Next Article in Journal
A Rare Pediatric Case of Severe Bird Fancier’s Lung Presented with Viral Pneumonitis-Like Picture
Next Article in Special Issue
Non-High-Risk Neuroblastoma: Classification and Achievements in Therapy
Previous Article in Journal
Gamma Glutamyl Transferase and Uric Acid Levels Can Be Associated with the Prognosis of Patients in the Pediatric Intensive Care Unit
Previous Article in Special Issue
Molecularly Targeted Therapy for Neuroblastoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Therapies for Relapsed and Refractory Neuroblastoma

1
Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego, La Jolla, CA 92093, USA
2
Peckham Center for Cancer and Blood Disorders, Rady Children’s Hospital, San Diego, CA 92123, USA
Children 2018, 5(11), 148; https://doi.org/10.3390/children5110148
Submission received: 11 September 2018 / Revised: 23 October 2018 / Accepted: 23 October 2018 / Published: 31 October 2018
(This article belongs to the Special Issue Recent Advances in Diagnosis and Treatment of Neuroblastoma)

Abstract

:
While recent increases in our understanding of the biology of neuroblastoma have allowed for more precise risk stratification and improved outcomes for many patients, children with high-risk neuroblastoma continue to suffer from frequent disease relapse, and despite recent advances in our understanding of neuroblastoma pathogenesis, the outcomes for children with relapsed neuroblastoma remain poor. These children with relapsed neuroblastoma, therefore, continue to need novel treatment strategies based on a better understanding of neuroblastoma biology to improve outcomes. The discovery of new tumor targets and the development of novel antibody- and cell-mediated immunotherapy agents have led to a large number of clinical trials for children with relapsed neuroblastoma, and additional clinical trials using molecular and genetic tumor profiling to target tumor-specific aberrations are ongoing. Combinations of these new therapeutic modalities with current treatment regimens will likely be needed to improve the outcomes of children with relapsed and refractory neuroblastoma.

1. Introduction

With new treatment strategies, including immunotherapy and therapies directed against tumor-specific molecular aberrations, the outcomes of children with high-risk neuroblastoma have significantly improved over the past two decades. Despite these recent successes, however, many of these children continue to suffer from refractory or relapsed disease, and to date, there are no established curative treatment options for many of these patients. Relapsed neuroblastoma has been considered invariably fatal in the past [1], with a median overall survival time for patients with refractory neuroblastoma of only 27.9 months and with an overall survival of only 11.0 months for patients with relapsed disease [2]. However, the reported 5-year overall survival rate for children after the initial relapse of neuroblastoma is 20% [3,4]. These outcomes were dependent on both the time of relapse and the initial patient tumor stage [3,4,5], suggesting that a subset of patients with recurrent neuroblastoma can be cured with appropriate treatment and also emphasizing the need for additional treatment options for these patients.
Recent data presented by Modak and colleagues and Manole and colleagues indicate that patients with isolated local recurrence can be treated successfully with surgical tumor resection and/or radiation therapy at sites of active disease [6,7]. These authors further recommend using chemotherapy regimens with demonstrated activity in children with recurrent neuroblastoma, rather than experimental therapy for localized relapses. However, children with neuroblastoma who suffer disease relapse often develop metastatic tumors resistant to standard therapies, and the underlying molecular profiles and signaling pathway activity in these relapsed tumors are likely significantly different from those in untreated tumors due to novel molecular and genetic aberrations induced or selected by prior therapy [8,9]. The goals of treatment in these patients historically have not been curative, but rather to prolong survival and minimize the toxicities of additional therapy. However, recent data have suggested that more aggressive treatment of these patients to achieve a complete or nearly complete response, followed by additional consolidation or maintenance therapy, can lead to durable disease remission and even cure [10], suggesting that the goals of treatment for many children with relapsed and refractory neuroblastoma may need to be modified.

2. Systemic Chemotherapy and Radiotherapy

2.1. Systemic Chemotherapy

Initial treatment regimens for patients with relapsed or refractory neuroblastoma typically include chemotherapy combinations distinct from those previously used. Topotecan and irinotecan have both demonstrated activity as single agents in preclinical and clinical studies of neuroblastoma via inhibition of the topoisomerase I enzyme [11,12,13,14,15,16] and are both commonly used for the treatment of children with relapsed neuroblastoma. An early phase II study using a combination of topotecan and cyclophosphamide in children with relapsed neuroblastoma demonstrated objective responses in 6 of 13 patients [17]. A follow-up study demonstrated that children with recurrent neuroblastoma who were treated with the combination of cyclophosphamide and topotecan had increased response rates compared to those who received topotecan as a single agent, although no difference in overall survival rates was observed in treated patients [18,19]. Using a combination of higher doses of cyclophosphamide and topotecan with vincristine, children with primary refractory neuroblastoma had an overall response rate of 19%, while the response rate was 52% for those with their initial neuroblastoma relapse [20]. A phase II study of topotecan combined with doxorubicin and vincristine (TVD) demonstrated a 64% overall response rate in 25 patients with relapsed neuroblastoma, with 4 patients having complete responses to the TVD combination [21]. The TVD regimen was subsequently incorporated into the SIOPEN HR-NBL-1 treatment protocol for high-risk neuroblastoma as salvage therapy for refractory patients with insufficient responses to induction therapy. Following two courses of TVD, 4 of 63 patients (6.4%) enrolled on this protocol had an overall complete response (CR), while 28 of 63 (44.4%) had a partial response (PR). Of note, 23 patients achieved sufficient response to be eligible to receive myeloablative therapy with stem cell rescue [22]. However, the long-term benefits of TVD in these patients will likely need to be established in further clinical trials.
Combinations of irinotecan with the alkylating agent temozolomide have also demonstrated efficacy in children with relapsed neuroblastoma. A single-institution study reported 2 complete responses to the combination of irinotecan and temozolomide among 19 patients with refractory disease, with 7 patients with mixed responses and 10 with stable disease (SD) [23]. Children with relapsed neuroblastoma treated on a separate multi-institutional study using lower doses of irinotecan and temozolomide had an overall response rate of 15%, while an additional 53% had SD [24]. Subsequent clinical trials have attempted to improve on these results with the addition of bevacizumab [25] and temsirolimus [26] to the irinotecan/temozolomide combination. Although these combinations were well tolerated by patients, the additional agents unfortunately did not significantly improve patient outcomes. A subsequent phase I study utilized irinotecan alone combined with the proteasome inhibitor bortezomib, and 2 of 17 evaluable children with refractory or relapsed neuroblastoma demonstrated responses (including one CR), and another 4 children had prolonged stable disease [27], suggesting that this combination might prove more effective in children with relapsed neuroblastoma and should be tested in further studies.
Combinations of ifosfamide, carboplatin, and etoposide (ICE) have also been routinely employed for children with relapsed neuroblastoma. In a single institution study, ICE was well tolerated and led to disease regression in 14 of 17 patients (82%) who were treated after their first neuroblastoma relapse. Disease regression was also seen in 13 of 26 patients (50%) with refractory neuroblastoma and in 12 of 34 patients (35%) who developed progressive disease during frontline treatment [28]. In a separate study, 37% of patients with relapsed or refractory neuroblastoma responded to the ICE regimen, while an additional 17% had SD [29]. Additionally, 15 of 16 children with high-risk neuroblastoma receiving ICE for upfront treatment had either partial or complete responses [30], further demonstrating the efficacy of ifosfamide-based chemotherapy regimens for both frontline treatment of high-risk neuroblastoma and for initial treatment of patients with relapsed neuroblastoma.
Prior studies have also suggested that some patients with relapsed or refractory neuroblastoma might benefit from more aggressive treatment consisting of myeloablative chemotherapy followed by autologous stem cell rescue (ASCR). One recent study demonstrated that children with relapsed neuroblastoma who received myeloablative therapy followed by ASCR had improved survival rates compared to those who received chemotherapy alone (43.5% vs. 9.6%), with 7 of 23 patients achieving either CR or PR after ASCR. However, 2 of the 23 patients experienced transplant-related mortality, suggesting that further studies to identify which patients would receive the most benefit from this approach are needed [31]. Additional studies have suggested that haploidentical stem cell transplantation may also be beneficial for children with relapsed and refractory neuroblastoma, with 5-year event-free survival (EFS) and overall survival (OS) rates of 19% and 23%, respectively, and no treatment-related mortality [32]. However, patients with residual disease who underwent these transplants had significantly worse outcomes than those in complete remission, and a large number of patients experienced acute or chronic graft-versus-host disease (GVHD), suggesting that further study is required.

2.2. Radiotherapy

Additional forms of therapy for children with relapsed and refractory neuroblastoma have directly targeted markers expressed on the surfaces of neuroblastoma tumor cells. The vast majority of neuroblastoma tumors demonstrate tumor cell surface expression of the norepinephrine transporter (NET), suggesting that the use of benzylguanidine analogues, such as meta-iodobenzylguanidine (MIBG), that bind to this transporter would selectively target these neuroblastoma tumor cells [33]. Approximately 90% of children with neuroblastoma have tumors that are detectable by radiolabeled MIBG imaging, and infusion of radiolabeled MIBG, therefore, allows for targeted delivery of radiation therapy directly to sites of active disease. Initial studies of 131I-MIBG used therapeutically in children with recurrent neuroblastoma showed overall response rates of 21–47% [34,35,36]. Although useful disease palliation and responses were seen in approximately one-third of patients, clinical benefit was often only temporary [37]. Subsequent studies used MIBG treatment followed by ASCR, as dose escalation was often associated with significant hematopoietic toxicity, particularly in heavily pretreated patients [38]. In a large multi-institutional phase II study, 36% of children with either relapsed or refractory neuroblastoma who were treated with increasing doses of 131I-MIBG had either complete or partial responses, while another 34% of patients demonstrated stable disease with a median time to progression of 6 months [39]. A subsequent retrospective analysis of 218 patients treated with 131I-MIBG therapy at a single institution demonstrated a 27% overall response rate. However, 24% of relapsed patients had progressive disease compared to only 9% of refractory patients, and 39% of relapsed patients had stable disease compared to 59% of refractory patients [40], suggesting patients with refractory neuroblastoma are most likely to benefit from 131I-MIBG therapy. The efficacy of 131I-MIBG therapy against neuroblastoma has led to subsequent clinical trials incorporating 131I-MIBG therapy as a component of frontline treatment for children with high-risk neuroblastoma, and ongoing clinical trials through the New Approaches to Neuroblastoma Therapy (NANT) consortium are evaluating the efficacy of additional anticancer agents combined with 131I-MIBG therapy (NCT02035137) in children with relapsed and refractory neuroblastoma. Despite the established efficacy of 131I-MIBG therapy in children with relapsed neuroblastoma, however, only a limited number of institutions have the resources to support the administration of radioactive iodine to pediatric patients, and the limited availability and accompanying need for available stem cells for ASCR remain major challenges to the more widespread use of 131I-MIBG therapy.
Prior studies have also shown that 77–89% of neuroblastoma cells express the somatostatin receptor (SSTR) [41,42,43], suggesting that agents targeting the SSTR may also be effective for neuroblastoma therapy. In adults with neuroendocrine tumors (NETs), peptide receptor radionuclide therapy (PRRT) with high-activity 111In-, 177Lu-, and 90Y-labeled somatostatin analogues has been successfully used [44,45]. In a single-institution study of PRRT, four children with relapsed neuroblastoma received 17 cycles of palliative PRRT using either 111In-DOTATATE, 177Lu-DOTATATE, or 90Y-DOTATATE. All four had objective responses, with two long-term survivors [46], supporting ongoing clinical trials.

3. Molecularly Targeted Therapy

While regimens incorporating chemotherapy and radiotherapy with 131I-MIBG have demonstrated efficacy in the treatment of children with relapsed and refractory neuroblastoma, treatment options for children who suffer from subsequent disease relapses and progression remain limited. However, ongoing investigations exploring the molecular mechanisms underlying the etiology and pathogenesis of neuroblastoma have identified a number of novel therapeutic targets, and several novel targeted agents have been shown to be highly active in preclinical models of neuroblastoma. Many of these agents have been tested or are currently being evaluated in early phase clinical trials for children with relapsed and refractory neuroblastoma (Table 1). Although untreated neuroblastoma tumors have a relative paucity of mutations in therapeutically relevant gene targets [47,48], relapsed neuroblastoma tumors have been found to have a number of actionable mutations [8,9], and initial clinical trials employing therapies selected based on genomic alterations identified in individual patient tumors have demonstrated both feasibility and efficacy, suggesting that molecularly targeted therapy and individualized treatment may lead to increased rates of response and improved outcomes for children with relapsed and refractory neuroblastoma.

3.1. Anaplastic Lymphoma Kinase (ALK)

Early efforts to identify gene mutations and other genetic aberrations in neuroblastoma tumors that could serve as therapeutic targets focused on familial neuroblastoma, which accounts for approximately 2% of all patients with neuroblastoma. Initial efforts determined that the majority of these cases are associated with activating germline mutations in the anaplastic lymphoma kinase (ALK) gene [49,50,51,52], and subsequent studies identified ALK gene mutations or gene amplifications in up to 15% of sporadic high-risk neuroblastoma tumors [49,53]. High-risk neuroblastoma tumors were also found to have increased ALK gene expression when compared to low-risk tumors [54], further suggesting a potential role for ALK inhibitors in neuroblastoma therapy. In a subsequent phase I trial, 79 children were enrolled and treated with the ALK inhibitor crizotinib, including 34 with neuroblastoma, 11 of which had known ALK mutations [55]. Despite an objective tumor response rate of 67% in children with other tumors with ALK mutations, only 1 of 11 children with neuroblastoma with ALK mutations (9%) demonstrated an objective response, suggesting that ALK inhibitors will likely need to be combined with other therapies for maximal benefit. Initial studies have identified synergistic combinations of ALK inhibitors with mTOR inhibitors [56] and with CDK4/6 inhibitors [57], and these combinations may serve to overcome some of the limitations of single-agent ALK inhibitor treatment for neuroblastoma. Additionally, novel second-generation ALK inhibitors, such as lorlatinib (PF06463922), ceritinib (LDK378), and ensartinib, that are effective against the crizotinib-resistant ALKF1174L mutant [58,59] are currently being evaluated in clinical trials for children with neuroblastoma (NCT01742286, NCT03107988, NCT03213652), with early results showing responses to ceritinib in six of nine patients with anaplastic large cell lymphoma (ALCL) and myofibroblastic tumors with ALK gene aberrations. To date, one patient with relapsed neuroblastoma with an ALKF1174L mutation had shrinkage of a retroperitoneal mass but concurrently experienced central nervous system (CNS) disease progression [60], suggesting that higher doses may be required to achieve adequate levels in neuroblastoma sanctuary sites such as the CNS.

