Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 116(24): 11579-11586, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31076554

ABSTRACT

Since the 1980s there has been a drive toward personalized targeted therapy for cancer. "Targeted cancer therapy" originally focused on inhibiting essential tumor survival factors, primarily protein tyrosine kinases. The complexity and rapid mutability of tumors, however, enable them to develop resistance to tyrosine kinase inhibitors (TKIs), even when these are multitargeted or applied in combination. This has led to the development of targeted cancer immunotherapy, to enhance immune surveillance against the tumor. In this paper, we provide a personal view of the development of targeted therapy, from TKIs to targeted immunotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Humans , Immunotherapy/methods , Molecular Targeted Therapy/methods , Neoplasms/metabolism
2.
Mol Cancer Ther ; 17(5): 931-942, 2018 05.
Article in English | MEDLINE | ID: mdl-29440449

ABSTRACT

The small-molecule drug NT157 has demonstrated promising efficacy in preclinical models of a number of different cancer types, reflecting activity against both cancer cells and the tumor microenvironment. Two known mechanisms of action are degradation of insulin receptor substrates (IRS)-1/2 and reduced Stat3 activation, although it is possible that others exist. To interrogate the effects of this drug on cell signaling pathways in an unbiased manner, we have undertaken mass spectrometry-based global tyrosine phosphorylation profiling of NT157-treated A375 melanoma cells. Bioinformatic analysis of the resulting dataset resolved 5 different clusters of tyrosine-phosphorylated peptides that differed in the directionality and timing of response to drug treatment over time. The receptor tyrosine kinase AXL exhibited a rapid decrease in phosphorylation in response to drug treatment, followed by proteasome-dependent degradation, identifying an additional potential target for NT157 action. However, NT157 treatment also resulted in increased activation of p38 MAPK α and γ, as well as the JNKs and specific Src family kinases. Importantly, cotreatment with the p38 MAPK inhibitor SB203580 attenuated the antiproliferative effect of NT157, while synergistic inhibition of cell proliferation was observed when NT157 was combined with a Src inhibitor. These findings provide novel insights into NT157 action on cancer cells and highlight how globally profiling the impact of a specific drug on cellular signaling networks can identify effective combination treatments. Mol Cancer Ther; 17(5); 931-42. ©2018 AACR.


Subject(s)
Protein-Tyrosine Kinases/metabolism , Proteomics/methods , Pyrogallol/analogs & derivatives , Signal Transduction/drug effects , Sulfonamides/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Interaction Maps/drug effects , Protein-Tyrosine Kinases/classification , Proto-Oncogene Proteins c-yes/genetics , Proto-Oncogene Proteins c-yes/metabolism , Pyrogallol/pharmacology , RNA Interference
3.
J Infect Dis ; 217(2): 288-297, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29149330

ABSTRACT

Superantigens (SAgs) are extremely potent bacterial toxins, which evoke a virulent immune response, inducing nonspecific T-cell proliferation, rapid cytokine release, and lethal toxic shock, for which there is no effective treatment. We previously developed a small molecule, S101, which potently inhibits proliferating T cells. In a severe mouse model of toxic shock, a single injection of S101 given together with superantigen challenge rescued 100% of the mice. Even when given 2 hours after challenge, S101 rescued 40% of the mice. S101 targets the T-cell receptor, inflammatory response, and actin cytoskeleton pathways. S101 inhibits the aryl hydrocarbon receptor, a ligand-activated transcription factor that is involved in the differentiation of T-helper cells, especially Th17, and regulatory T cells. Our results provide the rationale for developing S101 to treat superantigen-induced toxic shock and other pathologies characterized by T-cell activation and proliferation.


