Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Oct 1.
Published in final edited form as: Semin Oncol. 2014 Jul 21;41(5):589–612. doi: 10.1053/j.seminoncol.2014.07.003

GD2-targeted immunotherapy and radioimmunotherapy

Konstantin Dobrenkov 1, Nai-Kong Cheung 1
PMCID: PMC4254523  NIHMSID: NIHMS615855  PMID: 25440605

Abstract

Ganglioside GD2 is a tumor-associated surface antigen found in a broad spectrum of human cancers and stem cells. They include pediatric embryonal tumors (neuroblastoma, retinoblastoma, brain tumors, osteosarcoma, Ewing’s sarcoma, rhabdomyosarcoma), as well as adult cancers (small cell lung cancer, melanoma, soft tissue sarcomas). Because of its restricted normal tissue distribution, GD2 has been proven safe for antibody targeting. Anti-GD2 antibody is now incorporated into the standard of care for the treatment of high risk metastatic neuroblastoma. Building on this experience, novel combinations of antibody, cytokines, cells and genetically engineered products all directed at GD2 are rapidly moving into the clinic. In the review, past and present immunotherapy trials directed at GD2 will be summarized, highlighting the lessons learned and the future directions.

Keywords: GD2, neuroblastoma, immunotherapy, cytokines, cytotherapy

Introduction

Targeting tumor associated antigens (TAA) with immune effectors, including monoclonal antibodies (MAbs) and T-cells, has achieved substantial clinical benefits in cancer therapy, thereby energizing scientists and the pharmaceutical industry in the past decade. The goal is to find a target with ideal efficacy and safety profile, while continuing to optimize immune effectors. The context where TAA targeted therapy is applied can be critical for its success. For example, the leading cause of cancer treatment failure is minimal residual disease (MRD), i.e. the small number of cells that survive conventional therapy which in turn provide the nidus for regrowth or new metastasis. Immunotherapy targeting TAA holds promise for the eradication of MRD by virtue of its tumor-selectivity with toxicities that do not overlap with those of conventional chemotherapy or radiation therapy. Here, we review GD2-directed therapy, with an emphasis on immunotherapy and radioimmunotherapy applied to neuroblastoma and other GD2-positive tumors.

GD2, a disialoganglioside, is an oncofetal antigen that is expressed in the fetus. It is also found on neural stem cells,1 mesenchymal stem cells,2 and breast cancer stem cells.3,4 Postnatally it is detected on peripheral neurons, central nervous system and skin melanocytes.5 Its biological role is poorly understood. It is probably important in mediating attachment of tumor cells to extracellular matrix proteins.6 Since the first report on GD2 as a TAA in neuroblastoma,7 much is still unknown about its function in developmental biology or in oncogenesis. Compared to other antigens, GD2 expression is high and relatively homogeneous between and within neuroblastoma tumors, although heterogeneity has also been described.8 Based on multiple criteria (e.g. therapeutic function, immunogenicity, role of the antigen in oncogenicity, specificity, expression level and percent of antigen-positive cells, stem cell expression, number of patients with antigen-positive cancers, number of antigenic epitopes, and cellular location of antigen expression), the National Cancer Institute program for prioritization of cancer antigens ranked GD2 12th among a list of top 75 cancer antigens.9 Its rank becomes even higher if only directly targetable antigens are selected. In addition to neuroblastoma, other malignant tumors such as melanoma,10 soft tissue sarcomas,11 osteosarcoma,12 desmoplastic small round cell tumor (DSRCT),13 small cell lung cancer (SCLC)14 express GD2, although with more heterogeneity by immunohistochemistry.

Neuroblastoma is the most common extracranial solid tumor of childhood. About 90% of patients are diagnosed before their 6th birthday, with a median age of 19 months. In contrast to low and intermediate risk neuroblastoma (50% of cases), long term prognosis for high-risk neuroblastoma remains dismal.15 The introduction of murine monoclonal antibodies (e.g. 3F816 and 14G2a17) specific for the penta-saccharide moiety of GD2 initiated a new era of neuroblastoma immunotherapy more than 2 decades ago. The landmark randomized trial carried out by the Children’s Oncology Group (COG) definitively demonstrated an overall survival benefit of anti-GD2 antibody ch14.18 when combined with cytokines (interleukin-2 (IL-2) and granulocyte–macrophage colony stimulating factor (GM-CSF)), and anti-GD2 antibody therapy has now become the standard of care for patients with high-risk neuroblastoma.18 Humanized 3F8 (hu3F8)19 is now proven safe and non-immunogenic in phase I clinical trials.20,21 Granulocyte-mediated antibody dependent cell-mediated cytotoxicity (ADCC)22 and natural killer (NK) cell mediated NK-ADCC23 are important effector mechanisms. Despite the small number of patients for this orphan disease (650 new cases in the US each year and only 50% with high risk disease), important lessons in antibody immunotherapy have been learned.

Anti-GD2 monoclonal antibodies

Murine antibodies: IgG3 antibody 3F8 (see table 1 for clinical trials)

Table 1.

Clinical Trials of Murine Anti-GD2 Antibodies.

Type of antibody ClinicalTrials.gov identifier/reference Study type Type of therapy (number of patients) Dose, mg/m2/day Toxicity profile Best Response Long Term Survival (5 years from start of Mab therapy) Important observations

IgG3 3F8 −/[24][26] Phase I 3F8 antibody in patients with neuroblastoma (8) and melanoma (9) 5–100 (single dose per cycle), 1 cycle hypertension (dose-limiting), fever, pain, urticaria 2 CR (2 PR when reassessed using INRC) Not reported Specific binding to tumor; correlation between HAMA level, side effects and response
IgG3 3F8 and heat-modified 3F8 NCT00450307/[27] Phase I 3F8 antibody dose escalation plus GM-CSF in high-risk neuroblastoma (30) 20–160 (x 5 days per cycle), 2 cycles fever, pain, urticaria, hypertension 3 CR
2 PR
Not reported Heat-modified 3F8 antibodies decrease toxicity without blunting anti-NB activity
IgG3 3F8 NCT00037011/[37] Phase I 3F8 and barley-derived-β-D-glucan in chemoresistant neuroblastoma (24) 10 (x 10 days per cycle), upto 4 cycles pain, fever, urticaria, thrombocytopenia MIBG response 13/22 Not reported No DLT for barley glucan up to 80 mg/kg/dayx1 0 days; antineoplastic activity in advanced-stage neuroblastoma
IgG3 3F8 NCT00492167/[38] Phase I 3F8 and yeast-derived-β-D-glucan in chemoresistant neuroblastoma (44) 10 (x 10 days per cycle) pain, fever, urticaria MIBG response: 7/24
CR of BM: 3/5
Overall response: 1 CR, 1 PR, 22 SD, 17 PD
Not reported No DLT for yeast glucan up to 160 mg/kg/day x17 days; antineoplastic activity in advanced-stage neuroblastoma
IgG2a (14.G2a) −/[40] Phase I prolonged iv infusion of 14G2a antibody in patients with neuroectodermal tumors (18) 10–40 (x 5 days per cycle), 1–3 cycles pain, hyponatremia, fever, rash, paresthesias, weakness, chronic hypotension 2/5 PR in neuroblastoma Not reported modest antitumor activity at the expense of significant toxicity
IgG2a (14.G2a) −/[41] Phase I 14.G2a antibody plus interleukin-2 in GD2+ tumors (33) 2–60 (x 5 days per cycle), 1–3 cycles pain, fever, rash, allergic or anaphylactic reactions 1/31 PR in neuroblastoma, 1/2 CR in osteosarcoma Not reported Rare antitumor effect in bulky disease
IgG3 3F8 −/[28, 35] Phase II 3F8 antibody alone in treatment of minimal residual stage 4 neuroblastoma (34) 10 (x 10 days per cycle) upto 4 cycles pain, fever, urticaria, reversible decreases in blood counts MIBG response 6/16
CR of BM (immunocytology) 6/9
CR pf MRD 7/12
For patients treated in 1st remission: PFS at 5y = 44%
OS at 5y = 49%
long-term remission without autologous BMT can be achieved with 3F8; transient HAMA associated with significantly better survival

IgG3 3F8 NCT00002560/[30] Phase II 3F8 antibody and iv GM-CSF in refractory neuroblastoma (45) 10 (x 10 days per cycle), q4–8 weeks up to 2 years (range 1–14) Pain, rash CR of BM 12/19 (primary refractory), 5/10 (secondary refractory) neuroblastoma PFS at 5y (primary refractory) = 11%; OS at 5y = 17% 3F8/GM-CSF very effective against chemoresistant primary refractory neuroblastoma in bone marrow, but not PD or soft tissue masses
IgG3 3F8 NCT00072358/[33] Phase II 3F8 antibody and sc GM-CSF in high risk primary refractory stage 4 neuroblastoma (79) 20 (x 5 days per cycle), q1–3 months to 2 years (mean 7.7, median 7, range 1–25 cycles) Pain, fever, urticaria, hyperension, hypoxia, line infection, transient ALT/AST elevations, reversible decreases in WBC CR of BM: 40/46 (87%)
CR of MBIG: 10/26 (38%)
PFS at 5 y = 24%
OS at 5y = 65%
Favorable prognostic markers: HAMA, FCGR2A high affinity allele
Adverse prognostic markers: postive MRD after 2 cycles of 3F8/GMCSF
Not prognostic: MYCN amplifcation, prior stem cell transplant

IgG3 3F8 NCT00002560/[34] Phase II 3F8 antibody and iv GM-CSF in high risk stage 4 neuroblastoma in first remission (41) 20 (x 5 days per cycle), q1–3 months to 2 years (mean 4.6, median 4, range 1–13 cycles) Pain, fever, urticaria, hyperension, hypoxia, line infection, transient ALT/AST elevations, reversible decreases in WBC NA PFS at 5y = 56%
OS at 5y = 61%
Relapse mostly at isolated sites (CNS, soft issue and marrow/bon e).
Favorable prognostic factors: missing ligand for inhibitory killer immunoglobulin-like receptor (KIR), HAMA, and scGM-CSF.
Adverse prognostic factors: difficulty getting into first remission, MRD(+) after 2 cycles of immunotherapy.
Not prognostic: MYCN amplification, prior stem cell transplant
IgG3 3F8 NCT00072358/[34] Phase II 3F8 antibody and sc GM-CSF in high risk (57) and ultra high risk (28) stage 4 neuroblastoma in first remission 20 (x 5 days per cycle), q1–3 months to 2 years (mean 6.4, median 6, range 1–14 cycles) Pain, fever, urticaria, hyperension, hypoxia, line infection, transient ALT/AST elevations, reversible decreases in WBC NA HR: PFS at 5y = 62%, OS at 5y = 81%; UHR PF at 5 y = 36%, OS at 5 y = 75%

IgG3 3F8 NCT00072358/[35] Phase II 3F8 antibody and sc GM-CSF in high risk stage 4 neuroblastoma in second remission (101) 20 (x 5 days per cycle), q1–3 months to 2 years (mean 4.4, median 4, range 1–12 cycles) Pain, fever, urticaria, hyperension, hypoxia, line infection, transient ALT/AST elevations, reversible decreases in WBC NA PFS at 4 y = 33%
OS at 5y = 53%
Long-term PFS and OS are possible for after relapse if patients achieve >=2nd CR/VGPR
Favorable prognostic variables: ≤18 months at diagnosis, maintenance chemotherapy, rituximab for HAMA
Unfavorable prognostic marker: MRD(+) after 2 cycles of immunotherapy

In the first-in-human phase I study of anti-GD2 MAb, toxicity of murine IgG3 antibody 3F8 was assessed in 17 patients with neuroblastoma and malignant melanoma.24 Tumor response by using International Neuroblastoma Response Criteria (INRC)25 was observed in 2 of 17 patients with neuroblastoma.26 Importantly, this study demonstrated specific binding of 3F8 antibodies to tumor using in vitro immunostaining and in vivo radioimaging with 131I. Patients with significant levels of circulating human-anti-mouse antibody (HAMA) had minimal side effects, but also no therapeutic benefits from immunotherapy. Those with level of HAMA <1,000 U/mL had many side effects together with therapeutic responses. Toxicity profile included hypertension, fever, urticaria and pain. In subsequent studies it was shown that reduction of pain side effects could be achieved with pretreatment with heat-modified 3F8 to desensitize peripheral nerves. This approach was tested in Phase I clinical trial, showing feasibility of 3F8 dose-escalation without increase in analgesic requirements.27 In all subsequent trials 3F8, was only given if the HAMA test was <1,000 U/mL.

Clinical benefit was demonstrated in a phase II trial where 3F8 alone was used for remission consolidation in patients >1 year of age with stage 4 neuroblastoma.28 At study entry most of the patients had complete or very good partial remission (CR/VGPR), albeit 74% of these patients were found to have MRD when tested by non-conventional methods including immunoscintigraphy, immunocytology, quantitative (q)RT-PCR. This study demonstrated that long-term remission (>20 years) could be achieved with 3F8 treatment without myeloablative therapy (e.g. autologous marrow or stem cell transplantation).

