Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Pharmaceutics ; 13(10)2021 Oct 13.
Article in English | MEDLINE | ID: mdl-34683962

ABSTRACT

Autoimmune diseases such as rheumatoid arthritis are caused by immune system recognition of self-proteins and subsequent production of effector T cells that recognize and attack healthy tissue. Therapies for these diseases typically utilize broad immune suppression, which can be effective, but which also come with an elevated risk of susceptibility to infection and cancer. T cell recognition of antigens is driven by binding of T cell receptors to peptides displayed on major histocompatibility complex proteins (MHCs) on the cell surface of antigen-presenting cells. Technology for recombinant production of the extracellular domains of MHC proteins and loading with peptides to produce pMHCs has provided reagents for detection of T cell populations, and with the potential for therapeutic intervention. However, production of pMHCs in large quantities remains a challenge and a translational path needs to be established. Here, we demonstrate a fusion protein strategy enabling large-scale production of pMHCs. A peptide corresponding to amino acids 259-273 of collagen II was fused to the N-terminus of the MHC_II beta chain, and the alpha and beta chains were each fused to human IgG4 Fc domains and co-expressed. A tag was incorporated to enable site-specific conjugation. The cytotoxic drug payload, MMAF, was conjugated to the pMHC and potent, peptide-specific killing of T cells that recognize the collagen pMHC was demonstrated with tetramerized pMHC-MMAF conjugates. Finally, these pMHCs were incorporated into MMAF-loaded 3DNA nanomaterials in order to provide a biocompatible platform. Loading and pMHC density were optimized, and peptide-specific T cell killing was demonstrated. These experiments highlight the potential of a pMHC fusion protein-targeted, drug-loaded nanomaterial approach for selective delivery of therapeutics to disease-relevant T cells and new treatment options for autoimmune disease.

2.
Mol Ther ; 29(6): 2053-2066, 2021 06 02.
Article in English | MEDLINE | ID: mdl-33601052

ABSTRACT

RNA interference (RNAi) offers the potential to treat disease at the earliest onset by selectively turning off the expression of target genes, such as intracellular oncogenes that drive cancer growth. However, the development of RNAi therapeutics as anti-cancer drugs has been limited by both a lack of efficient and target cell-specific delivery systems and the necessity to overcome numerous intracellular barriers, including serum/lysosomal instability, cell membrane impermeability, and limited endosomal escape. Here, we combine two technologies to achieve posttranscriptional gene silencing in tumor cells: Centyrins, alternative scaffold proteins binding plasma membrane receptors for targeted delivery, and small interfering RNAs (siRNAs), chemically modified for high metabolic stability and potency. An EGFR Centyrin known to internalize in EGFR-positive tumor cells was site-specifically conjugated to a beta-catenin (CTNNb1) siRNA and found to drive potent and specific target knockdown by free uptake in cell culture and in mice inoculated with A431 tumor xenografts (EGFR amplified). The generalizability of this approach was further demonstrated with Centyrins targeting multiple receptors (e.g., BCMA, PSMA, and EpCAM) and siRNAs targeting multiple genes (e.g., CD68, KLKb1, and SSB1). Moreover, by installing multiple conjugation handles, two different siRNAs were fused to a single Centyrin, and the conjugate was shown to simultaneously silence two different targets. Finally, by specifically pairing EpCAM-binding Centyrins that exhibited optimized internalization profiles, we present data showing that an EpCAM Centyrin CTNNb1 siRNA conjugate suppressed tumor cell growth of a colorectal cancer cell line containing an APC mutation but not cells with normal CTNNb1 signaling. Overall, these data demonstrate the potential of Centyrin-siRNA conjugates to target cancer cells and silence oncogenes, paving the way to a new class of anticancer drugs.


Subject(s)
Gene Transfer Techniques , RNA Interference , RNA, Small Interfering/genetics , Animals , Cell Line, Tumor , Gene Knockdown Techniques , Gene Silencing , Genes, erbB-1 , Genetic Therapy , Humans , Ligands , Mice , RNA, Messenger , RNA, Small Interfering/administration & dosage , Tenascin/genetics , Xenograft Model Antitumor Assays , beta Catenin/genetics
3.
Article in English | MEDLINE | ID: mdl-23366734