Subject(s)
Immunologic Factors/administration & dosage , Shock, Septic/prevention & control , Shock, Septic/therapy , Superantigens/toxicity , T-Lymphocytes/drug effects , Animals , Disease Models, Animal , Female , Injections, Intravenous , Mice , Mice, Inbred BALB C , Survival Analysis , Treatment Outcome
4.
Proc Natl Acad Sci U S A ; 114(52): 13655-13660, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29229829

ABSTRACT

There is an urgent need for an effective treatment for metastatic prostate cancer (PC). Prostate tumors invariably overexpress prostate surface membrane antigen (PSMA). We designed a nonviral vector, PEI-PEG-DUPA (PPD), comprising polyethylenimine-polyethyleneglycol (PEI-PEG) tethered to the PSMA ligand, 2-[3-(1, 3-dicarboxy propyl)ureido] pentanedioic acid (DUPA), to treat PC. The purpose of PEI is to bind polyinosinic/polycytosinic acid (polyIC) and allow endosomal release, while DUPA targets PC cells. PolyIC activates multiple pathways that lead to tumor cell death and to the activation of bystander effects that harness the immune system against the tumor, attacking nontargeted neighboring tumor cells and reducing the probability of acquired resistance and disease recurrence. Targeting polyIC directly to tumor cells avoids the toxicity associated with systemic delivery. PPD selectively delivered polyIC into PSMA-overexpressing PC cells, inducing apoptosis, cytokine secretion, and the recruitment of human peripheral blood mononuclear cells (PBMCs). PSMA-overexpressing tumors in nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice with partially reconstituted immune systems were significantly shrunken following PPD/polyIC treatment, in all cases. Half of the tumors showed complete regression. PPD/polyIC invokes antitumor immunity, but unlike many immunotherapies does not need to be personalized for each patient. The potent antitumor effects of PPD/polyIC should spur its development for clinical use.


Subject(s)
Glutamate Carboxypeptidase II/antagonists & inhibitors , Poly I-C/pharmacology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Adoptive Transfer , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Bystander Effect , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Disease Models, Animal , Gene Expression , Glutamate Carboxypeptidase II/genetics , Glutamate Carboxypeptidase II/metabolism , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Mice , Poly I-C/chemistry , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Protein Binding , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
5.
Oncotarget ; 8(15): 24046-24062, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-28445962

ABSTRACT

The treatment of metastatic androgen-resistant prostate cancer remains a challenge. We describe a protein vector that selectively delivers synthetic dsRNA, polyinosinic/polycytidylic acid (polyIC), to prostate tumors by targeting prostate specific membrane antigen (PSMA), which is overexpressed on the surface of prostate cancer cells.The chimeric protein is built from the double stranded RNA (dsRNA) binding domain of PKR tethered to a single chain anti-PSMA antibody. When complexed with polyIC, the chimera demonstrates selective and efficient killing of prostate cancer cells. The treatment causes the targeted cancer cells to undergo apoptosis and to secrete toxic cytokines. In a "bystander effect", these cytokines kill neighboring cancer cells that do not necessarily overexpress PSMA, and activate immune cells that enhance the killing effect. The strong effects of the targeted polyIC are demonstrated on both 2D cell cultures and 3D tumor spheroids.


Subject(s)
Antigens, Surface/genetics , Bystander Effect/drug effects , Bystander Effect/genetics , Genetic Vectors/genetics , Glutamate Carboxypeptidase II/genetics , RNA, Double-Stranded/genetics , Recombinant Fusion Proteins/genetics , Animals , Antigens, Surface/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Chemotaxis, Leukocyte/drug effects , Cytokines/biosynthesis , Disease Models, Animal , Gene Expression , Genes, Reporter , Genetic Therapy , Genetic Vectors/administration & dosage , Glutamate Carboxypeptidase II/antagonists & inhibitors , Glutamate Carboxypeptidase II/metabolism , Humans , Mice , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Neoplasms/therapy , Poly I-C/chemistry , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/pharmacology , Spheroids, Cellular , Xenograft Model Antitumor Assays
6.
PLoS One ; 11(9): e0162321, 2016.
Article in English | MEDLINE | ID: mdl-27598772