In subsequent studies 3F8 was combined with cytokines, β-glucans, or cellular immune effectors. A phase I study of 3F8 combined with escalating doses of IL-2 was terminated after 12 patients because of capillary leak syndrome and unfavorable pharmacokinetics (unpublished results). Addition of intravenous (iv) GM-CSF led to enhancement of granulocyte-mediated ADCC.29 In a Phase I trial in patients with refractory chemo-resistant neuroblastoma in the bone marrow (BM), response was achieved in 12/19 patients with primary refractory, and 5/10 with secondary refractory neuroblastoma.30 It was shown that the use of qRT-PCR for detection of specific markers of MRD served as surrogate to achieve more precise evaluation of response to 3F8 therapy and patient stratification.31 Subsequent studies on the polymorphism of FCGR2A gene which encodes the Fcγ-receptor IIA (CD32), demonstrated a correlation between the high affinity allele and clinical outcome in patients treated with 3F8 and GM-CSF. These findings imply that genetic polymorphism can predict therapeutic efficacy and might be a potential tool for selection of patients who can most benefit from therapy.32 In a subsequent trial,33 79 patients with persistent osteomedullary neuroblastoma documented by histology and/or metaiodobenzyl-guanidine (MIBG) scan were treated with 3F8 plus subcutaneous (sc) GM-CSF, while isotretinoin was also included after remission was achieved. Complete response rates to 3F8+scGM-CSF were 87% by histology and 38% by MIBG. Five-year progression-free survival (PFS) was 24%±6%, which was significantly superior to 11%±7% with 3F8+ivGM-CSF (p=0.002); five-year overall survival (OS) was 65%±6%. In a multivariate analysis, significantly better PFS was associated with R/R or H/R FCGR2A polymorphism, sc route of GM-CSF, and early MRD response. MYCN amplification was not prognostic of outcome. Complement consumption was similar with either route of GM-CSF. Toxicities were manageable, allowing outpatient treatment.

A summary of 169 patients with high risk neuroblastoma, i.e. stage 4 diagnosed at ≥18 months of age or those with MYCN amplification treated in their first remission with 3F8 ± GM-CSF ± isotretinoin, showed that at 5 years from the start of immunotherapy, PFS improved from 44% (3F8 only) to 56% (3F8+ivGM-CSF) and 62% (3F8+scGM-CSF). OS was 49%, 61%, and 81%, respectively. Relapse was mostly at isolated sites (CNS, soft tissues and marrow/bone). Independent adverse prognostic factors included difficulty getting into first remission and positivity of marrow MRD by qRT-PCR after 2 cycles of immunotherapy (post-MRD), whereas favorable prognostic factors included missing ligand for inhibitory killer immunoglobulin-like receptor (KIR), positive HAMA response, and scGM-CSF.34 Phase II studies of high dose 3F8 (4-fold higher) in combination with GM-CSF (NCT01183429, NCT01183884, NCT01183897) currently recruit patients and the outcome analyses will be done.

The potential of 3F8 to control MRD was further tested in 101 patients with ultra high risk neuroblastoma, i.e. stage 4 neuroblastoma diagnosed after 18 months of age or with MYCN amplification, whose disease progressed before being salvaged back into ≥2nd remissions.35 They were treated with 3F8/GM-CSF plus isotretinoin. PFS at 48 months was 33%±5%. Time from 3F8 to progression was longer than the time to first relapse by >3–6.5 fold (n=17 patients), >1.5–1.9 fold (n=10), and >1–1.49 fold (n=10). Among the 33 patients who continued progression-free with long follow-up (>28–111 months), 18 had MYCN-amplified NB, 16 without maintenance chemotherapy and only two underwent autologous stem-cell transplantation (ASCT) during salvage. Patients sensitized to 3F8 were treated with rituximab to reduce HAMA to <1,000 U/mL before 3F8 treatment. By multivariate analysis, age, post-MRD response and maintenance chemotherapy were significant favorable prognostic factors for PFS. Variables significant for overall survival were post-MRD response and rituximab.

β-glucan, a natural polymer of glucose, interacts with CR3 and CR4 on myeloid cells, enhancing adhesion to their natural ligands including iC3b, the breakdown product of complement activation. Through activation of these receptors, anti-tumor effect of monoclonal antibodies can be enhanced in xenograft models.36 A phase I study of oral barley-derived β-glucan (10 to 80 mg/kg/day for 10 days) when combined with iv 3F8 (10 mg/m2/day × 10 days) demonstrated acceptable safety profile without reaching glucan maximum tolerated dose (MTD). Improvement was observed in 13 in 22 patients by MIBG scintigraphy.37 A subsequent phase I study using 10 to 200 mg/kg/day × 17 days of oral yeast-derived β-glucan (NCT00492167) did not reach glucan MTD, while the study did show improvement in MIBG scans in 7/24 and CR of BM in 3/5, with an overall response of 1 complete response (CR), 1 partial response (PR), 22 stable disease (SD), and 17 progressive disease (PD).38 The phase II combination study with 3F8, GM-CSF, β-glucan and isotretinoin (NCT00089258, completed) was discontinued because of a high incidence of HAMA (unpublished data).

A pilot study combining 3F8 with allogeneic NK cells (NCT00877110) used haploidentical NK-cells optimized for anti-neuroblastoma cytotoxicity by selecting NK donors: from licensed NK-cells responding to “missing self (MS)” or from unlicensed NK-cells responding to “missing ligand (ML)” (see Lessons Learned).39 Patients received a lymphodepleting regimen of high-dose cyclophosphamide, topotecan and vincristine (days 1–3) prior to infusion (day 5) of NK-cells isolated from donor leukophereses using CD3-depletion (to <2×104 CD3+ cells/kg) followed by CD56-enrichment. For subsequent cycles (≤3), conditioning was reduced to cyclophosphamide alone. 3F8 (20 mg/m2/day) was administered on days 8–12. 20 patients received 23 cycles: 1, 9, 5 and 5 at dose-level 0 (<1 × 106 CD56(+) cells/kg), I (1–4.99 × 106), II (5–9.99 × 106) and III (10–30 × 106), respectively. 7, 7 and 6 donors had ML, ML+MS and neither, respectively. MTD has not yet been reached. One patient had dose limiting toxicity (DLT): grade 4 hypertension and vomiting. The only other >grade 2 possibly-related toxicity was transient hepatic transaminitis. Neither graft versus host disease (GvHD) nor myeloablation was observed. Responses by INRC (n=19) were: 3 complete remission (n=3), 13 stable disease (7/13 had improved MIBG scores [median-4]), and 3 progressive disease (all donors without ML or MS).

Murine antibodies: IgG2a antibodies 14.G2a and ME36.1(see table 1 for clinical trials)

14.G2a is a class switch variant of 14.18 – IgG3 isotype of anti-GD2 antibody. Its phase I clinical trial in 18 patients demonstrated very modest antitumor activity40 at the expense of significant toxicities. A subsequent COG study tested anti-tumor activity of 14.G2a in combination with IL-2.41 Thirty-three pediatric patients with GD2(+) malignancies were included in the study. Partial response was observed in neuroblastoma (n=1/31) and CR in osteosarcoma (n=1/2). ME36.1 antibody is a mouse IgG3, class switched to IgG1 and IgG2a ME36.1. Unlike the other two anti-GD2 MAb 3F8 and 14.G2a, it cross-reacts with GD3.42 Preclinical studies demonstrated ani-tumor activity,43 but no clinical trials have been attempted.

Chimeric antibody ch14.8 (see table 2 for clinical trials)

Table 2.

Clinical Trials of Chimeric Anti-GD2 Antibodies.

Type of antibody ClinicalTrials.gov identifier/reference Study type Type of therapy (number of patients) Dose, mg/m2/day Toxicity profile Response Important observations
Ch14.18 −/[45] Phase I ch14.18 in patients with malignant melanoma (13) 5–100, 1–2 days pain No response antibody response directed at the variable region of ch14.18
Ch14.18 −/[46] Phase I ch14.18 in patients with neuroblastoma (9) 30–50, daily for 5 days, 1–4 courses pain, fever, allergic reactions, ocular symptoms 2 CR, 2 PR, 1 MR ocular toxicity, no antibody response to ch14.18
Ch14.18 −/[47] Phase I ch14.18 in patients with refractory neuroblastoma (10) and osteosarcoma (1) 10–200, daily for 1–4 days, 1–5 courses Pain, fever, hypertension, hypotension, serum sickness, rash 1 PR, 4 MR 7/11 developed antibody response to ch14.18; all patients with high titers of anti-chl4.18 had therapeutic benefits
Ch14.18 −/[48], [49] retrospective consolidation treatment with ch14.18 in children > 1 year with metastatic neuroblastoma (166) 20, daily for 5 days, 6 cycles fever, abnormal C-reactive protein, cough, rash, pain, ocular symptoms, capillary leak syndrome Improved OS in antibody – treated group no advantage of antibody treatment for EFS and OS in multivariate analysis; in followup study: ch14.18 may prevent late relapses
Ch14.18 −/[50] Phase I ch14.18 with GM-CSF in children with neuroblastoma after hematopoietic stem-cell transplantation (19) 20–50, daily for 4 days, up to 6 cycles pain, fever, nausea, vomiting, urticaria, hypotension, capillary leak syndrome, neurotoxicity No response HACA detected in 5/18 patients (28%); development of HACA response not noted until after the second course
Ch14.18 −/[51] Phase II chimeric anti-GD2 (ch14.18) and GM-CSF for refractory neuroblastoma (32) 50, daily for 5 days pain, fever, nausea, vomiting, diarrhea, urticaria, hypotension, hypertension, thrombocytopenia 1 CR, 3 PR, 1 MR anti-tumor response to ch14.18 and GM-CSF combination in refractory disease
Ch14.18 −/[52] Phase I ch14.18 and rhGM-CSF in metastatic melanoma (16) 15–60, day 1 pain, blood pressure changes, nausea, diarrhea, peripheral nerve dysesthesias, myalgias, and weakness No response toxicity similar to that observed with ch14.18 alone without improvement in tumor response
Ch14.18 −/[53] Phase I Ch14.18 plus IL-2 in melanoma patients (24) 2–10, daily for 5 days, 1–2 courses allergic reactions, weakness, pericardial effusion, decreased performance status 1 CR, 1PR Induced immune activation in all patients
Ch14.18/CHO NCT01704872/[54] Phase I Ch14.18 antibody produced in CHO cells in relapsed or refractory Stage 4 neuroblastoma patients: a SIOPEN Phase 1 study (16) 10–30, daily for 5 days, 1–3 courses similar to side effects in studies with ch14.18/SP2/0 2 PR in mice, comparable pharmacokinetics and biodistribution between ch14.18/CHO and ch14.18/SP2/0; similar in human (no direct comparison)
Ch14.18/CHO −/[55] not specified Ch14.18 antibody produced in CHO cells with sc IL-2 in high risk neuroblastoma patients (53) 100, continous infusion for 10 days not specified overall response following INRG guide lines − 30%. patients with KIR/KIRL mismatch and high affinity FCGR alleles showed a trend towards longer EFS (P= 0.08)
Ch14.18+R24 −/[56] Phase I Phase I trial of combined treatment with ch14.18 and R24 MAbs and IL-2 inpatients with melanoma and sarcoma (27) 2–7.5 (ch14.18) and 1–10 (R24), daily for 5 days, 1–3 courses severe allergic reactions, pain 2 PR MTD of ch14.18 and of R24 were lower than the MTD of each antibody
Ch14.18 NCT00026312/[18] Randomized phase III Ch14.18 with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma (113) 25, daily for 4 days, 5 cycles Pain, capillary leak syndrome, hypersensitivity reactions superior EFS and OS in immunotherapy group as compared to standard therapy ch14.18, GM-CSF, and interleukin-2 associated with a significantly improved outcome as compared with standard therapy in patients with high-risk neuroblastoma
hu14.18 NCT00743496 [57] Phase I humanized 14.18 antibody with a single point mutation (K322A) that reduces complement-dependent lysis (36) 2 to 70 mg/m2 dayly for 4 days every 28 days pain, fever, 4CR, cough, asthenia, sensory neuropathy, anorexia, serum sickness, hypertensive encephalopathy 2 PR recommended phase II dose 60 mg/m2/day for 4 days, adverse effects were manageable and improved with subsequent courses

Abbreviations: CR = complete response; PR = partial response; MR = mixed response; GM-CSF = granulocyte-macrophage colony-stimulating factor; HACA = human anti-chimeric antibody; HAMA = human anti-mouse antibody; iv = intravenous; sc = subcutaneous; MR = minor response, EFS = event-free survival

One limitation of murine MAb is their interference by HAMA. Chimeric antibody retains murine variable heavy (VH) and variable light (VL) chains grafted onto human IgG constant domains. In the case of humanized antibodies, only murine complementarity determining region (CDR) domains are retained, while most of the framework VH and VL sequences are of human origin. Chimeric 14.18 (ch14.18) antibodies demonstrated significantly higher ADCC when compared to its murine variant in experiments in vitro.44 Initial study in 13 patients with malignant melanoma did not show any anti-tumor response.45 Human anti-chimeric antibody (HACA) was detected. Further trials in 9 patients with neuroblastoma showed promising results: CR (n=2), PR (n=2), and MR (n=1).46 In another small study of 10 patients with neuroblastoma and 1 adult with osteosarcoma, one PR and 4 MR were seen. Seven patients developed HACA response.47 Interestingly, all patients who developed high titers of HACA had therapeutic benefits. Ch14.18 was further assessed in larger trials in patients with newly diagnosed stage 4 neuroblastoma. In a retrospective study that included 334 evaluable patients, no clear therapeutic advantage of ch14.18 was elicited.48 No statistically significant difference in event free survival (EFS) was observed in patients who received ch14.18 in comparison to those who were treated with oral low-dose maintenance therapy or did not receive any further treatment after initial therapy. An update of this study after 11 years of follow-up changed the original conclusion, observing a significantly better EFS and OS at 9 years for the immunotherapy group, although in the multivariate analysis, no difference was seen between immunotherapy and oral chemotherapy groups.49

