Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 290(21): 13017-27, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25851904

ABSTRACT

Ganglioside GD2 is highly expressed on neuroectodermal tumors and an attractive therapeutic target for antibodies that have already shown some clinical efficacy. To further improve the current antibodies, which have modest affinity, we sought to improve affinity by using a combined method of random mutagenesis and in silico assisted design to affinity-mature the anti-GD2 monoclonal antibody hu3F8. Using yeast display, mutants in the Fv with enhanced binding over the parental clone were FACS-sorted and cloned. In silico modeling identified the minimal key interacting residues involved in the important charged interactions with the sialic acid groups of GD2. Two mutations, D32H (L-CDR1) and E1K (L-FR1) altered the electrostatic surface potential of the antigen binding site, allowing for an increase in positive charge to enhance the interaction with the negatively charged GD2-pentasaccharide headgroup. Purified scFv and IgG mutant forms were then tested for antigen specificity by ELISA, for tissue specificity by immunohistochemistry, for affinity by BIACORE, for antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-mediated cytotoxicity in vitro, and for anti-tumor efficacy in xenografted humanized mice. The nearly 7-fold improvement in affinity of hu3F8 with a single D32H (L-CDR1) mutation translated into a ∼12-fold improvement in NK92MI-transfected CD16-mediated ADCC, a 6-fold improvement in CD32-mediated ADCC, and a 2.5-fold improvement in complement-mediated cytotoxicity while maintaining restricted normal tissue cross-reactivity and achieving substantial improvement in tumor ablation in vivo. Despite increasing GD2 affinity, the double mutation D32H (L-CDR1) and E1K (L-FR1) did not further improve anti-tumor efficacy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibody-Dependent Cell Cytotoxicity/immunology , Gangliosides/immunology , Neuroblastoma/immunology , Neuroblastoma/prevention & control , Static Electricity , Animals , Antibody Specificity , Cell Proliferation , Computer Simulation , Flow Cytometry , Humans , Immunoenzyme Techniques , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Models, Molecular , Mutagenesis, Site-Directed , Mutation/genetics , Neuroblastoma/pathology , Protein Conformation , Surface Properties , Tumor Cells, Cultured , Two-Hybrid System Techniques , Xenograft Model Antitumor Assays
2.
Pediatr Blood Cancer ; 63(10): 1780-5, 2016 10.
Article in English | MEDLINE | ID: mdl-27304202

ABSTRACT

BACKGROUND: GD2 and GD3 are the tumor-associated glycolipid antigens found in a broad spectrum of human cancers. GD2-specific antibody is currently a standard of care for high-risk neuroblastoma therapy. In this study, the pattern of GD2 and GD3 expression among pediatric/adolescent or young adult tumors was determined, providing companion diagnostics for targeted therapy. METHODS: Ninety-two specimens of human osteosarcoma (OS), rhabdomyosarcoma (RMS), Ewing family of tumors, desmoplastic small round cell tumor (DSRCT), and melanoma were analyzed for GD2/GD3 expression by immunohistochemistry. Murine monoclonal antibody 3F8 was used for GD2 staining, and R24 for GD3. Staining was scored according to both intensity and percentage of positive tumor cells from 0 to 4. RESULTS: Both gangliosides were highly prevalent in OS and melanoma. Among other tumors, GD3 expression was higher than GD2 expression. Most OS samples demonstrated strong staining for GD2 and GD3, whereas expression for other tumors was highly variable. Mean intensity of GD2 expression was significantly more heterogeneous (P < 0.001) when compared to GD3 across tumor types. When assessing the difference between GD2 and GD3 expression in all tumor types combined, GD3 expression had a significantly higher score (P = 0.049). When analyzed within each cancer, GD3 expression was significantly higher only in DSRCT (P = 0.002). There was no statistical difference in either GD2 or GD3 expression between primary and recurrent sarcomas. CONCLUSION: GD2/GD3 expression among pediatric solid tumors is common, albeit with variable level of expression. Especially for patients with sarcoma, these gangliosides can be potential targets for antibody-based therapies.


Subject(s)
Gangliosides/analysis , Sarcoma/chemistry , Adolescent , Adult , Child , Humans , Immunohistochemistry , Young Adult
3.
Genes Chromosomes Cancer ; 53(10): 803-14, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24898736

ABSTRACT

Central nervous system (CNS) is an increasingly common site of isolated metastasis for patients with Stage 4 neuroblastoma. To explore the microRNA (miRNA) profile of this metastatic process, miRNA sequencing was performed to identify miRNA sequence families with differential expression between tumor pairs (pre-CNS primary and CNS metastasis) from 13 patients with Stage 4 neuroblastoma. Seven miRNA sequence families had distinct expression in CNS metastases when compared with their corresponding pre-CNS primaries. MiR-7 was upregulated (3.75-fold), and miR-21, miR-22, miR-29a, miR-143, miR-199a-1-3p, and miR-199a-1-5p were downregulated (3.5-6.1-fold), all confirmed by quantitative reverse transcription-PCR. MiR-29a, previously shown to be downregulated in a broad spectrum of solid tumors including neuroblastoma, had the most significant decrease in all 13 CNS metastases (P = 0.001). Its known onco-targets CDC6, CDK6, and DNMT3A, as well as B7-H3, an inhibitory ligand for T cells, and natural killer cells, were found to have higher differential expression in these 13 CNS metastases when compared with their paired primaries. Additionally, miR-29a expression in primary tumors was significantly lower among patients who eventually relapsed in the CNS. Irrespective of the amplification status of MYCN, which is known to be associated with metastasis, pre-CNS primaries, and CNS metastases had significantly lower miR-29a expression than non-CNS primary tumors. Among MYCN amplified cell lines, those from CNS relapse also had lower miR-29a expression than non-CNS relapse. These findings raised the hypothesis that miR-29a could be a biomarker for neuroblastoma CNS metastasis, and its downregulation may play a pivotal role in CNS progression.


