo assess the feasibility and toxicity of infusing escalating doses of donor-derived activated NK cell donor lymphocyte infusions (NK-DLI) following human leukocyte antigen (HLA)-matched T cell depleted (TCD) peripheral blood stem cell transplant (PBSCT) in patients with metastatic or recurrent pediatric solid tumors and high risk leukemias who have unrelated donors or related donors. In addition, donor engraftment and acute graft versus host disease have been evaluated.
Shah NN et al. Blood. 2015.
NCT01386619NK DLI in patients after human leukocyte antigen (HLA)-haploidentical hematopoietic stem cell transplantation (HSCT)I/II201115CompletedTo evaluate the feasibility of NK-DLI production. The safety of NK DLI Infusion has been evaluated as transfusion associated adverse events (fever, fall in blood pressure, transfusion site reactions, etc) at the time of NK DLI infusion. The primary long-term safety measure is the absence of acute graft-versus-host disease 30 days after the last NK DLI infusion. The efficacy of NK DLI infusions has been assessed as rate of overall and disease free survival and disease relapse.
NCT01576692Combination chemotherapy, monoclonal antibody, and natural killer cells in treating young patients with recurrent or refractory neuroblastomaI201234CompletedTo evaluate the toxicity associated with humanized anti-GD2 antibody/chemotherapy associated or not with NK cells infusion. Clinical outcome was measured as response to therapy using response evaluation criteria in solid tumors, clearing of bone marrow and improvement in MIBG scans. Event-free and overall survival have been also analyzed.
NCT01701479Long term continuous infusion ch14.18/CHO plus s.c. aldesleukin (IL-2)I/II2012288Active, not recruitingTo find a way of giving ch14.18/CHO, in combination with subcutaneous aldesleukin (IL-2) and oral isotretinoin (13-cis-RA), to children and young people with primary refractory or relapsed neuroblastoma without intravenous morphine.
NCT01875601NK white blood cells and interleukin in children and young adults with advanced solid tumorsI201316CompletedTo evaluate the feasibility of harvesting and expanding activated NK cells and to assess the toxicity of infusing escalating doses of activated NK cells following lymphodepleting chemotherapy with or without escalating doses of rhIL15 in pediatric patients with refractory malignant solid tumors.
Kontny HU et al. Cell Death Differ. 2001.
Yu AL et al. N Engl J Med. 2010.
Dudley ME et al. Nat Rev Cancer. 2003.
NCT01857934Therapy for children with advanced stage neuroblastomaII2013153Active, not recruitingTo analyze event-free survival of patients with newly diagnosed high-risk NB treated with hu14.18K322A in addition to standard treatment. In addition, the tolerability of hu14.18K322A with allogeneic natural killer (NK) cells from an acceptable parent, in the immediate post-transplant period will be evaluated. Tolerability of hu14.18K322A with interleukin-2 and GM-CSF as treatment for minimal residual disease (MRD) will be assessed.
Nguyen R et al. J Immunother Cancer. 2020.
NCT01807468Haploidentical stem cell transplantation and NK cell therapy in patients with high-risk solid tumorsII201312Active, not recruitingTo evaluate feasibility and efficacy of haploidentical stem cell transplantation followed (or not) by NK cell infusion in patients with high-risk solid tumors who failed after tandem high-dose chemotherapy and autologous stem cell transplantation.
NCT02130869A pilot study of immunotherapy including haploidentical NK cell infusion following CD133+positively-selected autologous hematopoietic stem cells in children with high risk solid tumors or lymphomasI20148CompletedTo investigate the addition of haploidentical natural killer (NK) cell infusion to high dose chemotherapy and autologous stem cell transplantation in children with high-risk solid tumors. In patients with neuroblastoma, the anti-GD2 antibody hu14.18K322A has been also given. Survival of children treated with this approach has been analyzed.
NCT02100891Phase 2 STIR trial: Haploidentical transplant and donor natural killer cells for solid tumorsII201415Active, not recruitingIn this study, patients with high-risk solid tumors (Ewings sarcoma, neuroblastoma and rhabdomyosarcoma) underwent haploidentical hematopoietic cell transplantation (HCT) followed by an early, post-transplant infusion of donor natural killer (NK) cells. Safety and efficacy will be analyzed. Efficacy will be evaluated as complete (CR) and partial (PR) response and stable disease (SD).