Combination of ch14.18 with IL-2 and GM-CSF was subsequently evaluated in several clinical trials. In a COG phase I study, ch14.18 and GM-CSF were given to patients with neuroblastoma immediately after stem-cell transplantation. Nineteen patients were eligible for analysis. Disease progression occurred in 8 of 14 patients in first remission and 2 of 5 patients in second remission with a median follow-up of 40 months.50 Phase II study with ch14.18 and GM-CSF demonstrated 1 CR, 3 PR and 1 MR in 27 patients with recurrent or refractory neuroblastoma.51 Pharmacokinetics of ch14.18 when combined with GM-CSF is being studied in an ongoing trial (NCT01418495). Treatment with ch14.18 and rhGM-CSF in patients with metastatic melanoma demonstrated toxicity similar to that observed with ch14.18 alone without improvement in tumor response.52 Combination with IL-2 was tested in a phase I trial in adult patients with melanoma. Antitumor activity included 1 CR, 1 PR, 8 SD. Induction of lymphokine-activated killer (LAK) cell activity was observed in this study.53 In a phase III randomized study among patients with high risk neuroblastoma following autologous stem cell transplantation (NCT00026312), the combination of isotretinoin, ch14.18, IL-2 and GM-CSF was compared to standard maintenance therapy with isotretinoin alone. Randomization was stopped early because of the superiority of immunotherapy versus standard therapy with regard to EFS (66±5% vs. 46±5% at 2 years) and OS (86±4% vs. 75±5% at 2 years).18 Ch14.18 antibody produced in Chinese hamster ovary (CHO) cells (rather than in mouse SP2/0 cells - see below) was tested in a phase I study in relapsed or refractory stage 4 neuroblastoma patients. Side effects and pharmacokinetics similar to those with ch14.18/SP2/0 were observed.54 Another ongoing phase III randomized study of the European SIOP Neuroblastoma Group (SIOPEN) in high-risk neuroblastoma is testing the efficacy of isotretinoin and ch14.18/CHO, with or without IL-2 after autologous stem cell transplant (NCT01704716). Clinical response following long-term infusion of ch14.18/CHO in combination with sc IL-2 was assessed in high-risk neuroblastoma. The overall response following INRG guide lines was determined at 30% (12/40). The EFS was 32.4 % (observation 0.1 – 3.2 years, mean: 1.6 years) and an OS of 66.8% (observation 0.3 – 3.9 years, mean: 3.1 years).55 Continuous infusion of ch14.18/CHO antibodies with sc IL-2 is currently under investigation in phase I/II study (NCT01701479). A combination of ch14.18 with lenalidomide (analogue of thalidomide) and isotretinoin is also being tested in a phase I design (NCT01711554). In a phase I trial of concurrent therapy with ch14.18 and R24 (anti-GD3) antibodies and IL-2 in patients with melanoma and sarcoma, MTD were lower than the MTD of each antibody in prior studies.56

Humanized antibodies hu14.18 and hu3F8

Humanized 14.18 (hu14.18) was tested in a phase I trial in children and adolescents with neuroblastoma, osteosarcoma and melanoma (NCT00743496). Dose-limiting grade 3 or 4 toxicities were observed in four of 36 evaluable patients with refractory and recurrent neuroblastoma and were characterized by cough, asthenia, sensory neuropathy, anorexia, serum sickness, and hypertensive encephalopathy.57 Combination of hu14.18 with IL-2, GM-CSF and NK cells is the focus of a phase I study in children with relapsed or refractory neuroblastoma (NCT01576692). Efficacy of chemotherapy combined with hu14.18 plus GM-CSF is also being tested (NCT01857934).

3F8 was humanized (hu3F8) based on its crystal structure and manufactured in CHO cells.8 In a phase I dose escalation study (NCT01419834)20 hu3F8 (0.06 to 3 mg/kg/cycle) was given outpatient intravenously over 30 minutes, split as 2 and later amended to 3 doses per cycle; cycles were repeated every 3–6 weeks up to 24 months. Among 32 patients (neuroblastoma – 30, osteosarcoma – 2) two experienced a reversible but DLT of elevated liver transaminases; MTD has not been reached. At equivalent doses hu3F8 was less painful than murine 3F8. Human anti-human antibody (HAHA) was detectable in only 5 patients, despite their prior sensitization to murine 3F8 in 26 patients, and despite multiple cycles (up to 16) of hu3F8. Best responses on hu3F8 treatment were: CR (n=5) and PR (n=1), 21 (66%) maintained CR or SD (median 7 months, range 1–23+), and 5 (16%) developed PD.

In a second phase I dose escalation of hu3F8 in combination with GM-CSF (NCT01757626)21 patients with relapsed high risk neuroblastoma received cycles of hu3F8/GM-CSF in the absence of HAHA up to 24 months. MAb (0.9–3.6 mg/kg/cycle) was infused iv outpatient over 30 min three times a week (i.e., 3 days/cycle). Daily GM-CSF was administered subcutaneously, beginning 5 days pre-hu3F8, through the last dose of hu3F8. Of the 18 patients enrolled to date, 9 were in the 2nd remission; nine had osteomedullary metastases, and 16 had prior anti-GD2 immunotherapy with murine 3F8 (n=13), ch14.18 MAb (n=3), or both (n=1). PK studies showed dose-dependent increases in peak serum concentration, but not in terminal half-life (T1/2) (4.5±1.4 days). MTD has not been reached. HAHA developed in three patients (two previously treated with ch14.18).

In a third phase I dose escalation study of hu3F8 in combination with IL-2 (NCT01662804),58 hu3F8 (starting dose at 0.3mg/kg/cycle, split as day 1 and day 8 infusions) was combined with a fixed dosage of scIL-2 (6×106 units/m2/day x5 days/cycle starting on day 9), total of 4 cycles every 21 days, in patients with refractory or recurrent GD2(+) solid tumors. There was no DLT up to 0.9 mg/kg/cycle (n=8 patients). Side effects were grade 1/2 pain, allergic reactions, and fever. Pharmacokinetic studies showed dose-dependent increases in Cmax and Cmin. In 6/6 patients treated on the first two dosage levels (0.3 mg/kg and 0.6 mg/kg), significant discrepancies in hu3F8 T1/2 were seen between doses 1 and 2 of a cycle: T1/2 of dose 2 of hu3F8 (given with scIL-2) decreased by ~60% compared to T1/2 of dose 1 (hu3F8 given without scIL-2). However, for 2/2 patients treated at dosage level 3 (0.9mg/kg), T1/2 of dose 2 of hu3F8 was decreased by only ~5% compared to dose 1. Four out of 8 patients developed HAHA despite no prior anti-GD2 therapy. There have been no objective responses to date but 1 of 3 neuroblastoma and 2 of 4 osteosarcoma patients completed 3+ cycles without disease progression.

Non-compartmental pharmacokinetic analysis59 of serum concentrations over time during cycle 1 of the phase I trial of hu3F8 (NCT01419834) revealed a dose-independent T1/2 of 3.26±1.09 days. Both Cmax (R=0.95) and AUC (R=0.91) were strongly correlated with dose level, while Cmax was correlated with AUC (R=0.97). Pharmacokinetic parameters remained statistically unchanged during subsequent treatment cycles. When the PK of hu3F8 alone in the phase I trial (NCT01419834) was compared to that combined with IL-2 (NCT01662804),58 at comparable dosage level, there was a strong positive linear correlation between patient’s weight with AUC (R=0.95±0.11), Cmax (R=0.85±0.14), Cmin (R=0.88±0.14), and negative correlation with Cl (R=0.93±0.42). No correlation was observed between T1/2 and patient’s weight.

GD2 vaccines

Even though MAb therapy is often regarded as passive immunotherapy, the induction of a host anti-tumor or anti-idiotype network following MAb therapy has raised the hypothesis that vaccination against GD2 may achieve more long term tumor control.60 HAMA, an indirect measure of the host anti-idiotype network, was consistently correlated with long term survival.34,60 Because of these observations, anti-idiotypic antibody vaccines such as mouse IgG1 antibody 1A7 specific for ch14.1861 and rat IgG1 antibody A1G4 specific for 3F862 were tested in phase I trials (NCT00003023). The GD2 peptide mimotope63 and its DNA vaccine64 can induce serum antibodies and protective anti-GD2 IgG responses in mice. However, in contrast to whole-antigen vaccines, single-epitope vaccines will probably be limited by their narrow target coverage. The whole GD2 antigen has also been conjugated to keyhole limpet hemocyanin (KLH) to overcome the poor immunogenicity of carbohydrates.65 In a phase I trial, the MTD of the immunological adjuvant OPT-821 (50, 75, 100 and 150 μg/m2 per injection) was tested in combination with 30 μg each of GD2-KLH and of GD3-KLH, all administered subcutaneously (weeks 1–2–3–8–20–32–52) to 15 patients in ≥2nd remission (NCT00911560).66 Oral yeast β-glucan (40 mg/kg/day, 14 days on/14 days off) started week 6 and continued through 12 months. At 150 μg/m2 of OPT-821 (the dosing used in adults), no DLT was observed. 13/15 patients received the entire protocol treatment including 12 who remain relapse-free at 24+ to 39+ (median 32+) months and one who relapsed (single node) at 21 months. Relapse-free survival was 80±10% at 24 months. 12/15 patients had antibody responses against GD2 and/or GD3. Disappearance of MRD was documented in 6/10 patients assessable for response. The study was expanded to accrue more patients.

Immunocytokines

Fusion of cytokines to GD2-specific antibody has the potential to improve tumor delivery and help achieve a higher local concentration and reduce systemic toxicity. In phase I study of hu14.18/IL-2 in 28 children with refractory neuroblastoma and melanoma, no objective responses were observed. Hypersensitivity, hypotension and myelosuppression were dose-limiting toxicities.67 Phase II study included 39 children with refractory neuroblastoma. Of 13 patients with measurable disease, 3 had SD. Among 23 patients with evaluable disease (based on BM histology and/or MIBG scintigraphy), 5 achieved marrow CR. Anti-hu14.18 antibodies developed in 40% of patients.68 Toxicity profile was similar to that observed with IL-2 alone; they included capillary leak, hypoxia, pain, rash, allergic reaction, elevated transaminases, and hyperbilirubinemia. A Phase II study of hu14.18/IL-2 in combination with GM-CSF and isotretinoin in neuroblastoma (NCT01334515) and a phase II study among patients with minimal residual melanoma were recently completed (NCT00590824). Anti-GD2/GM-CSF immunocytokine showed enhanced ADCC in vitro when compared to antibody alone or in combination with systemic GM-CSF.69 Despite these compelling data in syngeneic mouse models,70 where anti-GD2 antibody targeted cytokines to tumor sites as immune stimulants when applied systemically68 or locally,71 iv hu14.18-IL-2 produced BM remission in patients only if neuroblastoma tumor burden was minimal,68 and no benefit was seen for soft-tissue tumors.68,72 The clinical advantage of hu14.18-IL-2 over ch14.18 plus IL-2 is uncertain, given the grade 3 and 4 capillary leak with abnormal liver functions seen in both settings.18,68 IL-15 is an alternative cytokine that activates NK and natural killer T-cells (NKT),73 but without the severe side effects of IL-2.

Lessons learned from clinical trials of anti-GD2 antibodies

Importance of NK cells and the role of inhibitory killer cell immunoglobulin-like (KIR) receptor

Anti-GD2 MAb is more than a passive linker of tumors to NK cells; they rescue NK cells from suppression by neuroblastoma. FcγRIII (CD16)-mediated signaling removes the inhibition by KIRs.74 Since 60% patients have KIRs on their NK cells with no corresponding KIR ligands during cellular maturation,75 these NK cells are “uneducated” and hyporeactive until their CD16 is activated during ADCC. On the other hand, while “educated” NK cells are trained to kill, they are inhibited by tumor HLA upregulated during ADCC. This inhibition by HLA explains why patients with “missing inhibitory KIR ligands” tend to have longer survival following anti-GD2 MAb therapy.23,34,75

Importance of Myeloid ADCC

Granulocyte-ADCC of neuroblastoma is unique among human cancers.34,69 This killing does not depend on oxidative intermediates, but requires azurophil (primary) granule exocytosis, and is enhanced by GM-CSF.29,69 In ADCC, FcγRIIA (CD32) is the responsible receptor on granulocytes and myeloid effectors76 and its high affinity allele (131-R, see Table 3) for MAb 3F8 is associated with a more favorable patient outcome.32,75 Since complement receptors CR3 and CR4 on granulocytes are also important adhesion molecules for ADCC,69,76 they can be manipulated to enhance tumor killing. One of their natural activating ligands is the membrane-bound C3bi, a breakdown product of C3 activation by MAb.77 When CR3 is activated, it acquires the conformational neoepitope CBRM1/5,78 which correlates with better patient survival.22,34 β-glucan binds and activates CR3, and when administered orally, enhances antibody therapy of NB.79 Tumor associated macrophages (TAM) regulate neuroblastoma growth, angiogenesis, and metastasis and is associated with dedifferentiation in MYCN amplified tumors.80 Depending on the microenvironment, TAMs can become polarized into type 1 antitumor or type 2 pro-tumor phenotypes.81 In the presence of anti-GD2 MAb, ADCC can transform pro-tumor M-CSF-activated macrophages into efficient antitumor effectors.82

Table 3.