3.2. Aurora A Kinase

Additional efforts to identify novel targets in neuroblastoma tumors have identified a critical role for mitotic spindle regulation in neuroblastoma pathogenesis, suggesting that regulators of the mitotic spindle represent potential therapeutic targets. Aurora A kinase represents one such potential target and is essential for appropriate completion of mitosis through regulation of the mitotic checkpoint complex [61]. Aberrant overexpression of aurora A kinase leads to tumor cell resistance to apoptosis and genomic instability [62], and, in neuroblastoma tumors, aurora A kinase expression correlates with high-risk disease and advanced tumor stage [63,64]. Inhibitors of aurora A kinase were shown to block neuroblastoma cell growth and to increase neuroblastoma cell responses to chemotherapy [63], and, in initial phase I trials, children with relapsed neuroblastoma treated with the aurora A kinase inhibitor MLN8237 (alisertib), both alone and in combination with irinotecan and temozolomide, demonstrated clinical responses [65,66]. More recent studies have identified polo-like kinase 4 (PLK4) as a potential target in neuroblastoma tumor cells [67], further implicating the process of mitotic spindle regulation in neuroblastoma pathogenesis and suggesting that children with relapsed neuroblastoma will benefit from the use of inhibitors of aurora A kinase and PLK4 for treatment.

3.3. Ornithine Decarboxylase (ODC1)

Ornithine decarboxylase (ODC1), the rate-limiting enzyme in polyamine synthesis, is frequently deregulated in neuroblastoma tumors [68,69] and represents another potential therapeutic target. ODC inhibitors, such as difluoromethylornithine (DFMO), have been shown to be effective in neuroblastoma preclinical models [70,71,72] and, although single-agent DFMO did not demonstrate efficacy in children with relapsed neuroblastoma in a recent phase I clinical trial [73], more recent studies have demonstrated that extended maintenance therapy with DFMO for children with neuroblastoma in second remission results in 2-year overall and event-free survival rates of 54% and 84% [74], respectively, suggesting that ODC1 inhibition is an effective strategy for prolonging survival in these patients. The efficacy of DFMO in combination with other anticancer agents, including cyclophosphamide, topotecan, and celecoxib (NCT02030964) and the proteasome inhibitor bortezomib (NCT02139397), is also currently being evaluated in clinical trials for children with relapsed neuroblastoma, in the hopes of observing synergistic efficacy.

3.4. PI3K/AKT/mTOR

Further studies in neuroblastoma preclinical models have confirmed a role for the PI-3 kinase/AKT/mTOR pathway in neuroblastoma pathogenesis. SF1126 is a pan-PI-3 kinase inhibitor that has been demonstrated to be effective against neuroblastoma in preclinical models [75], suggesting this pathway represents a therapeutic target in neuroblastoma, and clinical trials have been opened to test the safety and tolerability of SF1126 in children with relapsed neuroblastoma (NCT02337309). The AKT inhibitor perifosine has been tested in multiple phase I clinical trials, with 1 complete response and 8 of 27 children with relapsed neuroblastoma demonstrating prolonged stable disease in one phase I study [76], and response rates and disease control rates of 9% and 55% in 11 children with neuroblastoma with measurable disease [77]. Early studies also determined that mTOR inhibitors were effective in neuroblastoma models [78,79], and one of two patients with relapsed neuroblastoma had a CR in initial phase I studies with the mTOR inhibitor temsirolimus [80]. A follow-up phase II study did not meet predetermined efficacy criteria, but 7 of 19 patients with neuroblastoma demonstrated clinical benefit (1 PR, 6 SD) [81], and 2 patients with relapsed neuroblastoma had prolonged stable disease in response to the combination of vinblastine and sirolimus [82], suggesting that a subset of patients may benefit from the inhibition of this pathway.

3.5. Epigenetic Modifications

Epigenetic modifications to DNA involve heritable genomic modifications that are not due to changes in the DNA sequence, including DNA and histone methylation and histone acetylation. These modifications influence chromatin structure and play a key role in regulating gene expression [83]. Epigenetic regulation of gene expression is critical for both normal development and maintenance of tissue-specific gene expression, but aberrant epigenetic mechanisms can contribute to the malignant process, and epigenetic changes contribute to the genomic instability that is a hallmark of cancer [84]. The epigenomic landscape has been shown to contribute to neuroblastoma pathogenesis, leading to early phase clinical trials investigating the safety and efficacy of epigenetic agents [85,86,87], including drugs that target epigenetic writers, such as DNA methyltransferases, as well as epigenetic erasers, such as histone deacetylases [88]. An early phase I study using the demethylating agent decitabine combined with chemotherapy resulted in prolonged stable disease in 5 of 14 patients with relapsed neuroblastoma, although the doses of decitabine were limited due to toxicity [89]. A subsequent study of the combination of the HDAC inhibitor vorinostat combined with bortezomib did not show any responses in the three patients with relapsed neuroblastoma enrolled [90], and further combination studies using vorinostat in children with neuroblastoma are ongoing (NCT02035137, NCT02559778) to identify patient populations that may have improved responses.
Modulation of the epigenetic readers—proteins that recognize and bind to epigenetic chromatin modifications to modulate gene transcription—has recently emerged as a therapeutic strategy for anticancer treatment. Bromodomain and extraterminal domain (BET) proteins are epigenetic readers that regulate gene transcription, and BET inhibitors preferentially bind to super-enhancers—regions of DNA comprising multiple enhancers occupied by transcription factors and regulators of gene transcription that control the expression of genes critical for cell identity, growth, and survival [91]. Further studies have identified critical roles for chromatin structure and super-enhancers in neuroblastoma [92], and BET inhibitors have been shown to be effective against neuroblastoma via targeting of MYCN gene expression [93,94]. A number of BET inhibitors are in clinical development and early phase clinical trials, including a novel dual PI3K–BRD4 inhibitor that was effective in neuroblastoma preclinical models [95]. BET inhibitors therefore represent an exciting new class of therapeutic agents for children with relapsed neuroblastoma.

3.6. Reactive Oxygen Species (ROS)

Reactive oxygen species (ROS) have been shown to impact a variety of critical intracellular signaling pathways, and prior studies have identified a role for ROS in cancer pathogenesis [96]. However, the role of ROS in neuroblastoma tumors is less clear. Nifurtimox is a nitrofuran derivative that has been used for decades as a primary treatment for Chagas’ disease, a parasitic infection caused by the protozoan Trypanosoma cruzi [97,98]. Nifurtimox was shown to inhibit neuroblastoma cell growth both in vitro and in vivo in preclinical studies, most likely via inhibition of ROS production [99,100], and in early phase clinical trials, patients treated with nifurtimox both as a single agent and in combination with chemotherapy demonstrated responses [101], leading to an ongoing national phase II trial (NCT00601003). A separate study investigating the combination of melphalan with buthionine sulfoximine (BSO) resulted in 6 of 31 patients with relapsed neuroblastoma demonstrating objective responses (5PR, 1MR) [102], and the recent demonstration of synergy between nifurtimox and BSO in other solid tumor models [100] suggests that further testing of the combination of nifurtimox and BSO in neuroblastoma preclinical models is warranted.

3.7. Retinoids

Retinoid therapy has been shown to increase EFS rates when used as a component of maintenance therapy for high-risk neuroblastoma [103], and studies have demonstrated potential synergy of retinoids with epigenetic modifying agents [104,105]. An initial phase I study of the combination of the HDAC inhibitor vorinostat with 13-cis-retinoic acid had one patient with neuroblastoma who had a CR following treatment [106], and a more recent phase I study of vorinostat in combination with 13-cis-retinoic acid for children with relapsed neuroblastoma found no objective responses, but 11 of 29 evaluable patients had SD, with 7 of the 11 receiving at least 11 cycles of therapy [107]. Additionally, the RET tyrosine kinase, which is primarily expressed in cells and tissues derived from the neural crest, has been shown to be required for maturation of the peripheral nervous system, and RET was further shown to be required for neuroblastoma differentiation induced by retinoic acid [108]. RET inhibition has been found to be effective in in vitro and in vivo preclinical models of neuroblastoma [109,110,111], and a recently opened clinical trial (NCT03611595) will evaluate the efficacy of the combination of RET inhibition and retinoid therapy in children with neuroblastoma and other solid tumors.

3.8. Molecularly Guided Therapy

With the rapid advances in our understanding of the biology of cancer, the use of molecular tumor profiling to develop individualized treatment plans is increasingly employed for many adults with a variety of cancers [112]. Although the efficacy of this strategy in children with cancer remains mostly unknown, recently completed clinical trials have evaluated the potential feasibility and efficacy of utilizing molecular and genetic tumor profiling to develop personalized therapy for children with recurrent neuroblastoma. An initial pilot study demonstrated the feasibility of obtaining tumor biopsies from children with relapsed neuroblastoma and then performing DNA sequencing and RNA expression profiling on the relapsed tumor sample, which allowed investigators to generate individualized patient treatment plans in less than 12 days [113]. A follow-up multi-institutional phase I trial employing the same strategy showed that 64% of patients achieved either partial or complete response or disease stabilization for at least one cycle of therapy, with a 7% overall response rate and a progression-free survival time of 59 days [114]. A subsequent single-institution study also demonstrated that the utilization of DNA sequencing data from relapsed tumors to generate prospective treatment plans for children with neuroblastoma was feasible, with nearly half of patients enrolled in the study having potentially actionable genetic findings. Tumor DNA sequencing also led to changes in patient treatment and to genetic counseling for relatives and families of patients found to be at risk of other hereditary disorders [115]. Additional concurrent studies have also demonstrated the feasibility of using genomic data [116] and more comprehensive molecular profiling [117] to identify potential therapeutic targets in children with high-risk, relapsed, or refractory cancers. These results all clearly demonstrate the feasibility of employing molecular profiling to guide individualized treatment strategies for children with neuroblastoma. However, the lack of control groups in these and other studies has limited the ability to assess whether this individualized molecularly targeted treatment resulted in better clinical outcomes for patients when compared with outcomes using standard non-targeted treatment or molecularly targeted agents in an untargeted fashion. Clinical trials to further evaluate the efficacy of molecularly guided, individualized therapy in children with relapsed neuroblastoma and other pediatric solid tumors are currently being developed or are ongoing (NCT02162732, NCT02520713, NCT02638428, NCT03155620, NCT03434262).

4. Immunotherapy

Although a number of recent studies have demonstrated the efficacy of various forms of immunotherapy against neuroblastoma and other pediatric solid tumors, the role of immunotherapy in the treatment of patients with relapsed or refractory neuroblastoma is the focus of numerous prior and ongoing studies. Early studies demonstrated promising results for anti-GD2 antibody therapy in children with relapsed neuroblastoma [118,119,120], leading to the evaluation of the efficacy of the chimeric anti-GD2 antibody ch14.18 (dinutuximab) as a component of maintenance therapy for children with high-risk neuroblastoma [121]. Additional studies demonstrated significant efficacy of the mouse anti-GD2 antibody 3F8 in children with relapsed neuroblastoma, with 33% 5-year progression-free survival (PFS) in patients who were treated for relapsed neuroblastoma and achieved either a CR or very good partial response (VGPR) and were then treated with 3F8 plus GM-CSF and 13-cis-retinoic acid [10].

4.1. Antibody Immunotherapy

Further studies have attempted to expand the use of anti-GD2 antibody therapy in children with relapsed neuroblastoma by employing combinations with other therapies, in addition to the development and evaluation of modified forms of the antibodies themselves. A recent trial combining irinotecan and temozolomide with the anti-GD2 antibody dinutuximab demonstrated promising results, with 9 objective responses (including 5 with CR) among 17 patients receiving the combination [122]. Preliminary data from a follow-up study through the Children’s Oncology Group included data from 53 total eligible patients, with 21 patients (40%) experiencing objective responses, including 11 with CR [123], confirming this combination as a treatment regimen to be considered for children with neuroblastoma at the time of initial relapse. A phase I trial using a humanized version of the ch14.18 antibody with a mutation engineered to reduce side effects (hu14.18K322A) found 6 of 39 enrolled patients had either PR or CR, with 9 additional patients having prolonged SD [124]. A humanized version of the anti-GD2 antibody 3F8 also recently underwent phase I testing, with increased PFS among patients with a higher anti-GD2 antibody titer and reduced overall immunogenicity [125]. In a separate study, 39 patients with recurrent neuroblastoma were treated with the Hu14.18-IL-2 immunocytokine—a fusion protein combining the humanized 14.18 anti-GD2 antibody with IL-2. Of 13 patients with measurable soft tissue neuroblastoma tumors treated with Hu14.18-IL-2, no objective responses were seen, but in those with only MIBG-avid disease or with disease limited to the bone marrow, there were 5 complete responses out of 23 patients [126], suggesting that patients with minimal residual disease are most likely to benefit from this therapy. Further analyses have also shown that mismatches among natural killer (NK) cell KIR/KIR-ligand genotypes and polymorphisms in the Fcγ receptor are also associated with improved responses to anti-GD2 immunotherapy [127,128], suggesting that further strategies for improvement in antibody design and patient selection may result in improved outcomes.
One of the drawbacks to antibody therapy for neuroblastoma is that neuroblastoma tumors use a variety of strategies to evade the host immune response, including downregulation or weak immunogenicity of target antigens and creation of an immune-suppressive tumor environment. With the significant side effects experienced by patients receiving anti-GD2 antibody immunotherapy and with the challenges of antibody administration to patients, a number of alternative immunotherapy strategies are currently under investigation. Immunomodulatory checkpoint inhibitors, such as ipilimumab and nivolumab, have been employed in adult cancers to overcome the immune-suppressive tumor microenvironment. Although ipilimumab was well tolerated in a phase I study for children with relapsed solid tumors, only one patient enrolled in the study had neuroblastoma, and no objective tumor regressions were observed [129]. Ongoing studies evaluating the efficacy of nivolumab with and without added ipilimumab (NCT02304458), with the PD-1 inhibitor pembrolizumab (NCT02332668), and with the anti-PD-L1 antibody atezolizumab (NCT02541604) will hopefully provide more insight to their potential efficacy in children with relapsed neuroblastoma.
Anticancer vaccines have been tested in clinical trials for a number of different tumor types, and an early clinical trial of anti-neuroblastoma vaccine therapy showed the ability to induce an antitumor immune response, although the immune response was insufficient to induce tumor responses or prevent disease progression [130]. A subsequent trial using decitabine combined with a dendritic cell vaccine targeting MAGE-A1, MAGE-A3, and NY-ESO-1 resulted in 1 complete response out of 10 evaluable patients [131], and ongoing cancer vaccine trials include one trial exploring the efficacy of a vaccine using the GD2L and GD3L antigens linked to KLH and administered with the adjuvant OPT-821 and beta-glucan (NCT00911560).