ABSTRACT

Transgenic mice with Tie2- green fluorescent protein (GFP) are used as a model to study the kinetic distribution of the Cy5-siRNA delivered by lipid nanoparticles (LNP) into the liver. After the mouse is injected with the LNP, it undergoes a procedure of intra-vital multi-photon microscopy imaging over a period of two hours, during which the process for the nanoparticle to diffuse into the hepatocytes from the vasculature system is monitored. Since the images are obtained in-vivo, the quantification of Cy5 kinetics suffers from the moving field of view (FOV). A method is proposed to register the sequence of images through template matching. Based on the semi-automatic segmentations of the vessels in the common FOV, the registered images are segmented into three regions of interest (ROI) in which the Cy5 signals are quantified. Computation of the percentage signal strength in the ROIs over time allows for the analysis of the diffusion of Cy5-siRNA into the hepatocytes, and helps demonstrate the effectiveness of the Cy5-siRNA delivery vehicle.


Subject(s)
Carbocyanines/metabolism , Imaging, Three-Dimensional , Microscopy, Fluorescence, Multiphoton/methods , RNA, Small Interfering/metabolism , Signal Processing, Computer-Assisted , Animals , Green Fluorescent Proteins/metabolism , Mice , Mice, Transgenic
4.
Cancer Biol Ther ; 9(7): 493-503, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20139722

ABSTRACT

The PI3K-Akt pathway is dysregulated in the majority of solid tumors. Pharmacological inhibition of Akt is a promising strategy for treating tumors resistant to growth factor receptor antagonists due to mutations in PI3K or PTEN. We have developed allosteric, isozyme-specific inhibitors of Akt activity and activation, as well as ex vivo kinase assays to measure inhibition of individual Akt isozymes in tissues. Here we describe the relationship between PK, Akt inhibition, hyperglycemia and tumor efficacy for a selective inhibitor of Akt1 and Akt2 (AKTi). In nude mice, AKTi treatment caused transient insulin resistance and reversible, dose-dependent hyperglycemia and hyperinsulinemia. Akt1 and Akt2 phosphorylation was inhibited in mouse lung with EC50 values of 1.6 and 7 µM, respectively, and with similar potency in other tissues and xenograft tumors. Weekly subcutaneous dosing of AKTi resulted in dose-dependent inhibition of LNCaP prostate cancer xenografts, an AR-dependent tumor with PTEN deletion and constitutively activated Akt. Complete tumor growth inhibition was achieved at 200 mpk, a dose that maintained inhibition of Akt1 and Akt2 of greater than 80% and 50%, respectively, for at least 12 hours in xenograft tumor and mouse lung. Hyperglycemia could be controlled by reducing C(max), while maintaining efficacy in the LNCaP model, but not by insulin administration. AKTi treatment was well tolerated, without weight loss or gross toxicities. These studies supported the rationale for clinical development of allosteric Akt inhibitors and provide the basis for further refining of pharmacokinetic properties and dosing regimens of this class of inhibitors.


Subject(s)
Glucose/metabolism , Indazoles/pharmacology , Indoles/pharmacology , Insulin/metabolism , Naphthyridines/pharmacology , Prostatic Neoplasms/prevention & control , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Allosteric Regulation , Animals , Humans , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Indazoles/pharmacokinetics , Indoles/pharmacokinetics , Isoenzymes , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Naphthyridines/pharmacokinetics , PTEN Phosphohydrolase/metabolism , Phosphorylation/drug effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Tissue Distribution , Xenograft Model Antitumor Assays
5.
PLoS One ; 3(8): e3065, 2008 Aug 26.
Article in English | MEDLINE | ID: mdl-18725974

