Immunotherapy for Neuroblastoma


Acknowledgments

We thank Joe Olechnowicz for editorial assistance.

Introduction

Standard care for patients with high-risk neuroblastoma (NB) currently consists of aggressive multimodal therapy including high-dose chemotherapy, surgery, and radiotherapy . Immunotherapy utilizes mechanisms of action that are not cross-reactive with other antitumor modalities, and is thus, an attractive therapeutic option, particularly to eradicate minimal residual disease. Moreover, in the young children typically afflicted with NB, chemotherapy and radiotherapy, which have significant off-target effects on normal tissue, are associated with significant long-term morbidities. These include growth impairment and asymmetry, hearing deficits, learning difficulties, and secondary malignancies . Immunotherapy, by virtue of its specificity for identified tumor targets, has the potential to avoid or mitigate some of these toxicities. Indeed, NB is the only pediatric solid tumor for which immunotherapy is an established therapeutic modality given the approval of the anti-GD2 antibody dinutuximab for high-risk NB by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) in 2015 .

Targets for NB Immunotherapy

The properties for an ideal tumor antigen targetable by immunotherapy include specificity, role in oncogenesis, expression level, and immunogenicity, although none of the currently targeted antigens meet all criteria . In adults, the discovery of mutation-associated neoantigens has vastly expanded the pool of antigens that can be potentially used for immunotherapy . Most pediatric malignancies, including NB, have far fewer genetic mutations, thus restricting a strategy aimed at neoantigens . Immunotherapy for pediatric malignancies has mostly focused on targeting nonmutated antigens with a differential expression on malignant versus normal cells. Cell surface antigens, the most commonly targeted molecules in NB immunotherapy, are not major histocompatibility complex (MHC)-restricted and are optimal for targeting by monoclonal antibodies (MoAbs) or engineered immune effector cells. Intracellular tumor antigens usually require processing by antigen-presenting cells for presentation to cytotoxic lymphocytes and are MHC-restricted . MoAbs can target intracellular antigens if they internalize after binding. Tumor antigens targeted for immunotherapy in patients with NB thus far are chiefly expressed on the cell surface, the ganglioside GD2 being the most common ( Table 9.1 ). Other cell surface antigens include L1CAM and B7H3 . The intracellular cancer-testis antigens MAGE and NY-ESO-1 have also been clinically investigated ( Table 9.2 ).

Table 9.1
Selected Clinical Trials of GD2-Directed Immunotherapies With Published Results
Agent Combination Phase Summary of Results References
Murine Antibodies
m3F8 I
  • Toxicity profile established: common toxicities included pain and allergic reactions

m3F8 SC GMCSF II
  • Activity against chemorefractory NB

  • Improved PFS and OS compared to historical data for patients in first and subsequent remissions

  • Improved outcomes when GMCSF given SC rather than IV

  • Biomarkers influencing outcome identified

m3F8 barley- or yeast-derived-β-D- glucan I
  • No additional toxicities of BG

  • Activity against chemorefractory NB

14.G2a I
  • Toxicity profile established: common toxicities included pain and allergic reactions

Chimeric Antibodies
Dinutuximab I
  • Toxicity profile established: common toxicities included pain and allergic reactions.

Dinutuximab Randomized II/III
  • 3-year EFS superior for immunotherapy group compared to maintenance chemotherapy or no therapy

Dinutuximab IL-2 + GM-CSF; CRA Randomized III
  • 2-year EFS and OS superior for immunotherapy + CRA versus CRA

Dinutuximab I/T/GMCSF; temsirolimus Randomized II
  • Superior response rate for dinutuximab + I/T/GMCSF compared to dinutuximab + temsirolimus

Dinutuximab-beta I
  • Toxicity profile and pharmacokinetics similar to dinutuximab

Dinutuximab-beta SC IL-2 Randomized III
  • Higher toxicities in IL-2 treated group

  • EFS and OS similar for intent-to-treat analysis but favorable trend for combination in patients completing planned therapy

Humanized Antibodies
hu14.18K322 A I
  • Toxicity profile similar to dinutuximab though higher doses tolerable

Naxitamab (hu3F8) GM-CSF I
  • Toxicity profile similar to m3F8 though allergic reactions rare; higher doses tolerable compared to m3F8

  • Anti-NB activity observed

  • Lower immunogenicity compared to m3F8

Immunocytokines
Hu14.18-IL-2 I/II
  • Toxicity profile similar to dinutuximab + IL-2

  • Responses observed in osteomedullary NB

Radioimmunoconjugates
131 I- m3F8 I
  • Myelosuppression requiring bone marrow rescue required in most patients

  • Dosimetry data obtained; radiation exposure was well below tolerable doses for all organs