Binding kinetics of mouse 3F8 and hu3F8 antibodies to human FcγRII (CD32)

Antibody kon (1/Ms) SD koff (1/s) SD KA=kon/koff koff (relative to CD32A-131R) KA (relative to CD32A-131R)
mouse 3F8 (IgG3) CD32A-131R 2.59E+04 9.31E+03 3.63E-01 2.36E-02 7.15E+04 1 1
CD32A-131H 1.90E+04 3.58E+03 1.10E+00 8.27E-02 1.73E+04 3.02 0.24
CD32B 1.15E+04 1.60E+03 2.03E+00 3.78E-01 5.66E+03 5.6 0.08

hu3F8 (IgG1) CD32A-131R 1.49E+05 4.38E+04 4.06E-01 4.94E-02 3.68E+05 1.12 5.15
CD32A-131H 2.70E+05 9.48E+03 4.27E-01 3.59E-02 6.31E+05 1.18 8.83
CD32B 2.86E+04 1.63E+04 3.41E-01 2.63E-01 8.37E+04 0.94 1.17
*

Kinetics of FcγRII (CD32) interaction with mouse 3F8 and hu3F8 were measured using a Biacore T100 biosensor and CM5 sensor chips (Biacore AB of GE Healthcare, Uppsala, Sweden). Toward the CD32A-131R allele, 3F8 had 2.67 fold slower off rate (koff), and nearly 3-fold higher binding affinity (Ka) than toward CD32A-131H. For hu3F8, the high affinity allele was 131-H. Affinity for CD32B was much lower for mouse 3F8 than for hu3F8.

Role of lymphokines

Patients with high risk neuroblastoma have severely compromised lymphoid systems (NK, T and B cells) because of dose-intensive induction therapy, at a time when antibody immunotherapy is applied. By activating ADCC to kill NB, anti-GD2 MAb is most efficient when effector cell populations and functions are amplified by cytokines. IL-2, which activates NK cells, natural killer T (NKT) cells, T-cells, and the undesirable regulatory T-cells (Treg), have a modest anti-neuroblastoma effect as a single agent.83 In contrast, the consistent ability of GM-CSF and G-CSF to repopulate and activate the myeloid system, and their relative lack of toxicity, make them ideal cytokines to combine with anti-GD2 antibodies. Unlike GM-CSF, IL-2 is associated with substantial toxicity (e.g., 23% of patients experienced capillary leak18). Similar to IL-2, IL-15 activates NK, NKT, and CD8+ T-cells.73 However, it does not cause capillary leak, activation-induced cell death, or increased Treg activation in non-human primate studies.84 Another type of anti-neuroblastoma lymphocytes called NKT cells can infiltrate neuroblastoma, kill TAMs, and they are associated with superior patient survival.85 However, for NKT cells to survive the hypoxic neuroblastoma environment, IL-15 is essential.86 When complexed to its IL-15Rα receptor, the biological effects of IL15 are enhanced.87 IL15 fused to anti-GD2 antibody c.60C3 displayed strong antitumor activities in syngeneic cancer models.88 Preclinical evaluation of hu3F8 in combination with IL-15 has demonstrated similarly encouraging anti-tumor efficacy (unpublished data).

Compartmental efficacy of anti-GD2 antibodies

When the relapse pattern of patients following anti-GD2 immunotherapy was analyzed, it is striking to note that while diffuse relapse was no longer typical (only 34% of all relapses); isolated relapses in the BM or bone (27%), the brain parenchyma (23%), and soft tissues including nodes (16%) were increasingly frequent.34 This suggests that intravenous anti-GD2 antibody may be more efficacious in the blood and marrow compartment than the CSF and lymphatic sites. Nonetheless, the marrow compartment remains an important reservoir for disease relapse since marrow MRD early after consolidation with immunotherapy was the strongest predictor of PFS and OS.89

Biochemical differences among the anti-GD2 antibodies

There are substantial biochemical differences among the anti-GD2 antibodies; however, whether they translate into differences in clinical efficacy remains to be proven. For example, 3F8 and hu3F8 have a much slower koff (>10 fold) than 14G2a or ch14.18,19 which could translate into longer retention time on the target. Although murine and chimeric antibodies (ch14.18) are usually immunogenic over time, neutralizing antibodies against humanized antibodies (hu3F8, hu14.18) can still occur in some patients. Proteins produced in SP2/0 cell lines versus CHO cell lines can be biochemically different. For example, 3F8 and ch14.18 were made in SP2/0 cell lines which add α-linked galactose or N-acetylneuraminic acid (NeuAc) epitopes not present in proteins made by human or by CHO cells (ch14.18-CHO, hu3F8).90 High levels of natural anti-Gal91 and anti-NeuGc antibodies92 circulate in human blood, potentially capable to interfere with the pharmacokinetics of MAb, while causing immune complex disease or even anaphylactic reactions.93,94

Pain side effect, antibody dose, HAMA or HACA and MRD

GD2 is known to be expressed on neurons and peripheral nerves. CNS toxicity is rare partly because of the blood brain barrier to intravenous MAb. Moreover, anti-GD2 antibodies injected directly into the CSF space among patients with neuroblastoma and medulloblastoma have not produced any major long term CNS toxicity.95 Nonetheless, the acute pain side effect of anti-GD2 antibody necessitates heavy doses of analgesics and sedatives with their complications requiring inpatient administration (e.g. ch14.18). Because of pain, the dose of anti-GD2 is limited to 20–40 mg/m2/day and usually for a maximum of 5 consecutive days. Without the option of dose escalation, it is not surprising the clinical benefit of anti-GD2 antibody is primarily seen in patients with MRD. Soft tissue bulky tumors generally do not respond. Various attempts to lessen the pain side effects have been tried. A phase I human trial was completed to test the effect of mutation (K322A) in the Fc region on reducing complement activation and the presumed pain side effect, but pain was still observed.57 Premedication with a small dose of Fc-modified anti-GD2 antibody devoid of ADCC and CMC functions reduced significantly the doses of analgesics required, but pain side effects were not eliminated.27 MAbs that target 9-O-acetyl-GD2 but not GD2 could potentially reduce pain toxicities associated with binding to GD2 antigen on peripheral nerves.96 Although staff training and careful coordination in pain management has enabled safe outpatient administration (e.g. 3F8 and hu3F8), the identification of the novel receptor for this pain mechanism should provide a more rational and effective approach to reduce pain side effect and possibly to further dose escalate to improve anti-tumor effect.

Patients with HAMA have few to no pain side effects from MAb, as well as accelerated antibody clearance, and probably reduced or no therapeutic benefit. Hence, 3F8 treatments are postponed until HAMA falls below 1,000 U/mL. Unlike 3F8, HAMA and HACA are not part of treatment cycle prerequisite for 14G2a or ch14.18. How that translates into long term efficacy will require careful comparison. Interestingly, the ability to mount HAMA response (not the presence of HAMA during 3F8 administration) consistently correlated with long term overall survival for high risk neuroblastoma patients with stage 4 disease diagnosed after 18 months of age or with MYCN amplified tumors, irrespective if they were treated with primary refractory disease97 or in their first remission.34 We interpret this association as the result of active host anti-idiotypic network60 or an induced anti-tumor response following opsonization of tumors,82 responsible for the long term survival.

Antibody Drug Conjugates

Antibody immunoconjugates can selectively deliver the cytotoxic payload to the tumor. Such conjugates could be a drug (antibody drug conjugates, ADC) with known antitumor activity, radionuclide or toxin. There are several factors that can negatively affect clinical use of immunoconjugates. The conjugation chemistry can damage immunoreactivity, interfering with antibody binding to the target. Instability of the conjugate in serum can nullify its effect meant for the target, sometimes resulting in unintended systemic toxicities. Release of the active moiety at the tumor target can also be impaired by the microenvironment. The development of anti-conjugate antibodies represents another potential obstacle for immunoconjugates. Despite the success of antibody-toxin conjugates (Gemtuzumab ozogamicin) in AML and antibody-microtubule-poisons (Brentuximab Vedotin in Hodgkin’s lymphoma or Trastuzumab Emtansine in breast cancer), ADC targeting GD2 is in the exploratory phase in preclinical testing.98

Radionuclide immunoconjugates for radioimmunotherapy (RIT)

Radionuclides can exert antitumor effect not just on antigen(+) but also on surrounding antigen(−) tumor cells via a “cross-fire” effect and radiation-induced intercellular signaling.99

Intravenous anti-GD2 131I-3F8 has been tested in children with metastatic neuroblastoma at high radioisotope doses (6–28 mCi/kg).100 Although responses were seen in both soft tissue masses and bone marrows, the use of myeloablative 131I-3F8 (20 mCi/kg) did not add survival benefit compared to naked 3F8 among patients with high risk metastatic stage 4 neuroblastoma in first remission.34 In a subsequent phase I trial of 131I-3F8 in combination with bevacizumab (NCT00450827) in patients with relapsed or refractory NB, responses were similar to those with 131I-3F8 alone.

Intraventricular 131I-3F8 – compartmental RIT (cRIT)

While intravenous radiolabeled 3F8 did not improve survival, results with cRIT are highly promising. Brain metastasis is a common pathway for metastatic solid tumors; its biology is poorly understood, management inadequate, and cure is rare. Except for occasional tumor types, most patients with brain metastases die despite intensive therapy. Tumor cells from blood or from brain metastases can invade the CSF and disseminate throughout the neuroaxis by the constant flow of CSF from the ventricles to the spinal canal and over the cortical convexities, a condition called leptomeningeal (LM) carcinomatosis with significant morbidity and mortality. In contrast to leukemia, where LM metastasis is well controlled by intrathecal chemotherapy, the prognosis in solid tumors is extremely guarded despite the use of chemotherapy and radiation therapy. Neuroblastoma metastasis to the CNS was once considered rare. This entity is distinguishable from dural based metastases where direct extension from bony disease is the rule. As the natural history of neuroblastoma changed with systemic immunotherapy,22 isolated CNS afflicts up to 20% of the neuroblastoma patients at Memorial Sloan-Kettering Cancer Center (MSKCC), where their systemic disease was thought to have been eradicated.34 Conventional treatment modalities, including surgical resection, chemotherapy and radiation, have not improved survival.101

The CSF (thecal sac) space has unique characteristics suitable for cRIT: (1) the blood brain barrier prevents MAb recirculation, (2) compared to blood, CSF has few white cells, no Fc receptors (FcR), and ~1000-fold less IgG.102 MAb injected into the CSF compartment is better shielded from host immunity with less sequestration by FcR or degradation by enzymes, (3) the 200-fold lower protein content of CSF (versus serum) facilitates MAb binding, (4) since CSF volume is small (140 ml), MAb achieves a very high compartmental concentration, (5) CSF compartment is renewed every 7–8 hours, providing a built-in washing step, (6) CSF flow can be reduced pharmacologically, permitting longer MAb reaction time, (7) the apparent absence of anatomic barrier facilitates the movement of MAb between CSF and the extracellular space of brain, especially if there is damage to the meninges either by tumor itself or after surgical resection of tumor.

Intraventricular 131I-3F8103 when tested in cRIT for leptomeningeal cancers in both children and adults showed highly favorable therapeutic ratios. Toxicities included self-limited headache, fever, and vomiting. DLT was reached at the 20-mCi dose, when transient elevations in intracranial pressure and chemical meningitis were seen. Three of 13 assessable patients in the phase I study achieved objective radiographic and/or cytologic responses. No late toxicities have been seen in two patients who remain in remission off therapy for nearly 10 years. Among children with recurrent neuroblastoma metastatic to the CNS, when cRIT was incorporated into multi-modality therapies, long term remissions have been achieved (NCT00445965).104

131I-3F8 cRIT for relapsed medulloblastoma (MB, NCT00445965)95

Recurrent and high risk MB is challenging to cure. 36 patients with high risk (n=5) or recurrent (n=31) MB were treated on MSKCC IRB-approved protocol. Patients had <72 Gy (brain parenchyma) and 45 Gy (spinal cord) prior radiation. Patients received 2 mCi 131I-3F8 followed by 2–4 weekly injections (10 mCi 131I-3F8/injection), maximum CSF dose 2400 cGy, determined by CSF samplings and region of interest analyses on whole-body scintigraphy. 30 patients received >2 therapeutic injections. 32/36 (89%) remained alive ≥6 months after cRIT. Long term survivors in CR included 10 of 19 (53%) patients treated with 131I-3F8 as consolidative therapy, at a mean follow up of 49.6 months (6 months to 6 years); 4 of 17 (24%) with radiographic evidence of relapsed or refractory MB remain alive, 2 in CR 3.5 yrs and 5.5 yrs since relapse. Four of 5 patients with high risk MB remain in CR (mean follow up 54.4 months); 1 patient died of secondary glioma 5.3 years after initial diagnosis. Mean total absorbed CSF dose was 1250 cGy (237 – 2239) by sampling; average CSF dose was 64.1 cGy/mCi and blood was 2.9 cGy/mCi. Toxicities included self-limited headache, fever, and vomiting. No long term side effects directly attributable to 131I-3F8 have been observed.