Subject(s)
Biomarkers, Tumor/metabolism , Central Nervous System Neoplasms/secondary , Down-Regulation , MicroRNAs/genetics , Neuroblastoma/pathology , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Cell Line, Tumor , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/metabolism , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Killer Cells, Natural/metabolism , MicroRNAs/metabolism , N-Myc Proto-Oncogene Protein , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Staging , Neuroblastoma/genetics , Neuroblastoma/metabolism , Sequence Analysis, RNA , T-Lymphocytes/metabolism
4.
Int J Cancer ; 135(9): 2199-205, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-24644014

ABSTRACT

Anti-G(D2) murine antibody 3F8 plus subcutaneously (sc) administered granulocyte-macrophage colony-stimulating factor (GM-CSF) was used against primary refractory neuroblastoma in metastatic osteomedullary sites. Large study size and long follow-up allowed assessment of prognostic factors in a multivariate analysis not reported with other anti-G(D2) antibodies. In a phase II trial, 79 patients without prior progressive disease were treated for persistent osteomedullary neuroblastoma documented by histology and/or metaiodobenzyl-guanidine (MIBG) scan. In the absence of human antimouse antibody, 3F8 + scGM-CSF cycles were repeated up to 24 months. Minimal residual disease (MRD) in bone marrow was measured by quantitative reverse transcription-polymerase chain reaction pre-enrollment and post-cycle #2, before initiation of 13-cis-retinoic acid. Study endpoints were: (i) progression-free survival (PFS) compared with the predecessor trial of 3F8 plus intravenously administered (iv) GM-CSF (26 patients) and (ii) impact of MRD on PFS. Using all 105 patients from the two consecutive 3F8 + GM-CSF trials, prognostic factors were analyzed by multivariate Cox regression model. Complete response rates to 3F8 + scGM-CSF were 87% by histology and 38% by MIBG. Five-year PFS was 24 ± 6%, which was significantly superior to 11 ± 7% with 3F8 + ivGM-CSF (p = 0.002). In the 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. Complement consumption was similar with either route of GM-CSF. Toxicities were manageable, allowing outpatient treatment. 3F8 + scGM-CSF is highly active against chemoresistant osteomedullary neuroblastoma. MRD response may be an indicator of tumor sensitivity to anti-G(D2) immunotherapy. Correlative studies highlight the antineoplastic potency of myeloid effectors.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bone Neoplasms/drug therapy , Drug Resistance, Neoplasm , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunoglobulin G/therapeutic use , Myeloid Cells/drug effects , Neuroblastoma/drug therapy , Adolescent , Adult , Antibodies, Monoclonal, Murine-Derived , Antineoplastic Combined Chemotherapy Protocols , Bone Neoplasms/mortality , Bone Neoplasms/secondary , Child , Child, Preschool , Female , Follow-Up Studies , Gangliosides/immunology , Humans , Infant , Isotretinoin/therapeutic use , Male , Myeloid Cells/pathology , Neoplasm Staging , Neoplasm, Residual/drug therapy , Neoplasm, Residual/mortality , Neoplasm, Residual/pathology , Neuroblastoma/mortality , Neuroblastoma/pathology , Prognosis , Survival Rate , Young Adult
5.
Genes Chromosomes Cancer ; 52(12): 1150-66, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24123354

ABSTRACT

Stage 4 neuroblastomas have a high rate of local and metastatic relapse and associated disease mortality. The central nervous system (CNS) is currently one of the most common isolated relapse sites, yet the genomic alterations that contribute to these metastases are unknown. This study sought to identify recurrent DNA copy number alterations (CNAs) and target genes relating to neuroblastoma CNS metastases by studying 19 pre-CNS primary tumors and 27 CNS metastases, including 12 matched pairs. SNP microarray analyses revealed that MYCN amplified (MYCNA) tumors had recurrent CNAs different from non-MYCNA cohorts. Several CNAs known to be prevalent among primary neuroblastomas occurred more frequently in CNS metastases, including 4p-, 7q+, 12q+, and 19q- in non-MYCNA metastases, and 9p- and 14q- irrespective of MYCNA status. In addition, novel CNS metastases-related CNAs included 18q22.1 gains in non-MYCNA pre-CNS primaries and 5p15.33 gains and 15q26.1→tel losses in non-MYCNA CNS metastases. Based on minimal common regions, gene expression, and biological properties, TERT (5p), NR2F2 (15q), ALDH1A3 (15q), CDKN2A (9p), and possibly CDH7 and CDH19 (18q) were candidate genes associated with the CNS metastatic process. Notably, the 5p15 minimal common region contained only TERT, and non-MYCNA CNS metastases with focal 5p15 gains had increased TERT expression, similar to MYCNA tumors. These findings suggest that a specific genomic lesion (18q22.1 gain) predisposes to CNS metastases and that distinct lesions are recurrently acquired during metastatic progression. Among the acquired lesions, increased TERT copy number and expression appears likely to function in lieu of MYCNA to promote CNS metastasis.