4.2. Cell-Based Immunotherapy

CD8+ T lymphocytes play a key role in cell-mediated immunity and, when infused as a form of immunotherapy, offer the advantage of direct tumor targeting that can avoid or overcome the tumor cell strategies to evade the host’s own immune system. Cell therapy with CD8+ T cells has been employed in early phase clinical trials for children with relapsed neuroblastoma, with some notable successes. EBV-specific T lymphocytes engineered to express a chimeric antigen receptor (CAR) directed against GD2 resulted in 3 patients with complete responses out of 11 total treated patients with active disease [132,133]. CAR-T cell persistence was associated with improved responses and longer times to progression in these patients. Next-generation CAR-T cells, using constructs with modified costimulatory domains to regulate T cell activation, are currently being developed [134], and a number of national and international clinical trials are currently ongoing to further explore the efficacy of novel CAR-T cell products targeted against GD2 in children with relapsed neuroblastoma (NCT02239861, NCT02761915, NCT02765243, NCT02919046, NCT03373097). Other trials using CAR-T cells targeted against other cell surface markers, such as CD171, are also currently undergoing testing in clinical trials for children with neuroblastoma (NCT02311621).
Natural killer (NK) cells are another type of cytotoxic lymphocyte that can act in an MHC-unrestricted fashion to target cancer cells. Cell-based immunotherapy using NK cells for children with relapsed neuroblastoma has also been explored, particularly after the report of a child with relapsed neuroblastoma who received haploidentical donor NK cells combined with temozolomide, topotecan, and IL-2 and had a complete response [135]. Ongoing clinical trials are evaluating the efficacy of ex vivo expanded haploidentical NK cells infused after haploidentical allogeneic stem cell transplants for children with solid tumors, including neuroblastoma (NCT02100891), and a recently opened clinical trial will explore the efficacy of expanded autologous natural killer T (NKT) cells engineered to express the GD2-specific CAR and IL-15 (NCT03294954). A trial using the hu14.18K322A anti-GD2 antibody combined with chemotherapy, cytokines, and haploidentical NK cells demonstrated a 61% response rate in 13 heavily pretreated patients, with 4 complete responses, 1 very good partial response, and 3 partial responses, in addition to 5 patients with stable disease and 10 of the 13 patients (77%) surviving for at least 1 year [136], demonstrating the efficacy of treatment regimens that combine multiple immunotherapeutic strategies. A number of other trials are investigating the combination of haploidentical NK cells combined with anti-GD2 antibody therapy, including NK cells combined with dinutuxumab and lenalidomide (NCT02573896), humanized 3F8, cyclophosphamide, and IL-2 (NCT02650648), the immunocytokine Hu14.18-IL-2 (NCT03209869); and ch14.18/CHO (NCT03242603). Further studies of this approach are clearly warranted in patients with relapsed neuroblastoma.

5. Summary

The treatment of children with relapsed and refractory neuroblastoma remains a challenge, and the outcomes for these children remain poor despite decades of effort by clinicians and scientists. Recent advances in our understanding of the biology of neuroblastoma and of novel strategies to target tumor-specific pathways and antigens have led to a dramatic increase in the number of available treatment options for these patients and give hope that, in the future, novel treatment regimens will increase the responses of tumors to upfront therapy, limit the overall chances of relapse, and, in those hopefully rare cases of relapse, provide safe and effective therapies to eradicate neuroblastoma tumors. Continued development of novel therapies and therapeutic regimens directed against biologically relevant pathways and of novel approaches to harness the therapeutic potential of the innate immune system will provide new treatment strategies to improve the outcomes for these children.
The relative paucity of therapeutically actionable gene mutations has limited the development of individualized treatment regimens for children with neuroblastoma, but as we obtain increased knowledge of other mechanisms of altered gene and protein expression and aberrant function in neuroblastoma tumors, we are likely to identify additional relevant therapeutic targets. Studies to further delineate the critical genetic and proteomic aberrations that either contribute to neuroblastoma pathogenesis or influence neuroblastoma tumor responses to treatment are ongoing, and these aberrations will hopefully lead to the development of individualized patient treatment regimens and also hopefully serve as targets for future drug development. A number of novel therapies directed against recently identified molecular targets are currently being evaluated both in preclinical models and in early phase clinical trials, and established national and international collaborations and cooperative groups will provide opportunities to evaluate these new treatments in carefully controlled clinical trials, leading to more precise and effective therapeutic regimens.
The future holds promise for making considerable advances in our treatment of relapsed neuroblastoma, although continued difficulties in the management of metastatic, widespread relapse and in specific cases of isolated relapses, such as relapse in the CNS, represent ongoing challenges for the future. Although recent results need to be validated in future trials, these results do suggest that we should reconsider our treatment goals for many patients, particularly with the successes of treatments using extended maintenance therapy and immunotherapy for those patients who can achieve disease remission after relapse. Future treatment decisions need to be made based on not only the underlying diagnosis and clinical features of the patient, but also on the molecular features of the tumor itself and the feasibility and availability of tumor-specific treatment. The goals of future clinical and translational research should include identification and validation of the most effective treatment combinations, determination of the most appropriate patients and situations for use of effective therapies, and further delineation of the molecular subgroups of recurrent and refractory neuroblastoma to tailor treatment regimens to the patient populations most likely to benefit. Continued attempts to both develop novel therapeutic agents with efficacy against neuroblastoma tumors and to identify critical intracellular signaling pathways relevant for neuroblastoma pathogenesis and treatment resistance are underway, potentially leading to both individualized and improved treatment and to improved outcomes for children with relapsed and refractory neuroblastoma.

Author Contributions

Conceptualization, writing (draft preparation, review and editing) P.E.Z.

Funding

This manuscript is not supported by external funding.

Conflicts of Interest

The author receives research funding from Bayer, MetronomX, and Exelixis. The funders had no role in the writing of the manuscript or in the decision to publish the manuscript.

