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.
Mol Cancer Ther ; 21(10): 1510-1523, 2022 10 07.
Article in English | MEDLINE | ID: mdl-35876604

ABSTRACT

Pet dogs with naturally occurring cancers play an important role in studies of cancer biology and drug development. We assessed tolerability, efficacy, and pharmacokinetic/pharmacodynamic relationships with a first-in-class small molecule inhibitor of valosin-containing protein (VCP/p97), CB-5339, administered to 24 tumor-bearing pet dogs. Tumor types assessed included solid malignancies, lymphomas, and multiple myeloma. Through a stepwise dose and schedule escalation schema, we determined the maximum tolerated dose to be 7.5 mg/kg when administered orally on a 4 days on, 3 days off schedule per week for 3 consecutive weeks. Adverse events were minimal and mainly related to the gastrointestinal system. Pharmacokinetic/pharmacodynamic data suggest a relationship between exposure and modulation of targets related to induction of the unfolded protein response, but not to tolerability of the agent. An efficacy signal was detected in 33% (2/6) of dogs with multiple myeloma, consistent with a mechanism of action relating to induction of proteotoxic stress in a tumor type with abundant protein production. Clinical trials of CB-5339 in humans with acute myelogenous leukemia and multiple myeloma are ongoing.


Subject(s)
Antineoplastic Agents , Lymphoma , Multiple Myeloma , Valosin Containing Protein , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Dogs , Enzyme Inhibitors/therapeutic use , Lymphoma/drug therapy , Lymphoma/pathology , Lymphoma/veterinary , Maximum Tolerated Dose , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Multiple Myeloma/veterinary , Unfolded Protein Response , Valosin Containing Protein/antagonists & inhibitors
2.
Infect Dis Ther ; 10(4): 2765-2776, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34435336

ABSTRACT

INTRODUCTION: There is an increasing body of evidence surrounding the importance of a T cell-mediated response to SARS-CoV-2 infection and after COVID-19 vaccination. In this internal feasibility study, we evaluated both the total antibody (IgA, IgM, and IgG) and T cell responses in a cohort of COVID-19 convalescents and vaccinated individuals. METHODS: Whole blood specimens were collected weekly from 12 subjects at different time points within/after the COVID-19 mRNA vaccination regimen, and from 4 PCR-confirmed convalescent donors to measure durability of humoral and cell-mediated immune response. T cell and antibody responses were evaluated via the QuantiFERON SARS-CoV-2 research use only (QFN SARS-CoV-2) assay which is an interferon gamma release assay (IGRA) and QIAreach Anti-SARS-CoV-2 total (Anti-CoV-2) test, respectively. RESULTS: In a cohort of recently vaccinated individuals, subjects demonstrated robust total antibody and CD4+/CD8+ T cell response to SARS-CoV-2 mRNA vaccines when followed for 2 months post-2nd dose. In most individuals, T cell response declined between the 1st and 2nd doses suggesting a need for a booster or the completion of the 2-dose vaccine series. In a group of convalescent donors tested with QFN SARS-CoV-2 and Anti-CoV-2 tests, all patients had an antibody and T cell response up to 1 year after natural infection. CONCLUSION: This small feasibility study demonstrates that the QFN-SARS-CoV-2 test is able to identify CD4+ and CD8+ T cell-mediated responses in SARS-CoV-2-vaccinated subjects and those recovered from COVID-19, alongside a qualitative antibody response detectable via the QIAreach Anti-CoV2 test.

