Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Front Med (Lausanne) ; 7: 198, 2020.
Article in English | MEDLINE | ID: mdl-32582722

ABSTRACT

Background: Merkel cell carcinoma (MCC) is a rare neuroendocrine skin cancer. Prior to the advent of immunotherapy, treatment options were limited. In our study, we evaluate the impact of tumor cell PD-L1 expression and tumor immune microenvironment on survival in MCC patients who were not treated with immune checkpoint inhibitors. Methods: Clinical data and tissue samples were collected from 78 patients with confirmed MCC treated at Dana-Farber Cancer Institute. Specimens were analyzed for the distribution of PD-L1 by immunohistochemistry staining (IHC) and standardized analysis. Results were correlated with survival data. Results: In this study, membrane and cytoplasmic MCC tumor cell staining for PD-L1 was detected in 22.4% (15 of 67) of cases and PD-L1 staining of intratumoral microvessels and PD-L1 positive immune cells at the infiltrative margins of the tumor in 92.5% (62 of 67) of cases. In patients untreated with immune checkpoint inhibitors, median overall survival was not different for patients based on PD-L1 expression (PD-L1+ 64 months vs. PD-L1- not reached; HR = 1.26, 95% CI: 0.46-3.45; p = 0.60). Conclusion: PD-L1 expression is frequently detected in MCC tumor cells and tumor microenvironment. PD-L1 expression did not affect prognosis in this cohort that had not received PD-1/L1 blockade.

2.
PLoS One ; 10(6): e0129954, 2015.
Article in English | MEDLINE | ID: mdl-26091536

ABSTRACT

BACKGROUND: Interleukin (IL)-12 is a pro-inflammatory cytokine that mediates T-helper type 1 responses and cytotoxic T-cell activation, contributing to its utility as anti-cancer agent. Systemic administration of IL-12 often results in unacceptable toxicity; therefore, strategies to direct delivery of IL-12 to tumors are under investigation. The objective of this study was to assist the preclinical development of NHS-IL12, an immunocytokine consisting of an antibody, which targets necrotic tumor regions, linked to IL-12. Specifically this study sought to evaluate the safety, serum pharmacokinetics, anti-tumor activity, and immune modulation of NHS-IL12 in dogs with naturally occurring cancers. METHODOLOGY/PRINCIPAL FINDINGS: A rapid dose-escalation study of NHS-IL12 administered subcutaneously to dogs with melanoma was conducted through the Comparative Oncology Trials Consortium (COTC). Eleven dogs were enrolled in four dose-escalation cohorts; thereafter, an additional seven dogs were treated at the defined tolerable dose of 0.8 mg/m2. The expanded cohort at this fixed dose (ten dogs in total) was accrued for further pharmacokinetics and pharmacodynamics assessment. NHS-IL12 levels, serum cytokine concentrations, and peripheral blood mononuclear cell characterization (post-treatment) and draining lymph node immune profiling, and tumor biopsies (pre- and post-treatment) were collected. Adverse events included thrombocytopenia, liver enzymopathies, fever, and vasculitis. Correlation between interferon (IFN)-γ induction, adverse events, and NHS-IL12 exposure (maximum concentration and area under the concentration-time curve) were dose-dependent. Serum IL-10 levels and intratumoral CD8+ populations increased after treatment. Partial responses, according to Response Evaluation Criteria in Solid Tumors (RECIST) criteria, were observed in two dogs treated with NHS-IL12 0.8 mg/m2 and 1.6 mg/m2. CONCLUSIONS/SIGNIFICANCE: NHS-IL12 was administered safely to dogs with melanoma and both immunologic and clinical activity was observed. This study successfully defined a narrow therapeutic window for systemic delivery of NHS-IL12 via the subcutaneous route. Results will inform the design and implementation of first-in-human clinical trials of NHS-IL12 in cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Dog Diseases/drug therapy , Dog Diseases/pathology , Immunoglobulin G/pharmacology , Immunologic Factors/pharmacology , Interleukin-12/pharmacology , Melanoma/veterinary , Recombinant Fusion Proteins/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/blood , Dog Diseases/blood , Dogs , Female , Immunoglobulin G/administration & dosage , Immunologic Factors/administration & dosage , Immunologic Factors/pharmacokinetics , Immunophenotyping , Infusions, Subcutaneous , Interleukin-12/administration & dosage , Interleukin-12/pharmacokinetics , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Treatment Outcome
3.
Clin Cancer Res ; 17(11): 3673-85, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21531812

