Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Trends Pharmacol Sci ; 45(3): 243-254, 2024 03.
Article En | MEDLINE | ID: mdl-38383216

Lysine acetyltransferases (KATs) are a family of epigenetic enzymes involved in the regulation of gene expression; they represent a promising class of emerging drug targets. The frequent molecular dysregulation of these enzymes, as well as their mechanistic links to biological functions that are crucial to cancer, have led to exploration around the development of small-molecule inhibitors against KATs. Despite early challenges, recent advances have led to the development of potent and selective enzymatic and bromodomain (BRD) KAT inhibitors. In this review we discuss the discovery and development of new KAT inhibitors and their application as oncology therapeutics. Additionally, new chemically induced proximity approaches are presented, offering opportunities for unique target selectivity profiles and tissue-specific targeting of KATs. Emerging clinical data for CREB binding protein (CREBBP)/EP300 BRD inhibitors and KAT6 catalytic inhibitors indicate the promise of this target class in cancer therapeutics.


Lysine Acetyltransferases , Neoplasms , Humans , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Lysine Acetyltransferases/chemistry , Lysine Acetyltransferases/genetics , Lysine Acetyltransferases/metabolism , Histones/metabolism , Lysine/metabolism , Neoplasms/drug therapy
2.
Sci Rep ; 13(1): 7678, 2023 05 11.
Article En | MEDLINE | ID: mdl-37169829

Cell-cycle control is accomplished by cyclin-dependent kinases (CDKs), motivating extensive research into CDK targeting small-molecule drugs as cancer therapeutics. Here we use combinatorial CRISPR/Cas9 perturbations to uncover an extensive network of functional interdependencies among CDKs and related factors, identifying 43 synthetic-lethal and 12 synergistic interactions. We dissect CDK perturbations using single-cell RNAseq, for which we develop a novel computational framework to precisely quantify cell-cycle effects and diverse cell states orchestrated by specific CDKs. While pairwise disruption of CDK4/6 is synthetic-lethal, only CDK6 is required for normal cell-cycle progression and transcriptional activation. Multiple CDKs (CDK1/7/9/12) are synthetic-lethal in combination with PRMT5, independent of cell-cycle control. In-depth analysis of mRNA expression and splicing patterns provides multiple lines of evidence that the CDK-PRMT5 dependency is due to aberrant transcriptional regulation resulting in premature termination. These inter-dependencies translate to drug-drug synergies, with therapeutic implications in cancer and other diseases.


Neoplasms , Humans , Cell Cycle Checkpoints , Cell Cycle/genetics , Neoplasms/drug therapy , Protein-Arginine N-Methyltransferases/pharmacology
3.
Mol Cancer Ther ; 21(1): 3-15, 2022 01.
Article En | MEDLINE | ID: mdl-34737197

Protein arginine methyltransferase 5 (PRMT5) overexpression in hematologic and solid tumors methylates arginine residues on cellular proteins involved in important cancer functions including cell-cycle regulation, mRNA splicing, cell differentiation, cell signaling, and apoptosis. PRMT5 methyltransferase function has been linked with high rates of tumor cell proliferation and decreased overall survival, and PRMT5 inhibitors are currently being explored as an approach for targeting cancer-specific dependencies due to PRMT5 catalytic function. Here, we describe the discovery of potent and selective S-adenosylmethionine (SAM) competitive PRMT5 inhibitors, with in vitro and in vivo characterization of clinical candidate PF-06939999. Acquired resistance mechanisms were explored through the development of drug resistant cell lines. Our data highlight compound-specific resistance mutations in the PRMT5 enzyme that demonstrate structural constraints in the cofactor binding site that prevent emergence of complete resistance to SAM site inhibitors. PRMT5 inhibition by PF-06939999 treatment reduced proliferation of non-small cell lung cancer (NSCLC) cells, with dose-dependent decreases in symmetric dimethyl arginine (SDMA) levels and changes in alternative splicing of numerous pre-mRNAs. Drug sensitivity to PF-06939999 in NSCLC cells associates with cancer pathways including MYC, cell cycle and spliceosome, and with mutations in splicing factors such as RBM10. Translation of efficacy in mouse tumor xenograft models with splicing mutations provides rationale for therapeutic use of PF-06939999 in the treatment of splicing dysregulated NSCLC.


Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , S-Adenosylmethionine/metabolism , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Drug Resistance , Female , Humans , Lung Neoplasms/pathology , Mice
4.
Adv Ther ; 37(7): 3059-3082, 2020 07.
Article En | MEDLINE | ID: mdl-32445185

Epigenetic processes are essential for normal development and the maintenance of tissue-specific gene expression in mammals. Changes in gene expression and malignant cellular transformation can result from disruption of epigenetic mechanisms, and global disruption in the epigenetic landscape is a key feature of cancer. The study of epigenetics in cancer has revealed that human cancer cells harbor both genetic alterations and epigenetic abnormalities that interplay at all stages of cancer development. Unlike genetic mutations, epigenetic aberrations are potentially reversible through epigenetic therapy, providing a therapeutically relevant treatment option. Histone methyltransferase inhibitors are emerging as an epigenetic therapy approach with great promise in the field of clinical oncology. The recent accelerated approval of the enhancer of zeste homolog 2 (EZH2; also known as histone-lysine N-methyltransferase EZH2) inhibitor tazemetostat for metastatic or locally advanced epithelioid sarcoma marks the first approval of such a compound for the treatment of cancer. Many other histone methyltransferase inhibitors are currently in development, some of which are being tested in clinical studies. This review focuses on histone methyltransferase inhibitors, highlighting their potential in the treatment of cancer. We also discuss the role for such epigenetic drugs in overcoming epigenetically driven drug resistance mechanisms, and their value in combination with other therapeutic approaches such as immunotherapy.


Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Enhancer of Zeste Homolog 2 Protein/drug effects , Histone Methyltransferases/metabolism , Medical Oncology/standards , Neoplasms/drug therapy , Neoplasms/genetics , Pyridones/therapeutic use , Adult , Aged , Aged, 80 and over , Animals , Epigenesis, Genetic , Female , Humans , Male , Middle Aged , Practice Guidelines as Topic
5.
Cell Rep ; 30(6): 1935-1950.e8, 2020 02 11.
Article En | MEDLINE | ID: mdl-32049022

Alternative splicing is well understood to enhance proteome diversity as cells respond to stimuli. However, mechanistic understanding for how the spliceosome processes precursor messenger RNA (mRNA) transcripts to achieve template diversification is incomplete. We use recently developed enzymatic inhibitors of protein arginine methyltransferase 5 (PRMT5) and human naive T lymphocyte activation as a model system to uncover a precise set of mRNA transcripts that require symmetric arginine dimethylation. This methylation-dependent splicing selectivity is associated with a limited set of signaling pathways that are affected when PRMT5 is inhibited. Specifically, we identify a conserved role for symmetric arginine dimethylation in the induction of antiviral type I and type III interferon signaling following T cell receptor and pattern recognition receptor stimulation in human T lymphocytes and undifferentiated human THP-1 monocytes. Altogether, these findings reveal a mechanism by which cells may be enabled to precisely modulate transcript heterogeneity to orchestrate specific functional outcomes.


Alternative Splicing/genetics , Arginine/metabolism , Interferons/metabolism , RNA Splicing/genetics , Humans , Signal Transduction
6.
PLoS One ; 11(1): e0145705, 2016.
Article En | MEDLINE | ID: mdl-26756335