3.
Clin Cancer Res ; 22(4): 1000-10, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26446940

ABSTRACT

PURPOSE: To support clinical pharmacodynamic evaluation of the Smac mimetic TL32711 (birinapant) and other apoptosis-targeting drugs, we describe the development, validation, and application of novel immunoassays for 15 cytosolic and membrane-associated proteins indicative of the induction, onset, and commitment to apoptosis in human tumors. EXPERIMENTAL DESIGN: The multiplex immunoassays were constructed on the Luminex platform with apoptosis biomarkers grouped into three panels. Panel 1 contains Bak, Bax, total caspase-3, total lamin-B (intact and 45 kDa fragment), and Smac; panel 2 contains Bad, Bax-Bcl-2 heterodimer, Bcl-xL, Bim, and Mcl1; and panel 3 contains active (cleaved) caspase-3, Bcl-xL-Bak heterodimer, Mcl1-Bak heterodimer, pS99-Bad, and survivin. Antibody specificity was confirmed by immunoprecipitation and Western blot analysis. RESULTS: Two laboratories analytically validated the multiplex immunoassays for application with core-needle biopsy samples processed to control preanalytical variables; the biologic variability for each biomarker was estimated from xenograft measurements. Studies of TL32711 in xenograft models confirmed a dose-dependent increase in activated caspase-3 6 hours after dosing and provided assay fit-for-purpose confirmation. Coincident changes in cytosolic lamin-B and subsequent changes in Bcl-xL provided correlative evidence of caspase-3 activation. The validated assay is suitable for use with clinical specimens; 14 of 15 biomarkers were quantifiable in patient core-needle biopsies. CONCLUSIONS: The validated multiplex immunoassays developed for this study provided proof of mechanism data for TL32711 and are suitable for quantifying apoptotic biomarkers in clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Dipeptides/pharmacology , Indoles/pharmacology , Animals , Cell Line, Tumor , Female , Humans , Immunoassay , Intracellular Signaling Peptides and Proteins/chemistry , Mice, Nude , Mitochondrial Proteins/chemistry , Molecular Mimicry , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
PLoS One ; 6(5): e19605, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21573184

ABSTRACT

Evidence is presented for the nuclear presence of a functional heteromeric complex of epidermal growth factor (EGFR), Src and the Signal Transducer and Activator of Transcription (Stat)3 proteins in pancreatic cancer cells. Stat3 remains nuclear and associated with Src or EGFR, respectively, upon the siRNA knockdown of EGFR or Src, demonstrating the resistance of the complex to the modulation of EGFR or Src alone. Significantly, chromatin immunoprecipitation (ChIP) analyses reveal the nuclear EGFR, Src and Stat3 complex is bound to the c-Myc promoter. The siRNA knockdown of EGFR or Src, or the pharmacological inhibition of Stat3 activity only marginally suppressed c-Myc expression. By contrast, the concurrent modulation of Stat3 and EGFR, or Stat3 and Src, or EGFR and Src strongly suppressed c-Myc expression, demonstrating that the novel nuclear heteromeric complex intricately regulates the c-Myc gene. The prevalence of the transcriptionally functional EGFR, Src, and Stat3 nuclear complex provides an additional and novel mechanism for supporting the pancreatic cancer phenotype and explains in part the insensitivity of pancreatic cancer cells to the inhibition of EGFR, Src or Stat3 alone.


Subject(s)
Cell Nucleus/metabolism , ErbB Receptors/metabolism , Multiprotein Complexes/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins pp60(c-src)/metabolism , STAT3 Transcription Factor/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Endocytosis , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Immunoprecipitation , Light , Microscopy, Confocal , Nanoparticles , Nanotechnology , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Particle Size , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Scattering, Radiation
5.
J Biol Chem ; 285(46): 35855-65, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20807764

ABSTRACT

Given the role of constitutively active Signal Transducer and Activator of Transcription (Stat) 3 in human tumors, Stat3 inhibitors would be useful as novel therapeutics and as tools for probing Stat3-mediated tumor processes. We herein report that a 28-mer peptide, SPI, derived from the Stat3 SH2 domain, replicates Stat3 biochemical properties. Studies show SPI and Stat3 (or Stat3 SH2 domain) bind with similar affinities to known Stat3-binding phosphotyrosine (pY) peptide motifs, including those of the epidermal growth factor receptor (EGFR) and the high-affinity, IL-6R/gp130-derived pY-peptide, GpYLPQTV-NH(2). Consequently, SPI functions as a potent and selective inhibitor of Stat3 SH2 domain:pTyr interactions and disrupts the binding of Stat3 to the IL-6R/gp130 peptide, GpYLPQTV-NH(2). Fluorescence imaging and immunofluorescence staining/laser-scanning confocal microscopy show SPI is cell membrane-permeable, associates with the cytoplasmic tail of EGFR in NIH3T3/hEGFR, and is present in the cytoplasm, but strongly localized at the plasma membrane and in the nucleus in malignant cells harboring persistently active Stat3. Moreover, SPI specifically blocks constitutive Stat3 phosphorylation, DNA binding activity, and transcriptional function in malignant cells, with little or no effect on the induction of Stat1, Stat5, and Erk1/2(MAPK) pathways, or on general pTyr profile at the concentrations that inhibit Stat3 activity. Significantly, treatment with SPI of human breast, pancreatic, prostate, and non-small cell lung cancer cells harboring constitutively active Stat3 induced extensive morphology changes, associated with viability loss and apoptosis. Our study identifies SPI as a novel molecular probe for interrogating Stat3 signaling and that functions as a selective inhibitor of Stat3 activation with antitumor cell effects.