ABSTRACT

PURPOSE: The goal of the study was to engineer a form of interleukin 2 (IL-2) that, when delivered as a tumor-specific antibody fusion protein, retains the ability to stimulate an antitumor immune response via interaction with the high-affinity IL-2 receptor but has lower toxicity because of the reduced activation of the intermediate-affinity IL-2 receptor. EXPERIMENTAL DESIGN: We investigated changes in the proposed toxin motif of IL-2 by introducing a D20T mutation that has little effect on the activity of free IL-2. We expressed this IL-2 variant as a fusion protein with an antibody (NHS76) that targets the necrotic core of tumors and characterized this molecule (NHS-IL2LT) in vitro and in vivo. RESULTS: NHS-IL2LT was shown to have near normal biological activity in vitro by using T-cell lines expressing the high-affinity IL-2 receptor, but little or no activity by using cell lines expressing only the intermediate IL-2 receptor. Relative to the control antibody fusion protein containing wild-type IL-2, NHS-IL2LT retained antitumor activity against established neuroblastoma and non-small cell lung cancer metastases in syngeneic mouse tumor models but was much better tolerated in immune-competent mice and in cynomolgus monkeys. CONCLUSIONS: The qualities of low toxicity and single-agent efficacy shown suggest that NHS-IL2LT is a good candidate for therapeutic approaches combining standard cytotoxic and immune therapies. In fact, this molecule (also known as Selectikine or EMD 521873) is currently in phase I clinical trial.


Subject(s)
Interleukin-2/metabolism , Animals , Antibodies, Monoclonal/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Cell Line, Tumor , Clinical Trials as Topic , Immunoglobulin Variable Region/metabolism , Interleukin-2/chemistry , Interleukin-2/genetics , Interleukin-2/immunology , Macaca fascicularis , Mice , Multiple Myeloma/immunology , Mutation , Neuroblastoma/immunology , Protein Engineering , Receptors, Interleukin-2/metabolism , Recombinant Fusion Proteins/metabolism
4.
Cancer Immunol Immunother ; 56(4): 447-57, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16874486

ABSTRACT

IL-12 is a cytokine which showed anti-tumor effects in clinical trials, but also produced serious toxicity. We describe a fusion protein, huBC1-IL12, designed to achieve an improved therapeutic index by specifically targeting IL-12 to tumor and tumor vasculature. huBC-1 is a humanized antibody that targets a cryptic sequence of the human ED-B-containing fibronectin isoform, B-FN, present in the subendothelial extracellular matrix of most aggressive tumors. B-FN is oncofetal and angiogenesis-associated, and is undetectable in most normal adult tissues. The original murine BC-1 antibody has been used successfully for immunoscintigraphy to image brain tumor mass in glioblastoma patients. In huBC1-IL12, each of the IgG heavy chains is genetically fused to the N-terminus of the IL-12 p35 subunit, which in turn is disulfide-bonded to the p40 subunit, resulting in a hexameric molecule of MW of approximately 300 kDa. Since human IL-12 has no biological activity in mice, we produced huBC1-muIL12 as a surrogate molecule for animal tumor models. Despite the relatively poor PK profile of this molecule in mice and the apparent drawbacks of xenogeneic models in SCID mice, which lack T and B cells, one cycle of treatment with huBC1-muIL12 was efficacious in the PC3mm2, A431, and HT29 subcutaneous tumor models and PC3mm2 lung metastasis model. This molecule also was found to have surprisingly low toxicity in immunocompetent mice. A fusion protein that contains human IL-12 (huBC1-huIL12), which is a suitable molecule for investigation as a therapeutic, has also been produced. This protein has been shown to have a longer serum half-life than huBC1-muIL12 in mice, and retains both antigen binding and IL-12 activity in in vitro assays.