Lessons learned from clinical application of radioiodinated 3F8

Choice of radioisotope and immunoreactivity

Most clinical applications of RIT utilize β-emitting radioimmunoconjugates. β-particles have a relatively long range (0.8–5 mm) and low linear energy transfer (approximately 0.2 keV/μm). This long range results in the delivery of radiation not only to the antigen-positive, but also to antigen-negative tumor cells, as well as to the surrounding normal tissues. Thus, β-emitters can treat bulky diseases effectively, but are not optimal for eradicating single cells or micrometastasis. Most early human studies of RIT used iodine 131 (131I), a long-lived β-particle emitter. Because of its γ-particle emission, it is also suitable for dosimetry studies. However, γ-particle emission poses a radio-hazard at high treatment doses, necessitating patient isolation. In vivo dehalogenation can compromise tumor dose with subsequent thyroid damage from the released iodide. Yttrium 90 (90Y) is a pure β-emitter; its lack of γ-radiation allows outpatient treatment. However, 90Y has its limitations, including deposition in bone when dissociated from the MAb complex and its lack of γ-emissions, thereby requiring the use of indium 111 (111In) to estimate biodistribution and dosimetry. Lutetium-177 (177Lu), also a pure β-emitter, has a longer half-life of 6.6 days, more compatible with biologic half-life of MAb than 90Y (2.6 days). RIT with 177Lu may result in lower off-target radiation doses to surrounding normal tissues, compared with 90Y and 131I especially when multistep targeting (MST) strategies are used (see below).105

α-particles are helium nuclei; when compared with β-particles, they have a shorter range (50–80 μm) and a higher linear energy transfer (approximately 100 keV/μm). As few as one or two α-particles can destroy a target cell. RIT using α-emitters should result in less nonspecific toxicity to normal bystanders as well as more efficient single cell killing. This is ideal for controlling MRD. α-particle-emitting isotopes such as astatine-211 (211At, T½=7.2h, 1 α particle), bismuth-213 (213Bi, T½=46 min, 1 α particle), and actinium-225 (225Ac, T½=10d, 4 α particles) have proven safe in clinical trials.106 The relative lack of extramedullary toxicities should encourage further development of this targeting technique for micrometastases or neoplasms on the surface of body compartments, such as ovarian cancer and leptomeningeal metastasis.107,108 Radium-223 (223Ra, T½=11.4 d, 4 α particles) is now FDA approved for prostate cancer, and lead-212 (212Pb, T½=10.6h, 1 α particle) is another nanogenerator (giving rise to 212Bi, T½=25 min, 1 α particle), both are potential candidates for MAb targeted RIT. Modeling studies of 131I-3F8 cRIT clearly suggests that improving immunoreactivity from the current 50%19 to 90%109 could potentially bring about a major gain in the therapeutic index. MST offers an opportunity to maintain unimpaired immunoreactivity in the first step. Site directed conjugation using glycolinks and click chemistry present exciting alternative approaches. With the optimal choice of isotopes, skillful conjugation methods, RIT and cRIT could become important in the management of hard-to-cure neuroblastoma and other GD2(+) cancers.

Dosimetry using Theranostic agents

Quantitative methods for estimating radiation absorbed dose are now feasible and they are essential for individualizing patient treatment and avoiding excessive radiation toxicities. As theranostics, a radiolabeled MAb in tracer amounts serves as a diagnostics/dosimetry agent, and if calculations are favorable, can be given as a scale up for therapeutic benefit. Quantitative high resolution PET/CT imaging of antibodies (e.g. positron emitting 124I-3F8) provides accurate dosimetry and staging information. Since the dosimetry of positrons (β+) from 124I are similar to electrons (β−) from 131I, 124I labeled MAb can now be used as theranostic agents, e.g. in brain stem glioma (NCT01502917). Using these theranostic setups, patient and tumor selection will provide a rational basis for achieving durable therapeutic effects.

cRIT optimization110

Pharmacokinetic models have been used to evaluate the role of kinetics and transport parameters of cRIT in maximizing the therapeutic ratio, the ratio of area under the curve (AUC) for the concentration of the bound antibodies over time when compared to that for unbound antibodies. Using a single compartment model, it was predicted that increasing the affinity of antibodies to antigens greatly increased AUC for the same amount of isotope administered; smaller antibody dose and higher specific activity also improved therapeutic ratio. When the isotope half-life was 0.77 h, increasing the antibody association constant enhanced AUC much more than did decreasing the dissociation constant, even if overall affinity was unchanged. When isotope half-life reached 240 h, decreasing the dissociation constant would slightly enhance AUC. Other predictions were that decreasing the CSF bulk flow rate would increase AUC, with 3 mL/h being optimal; continuous infusion or split dose antibody can improve AUC by 1.8 fold; for antibody affinity of 10 nM, choosing an isotope with half-life of 64 h (instead of 0.77 h) could greatly enhance the therapeutic ratio.

In a subsequent two compartment model,109 fitting to patient data was improved when compared to the one-compartment model (R=0.92±0.11 versus 0.77±0.21, p=0.005). In addition, the following new predictions were made: (1) Increasing immunoreactivity of 131I-3F8 from 10% to 90% increased both AUC and therapeutic ratio by 7.4 fold, (2) When extrapolated to the clinical setting, the model predicted that if 131I-3F8 could be split into 4 doses of 1.4 mg each and given at ≥24 hours apart, an antibody affinity of 4nM at 50% immunoreactivity was adequate to deliver ≥100 Gy to tumor cells while keeping normal CSF exposure to <10 Gy.

Multistep targeting or pretargeting111,112

Suboptimal tumor radioactivity AUC versus blood or kidney AUC ratios (~3:1) has been the major limiting factor for RIT. cRIT took advantage of the fast clearance of unbound 131I-3F8 (half life of 3–12 h) out of the CSF, in contrast to 2–4 days for IgG in the blood. A promising solution to improve this therapeutic ratio uses a multistep procedure that pretargets the antibody before the binding of the cytotoxic ligand to the tumor. Generally, a tumor-specific antibody is conjugated to a ligand-binder, such as streptavidin (with high affinity for biotin)113 or ligand-specific antibody (binding to metal chelators, such as diethylene triamine pentaacetic acid [DTPA] or 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid [DOTA]).105,112 In the first step, these bispecific antibodies (172–200 kD) are allowed to localize to tumors in vivo, and any excess is cleared from the blood. The small radiolabeled ligand (DOTA-radiometal or its biotinylated form) is then injected intravenously. The ligand penetrates tissues rapidly and by virtue of the high affinity interaction, binds tightly to the antibody-conjugate at the tumor site. Unbound ligand is quickly excreted through the kidneys. Because of the short transit time of the toxic ligand (radionuclides or toxins), a substantial improvement in the therapeutic ratio is achievable without sacrificing the percent injected dose per gram in tumor.105 However, MST using streptavidin has been limited by immunogenicity, the absence of a clinical clearing agent, difficulty in manufacture and purification, interfering substances in human blood, and the nonversatility of the “hook” in terms of affinity variants. The modular (IgG-scFv) antibody form,105 with the IgG portion specific for tumor and the high affinity scFv (C825) specific for DOTA-metal has provided novel solutions. These bispecific bivalent constructs have high avidity for both the tumor and DOTA. The large molecular weight (~200 kD) ensures a long plasma half-life for optimal tumor targeting. More importantly, since the scFv affinity for DOTA depends on the chelated metal, ranging from 8 pM to 50 nM affinity, dextrans carrying DOTA-metal of low affinity for scFv can be exploited as clearing agents. Besides targeting 90Y (11 pM affinity for scFv) or 177Lu (11 pM affinity for scFv) in RIT, DOTA-metal provides a convenient hook to target nanoparticles. The proof of concept in animal studies was recently demonstrated using the hu3F8-C825 bispecific construct achieving a >20 AUC ratio for kidney and >100 ratio for blood, curing neuroblastoma without clinical or histologic toxicities.112

Emerging therapeutics targeting GD2

Nanoparticles98

Inorganic nanoparticles are delivery systems built to precise specifications for medical applications. They represent small particles (3–200 nm) made up of inert scaffolds. As therapeutics, they are built for their superior pharmacokinetics, thereby enhancing intracellular concentration of drugs in cancer cells while minimizing their systemic side effects. In neuroblastoma-bearing mice, fenretinide, a synthetic retinoic acid derivative,114 or doxorubin115 can be encapsulated in immunoliposomes targeted by 14G2a/ch14.18 anti-GD2 antibodies to completely inhibit the development of macroscopic and microscopic metastases. Antisense oligonucleotides can also be encapsulated in such liposomes to downregulate tumor associated gene expression, increase cytokines production and stimulate innate immune system (macrophages and NK cells).116 Ch14.18 conjugated to the surface of porous silica nanoparticles containing microRNA inhibited neuroblastoma xenograft growth,117 where miR-34a activates a caspase-mediated apoptotic pathway.118 It also targets multiple genetic pathways associated with oncogenesis including MYCN, BCL2, NOTCH1, JAG1, CCND1, CDK6, E2F3 and others.119,120 ALK-siRNA embedded in such liposomes can also inhibit neuroblastoma growth in animal models.121 Gold nanorods targeted by anti-GD2 antibodies induced thermolysis of neuroblastoma by releasing heat in the nanoenvironment when exposed to near infrared laser light – a process called thermoablation.122 Similar effects have been observed in anti-GD2-targeted carbon nanotubes.123

Chimeric Antigen Receptors (CARs)

T-cell mediated immunity can be extremely effective for cancer therapy if these highly potent effector cells can be harnessed, as shown in multiple preclinical studies and clinical trials. This potential was recently realized when polyclonal T-cell tumoricidal activity was redirected to surface TAA. Human cancers evade the classic T cell surveillance through a number of well established mechanisms, the most important one being the downregulation of MHC and co-stimulatory molecules. Tumor-specific CARs enable the recognition of TAA by T-lymphocytes in an HLA-independent manner. Introduction of co-stimulatory molecules (CD28, 4-1BB, OX40) provides essential “second signal” for further activation of T-cells and therefore obviates the need for “external” co-stimulation.124,125 To date, most clinical studies have been conducted in patients with leukemia, neuroblastoma, mesothelioma and glioblastoma multiforme. Attempts have been made to improve the kinetics of CAR-grafted cells, including their persistence and proliferation in vivo. To add a second stimulatory signal (through endogenous EBV virus), EBV-specific T-cells were used to make GD2-CAR modified T cells.126 In this seminal study, 4 of the 8 patients with evaluable tumors had evidence of tumor necrosis or regressions, including a complete remission. Importantly, no major side effects of immunotherapy were noted in this study. None of the patients had detectable antibody response to CAR-grafted T-cells. Impressive results were reported by the same group in 19 patients with high-risk neuroblastoma.127 Three of 11 patients with active disease at the time of treatment achieved complete remission. Interestingly, superior clinical outcome was associated with persistence of CAR-modified T-cells beyond 6 weeks after infusion. Encouraging results with GD2-specific CARs has prompted further clinical investigations using third generation CARs (NCT01822652), where the anti-GD2 scFv is fused with 4-1BB or CD28 in tandem with CD3ζ. The transgene used in this study also contains icaspase9 suicide switch that can be activated in case the transduced T cells need to be sacrificed. Another phase I study using VZV-specific CARs is also planned (NCT01953900). Efficacy of immunotherapy with GD2-redirected CARs in other than neuroblastoma GD2-positive tumors is being tested in preclinical studies.128 Besides T cells, NK modified to express GD2-CAR could overcome NK resistance.129 Feasibility and safety of allogeneic haploidentical NK cells administered together with hu3F8 in patients is planned.

Bispecific antibodies (BsAb) to engage T cells

The potential of exploiting cytotoxic potential of T-cells in conjunction with GD2 specific antibodies has led to the development of bispecific T-cell engagers (BiTE). Standard MAb are IgGs that kill by Fc-dependent mechanisms such as CMC and ADCC. T-lymphocytes have no Fc receptors, and are therefore under-utilized during standard MAb therapy. Moreover, with few exceptions, T cells are inefficient or incapable of targeting carbohydrate epitopes. BsAb recruit T cells for tumor cytotoxicity through HLA-non-restricted CD3-mediated activation. By engaging polyclonal T cells BsAb can overcome the low clonal frequency of classic T cell mediated anti-tumor immunity. CD3 engagement can also induce T cell proliferation and generation of effector cytokines to potentiate the anti-tumor effect. BiTEs, which are based on single-chain antibodies, can exhibit multiple rounds of target cell lysis by T cells.130 Clinical efficacy of low dose blinatumomab, a BiTE with dual specificity for CD19 and CD3, was highly encouraging among patients with relapsed non–Hodgkin’s B cell lymphoma.131 Anti-GD2 BsAb can kill a wide spectrum of GD2(+) tumors with femtomolar EC50 in the presence of expanded human T-cells, a potency orders of magnitude stronger than the nanomolar or picomolar EC50 for IgG mediated ADCC or CMC.132,133 GD2/CD3 BsAbs have demonstrated high efficiency in tumor ablation in xenograft models.132134 Both tandem scFv format (BiTE)132 as well as IgG-scFv modular formats133 have shown anti-tumor efficacy. Murine anti-GD2 antibody ME36.1 has also been used to engineer a trifunctional antibody TRBS07,135 a quadroma hybrid antibody with monovalent binding to GD2 and CD3, carrying a specially engineered chimeric mouse IgG2a x rat IgG2b Fc region with preferential binding to activating FcγR expressed on monocytes, macrophages, dendritic and natural killer cells.135 In mice models, it induced a polyvalent T-cell response and a vaccination effect.

Future directions

Anti-GD2 antibodies have been extensively evaluated in preclinical and clinical studies in the past two decades. Therapeutic efficacy and acceptable safety profile have made them an integral part of the current treatment of neuroblastoma. Curing high risk metastatic neuroblastoma diagnosed ≥18 months of age or with MYCN amplification was unthinkable 25 years ago. The addition of GD2-targeted treatments has greatly improved their outlook, where >50% progression-free survival beyond 5 years is now possible. Several key properties of antibodies and principles of therapeutic applications have been identified, improved, or to be optimized. These have encompassed antibody affinity, novel antibody forms including CAR and BsAb, multistep targeting, choice of isotopes, dosimetry, addition of novel cytokines, and alleviation of the pain side effects. With more effective delivery and less pain side effects, eradicating GD2-positive soft tissue tumors beyond the MRD setting may be possible. Retargeting T cells using CAR or BsAb holds great promise especially for tumors that are less sensitive to Fc-dependent cytotoxicities (ADCC or CMC). Importantly, these redirected T-cells can obviate tumor immune evasion, such as the down-regulation of HLA and co-stimulatory molecules. Although strategies are in place to reduce adverse cytokine storm, careful dosing and schedule will need to be tested in the upcoming clinical trials. Since GD2 is expressed on a wide spectrum of human tumors, including osteosarcoma, soft tissue sarcomas, SCLC, melanoma and retinoblastoma, GD2-targeted strategies could be beneficial in these other tumor types.