Hattinger CM et al. Expert Opin Emerg Drugs. 2019.
NCT02573896Immunotherapy of relapsed refractory neuroblastoma with expanded NK cellsI201524RecruitingTo determine the maximum tolerated dose of autologous expanded natural killer (NK) cells when combined with standard dosing of ch14.18 and will assess the feasibility of adding lenalidomide at the recommended Phase II dose of the expanded NK cells with ch14.18, for treatment of children with refractory or recurrent neuroblastoma.
NCT02650648Humanized anti-GD2 antibody Hu3F8 and allogeneic natural killer cells for high-risk neuroblastomaI201685Active, not recruitingTo see if it is safe and feasible to give the participant cyclophosphamide, natural killer (NK) cells and Hu3F8 antibody as a treatment for neuroblastoma.
NCT02508038Alpha/Beta CD19+Depleted haploidentical transplantation+zometa for pediatric hematologic malignancies and solid tumorsI201622RecruitingTo study safety of transplantation with a haploidentical donor peripheral blood stem cell graft depleted of TCRαβ+ and CD19+ cells in conjunction with the immunomodulating drug Zoledronate, given in the post-transplant period to treat pediatric patients with relapsed or refractory hematologic malignancies or high risk solid tumors.
NCT03242603Immunotherapy of Neuroblastoma Patients Using a Combination of Anti-GD2 and NK CellsI/II20175UnknownTo measure tumor response after infusion of expanded activated haploidentical NK cells with anti-GD2. Disease response will be defined as complete response/remission (CR), partial response (PR), minor response, stable disease (SD), or progressive disease (PD).
NCT03209869Treatment of relapsed or refractory neuroblastoma with expanded haploidentical NK cells and Hu14.18-IL2I20186SuspendedPatients with relapsed or refractory neuroblastoma received ex vivo expanded and activated natural killer (NK) cells from haploidentical donor with the immunocytokine, hu14.18-IL2. Safety has been evaluated as adverse event and GvHD rate and efficacy was evaluated as progression-free and overall survival and/or objective tumor response.
NCT04211675NK cells infusions with irinotecan, temozolomide, and dinutuximabI/II201931Not recruitingPhase 1: to assess the safety and tolerability of autologous expanded NK cells in combination with irinotecan, temozolomide and dinituximab.
Phase 2: to estimate the response to treatment.
Table 4. Ongoing clinical trials using CAR T cells.
Table 4. Ongoing clinical trials using CAR T cells.
IdentifierStudy TitlePhaseStart DatePatients EnrolledStatusPrimary Aims
NCT00085930Blood T cells and EBV specific CTLs expressing GD2 specific chimeric T cell receptors to neuroblastoma patientsI200419Active,
not recruiting
To evaluate the safety of escalating doses of 14g2a.zeta chimeric receptor transduced autologous EBV specific cytotoxic T-lymphocytes and 14g2a.zeta transduced autologous peripheral blood T cells
NCT018226523rd generation GD-2 chimeric antigen receptor and iCaspase suicide safety switch, neuroblastoma, GRAINI201311Active,
not recruiting
To define the dose limiting toxicities at six weeks post T cell infusion
NCT01953900iC9-GD2-CAR-VZV-CTLs/refractory or metastatic GD2-positive sarcoma and neuroblastomaI201326Active,
not recruiting
To evaluate the safety and feasibility of intravenous injections of autologous iC9-GD2-CAR-VZV-CTLs in combination with VZV vaccination in patients with advanced GD2-positive sarcomas or neuroblastoma
NCT02311621Engineered neuroblastoma cellular immunotherapy (ENCIT)-01I201440RecruitingPatients will be evaluated through day 28 for occurrence of dose limiting toxicity
NCT02761915A cancer research UK trial of anti-GD2 T-cells (1RG-CART)I201627RecruitingTo evaluate (i) the feasibility of 1RG-CART therapy in patients with relapsed or refractory neuroblastoma, (ii) safety and tolerability of 1RG-CART therapy and (iii) to assess the incidence, severity and causality of adverse events to 1RG-CART and/or the lymphodepleting regimen