131 I- m3F8 II
  • Patients received myeloablative doses after induction chemotherapy

  • Myelosuppression was reversible with autologous bone marrow rescue

  • Long-term EFS for HR-NB was 40%

131 I- m3F8 Bevacizumab I
  • Myelosuppression requiring stem cell rescue required in most patients

  • Bevacizumab did not interfere with 3F8 targeting to tumor sites

131 I- m3F8 IO I
  • Toxicity defined: headache, fever, vomiting

  • Recommended phase II dose established

Cell-Mediated Immunotherapy
Haploidentical NK cells + m3F8 Chemotherapy I
  • No unexpected toxicities (including GvHD) beyond those expected with chemotherapy and m3F8

  • NK-cell dose-dependent anti-NB activity observed

Haploidentical NK cells + hu14.18K322 A Myeloablative chemotherapy Component of phase II
  • Toxicities primarily related to chemotherapy and hu14.18K322 A

Haploidentical NK cells + hu14.18K322 A Combination chemotherapy Pilot
  • Toxicities primarily related to chemotherapy and hu14.18K322 A

  • Anti-NB activity noted

First generation CAR T-cells Chemotherapy I
  • Well-tolerated without neuropathic pain

  • Anti-NB activity noted

  • Persistence of CAR T cells for several years after infusion

Third generation CAR T-cells Chemotherapy I
  • Well-tolerated without neuropathic pain

Vaccines
Anti-m3F8 antiidiotype BCG I
  • Well tolerated; minimal toxicities

Anti-ch14.18 antiidiotype I
  • Well tolerated; minimal toxicities

Bivalent anti-GD2/GD3 KLH, OPT-821, yeast-derived-β-D- glucan I
  • Well tolerated; minimal toxicities

  • Prolonged EFS and OS for patients in second remission

BCG , Bacillus Calmette-Guerin; BG , β-glucan; CAR , Chimeric antigen receptors; CRA , cis-retinoic acid; EFS , Event-free survival; GM-CSF , Granulocyte macrophage colony stimulating factor; GvHD , Graft-versus-host disease; HR-NB , High-risk neuroblastoma; IL-2 , Interleukin-2; KLH , Keyhole limpet hemocyanin; NB , Neuroblastoma; NK-cell , Natural killer cell; OS , overall survival; PFS , Progression-free survival; SC , Subcutaneous

Table 9.2
Selected Clinical Trials of Immunotherapies Directed Against Targets Other Than GD2 With Published Results
Antigen Agent Phase Summary of Results References
B7H3 Radioimmunoconjugate 131 I-omburtamab (8H9) I/II
  • Toxicity profile studied: myelosuppression was main toxicity

  • Recommended phase II dose for NB established

  • Improved survival in patients with CNS relapse of NB

L1-CAM Radioimmunoconjugate 131 I-chCE7 Imaging
  • Radioimmunodetection of NB feasible

CAR T-cells I
  • Toxicity mild

  • No significant anti-NB activity

MAGE-A1, MAGE-A3, NY ESO1+ decitabine Dendritic cells I
  • Toxicity primarily related to decitabine

Various Autologous tumor cells secreting IL-2 ± lymphotactin I
  • Toxicity mild

  • Anti-NB immune responses observed

Allogeneic tumor cells secreting IL-2 I
  • Toxicity mild

  • Immune responses suboptimal

Dendritic cells pulsed with tumor RNA I
  • Toxicity mild

  • Modest anti-NB immune responses noted

CAR T-cells , chimeric antigen receptor T-cells; CNS , Central nervous system; NB , Neuroblastoma; RNA , Ribonucleic acid

GD2

Gangliosides, such as GD2, are complex, acidic glycolipids expressed on the outer cell membrane. GD2 is biosynthesized from precursor gangliosides GD3/GM3 by β -1,4-N-acetylgalactosaminyltransferase (GD2 synthase) . NB cells have high levels of the GM2/GD2 synthase transcript, enzyme activity, and GD2 expression with an estimated 5–10 million molecules/cell . In normal tissues, GD2 expression is largely limited to neurons, skin melanocytes, and peripheral pain fibers, making it well suited for targeted antitumor therapy . GD2 is not immunomodulated off the cell membrane and persists on NB cell membranes post-therapy . Circulating GD2 levels both in the blood and cerebrospinal fluid (CSF) are not high enough to interfere with binding to anti-GD2 specific MoAbs .

L1-CAM

L1-CAM (CD171) is a member of the L1 family of adhesion molecules within the immunoglobulin superfamily. It is known to be involved in axon guidance, neural cell migration, and differentiation, and induces constitutive nuclear factor-kappa B (NFκB) activation in tumor cells . In solid tumors in adults, L1-CAM overexpression confers metastatic capacity but does not appear to be associated with a poor prognosis in pediatric solid tumors . L1-CAM has been targeted by both MoAbs and chimeric antigen receptor T-cells (CAR-T cells) .