References

  1. Cole, K.A.; Maris, J.M. New Strategies in Refractory and Recurrent Neuroblastoma: Translational Opportunities to Impact Patient Outcome. Clin. Cancer Res. 2012, 18, 2423–2428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Moreno, L.; Rubie, H.; Varo, A.; Le Deley, M.C.; Amoroso, L.; Chevance, A.; Garaventa, A.; Gambart, M.; Bautista, F.; Valteau-Couanet, D.; et al. Outcome of children with relapsed or refractory neuroblastoma: A meta-analysis of ITCC/SIOPEN European phase II clinical trials. Pediatr. Blood Cancer 2017, 64, 25–31. [Google Scholar] [CrossRef] [PubMed]
  3. London, W.B.; Castel, V.; Monclair, T.; Ambros, P.F.; Pearson, A.D.J.; Cohn, S.L.; Berthold, F.; Nakagawara, A.; Ladenstein, R.L.; Iehara, T.; et al. Clinical and Biologic Features Predictive of Survival After Relapse of Neuroblastoma: A Report from the International Neuroblastoma Risk Group Project. J. Clin. Oncol. 2011, 29, 3286–3292. [Google Scholar] [CrossRef] [PubMed]
  4. London, W.B.; Bagatell, R.; Weigel, B.J.; Fox, E.; Guo, D.; Van Ryn, C.; Naranjo, A.; Park, J.R. Historical time to disease progression and progression-free survival in patients with recurrent/refractory neuroblastoma treated in the modern era on Children’s Oncology Group early-phase trials. Cancer 2017, 123, 4914–4923. [Google Scholar] [CrossRef] [PubMed]
  5. Garaventa, A.; Parodi, S.; De Bernardi, B.; Dau, D.; Manzitti, C.; Conte, M.; Casale, F.; Viscardi, E.; Bianchi, M.; D’Angelo, P.; et al. Outcome of children with neuroblastoma after progression or relapse. A retrospective study of the Italian neuroblastoma registry. Eur. J. Cancer 2009, 45, 2835–2842. [Google Scholar] [CrossRef] [PubMed]
  6. Modak, S. Generation and administration of autologous T cells transduced with a 3rd generation GD2 chimeric antigen receptor for patients with relapsed or refractory neuroblastoma. In Proceedings of the Advances in Neuroblastoma Research Conference, Cologne, Germany, 13–16 May 2014. [Google Scholar]
  7. Manole, B.A.; Wakefield, D.V.; Doubrovin, M.; Shulkin, B.B.; Merchant, T.E.; Davidoff, A.M.; Furman, W.L.; Krasin, M.; Lucas, J.T. Management of Local-Regional Failure in Children with High-Risk Neuroblastoma: A Single Institution Experience. Int. J. Radiat. Oncol. Biol. Phys. 2017, 99, E570–E571. [Google Scholar] [CrossRef]
  8. Eleveld, T.F.; Oldridge, D.A.; Bernard, V.; Koster, J.; Daage, L.C.; Diskin, S.J.; Schild, L.; Bentahar, N.B.; Bellini, A.; Chicard, M.; et al. Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations. Nat. Genet. 2015, 47, 864–871. [Google Scholar] [CrossRef] [PubMed]
  9. Padovan-Merhar, O.M.; Raman, P.; Ostrovnaya, I.; Kalletla, K.; Rubnitz, K.R.; Sanford, E.M.; Ali, S.J.; Miller, V.A.; Mosse, Y.P.; Granger, M.P.; et al. Enrichment of Targetable Mutations in the Relapsed Neuroblastoma Genome. PLoS Genet. 2016, 12, e1006501. [Google Scholar] [CrossRef] [PubMed]
  10. Kushner, B.H.; Ostrovnaya, I.; Cheung, I.Y.; Kuk, D.; Kramer, K.; Modak, S.; Yataghene, K.; Cheung, N.K.V. Prolonged Progression-Free Survival after Consolidating Second or Later Remissions of Neuroblastoma with Anti-GD2 Immunotherapy and Isotretinoin: A Prospective Phase II Study. Oncoimmunology 2015, 4, e1016704. [Google Scholar] [CrossRef] [PubMed]
  11. Houghton, P.J.; Cheshire, P.J.; Hallman, J.D.; Lutz, L.; Friedman, H.S.; Danks, M.K.; Houghton, J.A. Efficacy of topoisomerase I inhibitors, topotecan and irinotecan, administered at low dose levels in protracted schedules to mice bearing xenografts of human tumors. Cancer Chemother. Pharmacol. 1995, 36, 393–403. [Google Scholar] [CrossRef] [PubMed]
  12. Carol, H.; Houghton, P.J.; Morton, C.L.; Kolb, E.A.; Gorlick, R.; Reynolds, C.P.; Kang, M.H.; Maris, J.M.; Keir, S.T.; Watkins, A.; et al. Initial testing of topotecan by the pediatric preclinical testing program. Pediatr. Blood Cancer 2010, 54, 707–715. [Google Scholar] [CrossRef] [PubMed]
  13. Blaney, S.; Berg, S.L.; Pratt, C.; Weitman, S.; Sullivan, J.; Luchtman-Jones, L.; Bernstein, M. A phase I study of irinotecan in pediatric patients: A pediatric oncology group study. Clin. Cancer Res. 2001, 7, 32–37. [Google Scholar] [PubMed]
  14. Blaney, S.M.; Needle, M.N.; Gillespie, A.; Sato, J.K.; Reaman, G.H.; Berg, S.L.; Adamson, P.C.; Krailo, M.D.; Bleyer, W.A.; Poplack, D.G.; et al. Phase II trial of topotecan administered as 72-hour continuous infusion in children with refractory solid tumors: A collaborative Pediatric Branch, National Cancer Institute, and Children’s Cancer Group Study. Clin. Cancer Res. 1998, 4, 357–360. [Google Scholar] [PubMed]
  15. Hawkins, D.S.; Bradfield, S.; Whitlock, J.A.; Krailo, M.; Franklin, J.; Blaney, S.M.; Adamson, P.C.; Reaman, G. Topotecan by 21-day continuous infusion in children with relapsed or refractory solid tumors: A Children’s Oncology Group study. Pediatr. Blood Cancer 2006, 47, 790–794. [Google Scholar] [CrossRef] [PubMed]
  16. Bomgaars, L.R.; Bernstein, M.; Krailo, M.; Kadota, R.; Das, S.; Chen, Z.; Adamson, P.C.; Blaney, S.M. Phase II trial of irinotecan in children with refractory solid tumors: A Children’s Oncology Group Study. J. Clin. Oncol. 2007, 25, 4622–4627. [Google Scholar] [CrossRef] [PubMed]
  17. Saylors, R.L.; Stine, K.C.; Sullivan, J.; Kepner, J.L.; Wall, D.A.; Bernstein, M.L.; Harris, M.B.; Hayashi, R.; Vietti, T.J. Cyclophosphamide plus topotecan in children with recurrent or refractory solid tumors: A Pediatric Oncology Group phase II study. J. Clin. Oncol. 2001, 19, 3463–3469. [Google Scholar] [CrossRef] [PubMed]
  18. 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]
  19. Ashraf, K.; Shaikh, F.; Gibson, P.; Baruchel, S.; Irwin, M.S. Treatment with topotecan plus cyclophosphamide in children with first relapse of neuroblastoma. Pediatr. Blood Cancer 2013, 60, 1636–1641. [Google Scholar] [CrossRef] [PubMed]
  20. Kushner, B.H.; Kramer, K.; Modak, S.; Qin, L.X.; Cheung, N.K. Differential impact of high-dose cyclophosphamide, topotecan, and vincristine in clinical subsets of patients with chemoresistant neuroblastoma. Cancer 2010, 116, 3054–3060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Garaventa, A.; Luksch, R.; Biasotti, S.; Severi, G.; Pizzitola, M.R.; Viscardi, E.; Prete, A.; Mastrangelo, S.; Podda, M.; Haupt, R.; et al. A Phase II Study of Topotecan with Vincristine and Doxorubicin in Children with Recurrent/Refractory Neuroblastoma. Cancer 2003, 98, 2488–2494. [Google Scholar] [CrossRef] [PubMed]
  22. Amoroso, L.; Erminio, G.; Makin, G.; Pearson, A.D.J.; Brock, P.; Valteau-Couanet, D.; Castel, V.; Pasquet, M.; Laureys, G.; Thomas, C.; et al. Topotecan-Vincristine-Doxorubicin in Stage 4 High-Risk Neuroblastoma Patients Failing to Achieve a Complete Metastatic Response to Rapid COJEC: A SIOPEN Study. Cancer Res. Treat. 2018, 50, 148–155. [Google Scholar] [CrossRef] [PubMed]
  23. 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] [PubMed]
  24. 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] [PubMed]
  25. Modak, S.; Kushner, B.H.; Basu, E.; Roberts, S.S.; Cheung, N.K. Combination of bevacizumab, irinotecan, and temozolomide for refractory or relapsed neuroblastoma: Results of a phase II study. Pediatr. Blood Cancer 2017, 64, e26448. [Google Scholar] [CrossRef] [PubMed]
  26. Bagatell, R.; Norris, R.; Ingle, A.M.; Ahern, C.; Voss, S.; Fox, E.; Little, A.R.; Weigel, B.J.; Adamson, P.C.; Blaney, S. Phase 1 trial of temsirolimus in combination with irinotecan and temozolomide in children, adolescents and young adults with relapsed or refractory solid tumors: A Children’s Oncology Group Study. Pediatr. Blood Cancer 2014, 61, 833–839. [Google Scholar] [CrossRef] [PubMed]
  27. Mody, R.; Zhao, L.; Yanik, G.A.; Opipari, V. Phase I study of bortezomib in combination with irinotecan in patients with relapsed/refractory high-risk neuroblastoma. Pediatr. Blood Cancer 2017, 64, e26563. [Google Scholar] [CrossRef] [PubMed]
  28. Kushner, B.H.; Modak, S.; Kramer, K.; Basu, E.M.; Roberts, S.S.; Cheung, N.-K.V. Ifosfamide, carboplatin, and etoposide for neuroblastoma: A high-dose salvage regimen and review of the literature. Cancer 2013, 119, 665–667. [Google Scholar] [CrossRef] [PubMed]
  29. Adbel Rahman, H.; Moussa, E.A.H.; Zekri, W.Z.K.; El Debawy, E.; Mostafa, N.E.; Yones, A.; Ezzat, S.; Rayan, A.R. Did Salvage ICE Chemotherapy Improve the Outcome in Primary Resistant/Relapsing Stage III/IV Neuroblastoma? J. Egypt. Nat. Cancer Inst. 2011, 23, 47–53. [Google Scholar] [CrossRef] [PubMed]
  30. Donfrancesco, A.; Jenkner, A.; Castellano, A.; Ilari, I.; Milano, G.M.; De Sio, L.; Cozza, R.; Fidani, P.; Deb, G.; De Laurentis, C.; et al. Ifosfamide/carboplatin/etoposide (ICE) as front-line, topotecan/cyclophosphamide as second-line and oral temozolomide as third-line treatment for advanced neuroblastoma over one year of age. Acta Pediatr. 2004, 93, 6–11. [Google Scholar] [CrossRef]
  31. Simon, T.; Berthold, F.; Borkhardt, A.; Kremens, B.; De Carolis, B.; Hero, B. Treatment and Outcomes of Patients with Relapsed, High-Risk Neuroblastoma? Results of German Trials. Pediatr. Blood Cancer 2011, 56, 578–583. [Google Scholar] [CrossRef] [PubMed]
  32. Illhardt, T.; Toporski, J.; Feuchtinger, T.; Turkiewicz, D.; Teitschik, H.M.; Ebinger, M.; Schwarze, C.P.; Holzer, U.; Lode, H.N.; Albert, M.H.; et al. Haploidentical Stem Cell Transplantation for Refractory/Relapsed Neuroblastoma. Biol. Blood Marrow Transplant. 2018, 24, 1005–1012. [Google Scholar] [CrossRef] [PubMed]
  33. Pandit-Taskar, N.; Modak, S. Norepinephrine Transporter as a Target for Imaging and Therapy. J. Nucl. Med. 2017, 58, 39S–53S. [Google Scholar] [CrossRef] [PubMed]
  34. Hutchinson, R.J.; Sisson, J.C.; Shapiro, B.; Miser, J.S.; Normole, D.; Shulkin, B.L.; Francis, I.R.; Zasadny, K.; Carey, J.E.; Johnson, J.W.; et al. 131-I-metaiodobenzylguanidine treatment in patients with refractory advanced neuroblastoma. Am. J. Clin. Oncol. 1992, 15, 226–232. [Google Scholar] [CrossRef] [PubMed]
  35. Klingebiel, T.; Berthold, F.; Treuner, J.; Schwabe, D.; Fischer, M.; Feine, U.; Maul, F.D.; Waters, W.; Wehinger, H.; Niethammer, D. Metaiodobenzylguanidine (mIBG) in treatment of 47 patients with neuroblastoma: Results of the German Neuroblastoma Trial. Med. Pediatr. Oncol. 1991, 19, 84–88. [Google Scholar] [CrossRef] [PubMed]
  36. Lashford, L.S.; Lewis, I.J.; Fielding, S.L.; Flower, M.A.; Meller, S.; Kemshead, J.T.; Ackery, D. Phase I/II study of iodine 131 metaiodobenzylguanidine in chemoresistant neuroblastoma: A United Kingdom Children’s Cancer Study Group investigation. J. Clin. Oncol. 1992, 10, 1889–1896. [Google Scholar] [CrossRef] [PubMed]
  37. 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] [PubMed]
  38. Matthay, K.K.; Quach, A.; Huberty, J.; Franc, B.I.; Hawkins, R.A.; Jackson, H.; Groshen, S.; Shusterman, S.; Yanik, G.; Veach, J.; et al. Iodine-131—Metaiodobenzylguanidine double infusion with autologous stem-cell rescue for neuroblastoma: A new approaches to neuroblastoma therapy phase I study. J. Clin. Oncol. 2009, 27, 1020–1025. [Google Scholar] [CrossRef] [PubMed]
  39. Matthay, K.K.; Yanik, G.; Messina, J.; Quach, A.; Huberty, J.; Cheng, S.C.; Veach, 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]
  40. Zhou, M.J.; Doral, M.Y.; DuBois, S.G.; Villablanca, J.G.; Yanik, G.A.; Matthay, K.K. Different Outcomes for Relapsed vs Refractory Neuroblastoma after Therapy with 131I-Metaiodobenzylguanidine (131I-MIBG). Eur. J. Cancer 2015, 51, 2465–2472. [Google Scholar] [CrossRef] [PubMed]
  41. O’Dorisio, M.S.; Chen, F.; O’Dorisio, T.M.; Wray, D.; Qualman, S.J. Characterization of somatostatin receptors on human neuroblastoma tumors. Cell Growth Differ. 1994, 5, 1–8. [Google Scholar] [PubMed]
  42. Albers, A.R.; O’Dorisio, M.S.; Balster, D.A.; Caprara, M.; Gosh, P.; Chen, F.; Hoeger, C.; Rivier, J.; Wenger, G.; O’Dorisio, T.M.; et al. Somatostatin receptor gene expression in neuroblastoma. Regul. Pept. 2000, 88, 61–73. [Google Scholar] [CrossRef]
  43. Moertel, C.L.; Reubi, J.C.; Scheithauer, B.S.; Schaid, D.J.; Kvols, L.K. Expression of somatostatin receptors in childhood neuroblastoma. Am. J. Clin. Pathol. 1994, 102, 752–756. [Google Scholar] [CrossRef] [PubMed]
  44. Kwekkeboom, D.J.; Teunissen, J.J.; Bakker, W.H.; Kooij, P.P.; de Herder, W.W.; Feelders, R.A.; van Eijck, C.H.; Esser, J.P.; Kam, B.L.; Krenning, E.P. Radiolabeled somatostatin analog [177Lu-DOTA0,Tyr3]octreotate in patients with endocrine gastroenteropancreatic tumors. J. Clin. Oncol. 2005, 23, 2754–2762. [Google Scholar] [CrossRef] [PubMed]