Acknowledgments

Supported in part by a grant from the Department of Defense (PR111043), Enid A Haupt Endowed Chair, and grants from Kids Walk for Kids with Cancer NYC, Katie’s Find a Cure Foundation, Catie Hoch Foundation and the Robert Steel Foundation.

We want to thank Dr. Irene Cheung, Dr. Brian H. Kushner, Dr. Kim Kramer, and Dr. Shakeel Modak for their critical review of this manuscript and valuable suggestions.

Footnotes

Financial disclosure: NK Cheung was named as an inventor on patents filed by MSKCC on beta-glucan, hu3F8 and 8H9. Beta-glucan was licensed by MSKCC to Biotec Pharmacon.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Yanagisawa M, Yoshimura S, Yu RK. Expression of GD2 and GD3 gangliosides in human embryonic neural stem cells. ASN Neuro. 2011;3 doi: 10.1042/AN20110006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Martinez C, Hofmann TJ, Marino R, et al. Human bone marrow mesenchymal stromal cells express the neural ganglioside GD2: a novel surface marker for the identification of MSCs. Blood. 2007;109:4245–8. doi: 10.1182/blood-2006-08-039347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Battula VL, Shi Y, Evans KW, et al. Ganglioside GD2 identifies breast cancer stem cells and promotes tumorigenesis. J Clin Invest. 2012;122:2066–2078. doi: 10.1172/JCI59735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Liang YJ, Ding Y, Levery SB, et al. Differential expression profiles of glycosphingolipids in human breast cancer stem cells vs. cancer non-stem cells. Proc Natl Acad Sci U S A. 2013;110:4968–73. doi: 10.1073/pnas.1302825110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Lammie G, Cheung N, Gerald W, et al. Ganglioside gd(2) expression in the human nervous-system and in neuroblastomas - an immunohistochemical study. International journal of oncology. 1993;3:909–15. doi: 10.3892/ijo.3.5.909. [DOI] [PubMed] [Google Scholar]
  • 6.Cheresh DA, Pierschbacher MD, Herzig MA, et al. Disialogangliosides GD2 and GD3 are involved in the attachment of human melanoma and neuroblastoma cells to extracellular matrix proteins. J Cell Biol. 1986;102:688–96. doi: 10.1083/jcb.102.3.688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Shochat SJ, Abt AB, Schengrund CL. VCN-releasable sialic acid and gangliosides in human neuroblastomas. Journal of pediatric surgery. 1977;12:413–8. doi: 10.1016/0022-3468(77)90019-7. [DOI] [PubMed] [Google Scholar]
  • 8.Ahmed M, Goldgur Y, Hu J, et al. In silico Driven Redesign of a Clinically Relevant Antibody for the Treatment of GD2 Positive Tumors. PLoS One. 2013;8:e63359. doi: 10.1371/journal.pone.0063359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Cheever MA, Allison JP, Ferris AS, et al. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res. 2009;15:5323–37. doi: 10.1158/1078-0432.CCR-09-0737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Hamilton WB, Helling F, Lloyd KO, et al. Ganglioside expression on human malignant melanoma assessed by quantitative immune thin-layer chromatography. Int J Cancer. 1993;53:566–73. doi: 10.1002/ijc.2910530407. [DOI] [PubMed] [Google Scholar]
  • 11.Chang HR, Cordon-Cardo C, Houghton AN, et al. Expression of disialogangliosides GD2 and GD3 on human soft tissue sarcomas. Cancer. 1992;70:633–8. doi: 10.1002/1097-0142(19920801)70:3<633::aid-cncr2820700315>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]
  • 12.Heiner JP, Miraldi F, Kallick S, et al. Localization of GD2-specific monoclonal antibody 3F8 in human osteosarcoma. Cancer Res. 1987;47:5377–81. [PubMed] [Google Scholar]
  • 13.Modak S, Gerald W, Cheung NK. Disialoganglioside GD2 and a novel tumor antigen: potential targets for immunotherapy of desmoplastic small round cell tumor. Med Pediatr Oncol. 2002;39:547–51. doi: 10.1002/mpo.10151. [DOI] [PubMed] [Google Scholar]
  • 14.Grant SC, Kostakoglu L, Kris MG, et al. Targeting of small-cell lung cancer using the anti-GD2 ganglioside monoclonal antibody 3F8: a pilot trial. Eur J Nucl Med. 1996;23:145–9. doi: 10.1007/BF01731837. [DOI] [PubMed] [Google Scholar]
  • 15.Davidoff AM. Neuroblastoma. Semin Pediatr Surg. 2012;21:2–14. doi: 10.1053/j.sempedsurg.2011.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Cheung NK, Saarinen UM, Neely JE, et al. Monoclonal antibodies to a glycolipid antigen on human neuroblastoma cells. Cancer Res. 1985;45:2642–9. [PubMed] [Google Scholar]
  • 17.Mujoo K, Cheresh DA, Yang HM, et al. Disialoganglioside GD2 on human neuroblastoma cells: target antigen for monoclonal antibody-mediated cytolysis and suppression of tumor growth. Cancer Res. 1987;47:1098–1104. [PubMed] [Google Scholar]
  • 18.Yu AL, Gilman AL, Ozkaynak MF, et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N Engl J Med. 2010;363:1324–34. doi: 10.1056/NEJMoa0911123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Cheung NK, Guo H, Hu J, et al. Humanizing murine IgG3 anti-GD2 antibody m3F8 substantially improves antibody-dependent cell-mediated cytotoxicity while retaining targeting in vivo. OncoImmunology. 2012;1:477–486. doi: 10.4161/onci.19864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Basu EM, Kushner B, Modak S, et al. Phase I Study of Anti-GD2 Humanized 3F8 (hu3F8) Monoclonal Antibody (MAb) in Patients with Relapsed or Refractory Neuroblastoma (NB) or Other GD2-Positive Solid Tumors, Advances in Neuroblastoma Research 2014. Cologne. 2014:POC034. [Google Scholar]
  • 21.Kushner B, Cheung IY, Basu EM, et al. Phase I study of anti-GD2 humanized 3F8 (hu3F8) monoclonal antibody (MAb) plus granulocyte-macrophage colony-stimulating factor (GM-CSF) in patients with relapsed high-risk neuroblastoma (HR-NB), Advances in Neuroblastoma Research 2014. Cologne. 2014:POC032. [Google Scholar]
  • 22.Cheung IY, Hsu K, Cheung NK. Activation of Peripheral-Blood Granulocytes Is Strongly Correlated With Patient Outcome After Immunotherapy With Anti-GD2 Monoclonal Antibody and Granulocyte-Macrophage Colony-Stimulating Factor. J Clin Oncol. 2012;30:426–432. doi: 10.1200/JCO.2011.37.6236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Tarek N, Le Luduec JB, Gallagher MM, et al. Unlicensed NK cells target neuroblastoma following anti-GD2 antibody treatment. The Journal of clinical investigation. 2012;122:3260–70. doi: 10.1172/JCI62749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Cheung NK, Lazarus H, Miraldi FD, et al. Ganglioside GD2 specific monoclonal antibody 3F8: a phase I study in patients with neuroblastoma and malignant melanoma. J Clin Oncol. 1987;5:1430–40. doi: 10.1200/JCO.1987.5.9.1430. [DOI] [PubMed] [Google Scholar]
  • 25.Brodeur G, Seeger RC, Barrett A, et al. International criteria for diagnosis, staging, and response to treatment in patients with neuroblastoma. J Clin Oncol. 1988;6:1874–1881. doi: 10.1200/JCO.1988.6.12.1874. [DOI] [PubMed] [Google Scholar]
  • 26.Cheung NV, Lazarus H, Miraldi FD, et al. Reassessment of patient response to monoclonal antibody 3F8. J Clin Oncol. 1992;10:671–2. doi: 10.1200/JCO.1992.10.4.671. [DOI] [PubMed] [Google Scholar]
  • 27.Kushner BH, Kramer K, Modak S, et al. Successful multifold dose escalation of anti-GD2 monoclonal antibody 3F8 in patients with neuroblastoma: a phase I study. J Clin Oncol. 2011;29:1168–74. doi: 10.1200/JCO.2010.28.3317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Cheung NK, Kushner BH, Cheung IY, et al. Anti-G(D2) antibody treatment of minimal residual stage 4 neuroblastoma diagnosed at more than 1 year of age. J Clin Oncol. 1998;16:3053–60. doi: 10.1200/JCO.1998.16.9.3053. [DOI] [PubMed] [Google Scholar]
  • 29.Kushner BH, Cheung NK. GM-CSF enhances 3F8 monoclonal antibody-dependent cellular cytotoxicity against human melanoma and neuroblastoma. Blood. 1989;73:1936–41. [PubMed] [Google Scholar]
  • 30.Kushner BH, Kramer K, Cheung NK. Phase II trial of the anti-G(D2) monoclonal antibody 3F8 and granulocyte-macrophage colony-stimulating factor for neuroblastoma. J Clin Oncol. 2001;19:4189–94. doi: 10.1200/JCO.2001.19.22.4189. [DOI] [PubMed] [Google Scholar]
  • 31.Beiske K, Ambros PF, Burchill SA, et al. Detecting minimal residual disease in neuroblastoma patients-the present state of the art. Cancer Lett. 2005;228:229–40. doi: 10.1016/j.canlet.2005.02.053. [DOI] [PubMed] [Google Scholar]
  • 32.Cheung NK, Sowers R, Vickers AJ, et al. 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;24:2885–90. doi: 10.1200/JCO.2005.04.6011. [DOI] [PubMed] [Google Scholar]
  • 33.Cheung NK, Cheung IY, Kramer K, et al. Key Role for Myeloid Cells: Phase II Results of Anti-G Antibody 3F8 Plus Granulocyte-Macrophage Colony-Stimulating Factor for Chemoresistant Osteomedullary Neuroblastoma. Int J Cancer. 2014 doi: 10.1002/ijc.28851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Cheung NK, Cheung IY, Kushner BH, et al. Murine Anti-GD2 Monoclonal Antibody 3F8 Combined With Granulocyte-Macrophage Colony-Stimulating Factor and 13-Cis-Retinoic Acid in High-Risk Patients With Stage 4 Neuroblastoma in First Remission. J Clin Oncol. 2012;30:3264–70. doi: 10.1200/JCO.2011.41.3807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kushner BH, Cheung IY, Kuk D, et al. Anti-GD2 monoclonal antibody 3F8/GM-CSF plus 13-cis-reinotic acid for consolidation of second or later remission of neuroblastoma. Cancer. 2014 (submitted) [Google Scholar]
  • 36.Cheung NK, Modak S, Vickers A, et al. Orally administered beta-glucans enhance anti-tumor effects of monoclonal antibodies. Cancer Immunol Immunother. 2002;51:557–64. doi: 10.1007/s00262-002-0321-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Modak S, Kushner BH, Kramer K, et al. Anti-GD2 antibody 3F8 and barley-derived (1 --> 3),(1 --> 4)-beta--glucan: A Phase I study in patients with chemoresistant neuroblastoma. Oncoimmunology. 2013;2:e23402. doi: 10.4161/onci.23402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Modak S, Kushner BH, Kramer K, et al. Phase I Study of the Combination of Anti-GD2 Antibody 3F8 and Yeast-Derived (1 3), 1 6)-β-D-Glucan for Resistant Neuroblastoma. American Society of Pediatric Hematology/Oncology (22nd); 2008. Abstract #2346. [Google Scholar]
  • 39.Modak M, Cheung NK, Kushner B, et al. Phase I study of haploidentical natural killer (NK) cells plus monoclonal antibody 3F8 for resistant high-risk neuroblastom (HR-NB), Advances in Neuroblastoma Research 2014. Cologne. 2014:OR075. [Google Scholar]
  • 40.Murray JL, Cunningham JE, Brewer H, et al. Phase I trial of murine monoclonal antibody 14G2a administered by prolonged intravenous infusion in patients with neuroectodermal tumors. J Clin Oncol. 1994;12:184–93. doi: 10.1200/JCO.1994.12.1.184. [DOI] [PubMed] [Google Scholar]
  • 41.Frost JD, Hank JA, Reaman GH, et al. A phase I/IB trial of murine monoclonal anti-GD2 antibody 14.G2a plus interleukin-2 in children with refractory neuroblastoma: a report of the Children’s Cancer Group. Cancer. 1997;80:317–33. doi: 10.1002/(sici)1097-0142(19970715)80:2<317::aid-cncr21>3.0.co;2-w. [DOI] [PubMed] [Google Scholar]
  • 42.Thurin J, Thurin M, Kimoto Y, et al. Monoclonal antibody-defined correlations in melanoma between levels of GD2 and GD3 antigens and antibody-mediated cytotoxicity. Cancer Res. 1987;47:1229–33. [PubMed] [Google Scholar]
  • 43.Iliopoulos D, Ernst C, Steplewski Z, et al. Inhibition of metastases of a human melanoma xenograft by monoclonal antibody to the GD2/GD3 gangliosides. J Natl Cancer Inst. 1989;81:440–444. doi: 10.1093/jnci/81.6.440. [DOI] [PubMed] [Google Scholar]
  • 44.Mueller BM, Romerdahl CA, Gillies SD, et al. Enhancement of antibody-dependent cytotoxicity with a chimeric anti-GD2 antibody. J Immunol. 1990;144:1382–1386. [PubMed] [Google Scholar]