ABSTRACT

Selective delivery of drugs to tumor cells can increase potency and reduce toxicity. In this study, we describe a novel recombinant chimeric protein, dsRBEC, which can bind polyIC and deliver it selectively into EGFR over-expressing tumor cells. dsRBEC, comprises the dsRNA binding domain (dsRBD) of human PKR (hPKR), which serves as the polyIC binding moiety, fused to human EGF (hEGF), the targeting moiety. dsRBEC shows high affinity towards EGFR and triggers ligand-induced endocytosis of the receptor, thus leading to the selective internalization of polyIC into EGFR over-expressing tumor cells. The targeted delivery of polyIC by dsRBEC induced cellular apoptosis and the secretion of IFN-ß and other pro-inflammatory cytokines. dsRBEC-delivered polyIC is much more potent than naked polyIC and is expected to reduce the toxicity caused by systemic delivery of polyIC.


Subject(s)
Apoptosis/drug effects , Drug Delivery Systems , ErbB Receptors/genetics , Interferon Inducers/pharmacology , Poly I-C/pharmacology , Recombinant Fusion Proteins/genetics , Animals , Cell Line, Tumor , Chemokine CCL5/biosynthesis , Chemokine CCL5/metabolism , Cloning, Molecular , Endocytosis , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Interferon Inducers/chemistry , Interferon Inducers/metabolism , Interferon-beta/biosynthesis , Interferon-beta/metabolism , MCF-7 Cells , Poly I-C/chemistry , Poly I-C/metabolism , Protein Binding , Protein Domains , Recombinant Fusion Proteins/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
7.
Cancer Immunol Res ; 4(8): 688-97, 2016 08.
Article in English | MEDLINE | ID: mdl-27241844

ABSTRACT

The development of targeted therapies that affect multiple signaling pathways and stimulate antitumor immunity is greatly needed. About 20% of patients with breast cancer overexpress HER2. Small molecules and antibodies targeting HER2 convey some survival benefits; however, patients with advanced disease succumb to the disease under these treatment regimens, possibly because HER2 is not completely necessary for the survival of the targeted cancer cells. In the present study, we show that a polyinosine/polycytosine (pIC) HER2-homing chemical vector induced the demise of HER2-overexpressing breast cancer cells, including trastuzumab-resistant cells. Targeting pIC to the tumor evoked a number of cell-killing mechanisms, as well as strong bystander effects. These bystander mechanisms included type I IFN induction, immune cell recruitment, and activation. The HER2-targeted pIC strongly inhibited the growth of HER2-overexpressing tumors in immunocompetent mice. The data presented here could open additional avenues in the treatment of HER2-positive breast cancer. Cancer Immunol Res; 4(8); 688-97. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/immunology , Neoplasms/pathology , Poly I-C/administration & dosage , Receptor, ErbB-2/antagonists & inhibitors , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression , Humans , Inflammation Mediators/metabolism , Mice , Neoplasms/drug therapy , Neoplasms/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
8.
Bioconjug Chem ; 25(9): 1644-54, 2014 Sep 17.
Article in English | MEDLINE | ID: mdl-25121341

ABSTRACT

The delivery of nucleic acids into cells is an attractive approach for cancer therapy. Polyethylenimine (PEI) is among the most efficient nonviral carriers. Recent studies have demonstrated that PEI can be conjugated to targeting ligands, such as epidermal growth factor (EGF) and transferrin (Schaffert et al., 2011; Abourbeh et al., 2012; Ogris et al., 1999). Herein we present a simplified protocol for producing homogeneous preparations of PEGylated linear PEI: LPEI-PEG2k. We generated two well-characterized copolymers, with ratios of LPEI to PEG of 1:1 and 1:3. These copolymers were further conjugated through disulfide bonds to a Her-2 targeting moiety, Her-2 affibody. This reaction yielded two triconjugates that target Her-2 overexpressing tumors. Polyplexes were formed by complexing plasmid DNA with the triconjugates. We characterized the biophysical properties of the conjugates, and found that the triconjugate 1:3 polyplex had lower ζ potential, larger particle size, and more heterogeneous shape than the triconjugate 1:1 polyplex. Triconjugate 1:1 and triconjugate 1:3 polyplexes were highly selective toward cells that overexpress Her-2 receptors, but triconjugate 1:1 polyplex was more efficient at gene delivery. Our studies show that the biophysical and biological properties of the conjugates can be profoundly affected by the ratio of LPEI:PEG2k:ligand. The procedure described here can be adapted to generate a variety of triconjugates, simply by changing the targeting moiety.