  45. Kwekkeboom, D.J.; Kam, B.L.; van Essen, M.; Teunissen, J.J.; van Eijck, C.H.; Valkema, R.; de Jong, M.; de Herder, W.W.; Krenning, E.P. Somatostatin receptor-based imaging and therapy of gastroenteropancreatic neuroendocrine tumors. Endocr. Relat. Cancer 2010, 17, R53–R73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Kong, G.; Hofman, M.S.; Murray, W.K.; Wilson, S.; Wood, P.; Downie, P.; Super, L.; Hogg, A.; Eu, P.; Hicks, R.J. Initial Experience with Gallium-68 DOTA-Octreotate PET/CT and Peptide Receptor Radionuclide Therapy for Pediatric Patients with Refractory Metastatic Neuroblastoma. J. Pediatr. Hematol. Oncol. 2016, 38, 87–96. [Google Scholar] [CrossRef] [PubMed]
  47. Lawrence, M.S.; Stojanov, P.; Polak, P.; Kryukov, G.V.; Cibulskis, K.; Sivachenko, A.; Carter, S.I.; Stewart, C.; Mermel, C.H.; Roberts, S.A.; et al. Mutational Heterogeneity in Cancer and the Search for New Cancer Genes. Nature 2013, 499, 214–218. [Google Scholar] [CrossRef] [PubMed]
  48. Pugh, T.J.; Morozova, O.; Attiyeh, E.F.; Asgharzadeh, S.; Wei, J.S.; Auclair, D.; Carter, S.L.; Cibulskis, K.; Hanna, M.; Kiezun, A.; et al. The Genetic Landscape of High-Risk Neuroblastoma. Nat. Genet. 2013, 45, 279–284. [Google Scholar] [CrossRef] [PubMed]
  49. Mosse, Y.P.; Laudenslager, M.; Longo, L.; Cole, K.A.; Wood, A.; Attiyeh, E.F.; Laquaglia, M.J.; Sennett, R.; Lynch, J.E.; Perri, P.; et al. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature 2008, 455, 930–935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Janoueix-Lerosey, I.; Lequin, D.; Brugieres, L.; Ribeiro, A.; de Pontual, L.; Combaret, V.; Raynal, V.; Puisieux, A.; Schleiermacher, G.; Pierron, G.; et al. Somatic and germline activating mutations of the ALK kinase receptor in neuroblastoma. Nature 2008, 455, 967–970. [Google Scholar] [CrossRef] [PubMed]
  51. George, R.E.; Sanda, T.; Hanna, M.; Frohling, S.; Luther, W.; Zhang, J.; Ahn, Y.; Zhou, W.; London, W.B.; McGrady, P.; et al. Activating mutations in ALK provide a therapeutic target in neuroblastoma. Nature 2008, 455, 975–978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Chen, Y.; Takita, J.; Choi, Y.L.; Kato, M.; Ohira, M.; Sanada, M.; Wang, L.; Soda, M.; Kikuchi, A.; Igarashi, T.; et al. Oncogenic mutations of ALK kinase in neuroblastoma. Nature 2008, 455, 971–974. [Google Scholar] [CrossRef] [PubMed]
  53. Caren, H.; Abel, F.; Kogner, P.; Martinsson, T. High incidence of DNA mutations and gene amplifications of the ALK gene in advanced sporadic neuroblastoma tumors. Biochem. J. 2008, 416, 153–159. [Google Scholar] [CrossRef] [PubMed]
  54. Passoni, L.; Longo, L.; Collini, P.; Coluccia, A.M.L.; Bozzi, F.; Podda, M.; Gregorio, A.; Gambini, C.; Garaventa, A.; Pistoia, V.; et al. Mutation-independent anaplastic lymphoma kinase overexpression in poor prognosis neuroblastoma patients. Cancer Res. 2009, 69, 7338–7346. [Google Scholar] [CrossRef] [PubMed]
  55. Mosse, 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]
  56. Moore, N.F.; Azarova, A.M.; Bhatnagar, N.; Ross, K.N.; Drake, L.E.; Frumm, S.; Liu, Q.S.; Christie, A.L.; Sanda, T.; Chesler, L.; et al. Molecular rationale for the use of PI3K/AKT/mTOR pathway inhibitors in combination with crizotinib in ALK-mutated neuroblastoma. Oncotarget 2014, 5, 8737–8749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Wood, A.C.; Krytska, K.; Ryles, H.T.; Infarinato, N.R.; Sano, R.; Hansel, T.D.; Hart, L.S.; King, F.J.; Smith, T.R.; Ainscow, E.; et al. Dual ALK and CDK4/6 Inhibition Demonstrates Synergy Against Neuroblastoma. Clin. Cancer Res. 2017, 23, 2856–2868. [Google Scholar] [CrossRef] [PubMed]
  58. Sasaki, T.; Okuda, K.; Zheng, W.; Butrynski, J.; Capelletti, M.; Wang, L.; Gray, N.S.; Wilner, K.; Christensen, J.G.; Demetri, G.; et al. The neuroblastoma-associated F1174L ALK mutation causes resistance to an ALK kinase inhibitor in ALK-translocated cancers. Cancer Res. 2010, 70, 10038–10043. [Google Scholar] [CrossRef] [PubMed]
  59. Berry, T.; Luther, W.; Bhatnagar, N.; Jamin, Y.; Poon, E.; Sanda, T.; Pei, D.; Sharma, B.; Vetharoy, W.R.; Hallsworth, A.; et al. The ALK(F1174L) mutation potentiates the oncogenic activity of MYCN in neuroblastoma. Cancer Cell 2012, 22, 117–130. [Google Scholar] [CrossRef] [PubMed]
  60. Geoerger, B.; Schulte, J.; Zwaan, C.M.; Casanova, M.; Fischer, M.; Moreno, L.; Trahair, T.; Jimenez, I.; Kang, H.J.; Pappo, A.S.; et al. Phase I study of ceritinib in pediatric patients (Pts) with malignancies harboring a genetic alteration in ALK (ALK+): Safety, pharmacokinetic (PK), and efficacy results. J. Clin. Oncol. 2015, 15, 10005. [Google Scholar] [CrossRef]
  61. Zhou, H.; Kuang, J.; Zhong, L.; Kuo, W.L.; Gray, J.W.; Sahin, A.; Brinkley, B.R.; Sen, S. Tumour amplified kinase STK15/BTAK induces centrosome amplification, aneuploidy, and transformation. Nat. Genet. 1998, 20, 189–193. [Google Scholar] [CrossRef] [PubMed]
  62. Sen, S.; Katayama, H.; Sasai, K. Functional significance of aurora kinase A in centrosome amplification and genomic instability. Adv. Exp. Med. Biol. 2008, 617, 99–108. [Google Scholar] [PubMed]
  63. Shang, X.; Burlingame, S.M.; Okcu, M.F.; Ge, N.; Russell, H.V.; Egler, R.A.; David, R.D.; Vasudevan, S.A.; Yang, Y.; Nuchtern, J.G. Aurora A is a negative prognostic factor and a new therapeutic target in human neuroblastoma. Mol. Cancer Ther. 2009, 8, 2461–2469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Otto, T.; Horn, S.; Brockmann, M.; Eilers, U.; Schuttrumpf, L.; Popov, N.; Kenney, A.M.; Schulte, J.H.; Beijersbergen, R.; Christiansen, H.; et al. Stabilization of N-Myc is a critical function of Aurora A in human neuroblastoma. Cancer Cell 2009, 15, 67–78. [Google Scholar] [CrossRef] [PubMed]
  65. Mossé, Y.P.; Lipsitz, E.; Fox, E.; Teachey, D.T.; Maris, J.M.; Weigel, B.; Adamson, P.C.; Ingle, M.A.; Ahern, C.H.; Blaney, S.M. Pediatric phase I trial and pharmacokinetic study of MLN8237, an investigational oral selective small-molecule inhibitor of Aurora kinase A: A Children’s Oncology Group Phase I Consortium study. Clin. Cancer Res. 2012, 18, 6058–6064. [Google Scholar] [CrossRef] [PubMed]
  66. Dubois, S.G.; Marachelian, A.; Fox, E.; Kudgus, R.A.; Reid, J.M.; Groshen, S.; Malvar, J.; Bagatell, R.; Wagner, L.; Maris, J.M.; et al. Phase I Study of the Aurora A Kinase Inhibitor Alisertib in Combination with Irinotecan and Temozolomide for Patients with Relapsed or Refractory Neuroblastoma: A NANT (New Approaches to Neuroblastoma Therapy) Trial. J. Clin. Oncol. 2016, 34, 1368–1375. [Google Scholar] [CrossRef] [PubMed]
  67. Meitinger, F.; Davis, R.L.; Kabeche, R.; Anzola, J.V.; Wong, Y.L.; Shiau, A.K.; Desai, A.; Oegema, K. TRIM37 expression levels dictate susceptibility to centrosome removal, supporting Plk4 inhibition as a potential new strategy for targeting neuroblastoma. Cancer Res. 2018, 78. [Google Scholar] [CrossRef]
  68. Wallick, C.J.; Gamper, I.; Thorne, M.; Feith, D.J.; Takasaki, K.Y.; Wilson, S.M.; Seki, J.A.; Pegg, A.E.; Byus, C.V.; Bachmann, A.S. Key Role for p27Kip1, Retinoblastoma Protein Rb, and MYCN in Polyamine Inhibitor-Induced G1 Cell Cycle Arrest in MYCN-Amplified Human Neuroblastoma Cells. Oncogene 2005, 24, 5606–5618. [Google Scholar] [CrossRef] [PubMed]
  69. Hogarty, M.D.; Norris, M.D.; Davis, K.; Liu, X.; Evageliou, N.F.; Hayes, C.S.; Pawel, B.; Guo, R.; Zhao, H.; Sekyere, E.; et al. ODC1 Is a Critical Determinant of MYCN Oncogenesis and a Therapeutic Target in Neuroblastoma. Cancer Res. 2008, 68, 9735–9745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Koomoa, D.L.; Yco, L.P.; Borsics, T.; Wallick, C.J.; Bachmann, A.S. Ornithine Decarboxylase Inhibition by Alpha-Difluoromethylornithine Activates Opposing Signaling Pathways via Phosphorylation of both AKT/Protein Kinase B and p27Kip1 in Neuroblastoma. Cancer Res. 2008, 68, 9825–9831. [Google Scholar] [CrossRef] [PubMed]
  71. Rounbehler, R.J.; Li, W.; Hall, M.A.; Yang, C.; Fallahi, M.; Cleveland, J.L. Targeting Ornithine Decarboxylase Impairs Development of MYCN-Amplified Neuroblastoma. Cancer Res. 2009, 69, 547–553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Evageliou, N.F.; Haber, M.; Vu, A.; Laetsch, T.W.; Murray, J.; Gamble, L.D.; Cheng, N.C.; Liu, K.; Reese, M.; Corrigan, K.A.; et al. Polyamine Antagonist Therapies Inhibit Neuroblastoma Initiation and Progression. Clin. Cancer Res. 2016, 22, 4391–4404. [Google Scholar] [CrossRef] [PubMed]
  73. Saulnier Sholler, G.L.; Gerner, E.W.; Bergendahl, G.; MacArthur, R.B.; VanderWerff, A.; Ashikaga, T.; Bond, J.P.; Ferguson, W.; Roberts, W.; Wada, R.K.; et al. A Phase I Trial of DFMO Targeting Polyamine Addiction in Patients with Relapsed/Refractory Neuroblastoma. PLoS ONE 2015, 10, e0127246. [Google Scholar] [CrossRef] [PubMed]
  74. Saulnier Sholler, G.L.; Ferguson, W.; Bergendahl, G.; Bond, J.P.; Neville, K.; Eslin, D.; Brown, V.; Roberts, W.; Wada, R.K.; Oesterheld, J.; et al. Maintenance DFMO Increases Survival in High-Risk Neuroblastoma. Sci. Rep. 2018, 8, 1445. [Google Scholar] [CrossRef] [PubMed]
  75. Peirce, S.K.; Findley, H.W.; Prince, C.; Dasgupta, A.; Cooper, T.; Durden, D.L. The PI-3 kinase-Akt-MDM2-survivin signaling axis in high-risk neuroblastoma: A target for PI-3 kinase inhibitor intervention. Cancer Chemother. Pharmacol. 2011, 68, 325–335. [Google Scholar] [CrossRef] [PubMed]
  76. Becher, O.J.; Millard, N.E.; Modak, S.; Kushner, B.H.; Hague, S.; Spasojevic, I.; Trippett, T.M.; Gilheeney, S.W.; Khakoo, Y.; Lyden, D.C.; et al. A phase I study of single-agent perifosine for recurrent or refractory pediatric CNS and solid tumors. PLoS ONE 2017, 12, e0178593. [Google Scholar] [CrossRef] [PubMed]
  77. Matsumoto, K.; Shichino, H.; Kawamoto, H.; Kosaka, Y.; Chin, M.; Kato, K.; Mugishima, H. Phase I study of perifosine monotherapy in patients with recurrent or refractory neuroblastoma. Pediatr. Blood Cancer 2017, 64, e26623. [Google Scholar] [CrossRef] [PubMed]
  78. Misawa, A.; Hosoi, H.; Tsuchiya, K.; Sugimoto, T. Rapamycin inhibits proliferation of human neuroblastoma cells without suppression of MycN. Int. J. Cancer 2003, 104, 233–237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Johnsen, J.I.; Segerström, L.; Orrego, A.; Elfman, L.; Henriksson, M.; Kågedal, B.; Eksborg, S.; Sveinbjörnsson, B.; Kogner, P. Inhibitors of mammalian target of rapamycin downregulate MYCN protein expression and inhibit neuroblastoma growth in vitro and in vivo. Oncogene 2008, 27, 2910–2922. [Google Scholar] [CrossRef] [PubMed]
  80. Spunt, S.L.; Grupp, S.A.; Vik, T.A.; Santana, V.M.; Greenblatt, D.J.; Clancy, J.; Berkenbilt, A.; Krygowski, M.; Ananthakrishnan, R.; Boni, J.P.; et al. Phase I Study of Temsirolimus in Pediatric Patients with Recurrent/Refractory Solid Tumors. J. Clin. Oncol. 2011, 29, 2933–2940. [Google Scholar] [CrossRef] [PubMed]
  81. Geoerger, B.; Kieran, M.W.; Grupp, S.A.; Perek, D.; Clancy, J.; Krygowski, M.; Ananthakrishnan, R.; Boni, J.P.; Berkenblit, A.; Spunt, S.L. Phase II Trial of Temsirolimus in Children with High-Grade Glioma, Neuroblastoma, and Rhabdomyosarcoma. Eur. J. Cancer 2012, 48, 253–262. [Google Scholar] [CrossRef] [PubMed]
  82. Morgenstern, D.A.; Marzouki, M.; Bartels, U.; Irwin, M.S.; Sholler, G.L.S.; Gammon, J.; Yankanah, R.; Wu, B.; Samson, Y.; Baruchel, S. Phase I Study of Vinblastine and Sirolimus in Pediatric Patients with Recurrent or Refractory Solid Tumors. Pediatr. Blood Cancer 2014, 61, 128–133. [Google Scholar] [CrossRef] [PubMed]
  83. Yoo, C.B.; Jones, P.A. Epigenetic Therapy of Cancer: Past, Present, and Future. Nat. Rev. Drug Discov. 2006, 5, 37–50. [Google Scholar] [CrossRef] [PubMed]
  84. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Ram Kumar, R.M.; Schor, N.F. Methylation of DNA and chromatin as a mechanism of oncogenesis and therapeutic target in neuroblastoma. Oncotarget 2018, 9, 22184–22193. [Google Scholar] [PubMed]
  86. Jubierre, L.; Jimenez, C.; Rovira, E.; Soriano, A.; Sabado, C.; Gros, L.; Llort, A.; Hladun, R.; Roma, J.; de Toledo, J.S.; et al. Targeting of Epigenetic Regulators in Neuroblastoma. Exp. Mol. Med. 2018, 50, 51. [Google Scholar] [CrossRef] [PubMed]
  87. Durinck, K.; Speleman, F. Epigenetic Regulation of Neuroblastoma Development. Cell Tissue Res. 2018, 372, 309–324. [Google Scholar] [CrossRef] [PubMed]
  88. Falkenberg, K.J.; Johnstone, R.W. Histone Deacetylases and Their Inhibitors in Cancer, Neurological Diseases, and Immune Disorders. Nat. Rev. Drug Discov. 2014, 13, 673–691. [Google Scholar] [CrossRef] [PubMed]