  • 45.Saleh MN, Khazaeli MB, Wheeler RH, et al. Phase I trial of the chimeric anti-GD2 monoclonal antibody ch14.18 in patients with malignant melanoma. Hum Antibodies Hybridomas. 1992;3:19–24. [PubMed] [Google Scholar]
  • 46.Handgretinger R, Anderson K, Lang P, et al. A phase I study of human/mouse chimeric antiganglioside GD2 antibody ch14.18 in patients with neuroblastoma. Eur J Cancer. 1995;31A:261–7. doi: 10.1016/0959-8049(94)00413-y. [DOI] [PubMed] [Google Scholar]
  • 47.Yu AL, Uttenreuther-Fischer MM, Huang CS, et al. Phase I trial of a human-mouse chimeric anti-disialoganglioside monoclonal antibody ch14.18 in patients with refractory neuroblastoma and osteosarcoma. J Clin Oncol. 1998;16:2169–80. doi: 10.1200/JCO.1998.16.6.2169. [DOI] [PubMed] [Google Scholar]
  • 48.Simon T, Hero B, Faldum A, et al. Consolidation treatment with chimeric anti-GD2-antibody ch14.18 in children older than 1 year with metastatic neuroblastoma. J Clin Oncol. 2004;22:3549–57. doi: 10.1200/JCO.2004.08.143. [DOI] [PubMed] [Google Scholar]
  • 49.Simon T, Hero B, Faldum A, et al. Long term outcome of high-risk neuroblastoma patients after immunotherapy with antibody ch14.18 or oral metronomic chemotherapy. BMC Cancer. 2011;11:21. doi: 10.1186/1471-2407-11-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ozkaynak MF, Sondel PM, Krailo MD, et al. Phase I study of chimeric human/murine anti-ganglioside G(D2) monoclonal antibody (ch14.18) with granulocyte-macrophage colony-stimulating factor in children with neuroblastoma immediately after hematopoietic stem-cell transplantation: a Children’s Cancer Group Study. J Clin Oncol. 2000;18:4077–85. doi: 10.1200/JCO.2000.18.24.4077. [DOI] [PubMed] [Google Scholar]
  • 51.Yu ALBA, Alvarado C, Rao VJ, Castleberry RP. Usefulness of a chimeric anti-GD2 (ch14.18) and GM-CSF for refractory neuroblastoma: a POG phase II study. Proc Am Soc Clin Oncol. 1997;16 [Google Scholar]
  • 52.Murray JL, Kleinerman ES, Jia SF, et al. Phase Ia/Ib trial of anti-GD2 chimeric monoclonal antibody 14.18 (ch14.18) and recombinant human granulocyte-macrophage colony-stimulating factor (rhGM-CSF) in metastatic melanoma. J Immunother Emphasis Tumor Immunol. 1996;19:206–17. doi: 10.1097/00002371-199605000-00005. [DOI] [PubMed] [Google Scholar]
  • 53.Albertini MR, Hank JA, Schiller JH, et al. Phase IB trial of chimeric antidisialoganglioside antibody plus interleukin 2 for melanoma patients. Clin Cancer Res. 1997;3:1277–1288. [PubMed] [Google Scholar]
  • 54.Ladenstein R, Weixler S, Baykan B, et al. Ch14.18 antibody produced in CHO cells in relapsed or refractory Stage 4 neuroblastoma patients: a SIOPEN Phase 1 study. MAbs. 2013;5:801–9. doi: 10.4161/mabs.25215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Lode H, Muller I. Evaluation of Clinical Response and Survival Following Long-Term Infusion of Anti-GD2 Antibody ch14.18/CHO in Combination with Subcutaneous Interleukin-2 in a Single Center Treatment Program in High-Risk Neuroblastoma, Advances in Neuroblastoma Research 2014. Cologne. 2014:PL010. [Google Scholar]
  • 56.Choi BS, Sondel PM, Hank JA, et al. Phase I trial of combined treatment with ch14.18 and R24 monoclonal antibodies and interleukin-2 for patients with melanoma or sarcoma. Cancer Immunol Immunother. 2006;55:761–74. doi: 10.1007/s00262-005-0069-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Navid F, Sondel PM, Barfield R, 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;32:1445–52. doi: 10.1200/JCO.2013.50.4423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Roberts SS, Chou AJ, Basu EM, et al. Phase I Trial of Anti-GD2 Humanized 3F8 Monoclonal Antibody (MAb) Combined with Subcutaneous Interleukin-2 (scIL2) in Patients with Relapsed Neuroblastoma or Other GD2-Positive Solid Tumors, Advances in Neuroblastoma Research 2014. Cologne. 2014:POC027. [Google Scholar]
  • 59.Cheung IY, Basu EM, Modak M, et al. Pharmacokinetics of humanized anti-GD2 monoclonal antibody Hu3F8 in patients with metastatic GD2-positive tumors Advances in Neuroblastoma Research 2014. Cologne. 2014:POC037. [Google Scholar]
  • 60.Cheung NK, Guo HF, Heller G, et al. Induction of Ab3 and Ab3′ antibody was associated with long-term survival after anti-G(D2) antibody therapy of stage 4 neuroblastoma. Clin Cancer Res. 2000;6:2653–60. [PubMed] [Google Scholar]
  • 61.Foon KA, Lutzky J, Baral RN, et al. Clinical and immune responses in advanced melanoma patients immunized with an anti-idiotype antibody mimicking disialoganglioside GD2. J Clin Oncol. 2000;18:376–384. doi: 10.1200/JCO.2000.18.2.376. [DOI] [PubMed] [Google Scholar]
  • 62.Cheung NK, Canete A, Cheung IY, et al. Disialoganglioside GD2 anti-idiotypic monoclonal antibodies. Int J Cancer. 1993;54:499–505. doi: 10.1002/ijc.2910540324. [DOI] [PubMed] [Google Scholar]
  • 63.Bleeke M, Fest S, Huebener N, et al. Systematic amino acid substitutions improved efficiency of GD2-peptide mimotope vaccination against neuroblastoma. Eur J Cancer. 2009;45:2915–21. doi: 10.1016/j.ejca.2009.07.026. [DOI] [PubMed] [Google Scholar]
  • 64.Bolesta E, Kowalczyk A, Wierzbicki A, et al. DNA vaccine expressing the mimotope of GD2 ganglioside induces protective GD2 cross-reactive antibody responses. Cancer Res. 2005;65:3410–8. doi: 10.1158/0008-5472.CAN-04-2164. [DOI] [PubMed] [Google Scholar]
  • 65.Ragupathi G, Livingston PO, Hood C, et al. Consistent antibody response against ganglioside GD2 induced in patients with melanoma by a GD2 lactone-keyhole limpet hemocyanin conjugate vaccine plus immunological adjuvant QS-21. Clin Cancer Res. 2003;9:5214–20. [PubMed] [Google Scholar]
  • 66.Kushner BH, Cheung IY, Modak S, et al. Phase I Trial of a Bivalent Gangliosides Vaccine in Combination with beta-Glucan for High-Risk Neuroblastoma in Second or Later Remission. Clin Cancer Res. 2014 doi: 10.1158/1078-0432.CCR-13-1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Osenga KL, Hank JA, Albertini MR, et al. A phase I clinical trial of the hu14.18-IL2 (EMD 273063) as a treatment for children with refractory or recurrent neuroblastoma and melanoma: a study of the Children’s Oncology Group. Clin Cancer Res. 2006;12:1750–9. doi: 10.1158/1078-0432.CCR-05-2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Shusterman S, London WB, Gillies SD, 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;28:4969–75. doi: 10.1200/JCO.2009.27.8861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Metelitsa LS, Gillies SD, Super M, et al. Antidisialoganglioside/granulocyte macrophage-colony-stimulating factor fusion protein facilitates neutrophil antibody-dependent cellular cytotoxicity and depends on FcgammaRII (CD32) and Mac-1 (CD11b/CD18) for enhanced effector cell adhesion and azurophil granule exocytosis. Blood. 2002;99:4166–73. doi: 10.1182/blood.v99.11.4166. [DOI] [PubMed] [Google Scholar]
  • 70.Becker JC, Varki N, Gillies SD, et al. Long-lived and transferable tumor immunity in mice after targeted interleukin-2 therapy. J Clin Invest. 1996;98:2801–4. doi: 10.1172/JCI119107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Yang RK, Kalogriopoulos NA, Rakhmilevich AL, et al. Intratumoral hu14.18-IL-2 (IC) Induces Local and Systemic Antitumor Effects That Involve Both Activated T and NK Cells As Well As Enhanced IC Retention. J Immunol. 2012;189:2656–64. doi: 10.4049/jimmunol.1200934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Albertini MR, Hank JA, Gadbaw B, et al. Phase II trial of hu14.18-IL2 for patients with metastatic melanoma. Cancer Immunol Immunother. 2012 doi: 10.1007/s00262-012-1286-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Steel JC, Waldmann TA, Morris JC. Interleukin-15 biology and its therapeutic implications in cancer. Trends Pharmacol Sci. 2012;33:35–41. doi: 10.1016/j.tips.2011.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Leung W. Use of NK cell activity in cure by transplant. Br J Haematol. 2011;155:14–29. doi: 10.1111/j.1365-2141.2011.08823.x. [DOI] [PubMed] [Google Scholar]
  • 75.Delgado DC, Hank JA, Kolesar J, et al. Genotypes of NK cell KIR receptors, their ligands, and Fcgamma receptors in the response of neuroblastoma patients to Hu14.18-IL2 immunotherapy. Cancer Res. 2010;70:9554–61. doi: 10.1158/0008-5472.CAN-10-2211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Kushner BH, Cheung NK. Absolute requirement of CD11/CD18 adhesion molecules, FcRII and the phosphatidylinositol-linked FcRIII for monoclonal antibody-mediated neutrophil antihuman tumor cytotoxicity. Blood. 1992;79:1484–90. [PubMed] [Google Scholar]
  • 77.Ross GD, Vetvicka V, Yan J, et al. Therapeutic intervention with complement and beta-glucan in cancer. Immunopharmacology. 1999;42:61–74. doi: 10.1016/s0162-3109(99)00013-2. [DOI] [PubMed] [Google Scholar]
  • 78.Diamond MS, Springer TA. A subpopulation of Mac-1 (CD11b/CD18) molecules mediates neutrophil adhesion to ICAM-1 and fibrinogen. J Cell Biol. 1993;120:545–56. doi: 10.1083/jcb.120.2.545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Cheung NK, Modak S. Oral (1-->3),(1-->4)-beta-D-glucan synergizes with antiganglioside GD2 monoclonal antibody 3F8 in the therapy of neuroblastoma. Clin Cancer Res. 2002;8:1217–23. [PubMed] [Google Scholar]
  • 80.Ren Y, Chan HM, Li Z, et al. Upregulation of macrophage migration inhibitory factor contributes to induced N-Myc expression by the activation of ERK signaling pathway and increased expression of interleukin-8 and VEGF in neuroblastoma. Oncogene. 2004;23:4146–54. doi: 10.1038/sj.onc.1207490. [DOI] [PubMed] [Google Scholar]
  • 81.Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol. 2010;11:889–96. doi: 10.1038/ni.1937. [DOI] [PubMed] [Google Scholar]
  • 82.Munn DH, Cheung NK. Antibody-dependent antitumor cytotoxicity by human monocytes cultured with recombinant macrophage colony-stimulating factor. Induction of efficient antibody-mediated antitumor cytotoxicity not detected by isotope release assays. J Exp Med. 1989;170:511–26. doi: 10.1084/jem.170.2.511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Ladenstein R, Potschger U, Siabalis D, et al. Dose finding study for the use of subcutaneous recombinant interleukin-2 to augment natural killer cell numbers in an outpatient setting for stage 4 neuroblastoma after megatherapy and autologous stem-cell reinfusion. J Clin Oncol. 2011;29:441–8. doi: 10.1200/JCO.2009.23.5465. [DOI] [PubMed] [Google Scholar]
  • 84.Berger C, Berger M, Hackman RC, et al. Safety and immunologic effects of IL-15 administration in nonhuman primates. Blood. 2009;114:2417–26. doi: 10.1182/blood-2008-12-189266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Metelitsa LS, Wu HW, Wang H, et al. Natural killer T cells infiltrate neuroblastomas expressing the chemokine CCL2. J Exp Med. 2004;199:1213–21. doi: 10.1084/jem.20031462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Liu D, Song L, Wei J, et al. IL-15 protects NKT cells from inhibition by tumor-associated macrophages and enhances antimetastatic activity. J Clin Invest. 2012;122:2221–33. doi: 10.1172/JCI59535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Mortier E, Quemener A, Vusio P, et al. Soluble interleukin-15 receptor alpha (IL-15R alpha)-sushi as a selective and potent agonist of IL-15 action through IL-15R beta/gamma. Hyperagonist IL-15 x IL-15R alpha fusion proteins. J Biol Chem. 2006;281:1612–9. doi: 10.1074/jbc.M508624200. [DOI] [PubMed] [Google Scholar]
  • 88.Vincent M, Bessard A, Cochonneau D, et al. Tumor targeting of the IL-15 superagonist RLI by an anti-GD2 antibody strongly enhances its antitumor potency. Int J Cancer. 2013;133:757–65. doi: 10.1002/ijc.28059. [DOI] [PubMed] [Google Scholar]