Subject(s)
Central Nervous System Neoplasms/genetics , DNA Copy Number Variations , Neuroblastoma/genetics , Telomerase/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Central Nervous System Neoplasms/secondary , Child , Child, Preschool , Chromosomes, Human/genetics , Female , Humans , Male , Middle Aged , N-Myc Proto-Oncogene Protein , Neoplasm Recurrence, Local/genetics , Neuroblastoma/secondary , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Translocation, Genetic , Young Adult
6.
Vaccines (Basel) ; 12(6)2024 May 28.
Article in English | MEDLINE | ID: mdl-38932316

ABSTRACT

The carbohydrate ganglioside GD2/GD3 cancer vaccine adjuvanted by ß-glucan stimulates anti-GD2 IgG1 antibodies that strongly correlate with improved progression-free survival (PFS) and overall survival (OS) among patients with high-risk neuroblastoma. Thirty-two patients who relapsed on the vaccine (first enrollment) were re-treated on the same vaccine protocol (re-enrollment). Titers during the first enrollment peaked by week 32 at 751 ± 270 ng/mL, which plateaued despite vaccine boosts at 1.2-4.5 month intervals. After a median wash-out interval of 16.1 months from the last vaccine dose during the first enrollment to the first vaccine dose during re-enrollment, the anti-GD2 IgG1 antibody rose to a peak of 4066 ± 813 ng/mL by week 3 following re-enrollment (p < 0.0001 by the Wilcoxon matched-pairs signed-rank test). Yet, these peaks dropped sharply and continually despite repeated boosts at 1.2-4.5 month intervals, before leveling off by week 20 to the first enrollment peak levels. Despite higher antibody titers, patients experienced no pain or neuropathic side effects, which were typically associated with immunotherapy using monoclonal anti-GD2 antibodies. By the Kaplan-Meier method, PFS was estimated to be 51%, and OS was 81%. The association between IgG1 titer during re-enrollment and ß-glucan receptor dectin-1 SNP rs3901533 was significant (p = 0.01). A longer prime-boost interval could significantly improve antibody responses in patients treated with ganglioside conjugate cancer vaccines.

7.
Pediatr Blood Cancer ; 60(7): E32-4, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23335404

ABSTRACT

Molecular detection of minimal residual disease (MRD) measured by quantitative reverse transcription-polymerase chain reaction using a four-marker panel in the bone marrow (BM) after only two treatment cycles of anti-GD2 immunotherapy was a strong independent outcome predictor among high-risk patients with stage 4 neuroblastoma in first remission. While 32 of 46 MRD-negative patients relapsed within 2 years from immunotherapy, only four had marrow relapse; in three of these four patients, MRD turned positive in the subsequent BM. We conclude that negative MRD in the post-cycle two BM was rarely associated with BM relapse, but it did not exclude recurrences at other sites.


Subject(s)
Bone Marrow Neoplasms/secondary , Neoplasm Recurrence, Local/pathology , Neuroblastoma/pathology , Bone Marrow/pathology , Child , Child, Preschool , Female , Humans , Immunotherapy , Infant , Male , Neoplasm Staging , Neoplasm, Residual/pathology , Neuroblastoma/therapy , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors
8.
JAMA Oncol ; 9(2): 242-250, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36547975

ABSTRACT

Importance: Among patients with high-risk relapsed metastatic neuroblastoma, oral ß-glucan adjuvant during GD2/GD3 ganglioside vaccine boost has stimulated IgG antibody response, which was associated with improved survival; however, the effectiveness of oral ß-glucan during the vaccine priming phase remains unproven. Objective: To isolate the adjuvant effect of oral ß-glucan on antibody response to GD2/GD3 ganglioside vaccine in patients with high-risk neuroblastoma. Design, Setting, and Participants: In this phase 2 randomized clinical trial, enrolled patients with high-risk neuroblastoma were randomized to 2 groups to receive the GD2/GD3 vaccine at a large cancer center in a major metropolitan area from October 2018 to September 2020. Data were analyzed from October 7, 2021, to February 28, 2022. Interventions: Eligible patients receiving GD2/GD3 vaccine were randomly assigned to group 1 (n = 54) to receive no ß-glucan or group 2 (n = 53) to receive an oral ß-glucan regimen during the first 5 weeks of vaccine priming. From week 6 onwards, all 107 patients received oral ß-glucan during vaccine boost for 1 year or until disease progression. Main Outcomes and Measures: Primary end point was comparison of anti-GD2 IgG1 response before vaccine injection 6 (week 32) in group 1 vs group 2. Seroconversion rate and the association of antibody titer with ß-glucan receptor dectin-1 single nucleotide polymorphism (SNP) rs3901533 were also assessed. Results: In all, 107 patients with high-risk neuroblastoma were randomized to the 2 groups: 54 patients (median [range] age, 5.2 [1.0-17.3] years; 28 [52%] male and 26 [48%] female) in group 1; and 53 patients (median [range] age, 6.2 [1.9-18.4] years; 25 [47%] male and 28 [53%] female) in group 2; both groups were also comparable in their first remission status at study entry (70% vs 70%). Adding oral ß-glucan during the first 5 weeks of vaccine priming elicited a higher anti-GD2 IgG1 antibody response in group 2 (1.80; 90% CI, 0.12-3.39; P = .08; planned type I error, 0.10). Anti-GD2 IgG1 titer of 230 ng/mL or greater by week 8 was associated with statistically favorable PFS. Antibody titer correlated significantly with dectin-1 SNP. The genotype frequency, seroconversion rates, and vaccine-related toxic effects were similar in the 2 groups. Conclusions and Relevance: This phase 2 randomized clinical trial found that adding oral ß-glucan during vaccine priming increased anti-GD2 IgG1 titer among genetic responders without added toxic effects. Because responder dectin-1 SNP was identical in the 2 randomized groups, no difference was detected in seroconversion rates. Alternative or additional adjuvants may be needed to enhance seroconversion. Trial Registration: ClinicalTrials.gov Identifier: NCT00911560.