Subject(s)
Fibronectins/immunology , Interleukin-12/therapeutic use , Neoplasms, Experimental/therapy , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/therapeutic use , Animals , Electrophoresis, Polyacrylamide Gel , Female , Humans , Immunoglobulin G , Immunoglobulin Heavy Chains , Immunohistochemistry , Interleukin-12/immunology , Interleukin-12/pharmacokinetics , Male , Mice , Neoplasms, Experimental/immunology , Protein Isoforms/immunology , Protein Isoforms/pharmacokinetics , Protein Isoforms/therapeutic use , Recombinant Fusion Proteins/immunology
5.
Protein Eng Des Sel ; 18(3): 111-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15820978

ABSTRACT

Erythropoietin (Epo) is a cytokine that controls the production of red blood cells (RBCs). Epo acts continuously on RBC precursors to prevent apoptosis, so it is important to maintain high levels of Epo activity when treating anemic patients. We describe here modified human Epo [Epo(NDS)] with mutations His32Gly, Cys33Pro, Trp88Cys and Pro90Ala that result in the rearrangement of the disulfide bonding pattern from Cys29-Cys33 to Cys29-Cys88 and that, in the context of an Fc-Epo(NDS) fusion protein, lead to significantly improved properties. Fc-Epo was secreted from NS/0 myeloma cells as about 35% high molecular weight aggregates, was unstable upon removal of N-linked oligosaccharides and showed poor pharmacokinetics and little efficacy in mice. In contrast, a corresponding Fc-Epo(NDS) was secreted almost exclusively as a unit dimer, was relatively stable to removal of N-linked oligosaccharides, had much improved pharmacokinetic properties and had a significantly improved effect on RBC production. These results indicate that rearrangement of the disulfide bonding pattern in a therapeutic protein can have a significant effect on pharmacokinetics and, potentially, the dosing schedule of a protein drug.


Subject(s)
Erythropoietin/chemistry , Erythropoietin/pharmacokinetics , Protein Engineering/methods , Receptors, Fc/chemistry , Amino Acid Sequence , Animals , Apoptosis , Cattle , Cell Line, Tumor , Chromatography, High Pressure Liquid , Codon , Computer Simulation , Cysteine/chemistry , Dimerization , Disulfides/chemistry , Erythropoietin/genetics , Glycosylation , Humans , Kinetics , Mice , Models, Molecular , Molecular Sequence Data , Mutation , Oligosaccharides/chemistry , Peptides/chemistry , Protein Conformation , Protein Folding , Protein Structure, Tertiary , Rats , Receptors, Fc/genetics , Recombinant Fusion Proteins/chemistry , Sequence Homology, Amino Acid , Time Factors , Transfection , Trypsin/chemistry
6.
Protein Eng Des Sel ; 18(1): 1-10, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15790575

ABSTRACT

Leptin plays a central role in the homeostasis of body weight through its regulatory effects on appetite and energy expenditure, yet in trials as a therapeutic agent for the treatment of obesity in humans it has been disappointing. The poor clinical efficacy of leptin results from its short circulating half-life, low potency and poor solubility, necessitating large and frequent doses to obtain even modest clinical benefit. Engineered Fc-leptin immunofusins, consisting of the Fc fragment of an immunoglobulin gamma chain followed by leptin, exhibit improved pharmacological properties with very consistent and potent biological activities. Furthermore, in extending the circulating half-life of the protein in vivo from a few minutes for leptin to many hours for Fc-leptin, these proteins have the potential to reduce drastically the dosage and frequency of administration required to obtain clinical benefit. The results of this study show that the engineered leptin immunofusins described here have significantly enhanced pharmacological properties in comparison with the recombinant leptin that was used in clinical trials. As such, they could represent an important step towards a therapeutically superior form of leptin if the disappointing performance of leptin in early clinical trials was due to its poor pharmacological properties rather than any conceptual weakness in the strategy of using leptin for the treatment of obesity and its related disorders.


Subject(s)
Leptin/genetics , Leptin/therapeutic use , Obesity/drug therapy , Protein Engineering , Animals , Base Sequence , Blotting, Western , Cloning, Molecular , DNA Primers , Drug Administration Schedule , Humans , Leptin/administration & dosage , Leptin/pharmacokinetics , Mice , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...