Autoantibodies and the immunoreceptors to which they bind can contribute to the pathogenesis of autoimmune diseases such as rheumatoid arthritis (RA). Spleen Tyrosine Kinase (Syk) is a non-receptor tyrosine kinase with a central role in immunoreceptor (FcR) signaling and immune cell functionality. Syk kinase inhibitors have activity in antibody-dependent immune cell activation assays, in preclinical models of arthritis, and have progressed into clinical trials for RA and other autoimmune diseases. Here we describe the characterization of a novel triazolopyridine-based Syk kinase inhibitor, CC-509. This compound is a potent inhibitor of purified Syk enzyme, FcR-dependent and FcR-independent signaling in primary immune cells, and basophil activation in human whole blood. CC-509 is moderately selective across the kinome and against other non-kinase enzymes or receptors. Importantly, CC-509 was optimized away from and has modest activity against cellular KDR and Jak2, kinases that when inhibited in a preclinical and clinical setting may promote hypertension and neutropenia, respectively. In addition, CC-509 is orally bioavailable and displays dose-dependent efficacy in two rodent models of immune-inflammatory disease. In passive cutaneous anaphylaxis (PCA), CC-509 significantly inhibited skin edema. Moreover, CC-509 significantly reduced paw swelling and the tissue levels of pro-inflammatory cytokines RANTES and MIP-1α in the collagen-induced arthritis (CIA) model. In summary, CC-509 is a potent, moderately selective, and efficacious inhibitor of Syk that has a differentiated profile when compared to other Syk compounds that have progressed into the clinic for RA.


Indazoles/chemistry , Inflammation/drug therapy , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyridines/chemistry , Triazoles/chemistry , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/physiopathology , Basophils/cytology , Cell Line , Collagen/chemistry , Crystallography, X-Ray , Dose-Response Relationship, Drug , Edema/pathology , Eosinophils/cytology , Female , HEK293 Cells , Humans , Hypertension/drug therapy , Inflammation/physiopathology , Inhibitory Concentration 50 , Janus Kinase 2/antagonists & inhibitors , Male , Neutropenia/drug therapy , Neutrophils/cytology , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Receptors, Fc/chemistry , Skin/pathology , Syk Kinase , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
7.
Cancer Res ; 69(18): 7347-56, 2009 Sep 15.
Article En | MEDLINE | ID: mdl-19738071

Lenalidomide and pomalidomide have both been evaluated clinically for their properties as anticancer agents, with lenalidomide being available commercially. We previously reported that both compounds cause cell cycle arrest in Burkitt's lymphoma and multiple myeloma cell lines by increasing the level of p21(WAF-1) expression. In the present study, we unravel the molecular mechanism responsible for p21(WAF-1) up-regulation using Namalwa cells as a human lymphoma model. We show that the increase of p21(WAF-1) expression is regulated at the transcriptional level through a mechanism independent of p53. Using a combination of approaches, we show that several GC-rich binding transcription factors are involved in pomalidomide-mediated up-regulation of p21(WAF-1). Furthermore, we report that p21(WAF-1) up-regulation is associated with a switch from methylated to acetylated histone H3 on p21(WAF-1) promoter. Interestingly, lysine-specific demethylase-1 (LSD1) silencing reduced both pomalidomide and lenalidomide up-regulation of p21(WAF-1), suggesting that this histone demethylase is involved in the priming of the p21(WAF-1) promoter. Based on our findings, we propose a model in which pomalidomide and lenalidomide modify the chromatin structure of the p21(WAF-1) promoter through demethylation and acetylation of H3K9. This effect, mediated via LSD1, provides GC-rich binding transcription factors better access to DNA, followed by recruitment of RNA polymerase II and transcription activation. Taken together, our results provide new insights on the mechanism of action of pomalidomide and lenalidomide in the regulation of gene transcription, imply possible efficacy in p53 mutated and deleted cancer, and suggest new potential clinical uses as an epigenetic therapy.


Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Lymphoma/drug therapy , Multiple Myeloma/drug therapy , Oxidoreductases, N-Demethylating/metabolism , Thalidomide/analogs & derivatives , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Chromatin/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation, Neoplastic/drug effects , Histone Demethylases , Histones/genetics , Histones/metabolism , Humans , Lenalidomide , Lymphoma/genetics , Lymphoma/metabolism , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Oxidoreductases, N-Demethylating/genetics , Thalidomide/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
8.
J Clin Invest ; 118(1): 248-58, 2008 Jan.
Article En | MEDLINE | ID: mdl-18064299

Sickle-cell disease (SCD) and beta thalassemia constitute worldwide public health problems. New therapies, including hydroxyurea, have attempted to augment the synthesis of fetal hemoglobin (HbF) and improve current treatment. Lenalidomide and pomalidomide are members of a class of immunomodulators used as anticancer agents. Because clinical trials have demonstrated that lenalidomide reduces or eliminates the need for transfusions in some patients with disrupted blood cell production, we investigated the effects of lenalidomide and pomalidomide on erythropoiesis and hemoglobin synthesis. We used an in vitro erythropoiesis model derived from human CD34+ progenitor cells from normal and SCD donors. We found that both compounds slowed erythroid maturation, increased proliferation of immature erythroid cells, and regulated hemoglobin transcription, resulting in potent induction of HbF without the cytotoxicity associated with other HbF inducers. When combined with hydroxyurea, pomalidomide and, to a lesser extent, lenalidomide were found to have synergistic effects on HbF upregulation. Our results elucidate what we believe to be a new mechanism of action of pomalidomide and lenalidomide and support the hypothesis that pomalidomide, used alone or in combination with hydroxyurea, may improve erythropoiesis and increase the ratio of fetal to adult hemoglobin. These findings support the evaluation of pomalidomide as an innovative new therapy for beta-hemoglobinopathies.


Anemia, Sickle Cell/metabolism , Antigens, CD34 , Antineoplastic Agents/pharmacology , Erythropoiesis/drug effects , Fetal Hemoglobin/biosynthesis , Thalidomide/analogs & derivatives , beta-Thalassemia/metabolism , Anemia, Sickle Cell/therapy , Antineoplastic Agents/therapeutic use , Antisickling Agents/pharmacology , Antisickling Agents/therapeutic use , Blood Transfusion , Cells, Cultured , Clinical Trials as Topic , Drug Evaluation, Preclinical , Drug Synergism , Erythroid Cells/metabolism , Humans , Hydroxyurea/agonists , Hydroxyurea/pharmacology , Hydroxyurea/therapeutic use , Lenalidomide , Models, Biological , Thalidomide/agonists , Thalidomide/pharmacology , Thalidomide/therapeutic use , Up-Regulation/drug effects , beta-Thalassemia/therapy
9.
J Biol Chem ; 282(21): 15462-70, 2007 May 25.
Article En | MEDLINE | ID: mdl-17409098

p27, an important cell cycle regulator, blocks the G(1)/S transition in cells by binding and inhibiting Cdk2/cyclin A and Cdk2/cyclin E complexes (Cdk2/E). Ubiquitination and subsequent degradation play a critical role in regulating the levels of p27 during cell cycle progression. Here we provide evidence suggesting that both Cdk2/E and phosphorylation of Thr(187) on p27 are essential for the recognition of p27 by the SCF(Skp2/Cks1) complex, the ubiquitin-protein isopeptide ligase (E3). Cdk2/E provides a high affinity binding site, whereas the phosphorylated Thr(187) provides a low affinity binding site for the Skp2/Cks1 complex. Furthermore, binding of phosphorylated p27/Cdk2/E to the E3 complex showed positive cooperativity. Consistently, p27 is also ubiquitinated in a similarly cooperative manner. In the absence of p27, Cdk2/E and Cks1 increase Skp2 phosphorylation. This phosphorylation enhances Skp2 auto-ubiquitination, whereas p27 inhibits both phosphorylation and auto-ubiquitination of Skp2.