Subject(s)
Cancer Vaccines , Gangliosides , Neuroblastoma , beta-Glucans , Child , Child, Preschool , Female , Humans , Male , Antibody Formation , beta-Glucans/pharmacology , beta-Glucans/therapeutic use , Gangliosides/immunology , Gangliosides/therapeutic use , Immunoglobulin G , Neuroblastoma/therapy , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use
9.
Nat Genet ; 55(6): 1022-1033, 2023 06.
Article in English | MEDLINE | ID: mdl-37169874

ABSTRACT

Patients with high-risk neuroblastoma generally present with widely metastatic disease and often relapse despite intensive therapy. As most studies to date focused on diagnosis-relapse pairs, our understanding of the genetic and clonal dynamics of metastatic spread and disease progression remain limited. Here, using genomic profiling of 470 sequential and spatially separated samples from 283 patients, we characterize subtype-specific genetic evolutionary trajectories from diagnosis through progression and end-stage metastatic disease. Clonal tracing timed disease initiation to embryogenesis. Continuous acquisition of structural variants at disease-defining loci (MYCN, TERT, MDM2-CDK4) followed by convergent evolution of mutations targeting shared pathways emerged as the predominant feature of progression. At diagnosis metastatic clones were already established at distant sites where they could stay dormant, only to cause relapses years later and spread via metastasis-to-metastasis and polyclonal seeding after therapy.


Subject(s)
Neoplasm Recurrence, Local , Neuroblastoma , Humans , Neoplasm Recurrence, Local/genetics , Neuroblastoma/genetics , Clonal Evolution , Mutation , Neoplasm Metastasis
10.
FASEB J ; 25(12): 4138-49, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21856782

ABSTRACT

The BMI1 gene is overexpressed in ≈ 90% of human neuroblastomas. However, little is known about the regulation of BMI1 expression. Using microarray and immunohistochemical analysis, we show that BMI1 expression correlated with MYCN levels in MYCN-amplified human neuroblastomas, and with MYC levels in the MYCN-nonamplified group. We further demonstrated that BMI1 is a direct target gene of MYCN/MYC in 3 neuroblastoma cell lines: BE (2)-C, LAN1, and SH-SY5Y. Overexpression of MYCN or MYC transactivated the BMI1 promoter and up-regulated BMI1 gene expression. shRNA-mediated knockdown of MYCN or MYC decreased BMI1 gene expression. Chromatin immunoprecipitation and point-mutation assays revealed that both MYCN and MYC bind to the E-box within the BMI1 promoter. Overexpression of BMI1, MYCN, and MYC independently increased both cell proliferation and tumor growth. Conversely, specific inhibition of BMI1, MYCN, and MYC decreased tumor cell proliferation and tumor growth. Interestingly, BMI1 suppression in MYCN/MYC-overexpressing cells resulted in significantly greater inhibition compared to that in mock-transduced and parental cells. Our results indicate that MYCN and MYC regulate BMI1 gene expression at the transcriptional level and that dysregulation of the BMI1 gene mediated by MYCN or MYC overexpression, confers increased cell proliferation during neuroblastoma genesis and tumor progression.


Subject(s)
Genes, myc , Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Infant , Mice , Mice, Inbred BALB C , Mice, Nude , N-Myc Proto-Oncogene Protein , Neoplasm Transplantation , Neuroblastoma/metabolism , Neuroblastoma/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/metabolism , Polycomb Repressive Complex 1 , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering/genetics , Repressor Proteins/metabolism , Transcriptional Activation , Transplantation, Heterologous
11.
Pediatr Blood Cancer ; 58(3): 469-71, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21416584

ABSTRACT

Current therapies for high-risk neuroblastoma (NB) necessitate the availability of several aliquots of autologous peripheral blood stem cells to reverse-associated myelosuppression. Priming with the CXCR4 inhibitor plerixafor plus G-CSF was associated with successful stem cell harvest in 5/7 heavily prior-treated patients with stage 4 NB who had previously failed G-CSF priming. Minimal residual disease was not detected in harvested cells from any patient despite the presence of disease in bone/bone marrow in 6/7. Hematopoietic reconstitution was achieved in all three patients receiving plerixafor-primed stem cells after myeloablative therapy. Plerixafor is an effective and safe agent for stem cell collection in patients with NB.