  89. George, R.E.; Lahti, J.M.; Adamson, P.C.; Zhu, K.; Finkelstein, D.; Ingle, A.M.; Reid, J.M.; Krailo, M.; Neuberg, D.; Blaney, S.M.; et al. Phase I Study of Decitabine with Doxorubicin and Cyclophosphamide in Children with Neuroblastoma and Other Solid Tumors: A Children’s Oncology Group Study. Pediatr. Blood Cancer 2010, 55, 629–638. [Google Scholar] [CrossRef] [PubMed]
  90. Muscal, J.A.; Thompson, P.A.; Horton, T.M.; Ingle, A.M.; Ahern, C.H.; McGovern, R.M.; Reid, J.M.; Ames, M.M.; Espinoza-Delgado, I.; Weigel, B.; et al. A Phase I Trial of Vorinostat and Bortezomib in Children with Refractory or Recurrent Solid Tumors: A Children’s Oncology Group Phase I Consortium Study (ADVL0916). Pediatr. Blood Cancer 2013, 60, 390–395. [Google Scholar] [CrossRef] [PubMed]
  91. Hnisz, D.; Abraham, B.J.; Tee, T.I.; Lau, A.; Saint-Andre, V.; Sigova, A.A.; Hoke, H.A.; Young, R.A. Super-Enhancers in the Control of Cell Identity and Disease. Cell 2013, 155, 934–947. [Google Scholar] [CrossRef] [PubMed]
  92. Van Groningen, T.; Koster, J.; Valentijn, L.J.; Zwijnenburg, D.A.; Akogul, N.; Hasselt, N.E.; Broekmans, M.; Haneveld, F.; Nowakowska, N.E.; Bras, J.; et al. Neuroblastoma is Composed of Two Super-Enhancer-Associated Differentiation States. Nat. Genet. 2017, 49, 1261–1266. [Google Scholar] [CrossRef] [PubMed]
  93. Puissant, A.; Frumm, S.M.; Alexe, G.; Bassil, C.F.; Qi, J.; Chanthery, Y.H.; Nekritz, E.A.; Zeid, R.; Gustafson, W.C.; Greninger, P.; et al. Targeting MYCN in neuroblastoma by BET bromodomain inhibition. Cancer Discov. 2013, 3, 308–323. [Google Scholar] [CrossRef] [PubMed]
  94. Henssen, A.; Althoff, K.; Odersky, A.; Beckers, A.; Koche, R.; Speleman, F.; Schafers, S.; Bell, E.; Nortmeyer, M.; Westermann, F.; et al. Targeting MYCN-Driven Transcription by BET-Bromodomain Inhibition. Clin. Cancer Res. 2016, 22, 2470–2481. [Google Scholar] [CrossRef] [PubMed]
  95. Andrews, R.H.; Singh, A.R.; Joshi, S.; Smith, C.A.; Morales, G.A.; Garlich, J.R.; Durden, D.L.; Kutateladze, T.G. Dual-activity PI3K–BRD4 inhibitor for the orthogonal inhibition of MYC to block tumor growth and metastasis. Proc. Natl. Acad. Sci. USA 2017, 114, E1072–E1080. [Google Scholar] [CrossRef] [PubMed]
  96. Prasad, S.; Gupta, S.C.; Tyagi, A.K. Reactive Oxygen Species (ROS) and Cancer: Role of Antioxidative Nutraceuticals. Cancer Lett. 2017, 387, 95–105. [Google Scholar] [CrossRef] [PubMed]
  97. Bern, C. Antitrypanosomal Therapy for Chronic Chagas’ Disease. N. Engl. J. Med. 2011, 364, 2527–2534. [Google Scholar] [CrossRef] [PubMed]
  98. Rassi, A.; Rassi, A.; Marin-Neto, J.A. Chagas disease. Lancet 2010, 375, 1388–1402. [Google Scholar] [CrossRef]
  99. Saulnier Sholler, G.; Brard, L.; Straub, J.A.; Dorf, L.; Illeyne, S.; Koto, K.; Kalkunte, S.; Bosenberg, M.; Ashikaga, T.; Nishi, R. Nifurtimox induces apoptosis of neuroblastoma cells in vitro and in vivo. J. Pediatr. Hematol. Oncol. 2009, 31, 187–193. [Google Scholar] [CrossRef] [PubMed]
  100. Du, M.; Zhang, L.; Scorsone, K.A.; Woodfield, S.E.; Zage, P.E. Nifurtimox is Effective Against Neural Tumor Cells and Is Synergistic with Buthionine Sulfoximine. Sci. Rep. 2016, 6, 27458. [Google Scholar] [CrossRef] [PubMed]
  101. Saulnier Sholler, G.L.; Bergendahl, G.M.; Brard, L.; Singh, A.P.; Heath, B.W.; Bingham, P.M.; Ashikaga, T.; Kamen, B.A.; Homans, A.C.; Slavik, M.A.; et al. A Phase 1 Study of Nifurtimox in Patients with Relapsed/Refractory Neuroblastoma. J. Pediatr. Hematol. Oncol. 2011, 33, 25–30. [Google Scholar] [CrossRef] [PubMed]
  102. Anderson, C.P.; Matthay, K.K.; Perentesis, J.P.; Neglia, J.P.; Bailey, H.H.; Villablanca, J.G.; Groshen, S.; Hasenauer, B.; Maris, J.M.; Seeger, R.C.; et al. Pilot study of intravenous melphalan combined with continuous infusion L-S,R-buthionine sulfoximine for children with recurrent neuroblastoma. Pediatr. Blood Cancer 2015, 62, 1739–1746. [Google Scholar] [CrossRef] [PubMed]
  103. Matthay, K.K.; Reynolds, C.P.; Seeger, R.C.; Shimada, H.; Adkins, E.S.; Haas-Kogan, D.; Gerbing, R.B.; London, W.B.; Villablanca, J.G. Long-Term Results for Children with High-Risk Neuroblastoma Treated on a Randomized Trial of Myeloablative Therapy Followed by 13-cis-Retinoic Acid: A Children’s Oncology Group Study. J. Clin. Oncol. 2009, 27, 1007–1013. [Google Scholar] [CrossRef] [PubMed]
  104. Coffey, D.C.; Kutko, M.C.; Glick, R.D.; Butler, L.M.; Heller, G.; Rifkind, R.A.; Marks, P.A.; Richon, V.M.; La Quaglia, M.P. The histone deacetylase inhibitor, CBHA, inhibits growth of human neuroblastoma xenografts in vivo, alone and synergistically with all-trans retinoic acid. Cancer Res. 2001, 61, 3591–3594. [Google Scholar] [PubMed]
  105. Westerlund, I.; Shi, Y.; Toskas, K.; Feli, S.M.; Li, S.; Surova, O.; Sodersten, E.; Kogner, P.; Nyman, U.; Schlisio, S.; et al. Combined Epigenetic and Differentiation-Based Treatment Inhibits Neuroblastoma Tumor Growth and Links HIF2α to Tumor Suppression. Proc. Natl. Acad. Sci. USA 2017, 114, E6137–E6146. [Google Scholar] [CrossRef] [PubMed]
  106. Fouladi, M.; Park, J.R.; Stewart, C.F.; Gilbertson, R.J.; Schaiquevich, P.; Sun, J.; Reid, J.M.; Ames, M.M.; Speights, R.; Ingle, A.M. Pediatric Phase I Trial and Pharmacokinetic Study of Vorinostat: A Children’s Oncology Group Phase I Consortium Report. J. Clin. Oncol. 2010, 28, 3623–3629. [Google Scholar] [CrossRef] [PubMed]
  107. Pinto, N.; DuBois, S.G.; Marachelian, A.; Diede, S.J.; Taraseviciute, A.; Glade Bender, J.L.; Tsao-Wei, D.; Groshen, S.G.; Reid, J.M.; Haas-Kogan, D.A.; et al. Phase I study of vorinostat in combination with isotretinoin in patients with refractory/recurrent neuroblastoma: A new approaches to Neuroblastoma Therapy (NANT) trial. Pediatr. Blood Cancer 2018, 65, e27023. [Google Scholar] [CrossRef] [PubMed]
  108. Oppenheimer, O.; Cheung, N.-K.; Gerald, W.L. The RET oncogene is a critical component of transcriptional programs associated with retinoic acid-induced differentiation in neuroblastoma. Mol. Cancer Ther. 2007, 6, 1300–1309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Zage, P.E.; Zeng, L.; Palla, S.; Fang, W.; Nilsson, M.B.; Heymach, J.V.; Zweidler-McKay, P.A. A novel therapeutic combination for neuroblastoma: The VEGF/EGF/RET inhibitor vandetanib with 13-cis-retinoic acid. Cancer 2010, 116, 2465–2475. [Google Scholar] [PubMed]
  110. Zhang, L.; Scorsone, K.; Woodfield, S.E.; Zage, P.E. Sensitivity of Neuroblastoma to the Novel Kinase Inhibitor Cabozantinib Is Mediated by ERK Inhibition. Cancer Chemother. Pharmacol. 2015, 76, 977–987. [Google Scholar] [CrossRef] [PubMed]
  111. Chen, Z.; Zhao, Y.; Yu, Y.; Pang, J.C.; Woodfield, S.E.; Tao, L.; Guan, S.; Zhang, H.; Bieerkehazhi, S.; Shi, Y.; et al. Small Molecule Inhibitor Regorafenib Inhibits RET Signaling in Neuroblastoma Cells and Effectively Suppresses Tumor Growth in vivo. Oncotarget 2017, 8, 104090–104103. [Google Scholar] [CrossRef] [PubMed]
  112. Subbiah, V.; Kurzrock, R. Challenging Standard-of-Care Paradigms in the Precision Oncology Era. Trends Cancer 2018, 4, 101–109. [Google Scholar] [CrossRef] [PubMed]
  113. Saulnier Sholler, G.L.; Ferguson, W.; Bergendahl, G.; Currier, E.; Lenox, S.R.; Bond, J.; Slavik, M.; Roberts, W.; Mitchell, D.; Eslin, D.; et al. A Pilot Trial Testing the Feasibility of Using Molecular-Guided Therapy in Patients with Recurrent Neuroblastoma. J. Cancer Ther. 2012, 3, 602–612. [Google Scholar] [CrossRef]
  114. Saulnier Sholler, G.L.; Bond, J.P.; Bergendahl, G.; Dutta, A.; Dragon, J.; Neville, K.; Ferguson, W.; Roberts, W.; Eslin, D.; Kraveka, J.; et al. Feasibility of implementing molecular-guided therapy for the treatment of patients with relapsed or refractory neuroblastoma. Cancer Med. 2015, 4, 871–886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Mody, R.J.; Wu, Y.-M.; Lonigro, R.J.; Cao, X.; Roychowdhury, S.; Vats, P.; Frank, K.M.; Prensner, J.P.; Asangani, I.; Palanisamy, N.; et al. Integrative Clinical Sequencing in the Management of Refractory or Relapsed Cancer in Youth. JAMA 2015, 314, 913–925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Harris, M.H.; DuBois, S.G.; Glade Bender, J.L.; Kim, A.; Crompton, B.D.; Parker, E.; Dumont, I.P.; Hong, A.L.; Guo, D.; Church, A.; et al. Multicenter Feasibility Study of Tumor Molecular Profiling to Inform Therapeutic Decisions in Advanced Pediatric Solid Tumors: The Individualized Cancer Therapy (iCAT) Study. JAMA Oncol. 2016, 2, 608–615. [Google Scholar] [CrossRef] [PubMed]
  117. Worst, B.C.; van Tilburg, C.M.; Balasubramanian, G.P.; Fiesel, P.; Witt, R.; Freitag, A.; Boudalil, M.; Previti, C.; Wolf, S.; Schmidt, S.; et al. Next-Generation Personalised Medicine for High-Risk Pediatric Cancer Patients—The INFORM Pilot Study. Eur. J. Cancer 2016, 65, 91–101. [Google Scholar] [CrossRef] [PubMed]
  118. Cheung, N.K.; Lazarus, H.; Miraldi, F.D.; Abramowsky, C.R.; Kallick, S.; Saarinen, U.M.; Spitzer, T.; Strandjord, S.E.; Coccia, P.F.; Berger, N.A. Ganglioside GD2 Specific Monoclonal Antibody 3F8: A Phase I Study in Patients with Neuroblastoma and Malignant Melanoma. J. Clin. Oncol. 1987, 5, 1430–1440. [Google Scholar] [CrossRef] [PubMed]
  119. Handgretinger, R.; Anderson, K.; Lang, P.; Dopfer, R.; Klingebiel, T.; Schrappe, M.; Reuland, P.; Gillies, S.D.; Reisfeld, R.A.; Niethammer, D. A phase I study of human/mouse chimeric antiganglioside GD2 antibody ch14.18 in patients with neuroblastoma. Eur. J. Cancer 1995, 31, 261–267. [Google Scholar] [CrossRef]
  120. 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] [PubMed]
  121. Yu, A.L.; Gilman, A.L.; Ozkaynak, 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]
  122. 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]
  123. Mody, R.; Naranjo, A.; Yu, A.L.; Hibbitts, E.; London, W.B.; Shulkin, B.L.; Parisi, M.T.; Servaes, S.-E.-N.; Diccianni, M.B.; Sondel, P.M.; et al. Phase II Trial of Irinotecan/Temozolomide/Dinutuxumab/ Granulocyte-Macrophage Colony Stimulating factor (I/T/DIN/GMCSF) in Children with Relapsed/Refractory Neuroblastoma (NBL): A Report from the Children’s Oncology Group (COG). J. Clin. Oncol. 2018, 36, 10508. [Google Scholar] [CrossRef]
  124. Navid, F.; Sondel, P.M.; Barfield, R.; Shulkin, B.L.; Kaufman, R.A.; Allay, J.A.; Gan, J.; Hutson, P.; Seo, S.; Kim, K.; et al. Phase I Trial of a Novel Anti-GD2 Monoclonal Antibody, Hu14.18K322A, Designed to Decrease Toxicity in Children with Refractory or Recurrent Neuroblastoma. J. Clin. Oncol. 2014, 32, 1445–1452. [Google Scholar] [CrossRef] [PubMed]
  125. Cheung, I.Y.; Kushner, B.H.; Modak, S.; Basu, E.M.; Roberts, S.S.; Cheung, N.V. Phase I trial of anti-GD2 monoclonal antibody hu3F8 plus GM-CSF: Impact of body weight, immunogenicity and anti-GD2 response on pharmacokinetics and survival. Oncoimmunology 2017, 6, e1358331. [Google Scholar] [CrossRef] [PubMed]
  126. Shusterman, S.; London, W.B.; Gillies, S.D.; Hank, J.A.; Voss, S.D.; Seeger, R.C.; Reynolds, C.P.; Kimball, J.; Albertini, M.R.; Wagner, B.; et al. Antitumor activity of hu14.18-IL2 in patients with relapsed/refractory neuroblastoma: A Children’s Oncology Group (COG) phase II study. J. Clin. Oncol. 2010, 28, 4969–4975. [Google Scholar] [CrossRef] [PubMed]
  127. Delgado, D.C.; Hank, J.A.; Kolesar, J.; Lorentzen, D.; Gan, J.; Seo, S.; Kim, K.; Shusterman, S.; Gillies, S.D.; Reisfeld, R.A.; et al. Genotypes of NK cell KIR receptors, their ligands, and Fcγ receptors in the response of neuroblastoma patients to Hu14.18-IL2 immunotherapy. Cancer Res. 2010, 70, 9554–9561. [Google Scholar] [CrossRef] [PubMed]
  128. Cheung, N.K.; Sowers, R.; Vickers, A.J.; Cheung, I.Y.; Kushner, B.H.; Gorlick, R. FCGR2A polymorphism is correlated with clinical outcome after immunotherapy of neuroblastoma with anti-GD2 antibody and granulocyte macrophage colony stimulating factor. J. Clin. Oncol. 2006, 24, 2885–2890. [Google Scholar] [CrossRef] [PubMed]
  129. Merchant, M.S.; Wright, M.; Baird, K.; Wexler, L.H.; Rodriguez-Galindo, C.; Bernstein, D.; Delbrook, C.; Lodish, M.; Bishop, R.; Wolchok, J.D.; et al. Phase I Clinical Trial of Ipilimumab in Pediatric Patients with Advanced Solid Tumors. Clin. Cancer Res. 2016, 22, 1364–1370. [Google Scholar] [CrossRef] [PubMed]
  130. Russell, H.V.; Strother, D.; Mei, Z.; Rill, D.; Popek, E.; Biagi, E.; Yvon, E.; Brenner, M.; Rousseau, R. Phase I trial of vaccination with autologous neuroblastoma tumor cells genetically modified to secrete IL-2 and lymphotactin. J. Immunother. 2007, 30, 227–233. [Google Scholar] [CrossRef] [PubMed]