Carrier Proteins/chemistry , Cyclin-Dependent Kinases/chemistry , Multiprotein Complexes/chemistry , Protein Processing, Post-Translational , S-Phase Kinase-Associated Proteins/chemistry , Ubiquitin-Protein Ligases/chemistry , Animals , CDC2-CDC28 Kinases , Carrier Proteins/metabolism , Cell-Free System/chemistry , Cell-Free System/metabolism , Cyclin A/chemistry , Cyclin A/metabolism , Cyclin E/chemistry , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinases/metabolism , G1 Phase/physiology , Humans , Multiprotein Complexes/metabolism , Phosphorylation , Protein Processing, Post-Translational/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , S Phase/physiology , S-Phase Kinase-Associated Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
10.
J Clin Immunol ; 27(2): 210-20, 2007 Mar.
Article En | MEDLINE | ID: mdl-17308870

COX2 (prostaglandin G/H synthase, PTGS2) is a well-validated target in the fields of both oncology and inflammation. Despite their significant toxicity profile, non-steroidal anti-inflammatory drugs (NSAIDs) have become standard of care in the treatment of many COX2-mediated inflammatory conditions. In this report, we show that one IMiDs((R)) immunomodulatory drug, CC-4047, can reduce the levels of COX2 and the production of prostaglandins (PG) in human LPS-stimulated monocytes. The inhibition of COX2 by CC-4047 occurs at the level of gene transcription, by reducing the LPS-stimulated transcriptional activity at the COX2 gene. Because it is a transcriptional rather than an enzymatic inhibitor of COX2, CC-4047 inhibition of PG production is not susceptible to competition by exogenous arachadonic acid (AA). The distinct mechanisms of action allow CC-4047 and a COX2-selective NSAID to work additively to block PG secretion from monocytes. CC-4047 does not, however, block COX2 induction in or prostacyclin secretion from IL-1beta stimulated human umbilical vein endothelial cells (HUVEC) cells, nor does it inhibit COX1 in either monocytes or HUVEC cells. CC-4047 also inhibits COX2 and PG production in monocytes derived from patients with sickle cell disease (SCD). Taken together, the data in this manuscript suggest CC-4047 will provide important anti-inflammatory benefit to patients and will improve the safety of NSAIDs in the treatment of SCD or other inflammatory conditions.


Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/drug effects , Immunologic Factors/pharmacology , Membrane Proteins/drug effects , Thalidomide/analogs & derivatives , Transcription, Genetic/drug effects , Anemia, Sickle Cell/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blotting, Western , Drug Synergism , Gene Expression/drug effects , Humans , Immunoprecipitation , Lipopolysaccharides/pharmacology , Monocytes/drug effects , Monocytes/metabolism , Nitrobenzenes/pharmacology , Prostaglandins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Thalidomide/pharmacology
11.
Cancer Res ; 67(2): 746-55, 2007 Jan 15.
Article En | MEDLINE | ID: mdl-17234786

Clinical studies involving patients with myelodysplastic syndromes or multiple myeloma have shown the efficacy of lenalidomide by reducing and often eliminating malignant cells while restoring the bone marrow function. To better understand these clinical observations, we investigated and compared the effects of lenalidomide and a structurally related analogue, CC-4047, on the proliferation of two different human hematopoietic cell models: the Namalwa cancer cell line and normal CD34+ progenitor cells. Both compounds had antiproliferative effects on Namalwa cells and pro-proliferative effects on CD34+ cells, whereas p21WAF-1 expression was up-regulated in both cell types. In Namalwa cells, the up-regulation of p21WAF-1 correlated well with the inhibition of cyclin-dependent kinase (CDK) 2, CDK4, and CDK6 activity leading to pRb hypophosphorylation and cell cycle arrest, whereas in CD34+ progenitor cells the increase of p21WAF-1 did not inhibit proliferation. Similarly, antiproliferation results were observed in two B lymphoma cell lines (LP-1 and U266) but interestingly not in normal B cells where a protection of apoptosis was found. Finally, CC-4047 and lenalidomide had synergistic effects with valproic acid [a histone deacetylase (HDAC) inhibitor] by increasing the apoptosis of Namalwa cells and enhancing CD34+ cell expansion. Our results indicate that lenalidomide and CC-4047 have opposite effects in tumor cells versus normal cells and could explain, at least in part, the reduction of malignant cells and the restoration of bone marrow observed in patients undergoing lenalidomide treatment. Moreover, this study provides new insights on the cellular pathways affected by lenalidomide and CC-4047, proposes new potential clinical uses, such as bone marrow regeneration, and suggests that the combination of lenalidomide or CC-4047 with certain HDAC inhibitors may elevate the therapeutic index in the treatment of hematologic malignancies.