NCT02919046Study evaluating the efficacy and safety with CAR-T for relapsed or refractory neuroblastoma in childrenNA201622UnknownTo establish the overall efficiency of patients with neuroblastoma after autologous CAR-T cell therapy
NCT02765243Anti-GD2 4th generation CART cells targeting refractory and/or recurrent neuroblastomaI201620RecruitingTo determine the toxicity profile of the 4SCAR-GD2-modified T cells with common toxicity criteria for adverse effects
NCT03373097Anti-GD2 CAR T cells in pediatric patients affected by high risk and/or relapsed/refractory neuroblastoma or other GD2-positive solid tumorsI/II201742RecruitingPhase I—Identification of the dose limiting toxicity
Phase II—Assessment of Antitumor effect and best overall response
NCT03294954GD2 specific CAR and interleukin-15 expressing autologous NKT cells to treat children with neuroblastomaI201724RecruitingTo define the maximum tolerated dose of autologous NKTs expressing a second generation GD2-specific chimeric antigen receptor administered to patients with relapsed or refractory neuroblastoma.
NCT03721068Study of CAR T-Cells targeting the GD2 with IL-15+iCaspase9 for relapsed/refractory neuroblastomaI201818RecruitingTo establish the number of participants with adverse events as a measure of safety and tolerability of iC9.GD2.CAR.IL-15 T cells administered to pediatric subjects with relapsed or refractory neuroblastoma
NCT03635632C7R-GD2.CART cells for patients with relapsed or refractory Neuroblastoma and other GD2 positive cancers (GAIL-N)I201894RecruitingTo determine maximum tolerated dose of C7R-GD2.CART Cells and toxicity.
NCT03618381EGFR806 CAR T cell immunotherapy for recurrent/refractory solid tumors in children and young adultsI201836Recruiting
(1)
To estimate the maximum tolerated dose and dose limiting toxicities, and describe the full toxicity profile of the two CAR T cell products
(2)
The number of successfully manufactured EGFR806 and EGFR806xCD19 CAR T cell products will be assessed
(3)
To establish the safety, defined by adverse events, of EGFR806-specific CAR T cell infusions and of dual transduced EGFR806xCD19 CAR T cell infusions
NCT04483778B7H3 CAR T cell immunotherapy for recurrent/refractory solid tumors in children and young adultsI202068Recruiting
(1)
To assess the safety and tolerability of cellular immunotherapy utilizing ex-vivo expanded autologous T cells genetically modified to express B7H3-specific CAR
(2)
To assess the safety and tolerability of cellular immunotherapy utilizing ex vivo expanded autologous T cells genetically modified to express a bispecific B7H3xCD19 CAR.
(3)
To determine the maximum tolerated dose of B7H3-specific CAR.
(4)
To determine the maximum tolerated dose of bispecific B7H3xCD19 CAR.
(5)
To assess the dose limiting toxicities and describe the full toxicity profile for each study arm type, frequency, severity and duration of adverse events will be tabulated and summarized.
(6)
To assess the feasibility of manufacturing B7H3 specific CARs from patient-derived lymphocytes.
(7)
To assess the feasibility of manufacturing B7H3xCD19 bispecific CARs from patient-derived lymphocytes.
NCT046375034SCAR-T therapy targeting GD2, PSMA and CD276 for treating neuroblastomaI/II2020100RecruitingTo determine the number of patients with adverse events and the toxicity profile
NCT04539366Testing 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 PERSIST trialI202067Not yet recruitingTo define (i) feasibility of producing GD2-CAR-expressing autologous T-lymphocytes (GD2CART), (ii) incidence of adverse events and (iii) maximum tolerated dose and best response to GD2CART cells
Abbreviations: CAR T, Chimeric Antigen Receptor T cells.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Morandi, F.; Sabatini, F.; Podestà, M.; Airoldi, I. Immunotherapeutic Strategies for Neuroblastoma: Present, Past and Future. Vaccines 2021, 9, 43. https://doi.org/10.3390/vaccines9010043

AMA Style

Morandi F, Sabatini F, Podestà M, Airoldi I. Immunotherapeutic Strategies for Neuroblastoma: Present, Past and Future. Vaccines. 2021; 9(1):43. https://doi.org/10.3390/vaccines9010043

Chicago/Turabian Style

Morandi, Fabio, Federica Sabatini, Marina Podestà, and Irma Airoldi. 2021. "Immunotherapeutic Strategies for Neuroblastoma: Present, Past and Future" Vaccines 9, no. 1: 43. https://doi.org/10.3390/vaccines9010043

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