  131. Krishnadas, D.K.; Shusterman, S.; Bai, F.; Diller, L.; Sullivan, J.E.; Cheerva, A.C.; George, R.E.; Lucas, K.G. A phase I trial combining decitabine/dendritic cell vaccine targeting MAGE-A1, MAGE-A3 and NY-ESO-1 for children with relapsed or therapy-refractory neuroblastoma and sarcoma. Cancer Immunol. Immunother. 2015, 64, 1251–1260. [Google Scholar] [CrossRef] [PubMed]
  132. Pule, M.A.; Savoldo, B.; Myers, G.D.; Rossig, C.; Russell, H.V.; Dotti, G.; Huls, M.H.; Liu, E.; Gee, A.P.; Mei, Z.; et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: Persistence and antitumor activity in individuals with neuroblastoma. Nat. Med. 2008, 14, 1264–1270. [Google Scholar] [CrossRef] [PubMed]
  133. 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] [PubMed] [Green Version]
  134. Jena, B.; Dotti, G.; Cooper, L.J.N. Redirecting T-cell specificity by introducing a tumor-specific chimeric antigen receptor. Blood 2010, 116, 1035–1044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Kanold, J.; Paillard, C.; Tchirkov, A.; Lang, P.; Kelly, A.; Halle, P.; Isfan, F.; Merlin, E.; Marabelle, A.; Rochette, E.; et al. NK Cell immunotherapy for high-risk neuroblastoma relapse after haploidentical HSCT. Pediatr. Blood Cancer 2012, 59, 739–742. [Google Scholar] [CrossRef] [PubMed]
  136. 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. 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]
Table 1. Current open clinical trials for refractory and relapsed neuroblastoma.
Table 1. Current open clinical trials for refractory and relapsed neuroblastoma.
NCT NumberTitleDrugMolecular TargetTherapeutic ClassStudy Enrollment TargetEligibility Age RangePrimary Site/Group or SponsorStatus
NCT00089245Radiolabeled Monoclonal Antibody Therapy in Treating Patients with Refractory, Recurrent, or Advanced CNS or Leptomeningeal CancerIntrathecal 131I-8H9B7-H3Systemic Radiotherapy120anyNew York, NY (Memorial Sloan Kettering Cancer Center)Active, recruiting
NCT00107289Iodine 131I Metaiodobenzylguanidine in Treating Patients with Recurrent, Progressive, or Refractory Neuroblastoma or Malignant Pheochromocytoma or Paraganglioma131I-MIBGNETSystemic Radiotherapy80over 1 yearNew York, NY (Memorial Sloan Kettering Cancer Center)Active, recruiting
NCT00601003Study of Nifurtimox to Treat Refractory or Relapsed Neuroblastoma or MedulloblastomaNifurtimox, cyclophosphamide, topotecanReactive Oxygen SpeciesSystemic Chemotherapy100up to 21 yearsBeat Childhood Cancer (BCC)Active, recruiting
NCT00638898Busulfan, Melphalan, Topotecan Hydrochloride, and a Stem Cell Transplant in Treating Patients with Newly Diagnosed or Relapsed Solid TumorBusulfan, melphalan, topotecanN/ASystemic Chemotherapy256 months to 40 yearsDuarte, CA (City of Hope Cancer Center)Active, not recruiting
NCT00788125Dasatinib, Ifosfamide, Carboplatin, and Etoposide in Treating Young Patients with Metastatic or Recurrent Malignant Solid TumorsCarboplatin, dasatinib, etoposide, ifosfamideMultiple Kinases (Abl, Src, c-Kit, PDGFRβ)Molecularly Targeted Therapy1431–25 yearsDuarte, CA (City of Hope Cancer Center)Active, not recruiting
NCT00877110Anti-GD2 3F8 Antibody and Allogeneic Natural Killer Cells for High-Risk NeuroblastomaCyclophosphamide, vincristine, topotecan, NK cells, 3F8GD2Systemic Chemotherapy plus Immunotherapy71anyNew York, NY (Memorial Sloan Kettering Cancer Center)Active, not recruiting
NCT00911560Bivalent Vaccine with Escalating Doses of the Immunological Adjuvant OPT-821, in Combination with Oral β-glucan for High-Risk NeuroblastomaAdjuvant OPT-821 in a vaccine containing two antigens (GD2L and GD3L) covalently linked to KLHGD2L, GD3LImmunotherapy215up to 21 yearsNew York, NY (Memorial Sloan Kettering Cancer Center)Active, recruiting
NCT011838843F8/GM-CSF Immunotherapy Plus 13-Cis-Retinoic Acid for Consolidation of Second or Greater Remission of High-Risk Neuroblastoma3F8, GM-CSF, isotretinoinGD2Immunotherapy63over 18 monthsNew York, NY (Memorial Sloan Kettering Cancer Center)Active, not recruiting
NCT011838973F8/GM-CSF Immunotherapy Plus 13-Cis-Retinoic Acid for Primary Refractory Neuroblastoma in Bone Marrow3F8, GM-CSF, isotretinoinGD2Immunotherapy31over 18 monthsNew York, NY (Memorial Sloan Kettering Cancer Center)Active, not recruiting
NCT01331135Aflac ST0901 CHOANOME—Sirolimus in Solid Tumors (Aflac ST0901)SirolimusmTORMolecularly Targeted Therapy24up to 30 yearsAtlanta, GA (Children’s Healthcare of Atlanta)Active, not recruiting
NCT01419834Humanized 3F8 Monoclonal Antibody (Hu3F8) in Patients with High-Risk Neuroblastoma and GD2-Positive TumorsHu3F8GD2Immunotherapy74over 2 yearsNew York, NY (Memorial Sloan Kettering Cancer Center)Active, recruiting
NCT01467986Multimodal Molecular Targeted Therapy to Treat Relapsed or Refractory High-Risk NeuroblastomaDasatinib, sirolimus, irinotecan, temozolomideMultiple Kinases (Abl, Src, c-Kit, PDGFRβ), mTORMolecularly Targeted Therapy114up to 25 yearsRegensburg, Germany (University Hospital Regensburg)Active, recruiting
NCT01492673Cyclophosphamide, Topotecan, and Bevacizumab (CTB) in Patients with Relapsed/Refractory Ewing’s Sarcoma and NeuroblastomaCyclophosphamide, topotecan, bevacizumabAngiogenesisMolecularly Targeted Therapy9up to 21 yearsNew York, NY (Memorial Sloan Kettering Cancer Center)Active, not recruiting
NCT01576692Combination Chemotherapy, Monoclonal Antibody, and Natural Killer Cells in Treating Young Patients with Recurrent or Refractory NeuroblastomaCyclophosphamide, topotecan, Hu14.18K322A, cisplatin, etoposide, doxorubicin, Vincristine, busulfan, melphalan, natural killer cells, GM-CSF, interleukin-2GD2Systemic Chemotherapy plus Immunotherapy34up to 21 yearsMemphis, TN (St. Jude Children’s Research Hospital)Active, not recruiting
NCT01601535Study of MLN8237 in Combination with Irinotecan and TemozolomideAlisertib (MLN8237), irinotecan, temozolomideAurora A KinaseMolecularly Targeted Therapy41–30 yearsNew Approaches to Neuroblastoma Therapy (NANT)Active, not recruiting
NCT01606878Crizotinib and Combination Chemotherapy in Treating Younger Patients with Relapsed or Refractory Solid Tumors or Anaplastic Large Cell LymphomaCrizotinib, cyclophosphamide, topotecan, vincristine, doxorubicinALK, c-MetSystemic Chemotherapy plus Molecularly Guided Therapy651–21 yearsChildren’s Oncology GroupActive, not recruiting
NCT01625351A Study of CD45RA+ Depleted Haploidentical Stem Cell Transplantation in Children with Relapsed or Refractory Solid Tumors and LymphomasAlemtuzumab, fludarabine, sirolimus, busulfan, melphalanmTORSystemic Chemotherapy plus Immunotherapy232–21 yearsMemphis, TN (St. Jude Children’s Research Hospital)Active, not recruiting
NCT01662804Humanized 3F8 Monoclonal Antibody (Hu3F8) When Combined with Interleukin-2 in Patients with High-Risk Neuroblastoma and GD2-positive Solid TumorsHu3F8, IL-2GD2Immunotherapy14over 13 monthsNew York, NY (Memorial Sloan Kettering Cancer Center)Active, not recruiting
NCT01711554Lenalidomide and Dinutuximab With or Without Isotretinoin in Treating Younger Patients with Refractory or Recurrent NeuroblastomaLenalidomide, dinutuximab, isotretinoinGD2Immunotherapy62up to 21 yearsNew Approaches to Neuroblastoma Therapy (NANT)Active, recruiting
NCT01742286Phase I Study of LDK378 in Pediatric, Malignancies with a Genetic Alteration in Anaplastic Lymphoma Kinase (ALK)Ceritinib (LDK378)ALKMolecularly Guided Therapy831–17 yearsNovartis PharmaceuticalsActive, not recruiting
NCT01757626Combination Therapy of Antibody Hu3F8 With Granulocyte- Macrophage Colony Stimulating Factor (GM-CSF) in Patients with Relapsed/Refractory High-Risk NeuroblastomaHu3F8 with GM-CSFGD2Immunotherapy224anyNew York, NY (Memorial Sloan Kettering Cancer Center)Active, recruiting
NCT01804634A Phase II Trial of Reduced Intensity Conditioning and Haploidentical BMT for High-risk Solid TumorsCyclophosphamide, fludarabine, XRTN/ASystemic Chemotherapy plus Immunotherapy20up to 40 yearsBaltimore, MD (Sidney Kimmel Cancer Center at Johns Hopkins)Active, recruiting
NCT01956669A Phase II Study of Pazopanib GW786034, NSC# 737754 in Children, Adolescents, and Young Adults with Refractory Solid TumorsPazopanibMultiple Kinases (VEGFR-1, VEGFR-2, VEGFR-3, PDGFRβ, c-kit)Molecularly Targeted Therapy1541–18 yearsChildren’s Oncology GroupActive, recruiting
NCT02013336Phase 1 Study of MM-398 Plus Cyclophosphamide in Pediatric Solid TumorsMM-398 (Irinotecan sucrosofate liposomes), cyclophosphamideN/ASystemic chemotherapy301–20 yearsSouth Plains Oncology ConsortiumActive, recruiting
NCT02030964N2012-01: Phase 1 Study of Difluoromethylornithine (DFMO) and Celecoxib with Cyclophosphamide/Topotecan (DFMO)DFMO, celecoxib, cyclophosphamide, topotecanODCMolecularly Targeted Therapy302–30 yearsNew Approaches to Neuroblastoma Therapy (NANT)Active, not recruiting
NCT02034981Phase 2 Study Assessing Efficacy and Safety of Crizotinib in Patients Harboring an Alteration on ALK, MET or ROS1CrizotinibALK, c-MetMolecularly Guided Therapy246over 1 yearVillejuif, France (Institut Gustave Roussy)Active, not recruiting
NCT02035137131I-MIBG Alone VS. 131I-MIBG With Vincristine and Irinotecan VS. 131I-MIBG With Vorinostat (N2011-01)131I-MIBG, vorinostat, vincristine/irinotecanNETSystemic radiotherapy plus Chemotherapy1052–30 yearsNew Approaches to Neuroblastoma Therapy (NANT)Active, recruiting
NCT02076906MR-guided High Intensity Focused Ultrasound (HIFU) on Pediatric Solid TumorsMR-HIFUN/AN/A14up to 30 yearsWashington, DC (Children’s National Medical Center)Active, recruiting
NCT02095132WEE1 Inhibitor MK-1775 and Irinotecan Hydrochloride in Treating Younger Patients with Relapsed or Refractory Solid TumorsIrinotecan, AZD1775 (adavosertib)Wee1Molecularly Targeted Therapy1542–21 yearsChildren’s Oncology GroupActive, recruiting
NCT02100891Phase 2 STIR Trial: Haploidentical Transplant and Donor Natural Killer Cells for Solid Tumors (STIR)NK cellsN/AImmunotherapy20anyMilwaukee, WI (Medical College of Wisconsin)Active, recruiting
NCT02124772Study to Investigate Safety, Pharmacokinetic (PK), Pharmacodynamic (PD) and Clinical Activity of Trametinib in Subjects with Cancer or Plexiform Neurofibromas and Trametinib in Combination with Dabrafenib in Subjects with Cancers Harboring V600 MutationsTrametinib, dabrafenibMEKMolecularly Guided Therapy1421–17 yearsNovartis PharmaceuticalsActive, recruiting
NCT02139397Study of DFMO in Combination with Bortezomib for Relapsed or Refractory NeuroblastomaDFMO, bortezomibODC, ProteasomeMolecularly Targeted Therapy38up to 21 yearsBeat Childhood Cancer (BCC)Active, recruiting
NCT02162732Molecular-Guided Therapy for Childhood Cancer Precision MedicineMolecularly Guided Therapy20013 months to 21 yearsBeat Childhood Cancer (BCC)Active, recruiting
NCT02163356Fenretinide Lym-X-Sorb + Ketoconazole + Vincristine for Recurrent or Resistant Neuroblastoma (SPOC2013-001)Fenretinide, ketoconazole, vincristineN/ASystemic Chemotherapy42up to 30 yearsSouth Plains Oncology ConsortiumActive, recruiting
NCT02169609Safety Study of Dinutuximab Combined with Immunotherapy to Treat NeuroblastomaDinutuximab, GM-CSF, isotretinoin, IL-2GD2Immunotherapy25anyBarcelona, Spain (Hospital Sant Joan de Deu)Active, not recruiting
NCT02173093Activated T Cells Armed with GD2 Bispecific Antibody in Children and Young Adults with Neuroblastoma and OsteosarcomaIL-2, GD2Bi-aATC, GM-CSFGD2Immunotherapy4013 months to 29 yearsDetroit, MI (Children’s Hospital of Michigan)Active, recruiting
NCT02298348Sorafenib and Cyclophosphamide/Topotecan in Patients with Relapsed and Refractory Neuroblastoma (N2013-02)Sorafenib, cyclophosphamide, topotecanMultiple Kinases (Raf, VEGFR-2, VEGFR-3, c-Kit)Molecularly Targeted Therapy18up to 30 yearsNew Approaches to Neuroblastoma Therapy (NANT)Active, not recruiting
NCT02304458Nivolumab With or Without Ipilimumab in Treating Younger Patients with Recurrent or Refractory Solid Tumors or SarcomasIpilimumab, nivolumabPD-1, CTLA-4Immunotherapy4841–30 yearsChildren’s Oncology GroupActive, recruiting
NCT02308527Activity Study of Bevacizumab with Temozolomide ± Irinotecan for Neuroblastoma in ChildrenBevacizumab, temozolomide, irinotecan, topotecanAngiogenesisMolecularly Targeted Therapy1601–21 yearsCancer Research UKActive, recruiting
NCT02311621Engineered Neuroblastoma Cellular Immunotherapy (ENCIT)-01Anti-CD171 CAR-T cellsCD171Immunotherapy4018 months to 26 yearsSeattle, WA (Seattle Children’s Hospital)Active, recruiting
NCT02332668A Study of Pembrolizumab (MK-3475) in Pediatric Participants with an Advanced Solid Tumor or Lymphoma (MK-3475-051/KEYNOTE-051)PembrolizumabPD-1Immunotherapy3106 months to 17 yearsMerck Sharp & Dohme Corp.Active, recruiting