Antigens, CD34/biosynthesis , Antineoplastic Agents/pharmacology , B-Lymphocytes/drug effects , Hematopoietic Stem Cells/drug effects , Lymphoma, B-Cell/drug therapy , Thalidomide/analogs & derivatives , B-Lymphocytes/pathology , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans , Lenalidomide , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Phosphorylation/drug effects , Retinoblastoma Protein/metabolism , Thalidomide/pharmacology , Up-Regulation/drug effects
12.
Curr Pharm Biotechnol ; 3(1): 45-57, 2002 Mar.
Article En | MEDLINE | ID: mdl-11883506

We adapted filamentous phage vectors for targeted gene delivery to mammalian cells by inserting a mammalian reporter gene expression cassette (GFP) into the vector backbone and fusing the pIII coat protein to a cell targeting ligand (i.e. FGF2, EGF). Like transfection with animal viral vectors, targeted phage gene delivery is concentration, time, and ligand dependent. Importantly, targeted phage particles are specific for the appropriate target cell surface receptor. Phage have distinct advantages over existing gene therapy vectors because they are simple, economical to produce at high titer, have no intrinsic tropism for mammalian cells, and are relatively simple to genetically modify and evolve. Initially transduction by targeted phage particles was low resulting in foreign gene expression in 1-2% of transfected cells. We increased transduction efficiency by modifying both the transfection protocol and vector design. For example, we stabilized the display of the targeting ligand to create multivalent phagemid-based vectors with transduction efficiencies of up to 45% in certain cell lines when combined with genotoxic treatment. Taken together, these studies establish that the efficiency of phage-mediated gene transfer can be significantly improved through genetic modification. We are currently evolving phage vectors with enhanced cell targeting, increased stability, reduced immunogenicity and other properties suitable for gene therapy.


Bacteriophages/genetics , Genetic Therapy/methods , Genetic Vectors , Animals , Humans , Ligands , Peptide Library
13.
Cancer Res ; 62(4): 977-81, 2002 Feb 15.
Article En | MEDLINE | ID: mdl-11861367

Engineered phage-based vectors are an attractive alternative strategy for gene delivery because they possess no natural mammalian cell tropism and can be genetically modified for specific applications. Genotoxic treatments that increase the transduction efficiency of single-stranded adeno-associated virus were tested on cells transfected by single-stranded phage. Indeed, green fluorescent protein transgene expression by epidermal growth factor-targeted phagemid particles increased with heat shock, UV irradiation, and camptothecin (CPT) treatment. CPT resulted in transduction efficiencies of 30-45% in certain human carcinoma cell lines and reduced the minimal dose needed to detect green fluorescent protein-expressing cells to as low as 1-10 particles/cell. Targeted phage transduction was effective in many tumor cell lines and in prostate tumor xenografts with CPT treatment. Taken together, these data suggest the feasibility of using phage-based vectors for therapeutic gene delivery to cancer cells.


Antineoplastic Agents, Phytogenic/pharmacology , Bacteriophages/genetics , Camptothecin/pharmacology , Carcinoma/genetics , Genetic Therapy/methods , Neoplasms/genetics , Plasmids/genetics , Animals , Carcinoma/drug therapy , Carcinoma/therapy , Combined Modality Therapy , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Genetic Vectors/genetics , Humans , Male , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/therapy , Transduction, Genetic , Transfection/methods , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
...