Subject(s)
Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Mobilization/methods , Heterocyclic Compounds/administration & dosage , Neuroblastoma/therapy , Receptors, CXCR4/antagonists & inhibitors , Adolescent , Adult , Benzylamines , Child , Child, Preschool , Cyclams , Drug Therapy, Combination , Female , Heterocyclic Compounds/adverse effects , Humans , Male , Neuroblastoma/secondary , Retrospective Studies
12.
JAMA ; 307(10): 1062-71, 2012 Mar 14.
Article in English | MEDLINE | ID: mdl-22416102

ABSTRACT

CONTEXT: Neuroblastoma is diagnosed over a wide age range from birth through young adulthood, and older age at diagnosis is associated with a decline in survivability. OBJECTIVE: To identify genetic mutations that are associated with age at diagnosis in patients with metastatic neuroblastoma. DESIGN, SETTING, AND PATIENTS: Whole genome sequencing was performed on DNA from diagnostic tumors and their matched germlines from 40 patients with metastatic neuroblastoma obtained between 1987 and 2009. Age groups at diagnosis included infants (0-<18 months), children (18 months-<12 years), and adolescents and young adults (≥12 years). To confirm the findings from this discovery cohort, validation testing using tumors from an additional 64 patients obtained between 1985 and 2009 also was performed. Formalin-fixed, paraffin-embedded tumor tissue was used for immunohistochemistry and fluorescence in situ hybridization. Telomere lengths were analyzed using whole genome sequencing data, quantitative polymerase chain reaction, and fluorescent in situ hybridization. MAIN OUTCOME MEASURE: Somatic recurrent mutations in tumors from patients with neuroblastoma correlated with the age at diagnosis and telomere length. RESULTS: In the discovery cohort (n = 40), mutations in the ATRX gene were identified in 100% (95% CI, 50%-100%) of tumors from patients in the adolescent and young adult group (5 of 5), in 17% (95% CI, 7%-36%) of tumors from children (5 of 29), and 0% (95% CI, 0%-40%) of tumors from infants (0 of 6). In the validation cohort (n = 64), mutations in the ATRX gene were identified in 33% (95% CI, 17%-54%) of tumors from patients in the adolescent and young adult group (9 of 27), in 16% (95% CI, 6%-35%) of tumors from children (4 of 25), and in 0% (95% CI, 0%-24%) of tumors from infants (0 of 12). In both cohorts (N = 104), mutations in the ATRX gene were identified in 44% (95% CI, 28%-62%) of tumors from patients in the adolescent and young adult group (14 of 32), in 17% (95% CI, 9%-29%) of tumors from children (9 of 54), and in 0% (95% CI, 0%-17%) of tumors from infants (0 of 18). ATRX mutations were associated with an absence of the ATRX protein in the nucleus and with long telomeres. CONCLUSION: ATRX mutations were associated with age at diagnosis in children and young adults with stage 4 neuroblastoma. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT00588068.


Subject(s)
DNA Helicases/genetics , Mutation , Neoplasm Metastasis/genetics , Neuroblastoma/genetics , Neuroblastoma/pathology , Nuclear Proteins/genetics , Adolescent , Age of Onset , Child , Child, Preschool , Cohort Studies , DNA Mutational Analysis , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Infant , Infant, Newborn , Male , Sequence Analysis, DNA , Telomere Homeostasis , X-linked Nuclear Protein , Young Adult
13.
Clin Cancer Res ; 28(8): 1614-1627, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35078859

ABSTRACT

PURPOSE: Therapy-related myelodysplastic syndrome and acute leukemias (t-MDS/AL) are a major cause of nonrelapse mortality among pediatric cancer survivors. Although the presence of clonal hematopoiesis (CH) in adult patients at cancer diagnosis has been implicated in t-MDS/AL, there is limited published literature describing t-MDS/AL development in children. EXPERIMENTAL DESIGN: We performed molecular characterization of 199 serial bone marrow samples from 52 patients treated for high-risk neuroblastoma, including 17 with t-MDS/AL (transformation), 14 with transient cytogenetic abnormalities (transient), and 21 without t-MDS/AL or cytogenetic alterations (neuroblastoma-treated control). We also evaluated for CH in a cohort of 657 pediatric patients with solid tumor. RESULTS: We detected at least one disease-defining alteration in all cases at t-MDS/AL diagnosis, most commonly TP53 mutations and KMT2A rearrangements, including involving two novel partner genes (PRDM10 and DDX6). Backtracking studies identified at least one t-MDS/AL-associated mutation in 13 of 17 patients at a median of 15 months before t-MDS/AL diagnosis (range, 1.3-32.4). In comparison, acquired mutations were infrequent in the transient and control groups (4/14 and 1/21, respectively). The relative risk for development of t-MDS/AL in the presence of an oncogenic mutation was 8.8 for transformation patients compared with transient. Unlike CH in adult oncology patients, TP53 mutations were only detectable after initiation of cancer therapy. Last, only 1% of pediatric patients with solid tumor evaluated had CH involving myeloid genes. CONCLUSIONS: These findings demonstrate the clinical relevance of identifying molecular abnormalities in predicting development of t-MDS/AL and should guide the formation of intervention protocols to prevent this complication in high-risk pediatric patients.