Subject(s)
DNA/chemistry , Drug Carriers/chemistry , Polyethylene Glycols/chemistry , Polyethyleneimine/chemistry , Cell Line, Tumor , DNA/genetics , DNA/metabolism , Deoxyribonucleases/metabolism , Humans , Ligands , Molecular Weight , Protein Structure, Tertiary , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sulfhydryl Compounds/chemistry , Transfection
9.
PLoS One ; 8(3): e57748, 2013.
Article in English | MEDLINE | ID: mdl-23526950

ABSTRACT

Transformation is a complex process, involving many changes in the cell. In this work, we investigated the transcriptional changes that arose during the development of squamous cell carcinoma (SCC) in mice. Using microarray analysis, we looked at gene expression during different stages in cancer progression in 31 mice. By analyzing tumor progression in each mouse separately, we were able to define the global changes that were common to all 31 mice, as well as significant changes that occurred in fewer individuals. We found that different genes can contribute to the tumorigenic process in different mice, and that there are many ways to acquire the malignant properties defined by Hanahan and Weinberg as "hallmarks of cancer". Eventually, however, all these changes lead to a very similar cancerous phenotype. The finding that gene expression is strongly heterogeneous in tumors that were induced by a standardized protocol in closely related mice underscores the need for molecular characterization of human tumors and personalized therapy.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cell Transformation, Neoplastic/genetics , Skin Neoplasms/genetics , Animals , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/pathology , Cell Death/genetics , Disease Progression , Female , Gene Expression , Genes, cdc , Humans , Male , Mice , Oligonucleotide Array Sequence Analysis , Papilloma/etiology , Papilloma/genetics , Skin Neoplasms/etiology , Skin Neoplasms/pathology
10.
Anal Biochem ; 421(2): 750-4, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22197420

ABSTRACT

Protein-protein interactions (PPIs) govern nearly all processes in living cells. Peptides play an important role in studying PPIs. Peptides carrying photoaffinity labels that covalently bind the interacting protein can be used to obtain more accurate information regarding PPIs. Benzophenone (BP) is a useful photoaffinity label that is widely used to study PPIs. We developed a one-pot two-step synthesis for the preparation of novel BP units. To map the binding site more thoroughly, linkers of various lengths were attached to the BP moiety. These units can be incorporated into peptide sequences using well-established solid phase peptide synthesis (SPPS) protocols. As a proof of concept, we studied the interaction between protein kinase B (PKB/Akt) and its synthetic peptide inhibitor, PTR6154. The methodology is general and can be implemented to study PPIs in a variety of biological systems.


Subject(s)
Benzophenones/chemistry , Peptides/chemistry , Protein Interaction Maps , Proto-Oncogene Proteins c-akt/chemistry , Benzophenones/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Oligopeptides/chemistry , Oligopeptides/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors
11.
Chem Biol Drug Des ; 78(5): 887-92, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21824328

ABSTRACT

Linear peptides suffer from poor pharmacokinetic and pharmacodynamic properties. Peptidomimetics are designed to overcome these pharmacological drawbacks while maintaining the biological effects of the parent peptides. Aza-peptides, in which an alpha carbon is replaced with nitrogen, are promising peptidomimetic analogs; however, little is known about the stability of these analogs toward enzymatic degradation. We performed systematic aza and N-methyl scans of a PKB/Akt inhibitor, PTR6154. We evaluated the stability of the aza-scan and N-methyl scan libraries toward enzymatic degradation by trypsin/chymotrypsin. Our results indicate that the modification site is important for metabolic stability and that aza-peptides have a more global effect than N-methylation, affecting cleavage sites distant from the modification site.