  • 89.Howe BM, Johnson GB, Wenger DE. Current concepts in MRI of focal and diffuse malignancy of bone marrow. Semin Musculoskelet Radiol. 2013;17:137–44. doi: 10.1055/s-0033-1343069. [DOI] [PubMed] [Google Scholar]
  • 90.Qian J, Liu T, Yang L, et al. Structural characterization of N-linked oligosaccharides on monoclonal antibody cetuximab by the combination of orthogonal matrix-assisted laser desorption/ionization hybrid quadrupole-quadrupole time-of-flight tandem mass spectrometry and sequential enzymatic digestion. Anal Biochem. 2007;364:8–18. doi: 10.1016/j.ab.2007.01.023. [DOI] [PubMed] [Google Scholar]
  • 91.Galili U, Anaraki F, Thall A, et al. One percent of human circulating B lymphocytes are capable of producing the natural anti-Gal antibody. Blood. 1993;82:2485–93. [PubMed] [Google Scholar]
  • 92.Zhu A, Hurst R. Anti-N-glycolylneuraminic acid antibodies identified in healthy human serum. Xenotransplantation. 2002;9:376–81. doi: 10.1034/j.1399-3089.2002.02138.x. [DOI] [PubMed] [Google Scholar]
  • 93.Tronconi MC, Sclafani F, Rimassa L, et al. Fatal infusion reaction to cetuximab: the need for predictive risk factors and safer patient selection. J Clin Oncol. 2011;29:e680–1. doi: 10.1200/JCO.2011.35.5651. [DOI] [PubMed] [Google Scholar]
  • 94.Lammerts van Bueren JJ, Rispens T, Verploegen S, et al. Anti-galactose-alpha-1,3-galactose IgE from allergic patients does not bind alpha-galactosylated glycans on intact therapeutic antibody Fc domains. Nat Biotechnol. 2011;29:574–6. doi: 10.1038/nbt.1912. [DOI] [PubMed] [Google Scholar]
  • 95.Kramer K, Pandit-Taskar N, Zanzonico P, et al. International Society of Pediatric NeuroOncology. Singapore: 2014. High risk and Recurrent Medulloblastoma (MB): Lessons Learned Using Radioimmunotherapy. [Google Scholar]
  • 96.Alvarez-Rueda N, Desselle A, Cochonneau D, et al. A monoclonal antibody to O-acetyl-GD2 ganglioside and not to GD2 shows potent anti-tumor activity without peripheral nervous system cross-reactivity. PLoS ONE. 2011;6:e25220. doi: 10.1371/journal.pone.0025220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Cheung NK, Cheung IY, Kramer K, et al. Key Role for Myeloid Cells: Phase II Results of Anti-GD2 Antibody 3F8 Plus Granulocyte-Macrophage Colony-Stimulating Factor for Chemoresistant Osteomedullary Neuroblastoma. International Journal of cancer. 2014 doi: 10.1002/ijc.28851. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Ahmed M, Cheung NK. Engineering anti-GD2 monoclonal antibodies for cancer immunotherapy. FEBS Lett. 2014;588:288–97. doi: 10.1016/j.febslet.2013.11.030. [DOI] [PubMed] [Google Scholar]
  • 99.Prise KM, O’Sullivan JM. Radiation-induced bystander signalling in cancer therapy. Nat Rev Cancer. 2009;9:351–60. doi: 10.1038/nrc2603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Larson SM, Divgi C, Sgouros G, et al. Monoclonal antibodies: Basic prniciples - Radioisotope conjugates. In: DeVita VT, Hellman S, Rosenberg SA, editors. Biologic Therapy of Cancer - Principles and Practice. Philadelphia: J.B. Lippincott Co; 2000. pp. 396–412. [Google Scholar]
  • 101.Croog VJ, Kramer K, Cheung NK, et al. Whole Neuraxis Irradiation to Address Central Nervous System Relapse in High-Risk Neuroblastoma. Int J Radiat Oncol Biol Phys. 2010 doi: 10.1016/j.ijrobp.2009.09.005. [DOI] [PubMed] [Google Scholar]
  • 102.Davson H, Segal MB. Physiology of the CSF and blood-brain barriers. Boca Raton, FL: CRC Press; 1996. pp. 489–523. [Google Scholar]
  • 103.Kramer K, Humm JL, Souweidane MM, et al. Phase I study of targeted radioimmunotherapy for leptomeningeal cancers using intra-Ommaya 131-I-3F8. J Clin Oncol. 2007;25:5465–70. doi: 10.1200/JCO.2007.11.1807. [DOI] [PubMed] [Google Scholar]
  • 104.Kramer K, Kushner BH, Modak S, et al. Compartmental intrathecal radioimmunotherapy: results for treatment for metastatic CNS neuroblastoma. J Neurooncol. 2010;97:409–18. doi: 10.1007/s11060-009-0038-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Orcutt KD, Slusarczyk AL, Cieslewicz M, et al. Engineering an antibody with picomolar affinity to DOTA chelates of multiple radionuclides for pretargeted radioimmunotherapy and imaging. Nucl Med Biol. 2011;38:223–33. doi: 10.1016/j.nucmedbio.2010.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Elgqvist J, Frost S, Pouget JP, et al. The Potential and Hurdles of Targeted Alpha Therapy - Clinical Trials and Beyond. Front Oncol. 2014;3:324. doi: 10.3389/fonc.2013.00324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Miederer M, McDevitt MR, Borchardt P, et al. Treatment of neuroblastoma meningeal carcinomatosis with intrathecal application of alpha-emitting atomic nanogenerators targeting disialo-ganglioside GD2. Clin Cancer Res. 2004;10:6985–92. doi: 10.1158/1078-0432.CCR-04-0859. [DOI] [PubMed] [Google Scholar]
  • 108.Rotmensch J, Roeske J, Chen G, et al. Estimates of dose to intraperitoneal micrometastases from alpha and beta emitters in radioimmunotherapy. Gynecol Oncol. 1990;38:478–85. doi: 10.1016/0090-8258(90)90095-3. [DOI] [PubMed] [Google Scholar]
  • 109.He P, Kramer K, Smith-Jones P, et al. Two-compartment model of radioimmunotherapy delivered through cerebrospinal fluid. Eur J Nucl Med Mol Imaging. 2011;38:334–42. doi: 10.1007/s00259-010-1633-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Lv Y, Cheung NK, Fu BM. A pharmacokinetic model for radioimmunotherapy delivered through cerebrospinal fluid for the treatment of leptomeningeal metastases. J Nucl Med. 2009;50:1324–31. doi: 10.2967/jnumed.108.060798. [DOI] [PubMed] [Google Scholar]
  • 111.Goldenberg DM, Sharkey RM, Paganelli G, et al. Antibody pretargeting advances cancer radioimmunodetection and radioimmunotherapy. J Clin Oncol. 2006;24:823–34. doi: 10.1200/JCO.2005.03.8471. [DOI] [PubMed] [Google Scholar]
  • 112.Cheal SM, Xu H, Guo HF, et al. Preclinical Evaluation of Multistep Targeting of Diasialoganglioside GD2 Using an IgG-scFv Bispecific Antibody with High Affinity for GD2 and DOTA Metal Complex. Mol Cancer Ther. 2014;13:1803–12. doi: 10.1158/1535-7163.MCT-13-0933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Cheung NK, Modak S, Lin Y, et al. Single-chain Fv-streptavidin substantially improved therapeutic index in multistep targeting directed at disialoganglioside GD2. J Nucl Med. 2004;45:867–77. [PubMed] [Google Scholar]
  • 114.Raffaghello L, Pagnan G, Pastorino F, et al. In vitro and in vivo antitumor activity of liposomal Fenretinide targeted to human neuroblastoma. Int J Cancer. 2003;104:559–67. doi: 10.1002/ijc.10991. [DOI] [PubMed] [Google Scholar]
  • 115.Pastorino F, Brignole C, Marimpietri D, et al. Doxorubicin-loaded Fab’ fragments of anti-disialoganglioside immunoliposomes selectively inhibit the growth and dissemination of human neuroblastoma in nude mice. Cancer Res. 2003;63:86–92. [PubMed] [Google Scholar]
  • 116.Brignole C, Pastorino F, Marimpietri D, et al. Immune cell-mediated antitumor activities of GD2-targeted liposomal c-myb antisense oligonucleotides containing CpG motifs. J Natl Cancer Inst. 2004;96:1171–80. doi: 10.1093/jnci/djh221. [DOI] [PubMed] [Google Scholar]
  • 117.Tivnan A, Orr WS, Gubala V, et al. Inhibition of neuroblastoma tumor growth by targeted delivery of microRNA-34a using anti-disialoganglioside GD2 coated nanoparticles. PLoS ONE. 2012;7:e38129. doi: 10.1371/journal.pone.0038129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Welch C, Chen Y, Stallings RL. MicroRNA-34a functions as a potential tumor suppressor by inducing apoptosis in neuroblastoma cells. Oncogene. 2007;26:5017–22. doi: 10.1038/sj.onc.1210293. [DOI] [PubMed] [Google Scholar]
  • 119.Sun F, Fu H, Liu Q, et al. Downregulation of CCND1 and CDK6 by miR-34a induces cell cycle arrest. FEBS Lett. 2008;582:1564–8. doi: 10.1016/j.febslet.2008.03.057. [DOI] [PubMed] [Google Scholar]
  • 120.Pang RT, Leung CO, Ye TM, et al. MicroRNA-34a suppresses invasion through downregulation of Notch1 and Jagged1 in cervical carcinoma and choriocarcinoma cells. Carcinogenesis. 2010;31:1037–44. doi: 10.1093/carcin/bgq066. [DOI] [PubMed] [Google Scholar]
  • 121.Di Paolo D, Pastorino F, Zuccari G, et al. Enhanced anti-tumor and anti-angiogenic efficacy of a novel liposomal fenretinide on human neuroblastoma. J Control Release. 2013;170:445–51. doi: 10.1016/j.jconrel.2013.06.015. [DOI] [PubMed] [Google Scholar]
  • 122.Peng CA, Wang CH. Anti-Neuroblastoma Activity of Gold Nanorods Bound with GD2 Monoclonal Antibody under Near-Infrared Laser Irradiation. Cancers (Basel) 2011;3:227–40. doi: 10.3390/cancers3010227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Wang CH, Huang YJ, Chang CW, et al. In vitro photothermal destruction of neuroblastoma cells using carbon nanotubes conjugated with GD2 monoclonal antibody. Nanotechnology. 2009;20:315101. doi: 10.1088/0957-4484/20/31/315101. [DOI] [PubMed] [Google Scholar]
  • 124.Kowolik CM, Topp MS, Gonzalez S, et al. CD28 costimulation provided through a CD19-specific chimeric antigen receptor enhances in vivo persistence and antitumor efficacy of adoptively transferred T cells. Cancer Res. 2006;66:10995–1004. doi: 10.1158/0008-5472.CAN-06-0160. [DOI] [PubMed] [Google Scholar]
  • 125.Carpenito C, Milone MC, Hassan R, et al. Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc Natl Acad Sci U S A. 2009;106:3360–5. doi: 10.1073/pnas.0813101106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Pule MA, Savoldo B, Myers GD, et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med. 2008;14:1264–70. doi: 10.1038/nm.1882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Louis CU, Savoldo B, Dotti G, et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood. 2011;118:6050–6. doi: 10.1182/blood-2011-05-354449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Liebsch L, Kailayangiri S, Beck L, et al. Ewing sarcoma dissemination and response to T-cell therapy in mice assessed by whole-body magnetic resonance imaging. Br J Cancer. 2013;109:658–66. doi: 10.1038/bjc.2013.356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Esser R, Muller T, Stefes D, et al. NK cells engineered to express a GD2 -specific antigen receptor display built-in ADCC-like activity against tumour cells of neuroectodermal origin. J Cell Mol Med. 2012;16:569–81. doi: 10.1111/j.1582-4934.2011.01343.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Hoffmann P, Hofmeister R, Brischwein K, et al. Serial killing of tumor cells by cytotoxic T cells redirected with a CD19-/CD3-bispecific single-chain antibody construct. Int J Cancer. 2005;115:98–104. doi: 10.1002/ijc.20908. [DOI] [PubMed] [Google Scholar]
  • 131.Bargou R, Leo E, Zugmaier G, et al. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science. 2008;321:974–7. doi: 10.1126/science.1158545. [DOI] [PubMed] [Google Scholar]
  • 132.Cheng M, Ahmed M, Xu H, et al. Structural design of disialoganglioside GD2 and CD3-bispecific antibodies to redirect T cells for tumor therapy. Int J Cancer. 2014 doi: 10.1002/ijc.29007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Xu H, Cheng M, Guo HF, et al. Hu3F8 Bispecific Antibody to Engage T cells against Neuroblastoma, Advances in Neuroblastoma Research 2014. Cologne, Germany. 2014:POT020. [Google Scholar]
  • 134.Yankelevich M, Kondadasula SV, Thakur A, et al. Anti-CD3 x anti-GD2 bispecific antibody redirects T-cell cytolytic activity to neuroblastoma targets. Pediatr Blood Cancer. 2012 doi: 10.1002/pbc.24237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Eissler N, Ruf P, Mysliwietz J, et al. Trifunctional bispecific antibodies induce tumor-specific T cells and elicit a vaccination effect. Cancer Res. 2012;72:3958–66. doi: 10.1158/0008-5472.CAN-12-0146. [DOI] [PubMed] [Google Scholar]

RESOURCES