Subject(s)
Cancer Survivors , Leukemia, Myeloid, Acute , Neuroblastoma , Adult , Bone Marrow/pathology , Child , Clone Cells , Humans , Leukemia, Myeloid, Acute/genetics , Neuroblastoma/pathology
14.
Int J Cancer ; 129(8): 1953-62, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21154747

ABSTRACT

Checkpoint kinase inhibitors can enhance the cancer killing action of DNA-damaging chemotherapeutic agents by disrupting the S/G(2) cell cycle checkpoints. The in vitro and in vivo effects of the Chk1/2 inhibitor AZD7762 when combined with these agents were examined using neuroblastoma cell lines with known p53/MDM2/p14(ARF) genomic status. Four of four p53 mutant lines and three of five MDM2/p14(ARF) abnormal lines were defective in G(1) checkpoint, correlating with failure to induce endogenous p21 after treatment with DNA-damaging agents. In cytotoxicity assays, these G(1) checkpoint-defective lines were more resistant to DNA-damaging agents when compared to G(1) checkpoint intact lines, yet becoming more sensitive when AZD7762 was added. Moreover, AZD7762 abrogated DNA damage-induced S/G(2) checkpoint arrest both in vitro and in vivo. In xenograft models, a significant delay in tumor growth accompanied by histological evidence of increased apoptosis was observed, when AZD7762 was added to the DNA-damaging drug gemcitabine. These results suggest a therapeutic potential of combination therapy using checkpoint kinase inhibitor and chemotherapy to reverse or prevent drug resistance in treating neuroblastomas with defective G(1) checkpoints.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Neuroblastoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Thiophenes/pharmacology , Urea/analogs & derivatives , Animals , Cell Line, Tumor , Checkpoint Kinase 1 , Checkpoint Kinase 2 , DNA Damage , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Drug Resistance, Neoplasm , Drug Synergism , Female , Humans , Mice , Mice, Nude , Protein Serine-Threonine Kinases/metabolism , Urea/pharmacology , Xenograft Model Antitumor Assays , Gemcitabine
15.
Cancers (Basel) ; 13(24)2021 Dec 14.
Article in English | MEDLINE | ID: mdl-34944886

ABSTRACT

Beta glucans, complex polysaccharides, prime leukocyte dectin-1 and CR3-receptors and enhance anti-tumor cytotoxicity of complement-activating monoclonal antibodies. We conducted a phase I study (clinicaltrials.gov NCT00492167) to determine the safety of the combination of yeast-derived beta glucan (BG) and anti-GD2 murine monoclonal antibody 3F8 in patients with relapsed or refractory high-risk neuroblastoma. Patients received intravenous 3F8 (fixed dose of 10 mg/m2/day × 10 days) and oral BG (dose-escalated from 10-200 mg/kg/day × 17 days in cohorts of 3-6 patients each). Forty-four patients completed 141 cycles. One patient developed DLT: transient self-limiting hepatic transaminase elevation 5 days after starting BG (120 mg/kg/day). Overall, 1, 3, 12 and 24 evaluable patients had complete response, partial response, stable and progressive disease, respectively, at the end of treatment. Positive human anti-mouse antibody response and dectin-1 rs3901533 polymorphism were associated with better overall survival. BG dose level and serum BG levels did not correlate with response. Progression-free and overall survival at 2 years were 28% and 61%, respectively. BG lacked major toxicity. Treatment with 3F8 plus BG was associated with anti-neuroblastoma responses in patients with resistant disease. Although the maximal tolerated dose for yeast BG was not reached, considering the large volume of oral BG, we recommended 40 mg/kg/day as the phase II dose.