B7H3

B7H3 (CD276) is an immune checkpoint member of the B7 and CD28 families and is widely expressed on a range of solid tumors including NB with a restricted expression on normal tissues . Induced on antigen presenting cells, B7H3 plays an important role in regulating the innate immune system as well as adoptive immunotherapy , and its expression appears to be mediated by the micro-RNA mir-29 . B7H3 has been targeted clinically by MoAbs as a tumor antigen and for its putative immunomodulatory effect .

Oncofetal Antigens

Oncofetal antigens, a group of antigens of embryonic origin, are normally expressed only during normal fetal development but are abnormally present in tumors. NB expresses several such oncofetal antigens including melanoma antigen encoding gene (MAGE)-1 seen in the absence of MYCN amplification, MAGE-3 (which is associated with the absence of metastasis), and NY-ESO-1 . These are potential targets for native T cell receptors and have been targets for immunotherapy .

Others

Other antigens that have been clinically targeted on pediatric solid tumors, including but not specifically NB, include vascular endothelial growth factor (VEGF) , insulin-like growth factor-1 receptor (IGF-1R) , neural cell adhesion molecule , and endosialin .

Effectors of Immunotherapy for NB

NB can be potentially targeted by all the major effector cells and proteins of the immune system ( Fig. 9.1 ). Innate immune system components including complement, MoAbs, macrophages, polymorphonuclear leukocytes, and natural killer cells have been recruited for passive anti-NB immunotherapy in the clinic. Natural killer T cells (NKT) and γδ T cells can play roles as both innate and adaptive components and are being studied in the early phase trials. The adaptive immune system requires direct activation through antigen presentation by antigen-presenting cells (APCs). Upon antigen presentation and activation, antigen-specific T and B cells are generated. B-cell activation can generate humoral immunity against NB . However, expression of human leukocyte antigen (HLA) and PD-L-1 is absent or low in NB, limiting the effect of potent T-cell-mediated active immunotherapy. Strategies to redirect and harness T-cells in NB immunotherapy include MHC-independent approaches such as chimeric antigen receptor-transduced T-cells and bispecific MoAbs .

Figure 9.1, Effectors of immunotherapy for neuroblastoma [30] .

Cytokines

MoAb-based NB cell lysis involves recognition and binding to the tumor cell, followed by recruitment of immune effectors, which then release cytotoxic molecules for NB cell destruction . However, effector function, particularly antibody-dependent cellular cytotoxicity (ADCC), is often compromised in cancer patients due to immune suppression from both the underlying disease and the myelosuppressive treatment regimens . Cytokines can augment effector cell function and enhance ADCC . In general, these immunomodulators stimulate a broad range of immune cells, including T and B lymphocytes, monocytes, macrophages, and NK cells. The cytokines interleukin-2 (IL-2) and granulocyte macrophage colony stimulating factor (GM-CSF) have been used extensively for NB immunotherapy in conjunction with MoAbs. Other cytokines have the potential to be more effective but have not yet been tested in patients with NB.

Interleukin-2

IL-2 is a pro-inflammatory cytokine with effects on both innate and adaptive immunity. It stimulates growth, proliferation, and activation of the T cell, B cell, and NK cell lineage . However, as a single agent, it has only modest anti-NB activity . Adverse events include capillary leak, hypoxia, pain, rash, allergic reaction, elevated transaminases, and hyperbilirubinemia particularly at high doses and when administered intravenously (IV). A negative impact on MoAb pharmacokinetics has also been observed .

Granulocyte Macrophage Colony Stimulating Factor

GM-CSF directly activates the clonal expansion and maturation of partially committed progenitor cells in the granulocyte-macrophage pathways to form monocytes, macrophages, and dendritic cells . It has been shown both in vitro and in vivo to enhance antitumor immunity through direct activation of monocytes, macrophages, dendritic cells, and ADCC . In addition, it indirectly activates T cells via secretion of tumor necrosis factor, interferon, and IL-1 . GM-CSF increases the number of circulating neutrophils and eosinophils but does not affect complement levels . Exposure in vitro or in vivo to GM-CSF also primes monocytes/macrophages for greater antineoplastic cytotoxicity and enhances the proliferation, maturation, and function of APCs . This includes antigen processing and presentation by macrophages and dendritic cells effects that might promote induction of antitumor activity, through an idiotypic network. It is well tolerated, without significantly enhancing the known toxicities of anti-GD2 MoAbs .

Other Cytokines

Other cytokines being considered for clinical development include IL-15 and IL-21. IL-15 stimulates the proliferation and cytotoxicity of the NK cells and CD8-T cells. Recent efforts to enhance pharmacokinetics of IL-15 involve the production of IL-15/IL15-receptor conjugates . Anti-NB activity has been demonstrated in several pre-clinical NB models . IL-21 is a pleiotropic cytokine that stimulates multiple immune cells . In murine models, IL-21 combined with other immunotherapies showed anti-NB activity, producing a whole-cell vaccine given to treat mice previously challenged with wild-type tumor cells .

Monoclonal Antibodies

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here