Subject(s)
Antineoplastic Agents/metabolism , Peptides/metabolism , STAT3 Transcription Factor/metabolism , src Homology Domains , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Membrane Permeability , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytoplasm/metabolism , Humans , Immunoblotting , Mice , Microscopy, Confocal , Models, Molecular , Molecular Sequence Data , NIH 3T3 Cells , Peptides/chemistry , Peptides/pharmacology , Phosphorylation/drug effects , Protein Binding , Protein Conformation , Protein Structure, Tertiary , STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/genetics
6.
J Pharmacol Exp Ther ; 333(2): 373-81, 2010 May.
Article in English | MEDLINE | ID: mdl-20100905

ABSTRACT

Many molecular aberrations occur in pancreatic cancer. Although aberrant epidermal growth factor receptor (EGFR), Src, and signal transducer and activator of transcription 3 (Stat3) are implicated in pancreatic cancer, therapies that target only one of these entities are undermined by signaling cross-talk. In the human pancreatic cancer lines, Panc-1 and Colo-357, pY845EGFR, pY1068EGFR, pY1086EGFR, and pY1173EGFR levels and pY416c-Src are concurrently elevated with aberrantly active Stat3 in a complex signaling cross-talk. Thus, understanding the signaling integration would facilitate the design of effective multiple-targeted therapeutic modalities. In Panc-1 and Colo-357 lines, pY845EGFR, pY1068EGFR, and pY1086EGFR levels are responsive to c-Src inhibition in contrast to pY1173EGFR, which is EGFR kinase-dependent. Constitutively active Stat3 is sensitive to both EGFR and Src inhibition, but the early suppression of aberrantly active Stat3 in response to the inhibition of EGFR and Src is countered by a Janus kinase (Jaks)-dependent reactivation, suggesting that Jaks activity is a compensatory mechanism for Stat3 induction. The inhibition of EGFR, Src, or Stat3 alone induced weak biological responses. By contrast, the concurrent inhibition of Stat3 and EGFR or Src induced greater viability loss and apoptosis and decreased the migration/invasion of pancreatic cancer cells in vitro. Significantly, the concurrent inhibition, compared with monotargeting modality, induced stronger human pancreatic tumor growth inhibition in xenografts. We infer that the tumor growth inhibition in vivo is caused by the simultaneous suppression of the abnormal functions of Stat3 and EGFR or Src. These studies strongly suggest that the concurrent targeting of Stat3 and EGFR or Src could be a beneficial therapeutic approach for pancreatic cancer.


Subject(s)
ErbB Receptors/drug effects , Pancreatic Neoplasms/physiopathology , Protein-Tyrosine Kinases/drug effects , Proto-Oncogene Proteins/drug effects , STAT3 Transcription Factor/antagonists & inhibitors , Animals , CSK Tyrosine-Protein Kinase , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , ErbB Receptors/genetics , ErbB Receptors/physiology , Flow Cytometry , Humans , Immunoblotting , Mice , Mitogen-Activated Protein Kinase Kinases/drug effects , Oncogene Protein v-akt/drug effects , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Receptor Cross-Talk/drug effects , Receptor Cross-Talk/physiology , STAT3 Transcription Factor/genetics , src-Family Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...