16.
J Clin Oncol ; 39(3): 215-226, 2021 01 20.
Article in English | MEDLINE | ID: mdl-33326254

ABSTRACT

PURPOSE: Anti-GD2 monoclonal antibody (mAb) has proven efficacy in high-risk neuroblastoma (HR-NB). A small phase I GD2/GD3 vaccine trial (n = 15) described long-term survival and a favorable safety profile among patients with a history of disease progression (PD). The kinetics of mounting antibody response to vaccine and its prognostic impact on survival are now investigated in a phase II study (ClinicalTrials.gov identifier: NCT00911560). PATIENTS AND METHODS: One hundred two patients with HR-NB who achieved remission after salvage therapies were enrolled in this trial. They received seven subcutaneous injections of GD2/GD3 vaccine spanning 1 year plus oral ß-glucan starting at week 6 after the third dose of vaccine. Serum anti-vaccine antibody titers were quantified by enzyme-linked immunosorbent assay. Single nucleotide polymorphisms (SNPs) were determined by quantitative polymerase chain reaction. Kaplan-Meier and landmark Cox Regression models were used for survival estimates. RESULTS: Patients had a history of one (63%), two (21%), or three to six (16%) episodes of PD. 82% of them progressed following anti-GD2 mAb (m3F8/dinutuximab/naxitamab) therapy. Vaccine-related toxicities were self-limited injection-associated local reactions and fever without any > grade 3 toxicities. The progression-free survival (PFS) was 32% ± 6%, and the overall survival (OS) was 71% ± 7% at 5 years. Serum anti-GD2 (immunoglobulin G1 [IgG1] and IgM) and anti-GD3 (IgG1) titers showed notable increases following the initiation of ß-glucan at week 6. There was an association between IgG1 titer and SNP rs3901533 of dectin-1, the ß-glucan receptor. Multivariable analyses showed that anti-GD2-IgG1 titer ≥ 150 ng/mL by week 8 was associated with favorable PFS and OS, while having prior episodes of PD and the time from last PD to vaccine were associated with PFS. CONCLUSION: GD2/GD3 vaccine plus ß-glucan elicited robust antibody responses in patients with HR-NB with prior PD. Higher anti-GD2-IgG1 titer was associated with improved survival.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Brain Neoplasms/drug therapy , Cancer Vaccines/therapeutic use , Gangliosides/immunology , Glioblastoma/drug therapy , Immunogenicity, Vaccine , Immunoglobulin G/blood , beta-Glucans/therapeutic use , Adjuvants, Immunologic/adverse effects , Biomarkers/blood , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Cancer Vaccines/adverse effects , Child , Child, Preschool , Disease Progression , Female , Glioblastoma/genetics , Glioblastoma/immunology , Glioblastoma/mortality , Humans , Infant , Lectins, C-Type/genetics , Male , Polymorphism, Single Nucleotide , Progression-Free Survival , Time Factors , beta-Glucans/adverse effects
17.
J Clin Oncol ; 38(36): 4283-4291, 2020 12 20.
Article in English | MEDLINE | ID: mdl-33119478

ABSTRACT

PURPOSE: Desmoplastic small round cell tumor (DSRCT), a rare sarcoma of adolescents/young adults primarily involving the peritoneum, has a long-term survival of < 20% despite aggressive multimodality treatment. B7H3 is expressed on DSRCT cell surface, providing a target for antibody-based immunotherapy. PATIENTS AND METHODS: In this phase I study, we evaluated the safety, pharmacokinetics, and biodistribution of intraperitoneal (IP) radioimmunotherapy (RIT) with the anti-B7H3 murine monoclonal antibody 131I-omburtamab in patients with DSRCT or other B7H3-expressing tumors involving the peritoneum. After thyroid blockade, patients received 131I-omburtamab as a single IP injection at escalated activities from 1.11 to 3.33/GBq/m2. A prior tracer dose of IP 74 MBq124I-omburtamab was used for radioimmuno-positron emission tomography imaging. Each injection was followed by IP saline infusion. RESULTS: Fifty-two patients (48, three, and one with DSRCT, peritoneal rhabdomyosarcoma, and Ewing sarcoma, respectively) received IP 131I-omburtamab administered on an outpatient basis. Maximum tolerated dose was not reached; there were no dose-limiting toxicities. Major related adverse events were transient: grade 4 neutropenia (n = 2 patients) and thrombocytopenia (n = 1), and grade 1 (10%) and grade 2 (52%) pain lasting < 2 hours related to saline infusion. Hypothyroidism was not observed, and antidrug antibody was elicited in 5%. Mean (± SD) projected peritoneal residence time was 22.4 ± 7.9 hours. Mean projected absorbed doses for 131I-omburtamab based on 124I-omburtamab dosimetry to normal organs were low and well within tolerable limits. More than 80% 131I remained protein bound in blood 66 hours after RIT. On the basis of peritoneal dose and feasibility for outpatient administration, the recommended phase II activity was established at 2.96 GBq/m2. Patients with DSRCT receiving standard whole-abdominal radiotherapy after RIT did not experience unexpected toxicity. CONCLUSION: IP RIT 131I-omburtamab was well tolerated with minimal toxicities. Radiation exposure to normal organs was low, making combination therapy with other anticancer therapies feasible.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/therapeutic use , Desmoplastic Small Round Cell Tumor/drug therapy , Iodine Radioisotopes/therapeutic use , Peritoneal Neoplasms/drug therapy , Radioimmunotherapy/methods , Adolescent , Adult , Antibodies, Monoclonal, Murine-Derived/pharmacology , Child , Child, Preschool , Female , Humans , Iodine Radioisotopes/pharmacology , Male , Young Adult
18.
Clin Cancer Res ; 14(21): 7020-7, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18980998

ABSTRACT

PURPOSE: Minimal residual disease (MRD) presents a significant hurdle to curing metastatic neuroblastoma. Biological therapies directed against MRD can improve outcome. Evaluating treatment efficacy requires MRD measurement, which serves as surrogate endpoint. Because of tumor heterogeneity, no single marker will likely be adequate. Genome-wide expression profiling can uncover potential MRD markers differentially expressed in tumors over normal marrow/blood. EXPERIMENTAL DESIGN: Gene expression array was carried out on 48 stage 4 tumors and 9 remission marrows using the Affymetrix U95 gene chip. Thirty-four genes with a tumor-to-marrow expression ratio higher than tyrosine hydroxylase were identified. Quantitative reverse transcription-PCR was done on all 34 genes to study the dynamic range of tumor cell detection and the expression of these genes in normal marrow/blood samples and in stage 4 neuroblastoma tumors. Top ranking markers were then tested for prognostic significance in the marrows of stage 4 patients collected from the same treatment protocol after two cycles of immunotherapy. RESULTS: Based on sensitivity assays, 8 top-ranking markers were identified: CCND1, CRMP1, DDC, GABRB3, ISL1, KIF1A, PHOX2B, and TACC2. They were abundantly expressed in stage IV neuroblastoma tumors (n=20) and had low to no detection in normal marrow/blood samples (n=20). Moreover, expression of CCND1, DDC, GABRB3, ISL1, KIF1A, and PHOX2B in 116 marrows sampled after two treatment cycles was highly prognostic of progression-free and overall survival (P<0.001). CONCLUSIONS: Marker discovery based on differential gene expression profiling, stringent sensitivity and specificity assays, and well-annotated patient samples can rapidly prioritize and identify potential MRD markers of neuroblastoma.