ABSTRACT

BACKGROUND: Dysregulated PI3K/Akt signaling occurs commonly in breast cancers and is due to HER2 amplification, PI3K mutation or PTEN inactivation. The objective of this study was to determine the role of Akt activation in breast cancer as a function of mechanism of activation and whether inhibition of Akt signaling is a feasible approach to therapy. METHODOLOGY/PRINCIPAL FINDINGS: A selective allosteric inhibitor of Akt kinase was used to interrogate a panel of breast cancer cell lines characterized for genetic lesions that activate PI3K/Akt signaling: HER2 amplification or PI3K or PTEN mutations in order to determine the biochemical and biologic consequences of inhibition of this pathway. A variety of molecular techniques and tissue culture and in vivo xenograft models revealed that tumors with mutational activation of Akt signaling were selectively dependent on the pathway. In sensitive cells, pathway inhibition resulted in D-cyclin loss, G1 arrest and induction of apoptosis, whereas cells without pathway activation were unaffected. Most importantly, the drug effectively inhibited Akt kinase and its downstream effectors in vivo and caused complete suppression of the growth of breast cancer xenografts with PI3K mutation or HER2 amplification, including models of the latter selected for resistance to Herceptin. Furthermore, chronic administration of the drug was well-tolerated, causing only transient hyperglycemia without gross toxicity to the host despite the pleiotropic normal functions of Akt. CONCLUSIONS/SIGNIFICANCE: These data demonstrate that breast cancers with PI3K mutation or HER2 amplification are selectively dependent on Akt signaling, and that effective inhibition of Akt in tumors is feasible and effective in vivo. These findings suggest that direct inhibition of Akt may represent a therapeutic strategy for breast and other cancers that are addicted to the pathway including tumors with resistant to Herceptin.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Gene Amplification , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/physiology , Receptor, ErbB-2/genetics , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Cell Cycle/drug effects , Dimethyl Sulfoxide/pharmacology , Female , G1 Phase , Humans , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction , Tumor Cells, Cultured
6.
Bioorg Med Chem Lett ; 18(11): 3178-82, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18479914

ABSTRACT

A series of naphthyridine and naphthyridinone allosteric dual inhibitors of Akt1 and 2 have been developed. These compounds have been optimized to have potent dual activity against the activated kinase as well as the activation of Akt in cells. One molecule in particular, compound 17, has potent inhibitory activity against Akt1 and 2 in vivo in a mouse lung and efficacy in a tumor xenograft model.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Naphthyridines/chemical synthesis , Naphthyridines/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Combinatorial Chemistry Techniques , Disease Models, Animal , Drug Design , Drug Screening Assays, Antitumor , Humans , Mice , Naphthyridines/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Structure-Activity Relationship
7.
Mol Cancer Ther ; 4(2): 271-9, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15713898

ABSTRACT

Recent studies indicate that dysregulation of the Akt/PKB family of serine/threonine kinases is a prominent feature of many human cancers. The Akt/PKB family is composed of three members termed Akt1/PKBalpha, Akt2/PKBbeta, and Akt3/PKBgamma. It is currently not known to what extent there is functional overlap between these family members. We have recently identified small molecule inhibitors of Akt. These compounds have pleckstrin homology domain-dependent, isozyme-specific activity. In this report, we present data showing the relative contribution that inhibition of the different isozymes has on the apoptotic response of tumor cells to a variety of chemotherapies. In multiple cell backgrounds, maximal induction of caspase-3 activity is achieved when both Akt1 and Akt2 are inhibited. This induction is not reversed by overexpression of functionally active Akt3. The level of caspase-3 activation achieved under these conditions is equivalent to that observed with the phosphatidylinositol-3-kinase inhibitor LY294002. We also show that in different tumor cell backgrounds inhibition of mammalian target of rapamycin, a downstream substrate of Akt, is less effective in inducing caspase-3 activity than inhibition of Akt1 and Akt2. This shows that the survival phenotype conferred by Akt can be mediated by signaling pathways independent of mammalian target of rapamycin in some tumor cell backgrounds. Finally, we show that inhibition of both Akt1 and Akt2 selectively sensitizes tumor cells, but not normal cells, to apoptotic stimuli.


Subject(s)
Apoptosis/physiology , Neoplasms/enzymology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , 3-Phosphoinositide-Dependent Protein Kinases , Antibiotics, Antineoplastic/pharmacology , Caspase 3 , Caspases/metabolism , Chromones/pharmacology , Drug Resistance, Neoplasm/drug effects , Enzyme Activation , Humans , Morpholines/pharmacology , Protein Isoforms/chemistry , Protein Isoforms/pharmacology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Sirolimus/pharmacology , Tumor Cells, Cultured
8.
Biochem J ; 385(Pt 2): 399-408, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15456405