Subject(s)
Aza Compounds/chemistry , Oligopeptides/chemistry , Peptidomimetics , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , 3-Phosphoinositide-Dependent Protein Kinases , Amino Acid Sequence , Chymotrypsin/metabolism , Oligopeptides/metabolism , Protein Kinase Inhibitors/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Trypsin/metabolism
12.
J Med Chem ; 54(14): 5154-64, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21650457

ABSTRACT

Elevated levels of activated protein kinase B (PKB/Akt) have been detected in many types of cancer. Substrate-based peptide inhibitors have the advantage of selectivity due to their extensive interactions with the kinase-specific substrate binding site but often lack necessary pharmacological properties. Chemical modifications of potent peptide inhibitors, such as cyclization, may overcome these drawbacks while maintaining potency. We present an extensive structure-activity relationship (SAR) study of a potent peptide-based PKB/Akt inhibitor. Two backbone cyclic (BC) peptide libraries with varying modes of cyclization, bridge chemistry, and ring size were synthesized and evaluated for in vitro PKB/Akt inhibition. Backbone-to-backbone urea BC peptides were more potent than N-terminus-to-backbone amide BC peptides. Several analogues were up to 10-fold more active than the parent linear peptide. Some activity trends could be rationalized using computational surface mapping of the PKB/Akt kinase catalytic domain. The novel molecules have enhanced pharmacological properties which make them promising lead candidates.


Subject(s)
Models, Molecular , Peptides, Cyclic/chemical synthesis , Peptidomimetics/chemical synthesis , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Amides/chemical synthesis , Amides/chemistry , Arginine/analogs & derivatives , Arginine/chemical synthesis , Arginine/chemistry , Catalytic Domain , Magnetic Resonance Spectroscopy , Molecular Conformation , Peptide Library , Peptides, Cyclic/chemistry , Peptidomimetics/chemistry , Proline/analogs & derivatives , Proline/chemical synthesis , Proline/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Structure-Activity Relationship , Thermodynamics , Urea/analogs & derivatives , Urea/chemical synthesis , Urea/chemistry
13.
Anticancer Agents Med Chem ; 11(8): 693-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21707501

ABSTRACT

Nucleic acid based therapeutics offer the possibility of tailor-made treatment of malignant diseases. For recurrent glioblastoma multiforme (GBM), the most aggressive type of brain tumor, no accepted treatment exists, making therapeutically active nucleic acids a viable option. In this review, current preclinical and clinical studies harnessing the potential of antitumoral nucleic acids for GBM treatment will be considered. These include gene therapy to over-express antitumoral gene products, RNA interference to knock down components that promote tumor progression, and the tumor-targeted delivery of antitumoral double stranded RNA. Vectors applied in GBM for the delivery of nucleic acids will be discussed. These include non-replicating and replicating (oncolytic) viruses, as well as non-viral delivery vectors based on polycations or cationic lipids.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Gene Transfer Techniques , Glioblastoma/drug therapy , Nucleic Acids/administration & dosage , Animals , Brain Neoplasms/genetics , Drug Delivery Systems/methods , Glioblastoma/genetics , Humans , Nucleic Acids/genetics
14.
J Org Chem ; 76(9): 3078-85, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21410283

ABSTRACT

Aza-peptides are peptidomimetics in which one or more of the α-carbons, bearing the side-chain residues, has been replaced by a nitrogen. These peptidomimetics have been shown to be promising for the generation of drug leads and for structure-activity relationship studies. Aza-scan is the systematic replacement of amino acid residues in a given peptide with their aza counterparts. We report here an aza-scan of a potent, peptide-based PKB/Akt inhibitor, PTR6154. Procedures for microwave-assisted, Fmoc/t-Bu chemistry, solid-phase aza-peptide synthesis were developed which significantly reduce standard reaction time and are suitable for automation. Novel substituted hydrazines have been prepared for the straightforward incorporation of aza-arginine and aza-proline residues. This work will enable aza-scan to become a more common and standard method for structure-activity relationship studies of peptides.


Subject(s)
Aza Compounds/chemistry , Microwaves , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Proline/chemistry , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Amino Acid Sequence , Arginine/chemistry , Fluorenes/chemistry , Hydrazines/chemical synthesis , Hydrazines/chemistry , Oligopeptides/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship
15.
Mol Biosyst ; 7(5): 1501-11, 2011 May.
Article in English | MEDLINE | ID: mdl-21350750

ABSTRACT

Cervical cancer is initiated by infection with high-risk human papillomavirus (HPV). The viral E6 and E7 oncogenes are required for the initiation of cervical epithelial cell immortalization, but do not suffice to cause cervical cancer. Human oncogenes and tumor suppressor genes with altered activity in cervical carcinoma have been recognized, but none of these appears to be an absolute precondition for the development of the cancer. To examine the contribution of chromosomal aberrations to the development of cervical carcinoma, we used an in vitro model system consisting of primary keratinocytes (K) and papillomavirus transformed keratinocytes from early (E) and late (L) passages and from benzo[a]pyrene treated L cells (BP). Using DIGMAP software we compared alterations in gene expression as a function of chromosomal location. SKY chromosomal painting confirmed that the alterations identified by DIGMAP include gross chromosomal aberrations. Nearly half of the transcripts that were significantly reduced or increased in BP cells mapped to chromosomal regions showing coordinate changes in expression. These included transcripts involved in previously characterized phenotypic changes involved in transformation, such as the switch from apoptosis to necrosis and the reduction in cap-dependent translation. The global chromosomal changes that occurred during transformation are highly correlated with the phenotypic changes in HPV transformed keratinocytes. Our data are consistent with the theory that global chromosomal change is a necessary step in the process of cervical transformation.


Subject(s)
Cell Transformation, Viral/genetics , Chromosome Aberrations , Human papillomavirus 16/physiology , Keratinocytes/metabolism , Keratinocytes/virology , Apoptosis/genetics , Cell Line, Transformed , Cells, Cultured , Female , Gene Expression Profiling , Host-Pathogen Interactions , Humans , Keratinocytes/cytology , Necrosis/genetics , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Spectral Karyotyping/methods , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/virology
16.
J Biol Chem ; 286(17): 15268-77, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21245141

ABSTRACT

We report that the activation level of AMP-dependent protein kinase AMPK is elevated in cancer cell lines as a hallmark of their transformed state. In OVCAR3 and A431 cells, c-Src signals through protein kinase Cα, phospholipase Cγ, and LKB1 to AMPK. AMPK controls internal ribosome entry site (IRES) dependent translation in these cells. We suggest that AMPK activation via PKC might be a general mechanism to regulate IRES-dependent translation in cancer cells.


Subject(s)
AMP-Activated Protein Kinases/genetics , Cell Transformation, Neoplastic , Protein-Tyrosine Kinases/metabolism , Up-Regulation/genetics , CSK Tyrosine-Protein Kinase , Cell Line, Tumor , Enzyme Activation/physiology , Humans , Protein Biosynthesis , Signal Transduction , src-Family Kinases
17.
IUBMB Life ; 62(10): 757-63, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20931634

ABSTRACT

We explored the crosstalk between protein degradation and synthesis in cancer cells. The tumorigenic cell line, MCF7, showed enhanced proteasome activity compared to the nontumorigenic line, MCF10A. Although there was no difference in the sensitivity of MCF7 and MCF10A cells to proteasome inhibition in complete growth medium, combining proteasome inhibition with amino acid deprivation led to reduced protein synthesis and survival of MCF7 cells, with a lesser effect on MCF10A cells. Additional cancer cell lines (including CAG and A431) could be strongly sensitized to proteasome inhibition by concomitant amino acid deprivation, whereas others were completely resistant to proteasome inhibition. We hypothesize that protein catabolism contributes to the pool of free amino acids available for protein synthesis, leading to a crucial role of the proteasome in cell survival during amino acid depletion, in some tumor cell lines.


Subject(s)
Amino Acids/pharmacology , Boronic Acids/pharmacology , Proteasome Inhibitors , Pyrazines/pharmacology , Bortezomib , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Humans , Neoplasms/metabolism , Neoplasms/pathology , Protease Inhibitors/pharmacology , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism
18.
Mol Aspects Med ; 31(4): 287-329, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20451549

ABSTRACT

Signal transduction therapy for cancer targets signaling elements with key roles in cancer cell survival and proliferation, but with more minor roles in the survival of healthy cells. Cancer cells have shrunken signaling networks, and therefore tend to be dependent on fewer signaling modules than non-cancerous cells. Thus, targeted therapy holds the promise of efficacy with minimal toxicity. Yet, with the notable exception of Gleevec for the treatment of early chronic myelogenous leukemia (CML), targeted therapies have so far had minimal success. Unlike early CML, which is dependent upon BCR-ABL, most cancers are not dependent on a single survival factor. Furthermore, tumors are constantly evolving entities, and are heterogeneous in their cellular makeup, compounding the challenge. "Smart cocktails", comprising rational combinations of therapies, need to be developed to meet this challenge. What are the best pathways to target, and why? What types of molecules can be developed into effective therapeutics? What combinations are likely to be successful? Here we present an overview of the principles that need to be considered in designing effective targeted therapy for cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/physiopathology , Neoplasms/therapy , Signal Transduction , Animals , Antineoplastic Agents/chemistry , Benzamides , Cancer Vaccines , Clinical Trials as Topic , Drug Discovery , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , Genetic Therapy , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Molecular Structure , Piperazines/chemistry , Piperazines/therapeutic use , Proteasome Inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/chemistry , Pyrimidines/therapeutic use
19.
J Pept Sci ; 16(4): 178-85, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20196085

ABSTRACT

Cyclization of bioactive peptides, utilizing functional groups serving as natural pharmacophors, is often accompanied with loss of activity. The backbone cyclization approach was developed to overcome this limitation and enhance pharmacological properties. Backbone cyclic peptides are prepared by the incorporation of special building units, capable of forming amide, disulfide and coordinative bonds. Urea bridge is often used for the preparation of cyclic peptides by connecting two amine functionalized side chains. Here we present urea backbone cyclization as an additional method for the preparation of backbone cyclic peptide libraries. A straightforward method for the synthesis of crystalline Fmoc-N(alpha) [omega-amino(Alloc)-alkyl] glycine building units is presented. A set of urea backbone cyclic Glycogen Synthase Kinase 3 analogs was prepared and assessed for protein kinase B inhibition as anticancer leads.


Subject(s)
Glycine/chemistry , Peptides, Cyclic/chemical synthesis , Urea/chemistry , Chromatography, High Pressure Liquid , Cyclization , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Peptides, Cyclic/chemistry , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
20.
Bioorg Med Chem ; 18(8): 2976-85, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20347317

ABSTRACT

Elevated levels of activated Protein Kinase B (PKB/Akt) have been detected in many types of human cancer. In contrast to ATP site inhibitors, substrate-based inhibitors are more likely to be selective because of extensive interactions with the specific substrate binding site. Unfortunately, peptide-based inhibitors lack important pharmacological properties that are required of drug candidates. Chemical modifications of potent peptide inhibitors, such as peptoids and N(alpha)-methylated amino acids, may overcome these drawbacks, while maintaining potency. We present a structure-activity relationship study of a potent, peptide-based PKB/Akt inhibitor, PTR6154. The study was designed to evaluate backbone modifications on the inhibitory activity of PTR6154. Two peptidomimetic libraries, peptoid and N(alpha)-methylation, based on PTR6154, were synthesized and evaluated for in vitro PKB/Akt inhibition efficiency. All the peptoid analogs reduced potency significantly, as well as most of the members of the N-methyl library, suggesting that the backbone conformation and/or hydrogen bond interactions of PTR6154 derivatives are essential for inhibition activity. Two N-terminal members of the N-methyl library did not decrease potency and can be used as future drug leads.


Subject(s)
Antineoplastic Agents/chemical synthesis , Oligopeptides/chemical synthesis , Peptoids/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Amino Acid Sequence , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Oligopeptides/chemistry , Oligopeptides/pharmacology , Peptoids/chemistry , Peptoids/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...