Subject(s)
Gene Expression Profiling , Neoplasm, Residual/genetics , Neuroblastoma/genetics , Biomarkers, Tumor/genetics , Bone Marrow , Humans , Infant , Microarray Analysis , Prognosis , Sensitivity and Specificity
19.
Neoplasia ; 21(7): 689-701, 2019 07.
Article in English | MEDLINE | ID: mdl-31128432

ABSTRACT

Telomeres play important roles in genome stability and cell proliferation. High risk neuroblastoma (HRNB), an aggressive childhood cancer, is especially reliant on telomere maintenance. Three recurrent genetic aberrations in HRNB (MYCN amplification, TERT re-arrangements, and ATRX mutations) are mutually exclusive and each capable of promoting telomere maintenance mechanisms (i.e., through telomerase or ALT). We analyzed a panel of 5 representative HRNB cell lines and 30 HRNB surgical samples using assays that assess average telomere lengths, length distribution patterns, single-stranded DNA on the G- and C-strand, as well as extra-chromosomal circular telomeres. Our analysis pointed to substantial and variable degrees of telomere DNA damage in HRNB, including pervasive oxidative lesions. Moreover, unlike other cancers, neuroblastoma consistently harbored high levels of C-strand ssDNA overhangs and t-circles, which are consistent with active "telomere trimming". This feature is observed in both telomerase- and ALT-positive tumors and irrespective of telomere length distribution. Moreover, evidence for telomere trimming was detected in normal neural tissues, raising the possibility that TMMs in HRNB evolved in the face of a canonical developmental program of telomere shortening. Telomere trimming by itself appears to distinguish neuroectodermal derived tumors from other human cancers, a distinguishing characteristic with both biologic and therapeutic implications.


Subject(s)
DNA Damage/genetics , Neuroblastoma/genetics , Telomere Homeostasis/genetics , Telomere/genetics , Cell Proliferation/genetics , Female , Genomic Instability/genetics , HeLa Cells , Humans , Male , Mutation/genetics , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/pathology , Telomerase/genetics , X-linked Nuclear Protein/genetics
20.
Clin Cancer Res ; 13(23): 6978-83, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18056173

ABSTRACT

PURPOSE: Targeting subclinical disease in the bone marrow is particularly relevant in metastatic Ewing family tumors (EFT) where cure is difficult. Genome-wide expression arrays can uncover novel genes differentially expressed in tumors over normal marrow/blood, which may have potentials as markers of subclinical disease. EXPERIMENTAL DESIGN: Gene expression array data were obtained on 28 EFT tumors using the Affymetrix U133 gene chip and compared with 10 normal blood samples. Ten genes with high tumor to blood ratios were identified. Quantitative reverse transcription-PCR was done to study (a) the dynamic range of detection of rare tumor cells, (b) the gene expression in normal blood/marrow samples, (c) the gene expression among EFT tumors, and (d) the detection and prognostic impact of marker positivity in histology-negative diagnostic marrows of EFT patients. RESULTS: Five of 10 genes (i.e., six-transmembrane epithelial antigen of the prostate 1 [STEAP1], cyclin D1 [CCND1], NKX2-2 transcription factor [NKX2-2], plakophilin 1 [PKP1], and transmembrane protein 47 [TMEM47]) were chosen for further analyses based on their steep linear dynamic range in detecting tumor cells seeded in normal mononuclear cells and on their homogeneous expression among EFT tumors. Prognostic effect was evaluated in 35 histology-negative diagnostic marrows. Marker negativity of STEAP1, CCND1, or NKX2-2, as well as three markers in combination, was strongly correlated with patient survival as well as survival without new metastases. CONCLUSIONS: This gene expression array-based approach identified novel markers that may be informative at diagnosis for risk group assessment. Their clinical utility needs to be tested in large patient cohorts.


Subject(s)
Biomarkers, Tumor/genetics , Sarcoma, Ewing/genetics , Adolescent , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/blood , Bone Marrow/pathology , Cell Line, Tumor , Cohort Studies , Female , Gene Expression Profiling , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Kaplan-Meier Estimate , Male , Nuclear Proteins , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/biosynthesis , Oncogene Proteins, Fusion/blood , Oncogene Proteins, Fusion/genetics , Prognosis , Proto-Oncogene Protein c-fli-1/biosynthesis , Proto-Oncogene Protein c-fli-1/blood , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction/methods , Sarcoma, Ewing/blood , Sensitivity and Specificity , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL