Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunother Cancer ; 8(1)2020 03.
Article in English | MEDLINE | ID: mdl-32169872

ABSTRACT

PURPOSE: Patients with cancer receiving tumor-reactive humanized monoclonal antibody (mAb) therapy can develop a human antihuman antibody (HAHA) response against the therapeutic mAb. We evaluated for HAHA in patients with neuroblastoma treated in a phase I study of humanized anti-GD2 mAb (immunoglobulin (Ig)G1 isotype), hu14.18K322A (NCT00743496). The pretreatment sera (collected prior to mAb treatment) from 9 of 38 patients contained antitherapeutic antibodies, even though they had no prior mAb exposure. We sought to characterize these pre-existing antitherapeutic antibodies (PATA). EXPERIMENTAL DESIGN: The PATA+ pretreatment samples were characterized via ELISA; clinical associations with PATA status were evaluated. RESULTS: Pretreatment sera from eight of nine PATA+ patients also bound rituximab and demonstrated preferential ELISA reactivity against the Fc portions of hu14.18K322A and rituximab as compared with the Fab portions of these mAbs. These PATA+ sera also recognized dinutuximab (human IgG1 isotype) and mouse IgG2a isotype mAbs, but not a mouse IgG1 isotype or the fully human panitumumab (IgG2 isotype) mAb. Of the 38 treated patients, only 4 patients (all in the PATA+ cohort) demonstrated no disease progression for >2.5 years without receiving further therapy (p=0.002). CONCLUSIONS: This study demonstrates an association between clinical outcome and the presence of PATA against determinant(s) on the Fc component of the therapeutic mAb, suggesting that the PATA may be playing a role in augmenting mAb-based antitumor effects. Further analyses for the presence of PATA in a larger cohort of patients with relapsed neuroblastoma, analyses of their clinical correlates, identification of their immunological targets, and potential antitumor mechanisms are warranted.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Female , Humans , Mice , Neoplasm Recurrence, Local , Neuroblastoma , Treatment Outcome
2.
Clin Cancer Res ; 25(20): 6044-6051, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31358541

ABSTRACT

PURPOSE: Combining anti-GD2 (disialoganglioside) mAb with GM-CSF, IL2, and isotretinoin is now FDA-approved for high-risk neuroblastoma minimal residual disease (MRD) therapy. The humanized anti-GD2 antibody conjugated to IL2 (hu14.18-IL2) has clinical activity in neuroblastoma and is more effective in neuroblastoma-bearing mice than antibody and cytokine given separately. We therefore evaluated the safety, tolerability, and antitumor activity of hu14.18-IL2 given with GM-CSF and isotretinoin in a schedule similar to standard MRD therapy. PATIENTS AND METHODS: Hu14.18-IL2 was given at the recommended phase II dose of 12 mg/m2/day on days 4-6 of a 28-day cycle with GM-CSF (250 mg/m2/dose, days 1-2 and 8-14) and isotretinoin (160 mg/m2/day, days 11-25). Tolerability was determined on the basis of the number of unacceptable toxicities observed. Response was evaluated separately for patients with disease measurable by standard radiologic criteria (stratum 1), and for patients with disease evaluable only by I123-metaiodobenzylguanidine (I123-MIBG) scan and/or bone marrow histology (stratum 2). RESULTS: Fifty-two patients with recurrent or refractory neuroblastoma were enrolled; 51 were evaluable for toxicity and 45 were evaluable for response. Four patients had unacceptable toxicities, well below the protocol-defined rule for tolerability. Other grade 3 and 4 nonhematologic toxicities were expected and reversible. No responses were seen in stratum 1 (n = 14). In stratum 2 (n = 31), 5 objective responses were confirmed by central review (3 complete, 2 partial). CONCLUSIONS: Hu14.18-IL2 given in combination with GM-CSF and isotretinoin is safe and tolerable. Patients with MIBG and/or bone marrow-only disease had a 16.1% response rate, confirming activity of the combination.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Interleukin-2/administration & dosage , Isotretinoin/administration & dosage , Neoplasm Recurrence, Local/drug therapy , Neuroblastoma/drug therapy , Adolescent , Adult , Antibodies, Monoclonal/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Child, Preschool , Drug Administration Schedule , Drug Resistance, Neoplasm , Feasibility Studies , Female , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Humans , Interleukin-2/adverse effects , Isotretinoin/adverse effects , Male , Neoplasm Recurrence, Local/pathology , Neuroblastoma/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Treatment Outcome , Young Adult
3.
N Engl J Med ; 363(14): 1324-34, 2010 Sep 30.
Article in English | MEDLINE | ID: mdl-20879881

ABSTRACT

BACKGROUND: Preclinical and preliminary clinical data indicate that ch14.18, a monoclonal antibody against the tumor-associated disialoganglioside GD2, has activity against neuroblastoma and that such activity is enhanced when ch14.18 is combined with granulocyte-macrophage colony-stimulating factor (GM-CSF) or interleukin-2. We conducted a study to determine whether adding ch14.18, GM-CSF, and interleukin-2 to standard isotretinoin therapy after intensive multimodal therapy would improve outcomes in high-risk neuroblastoma. METHODS: Patients with high-risk neuroblastoma who had a response to induction therapy and stem-cell transplantation were randomly assigned, in a 1:1 ratio, to receive standard therapy (six cycles of isotretinoin) or immunotherapy (six cycles of isotretinoin and five concomitant cycles of ch14.18 in combination with alternating GM-CSF and interleukin-2). Event-free survival and overall survival were compared between the immunotherapy group and the standard-therapy group, on an intention-to-treat basis. RESULTS: A total of 226 eligible patients were randomly assigned to a treatment group. In the immunotherapy group, a total of 52% of patients had pain of grade 3, 4, or 5, and 23% and 25% of patients had capillary leak syndrome and hypersensitivity reactions, respectively. With 61% of the number of expected events observed, the study met the criteria for early stopping owing to efficacy. The median duration of follow-up was 2.1 years. Immunotherapy was superior to standard therapy with regard to rates of event-free survival (66±5% vs. 46±5% at 2 years, P=0.01) and overall survival (86±4% vs. 75±5% at 2 years, P=0.02 without adjustment for interim analyses). CONCLUSIONS: Immunotherapy with ch14.18, GM-CSF, and interleukin-2 was associated with a significantly improved outcome as compared with standard therapy in patients with high-risk neuroblastoma. (Funded by the National Institutes of Health and the Food and Drug Administration; ClinicalTrials.gov number, NCT00026312.)


Subject(s)
Antibodies, Monoclonal/therapeutic use , Gangliosides/immunology , Immunotherapy , Neuroblastoma/therapy , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/therapeutic use , Child , Child, Preschool , Combined Modality Therapy , Drug Therapy, Combination , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Immunotherapy/adverse effects , Infant , Intention to Treat Analysis , Interleukin-2/therapeutic use , Isotretinoin/therapeutic use , Neuroblastoma/drug therapy , Neuroblastoma/mortality , Stem Cell Transplantation , Survival Analysis
4.
Cancer Biol Ther ; 3(6): 551-8, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15044850

ABSTRACT

Prostate-Specific Membrane Antigen (PSMA) is a glutamate carboxypeptidase II that is highly expressed by both normal and malignant prostate epithelial cells and by the neovasculature of many tumor types but is not expressed by endothelial cells in normal tissue. PSMA possesses the hydrolytic properties of an N-acetylated alpha-linked acidic dipeptidase (NAALADase) and also functions as a pteroyl poly-gamma-glutamyl carboxypeptidase (i.e., folate hydrolase). Therefore, PSMA can be targeted for activation of peptide-based prodrugs within the extracellular fluid of prostate cancers. In this study, methotrexate-based peptide analogs were evaluated to identify PSMA selective substrates that are also stable to nonspecific hydrolysis in human and mouse plasma. These methotrexate analogs were also characterized for in vitro toxicity against PSMA and nonPSMA producing human cancer cell lines. Analogs containing gamma-linked glutamate residues were most efficiently hydrolyzed by PSMA, but were unstable in plasma. Analogs containing both alpha- and gamma-linked acidic amino acids were less efficiently hydrolyzed by PSMA but were most stable in plasma. Analogs were 5-10 fold more selectively toxic in vitro in the presence of active PSMA. These studies have identified PSMA selective, plasma stable peptide substrates that can be incorporated into prodrugs targeted for activation by PSMA within prostate cancer sites.


Subject(s)
Antigens, Surface/metabolism , Glutamate Carboxypeptidase II/metabolism , Methotrexate/metabolism , Peptide Fragments/metabolism , Prodrugs , Prostatic Neoplasms/metabolism , Animals , Antigens, Surface/genetics , Aspartic Acid/metabolism , Blotting, Western , Extracellular Fluid , Glutamate Carboxypeptidase II/genetics , Glutamic Acid/metabolism , Humans , Immunoglobulin gamma-Chains/genetics , Immunoglobulin kappa-Chains/genetics , Male , Mice , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Recombinant Fusion Proteins/metabolism , Substrate Specificity , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...