NCT02343718Vinblastine and Temsirolimus in Pediatric Patients with Recurrent or Refractory Lymphoma or Solid Tumours Including CNS TumoursTemsirolimus, vinblastinemTORMolecularly Targeted Therapy71–18 yearsToronto, ON (Hospital for Sick Children)Active, not recruiting
NCT02378428MIBG Therapy for Patients with MIBG Avid Tumors (MIBG)131I-MIBGNETSystemic Radiotherapy651–40 yearsColumbus, OH (Nationwide Children’s Hospital)Active, recruiting
NCT02390843Simvastatin with Topotecan and Cyclophosphamide in Relapsed and/or Refractory Pediatric Solid and CNS Tumors (AflacST1402)Simvastatin, cyclophosphamide, topotecanHMG-coA ReductaseSystemic Chemotherapy361–29 yearsAtlanta, GA (Children’s Healthcare of Atlanta)Active, recruiting
NCT02441088Theranostics: 68GaDOTATOC and 90YDOTATOC (PRRT)90Y-DOTA-tyr3-Octreotide (90Y-DOTATOC)SSTRSystemic Radiotherapy25over 6 monthsIowa City, IA (University of Iowa)Active, not recruiting
NCT02452554Lorvotuzumab Mertansine in Treating Younger Patients with Relapsed or Refractory Wilms Tumor, Rhabdomyosarcoma, Neuroblastoma, Pleuropulmonary Blastoma, Malignant Peripheral Nerve Sheath Tumor, or Synovial SarcomaLorvotuzumab mertansine (IMGN-901)CD56Immunotherapy1141–30 yearsChildren’s Oncology GroupActive, not recruiting
NCT02508038TCRαβ+/CD19+ Depleted Haploidentical HSCT + ZoledronateZoledronate, TCRαβ+/CD19+ depleted Haploidentical HSCTN/AImmunotherapy217 months to 21 yearsMadison, WI (University of Wisconsin)Active, recruiting
NCT02520713The iCat2, GAIN (Genomic Assessment Informs Novel Therapy) Consortium Study Precision MedicineMolecularly Guided Therapy825up to 30 yearsBoston, MA (Dana-Farber Cancer Institute)Active, recruiting
NCT02536183A Phase I Study of Lyso-thermosensitive Liposomal Doxorubicin and MR-HIFU for Pediatric Refractory Solid TumorsMR-HIFU hyperthermia + lyso-thermosensitive liposomal doxorubicinN/ASystemic Chemotherapy34up to 30 yearsWashington, DC (Children’s National Medical Center)Active, recruiting
NCT02541604A Study to Evaluate the Safety, Tolerability, Pharmacokinetics, Immunogenicity, and Preliminary Efficacy of Atezolizumab (Anti-Programmed Death-Ligand 1 [PD-L1] Antibody) in Pediatric and Young Adult Participants with Solid TumorsAtezolizumabPD-L1Immunotherapy90up to 30 yearsHoffmann-La RocheActive, not recruiting
NCT02557854HIFU Hyperthermia with Liposomal Doxorubicin (DOXIL) for Relapsed or Refractory Pediatric and Young Adult Solid TumorsDoxil + MR-HIFU HyperthermiaN/ASystemic Chemotherapy141–40 yearsDallas, TX (University of Texas Southwestern Medical Center)Active, recruiting
NCT02573896Immunotherapy of Relapsed Refractory Neuroblastoma with Expanded NK CellsNK cells, dinutuximab, lenalidomideGD2Immunotherapy241 month to 30 yearsNew Approaches to Neuroblastoma Therapy (NANT)Active, not yet recruiting
NCT02574728Sirolimus in Combination with Metronomic Chemotherapy in Children with Recurrent and/or Refractory Solid and CNS TumorsSirolimus, celecoxib, etoposide, cyclophosphamidemTORMolecularly Targeted Therapy601–30 yearsAtlanta, GA (Children’s Healthcare of Atlanta)Active, recruiting
NCT02624388Study of Genistein in Pediatric Oncology Patients (UVA-Gen001) (UVA-Gen001)GenisteinN/ASystemic Chemotherapy501–21 yearsCharlottesville, VA (University of Virginia)Active, recruiting
NCT02630043Trial of Tolcapone With Oxaliplatin for NeuroblastomaTolcapone, oxaliplatinN/ASystemic Chemotherapy21up to 21 yearsBeat Childhood Cancer (BCC)Active, recruiting
NCT02638428Genomics-Based Target Therapy for Children with Relapsed or Refractory MalignancyIfosfamide, carboplatin, etoposide, axitinib, crizotinib, dasatinib, erlotinib, everolimus, imatinib, pazopanib, ruxolitinib, sorafenib, vandetanib, vemurafenib, trastuzumab, pazopanib, sorafenibPrecision MedicineMolecularly Guided Therapy90up to 18 yearsSeoul, Korea (Samsung Medical Center)Active, recruiting
NCT02639546Safety and Pharmacokinetics of Cobimetinib in Pediatric and Young Adult Participants with Previously Treated Solid Tumors (iMATRIXcobi)CobimetinibMEKMolecularly Targeted Therapy506 months to 30 yearsHoffmann-La RocheActive, recruiting
NCT02641314Metronomic Treatment in Children and Adolescents with Recurrent or Progressive High Risk NeuroblastomaPropranolol, celecoxib, cyclophosphamide, vinblastine, etoposideN/ASystemic Chemotherapy262–20 yearsCologne, Germany (University of Cologne)Active, recruiting
NCT02644460Abemaciclib in Children with DIPG or Recurrent/Refractory Solid Tumors (AflacST1501)Abemaciclib (LY2835219)CDK4/6Molecularly Targeted Therapy502–25 yearsAtlanta, GA (Children’s Healthcare of Atlanta)Active, recruiting
NCT02650401Study of RXDX-101 in Children with Recurrent or Refractory Solid Tumors and Primary CNS Tumors, With or Without TRK, ROS1, or ALK FusionsRXDX-101 (Entrectinib)TRK, ROS1, ALKMolecularly Guided Therapy1902–22 yearsHoffmann-La RocheActive, recruiting
NCT02650648Humanized Anti-GD2 Antibody Hu3F8 and Allogeneic Natural Killer Cells for High-Risk NeuroblastomaHu3F8, cyclophosphamide, NK cells, IL-2GD2Immunotherapy36anyNew York, NY (Memorial Sloan Kettering Cancer Center)Active, recruiting
NCT02743429Phase II Study of Monoclonal Antibody ch14.18/CHO Continuous Infusion in Patients with Primary Refractory or Relapsed Neuroblastomach14.18/CHOGD2Immunotherapy401–21 yearsGreifswald, Germany (University Medicine Greifswald)Active, recruiting
NCT02748135A Two-Part Study of TB-403 in Pediatric Subjects with Relapsed or Refractory MedulloblastomaTB-403N/ASystemic Chemotherapy366 months to 18 yearsBeat Childhood Cancer (BCC)Active, recruiting
NCT02761915A Cancer Research UK Trial of Anti-GD2 T-cells (1RG-CART)Anti-GD2 CAR-T cells, cyclophosphamide, fludarabineGD2Immunotherapy27over 1 yearLondon, UK (University College London)Active, recruiting
NCT02765243Anti-GD2 4th Generation CART Cells Targeting Refractory and/or Recurrent NeuroblastomaAnti-GD2 CAR-T cellsGD2Immunotherapy301–14 yearsChina (Zhujiang Hospital)Active, recruiting
NCT02780128Next Generation Personalized Neuroblastoma Therapy (NEPENTHE)Ceritinib, trametinib, HDM201, ribociclibALK, RAS-MAPK, p53Molecularly Guided Therapy1051–21 yearsPhiladelphia, PA (Children’s Hospital of Philadelphia)Active, recruiting
NCT02813135European Proof-of-Concept Therapeutic Stratification Trial of Molecular Anomalies in Relapsed or Refractory Tumors (ESMART)Ribociclib, topotecan, temozolomide, everolimus, AZD1775, carboplatin, olaparib, irinotecan, AZD2014, nivolumab, Vistusertib, selumetinib, enasidenib, lirilumabPrecision MedicineMolecularly Guided Therapy397up to 18 yearsVillejuif, France (Institut Gustave Roussy)Active, recruiting
NCT02909777Trial of CUDC-907 in Children and Young Adults with Relapsed or Refractory Solid Tumors, CNS Tumors, or LymphomaCUDC-907 (fimepinostat)PI3K, HDACMolecularly Targeted Therapy441–21 yearsBoston, MA (Dana-Farber Cancer Institute)Active, recruiting
NCT02919046Study Evaluating the Efficacy and Safety With CAR-T for Relapsed or Refractory Neuroblastoma in ChildrenAnti-GD2 CAR-T cellsGD2Immunotherapy221–14 yearsChina (Nanjing Children’s Hospital)Active, recruiting
NCT02982941Enoblituzumab (MGA271) in Children with B7-H3-expressing Solid TumorsEnoblituzumab (MGA271)B7-H3Immunotherapy1121–35 yearsMacroGenicsActive, recruiting
NCT02998983Racotumomab in Patients with High risk NeuroblastomaRacotumomabN-glycolil (NGc) GM3 (NGcGM3)Immunotherapy391–12 yearsBuenos Aires, Argentina (Hospital Universitario Austral)Active, recruiting
NCT03107988Study of Lorlatinib (PF-06463922)Lorlatinib, cyclophosphamide, topotecanALK, ROS1Molecularly Targeted Therapy401–90 yearsNew Approaches to Neuroblastoma Therapy (NANT)Active, recruiting
NCT03155620Targeted Therapy Directed by Genetic Testing in Treating Pediatric Patients with Relapsed or Refractory Advanced Solid Tumors, Non-Hodgkin Lymphomas, or Histiocytic Disorders (Pediatric MATCH)Palbociclib, selumetinib, ensartinib, vemurafenib, olaparib, larotrectinib, LY3023414, erdafitinibPrecision MedicineMolecularly Guided Therapy491–21 yearsChildren’s Oncology GroupActive, recruiting
NCT03189706Pilot Study of Chemoimmunotherapy for High-Risk NeuroblastomaHu3F8, irinotecan/temozolomide, GM-CSFGD2Immunotherapy20anyNew York, NY (Memorial Sloan Kettering Cancer Center)Active, recruiting
NCT03209869Treatment of Relapsed or Refractory Neuroblastoma with Expanded Haploidentical NK Cells and Hu14.18-IL2Hu14.18-IL2, NK cellsGD2Immunotherapy67 months to 21 yearsMadison, WI (University of Wisconsin)Active, recruiting
NCT03236857A Study of the Safety and Pharmacokinetics of Venetoclax in Pediatric and Young Adult Patients with Relapsed or Refractory MalignanciesVenetoclaxBCL-2Molecularly Targeted Therapy135up to 25 yearsAbbVieActive, recruiting
NCT03242603Immunotherapy of Neuroblastoma Patients Using a Combination of Anti-GD2 and NK Cells (NKEXPGD2)Haploidentical NK cells, ch14.18/CHOGD2Immunotherapy56 months to 25 yearsSingapore (National University Hospital)Active, recruiting
NCT03273712Dosimetry-Guided, Peptide Receptor Radiotherapy (PRRT) With 90Y-DOTA-tyr3-Octreotide (90Y-DOTATOC)90Y-DOTA-tyr3-Octreotide (90YDOTATOC)SSTRSystemic Radiotherapy206 months to 90 yearsIowa City, IA (University of Iowa)Active, recruiting
NCT03275402131I-burtomab Radioimmunotherapy for Neuroblastoma Central Nervous System/Leptomeningeal Metastases131I-burtomab (8H9)B7-H3Systemic Radiotherapy32up to 18 yearsY-mAbs TherapeuticsActive, not yet recruiting
NCT03294954GD2 Specific CAR and Interleukin-15 Expressing Autologous NKT Cells to Treat Children with Neuroblastoma (GINAKIT2)NKT cells, cyclophosphamide, fludarabineGD2Immunotherapy241–21 yearsHouston, TX (Baylor College of Medicine)Active, recruiting
NCT03332667MIBG With Dinutuximab131I-MIBG dinutuximabNET, GD2Systemic Radiotherapy plus Immunotherapy241–30 yearsNew Approaches to Neuroblastoma Therapy (NANT)Active, not yet recruiting
NCT03373097Anti-GD2 CAR T Cells in Pediatric Patients Affected by High Risk and/or Relapsed/Refractory NeuroblastomaAnti-GD2 CAR-T cellsGD2Immunotherapy421–18 yearsItaly (Bambino Gesù Hospital and Research Institute)Active, recruiting
NCT03434262SJDAWN: St. Jude Children’s Research Hospital Phase 1 Study Evaluating Molecularly-Driven Doublet Therapies for Children and Young Adults with Recurrent Brain TumorsRibociclib, gemcitabine, trametinib, sonidegibPrecision MedicineMolecularly Guided Therapy1081–39 yearsMemphis, TN (St. Jude Children’s Research Hospital)Active, recruiting
NCT03458728Safety, Tolerability, Efficacy and Pharmacokinetics of Copanlisib in Pediatric PatientsCopanlisib (BAY806946)PI3KMolecularly Targeted Therapy1306 months to 21 yearsBayer PharmaActive, recruiting
NCT03478462Dose Escalation Study of CLR 131 in Children and Adolescents with Relapsed or Refractory Malignant Brain Cancer, Neuroblastoma, Rhabdomyosarcoma, Ewings Sarcoma, and OsteosarcomaCLR 131N/ASystemic Radiotherapy302–21 yearsMadison, WI (University of Wisconsin)Active, not yet recruiting
NCT03507491Nab-paclitaxel in Combination with Gemcitabine for Pediatric Relapsed and Refractory Solid TumorsNab-paclitaxel, gemcitabineN/ASystemic Chemotherapy246 months to 30 yearsAtlanta, GA (Children’s Healthcare of Atlanta)Active, not yet recruiting
NCT03561259A Study of Therapeutic Iobenguane (131I) for High-Risk Neuroblastoma at the Time of First Relapse (OPTIMUM)131I-MIBGNETSystemic Radiotherapy65over 1 yearJubilant DraxImage Inc.Active, recruiting
Molecularly Targeted Therapy—Targeted agent used in non-targeted fashion; Molecularly Guided Therapy—Targeted agent used in targeted patient population based on molecular aberration or phenotype; NET—norepinephrine transporter; SSTR—somatostatin receptor; XRT—radiation therapy; ODC—ornithine decarboxylase, HIFU—high-intensity focused ultrasound, HDAC—histone deacetylase; MIBG—meta-iodobenzylguanidine; CAR-T cells—chimeric antigen receptor T cells; ALK—anaplastic lymphoma kinase; NK cells—natural killer cells.

Share and Cite

MDPI and ACS Style

Zage, P.E. Novel Therapies for Relapsed and Refractory Neuroblastoma. Children 2018, 5, 148. https://doi.org/10.3390/children5110148

AMA Style

Zage PE. Novel Therapies for Relapsed and Refractory Neuroblastoma. Children. 2018; 5(11):148. https://doi.org/10.3390/children5110148

Chicago/Turabian Style

Zage, Peter E. 2018. "Novel Therapies for Relapsed and Refractory Neuroblastoma" Children 5, no. 11: 148. https://doi.org/10.3390/children5110148

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