ABSTRACT

We developed a high-throughput HTRF (homogeneous time-resolved fluorescence) assay for Akt kinase activity and screened approx. 270000 compounds for their ability to inhibit the three isoforms of Akt. Two Akt inhibitors were identified that exhibited isoenzyme specificity. The first compound (Akt-I-1) inhibited only Akt1 (IC50 4.6 microM) while the second compound (Akt-I-1,2) inhibited both Akt1 and Akt2 with IC50 values of 2.7 and 21 microM respectively. Neither compound inhibited Akt3 nor mutants lacking the PH (pleckstrin homology) domain at concentrations up to 250 microM. These compounds were reversible inhibitors, and exhibited a linear mixed-type inhibition against ATP and peptide substrate. In addition to inhibiting kinase activity of individual Akt isoforms, both inhibitors blocked the phosphorylation and activation of the corresponding Akt isoforms by PDK1 (phosphoinositide-dependent kinase 1). A model is proposed in which these inhibitors bind to a site formed only in the presence of the PH domain. Binding of the inhibitor is postulated to promote the formation of an inactive conformation. In support of this model, antibodies to the Akt PH domain or hinge region blocked the inhibition of Akt by Akt-I-1 and Akt-I-1,2. These inhibitors were found to be cell-active and to block phosphorylation of Akt at Thr308 and Ser473, reduce the levels of active Akt in cells, block the phosphorylation of known Akt substrates and promote TRAIL (tumour-necrosis-factor-related apoptosis-inducing ligand)-induced apoptosis in LNCap prostate cancer cells.


Subject(s)
Blood Proteins/chemistry , Blood Proteins/genetics , Peptides/chemistry , Peptides/genetics , Phosphoproteins/chemistry , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Sequence Homology, Amino Acid , 3-Phosphoinositide-Dependent Protein Kinases , Adenosine Triphosphate/metabolism , Apoptosis Regulatory Proteins , Benzylamines/pharmacology , Binding, Competitive , Blood Proteins/immunology , Carcinoma/chemistry , Carcinoma/metabolism , Carcinoma/pathology , Caspases/metabolism , Cell Line, Tumor , Cloning, Molecular , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Female , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Male , Membrane Glycoproteins/pharmacology , Molecular Structure , Peptides/immunology , Peptides/metabolism , Phosphoproteins/immunology , Phosphorylation/drug effects , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Quinoxalines/pharmacology , Signal Transduction/physiology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/pharmacology , Uterine Cervical Neoplasms/chemistry , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
9.
Mol Cancer Ther ; 1(7): 451-9, 2002 May.
Article in English | MEDLINE | ID: mdl-12479263

ABSTRACT

Currently, there is no therapy for men with androgen-refractory prostate cancer that substantially extends survival. This report characterizes by in vitro and in vivo techniques a new chemotherapeutic that is composed of desacetyl-vinblastine covalently linked to a peptide that contains a peptide bond that can be hydrolyzed by prostate-specific antigen (PSA). This compound (referred to as vinblastine-conjugate) is minimally toxic to cells in culture which do not express PSA. In the presence of PSA, the peptide moiety is hydrolyzed, generating several highly toxic metabolites that contain vinblastine. Animals bearing PSA-positive human prostate tumors that were treated with the vinblastine-conjugate experienced a >99% reduction in PSA serum level. In contrast, animals bearing PSA-positive human prostate tumors treated with the cytotoxic metabolites derived from the PSA hydrolysis of the vinblastine-conjugate showed a nonsignificant change in both PSA and tumor weight values. The cell killing activity of the vinblastine-conjugate is PSA dependent because animals bearing non-PSA-producing human tumor xenografts had a nonsignificant increase in tumor weight after vinblastine-conjugate treatment. Exploratory efficacy/toxicity studies in LNCaP tumor-bearing nude mice were conducted with animals treated for 5 consecutive days with various doses of either the vinblastine-conjugate or a PSA-generated toxic metabolite (desacetyl-vinblastine). The desacetyl-vinblastine treatment resulted in 10-70% mortality with a very slight effect on tumor growth. In contrast, vinblastine-conjugate treatments resulted in no mortality, good to excellent antitumor efficacy, very slight to slight peripheral neuropathy and myelopathy, and slight to severe testicular degeneration. Similar treatment of beagle dogs with the vinblastine-conjugate showed even less toxicity. These data support the use of the PSA-hydrolyzable vinblastine-conjugate as an experimental therapy for prostate cancer in man.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Prodrugs/therapeutic use , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/therapy , Vinblastine/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents, Phytogenic/metabolism , Dogs , Doxorubicin/therapeutic use , Humans , Male , Mice , Mice, Nude , Models, Chemical , Neoplasm Transplantation , Prodrugs/metabolism , Prostatic Neoplasms/pathology , Species Specificity , Tissue Distribution , Tumor Cells, Cultured , Vinblastine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...