Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 19(9): 1833-1843, 2020 09.
Article in English | MEDLINE | ID: mdl-32669315

ABSTRACT

First-generation antibody-drug conjugates (ADC) are heterogeneous mixtures that have shown clinical benefit, but generally exhibited safety issues and a narrow therapeutic window due, in part, to off-target toxicity caused by ADC instability. ARX788 is a next-generation, site-specific anti-HER2 ADC that utilizes a unique nonnatural amino acid-enabled conjugation technology and a noncleavable Amberstatin (AS269) drug-linker to generate a homogeneous ADC with a drug-to-antibody ratio of 1.9. ARX788 exhibits high serum stability in mice and a relatively long ADC half-life of 12.5 days. When compared in vitro against T-DM1 across a panel of cancer cell lines, ARX788 showed superior activity in the lower HER2-expressing cell lines and no activity in normal cardiomyocyte cells. Similarly, ARX788 significantly inhibited tumor growth, and generally outperformed T-DM1 in HER2-high and HER2-low expression xenograft models. Breast and gastric cancer patient-derived xenograft studies confirmed strong antitumor activity of ARX788 in HER2-positive and HER2-low expression tumors, as well as in a T-DM1-resistant model. The encouraging preclinical data support the further development of ARX788 for treatment of patients with HER2-positive breast and gastric cancer, including those who have developed T-DM1 resistance, and patients with HER2-low expression tumors who are currently ineligible to receive HER2-targeted therapy.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Oligopeptides/administration & dosage , Receptor, ErbB-2/metabolism , Stomach Neoplasms/drug therapy , Ado-Trastuzumab Emtansine/pharmacology , Ado-Trastuzumab Emtansine/therapeutic use , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Breast Neoplasms/blood , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Oligopeptides/pharmacokinetics , Oligopeptides/pharmacology , Stomach Neoplasms/blood , Stomach Neoplasms/metabolism , Xenograft Model Antitumor Assays
2.
Cureus ; 10(8): e3102, 2018 Aug 04.
Article in English | MEDLINE | ID: mdl-30338178

ABSTRACT

There remains a lack of understanding of how wound closure methods perform comparatively when exposed to patient-induced movement during healing and how they may contribute to bacterial infiltration in the wound site. The present study attempts to objectively quantify this gap. The study evaluates bacterial penetration and subsequent symptoms of infection of traditional sutures and an emerging tape-based, zip-type wound closure technology under physiologically relevant loading. In an in vivo model to simulate real-world conditions, the latter demonstrates better performance compared to commonly used sutures, holding the wound intact and minimizing bacterial penetration when subjected to simulated patient movement-induced stress.

3.
Proc Natl Acad Sci U S A ; 111(5): 1766-71, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24443552

ABSTRACT

Using an expanded genetic code, antibodies with site-specifically incorporated nonnative amino acids were produced in stable cell lines derived from a CHO cell line with titers over 1 g/L. Using anti-5T4 and anti-Her2 antibodies as model systems, site-specific antibody drug conjugates (NDCs) were produced, via oxime bond formation between ketones on the side chain of the incorporated nonnative amino acid and hydroxylamine functionalized monomethyl auristatin D with either protease-cleavable or noncleavable linkers. When noncleavable linkers were used, these conjugates were highly stable and displayed improved in vitro efficacy as well as in vivo efficacy and pharmacokinetic stability in rodent models relative to conventional antibody drug conjugates conjugated through either engineered surface-exposed or reduced interchain disulfide bond cysteine residues. The advantages of the oxime-bonded, site-specific NDCs were even more apparent when low-antigen-expressing (2+) target cell lines were used in the comparative studies. NDCs generated with protease-cleavable linkers demonstrated that the site of conjugation had a significant impact on the stability of these rationally designed prodrug linkers. In a single-dose rat toxicology study, a site-specific anti-Her2 NDC was well tolerated at dose levels up to 90 mg/kg. These experiments support the notion that chemically defined antibody conjugates can be synthesized in commercially relevant yields and can lead to antibody drug conjugates with improved properties relative to the heterogeneous conjugates formed by nonspecific chemical modification.


Subject(s)
Antibodies/metabolism , Immunoconjugates/metabolism , Pharmaceutical Preparations/chemical synthesis , Protein Engineering/methods , Animals , Antibodies/blood , Antibodies/chemistry , Antibodies/toxicity , Batch Cell Culture Techniques , CHO Cells , Cell Death/drug effects , Cell Line , Cricetinae , Cricetulus , Cysteine/metabolism , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/toxicity , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/chemistry , Protein Stability/drug effects , Rats
4.
Proc Natl Acad Sci U S A ; 110(44): 17796-801, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24127589

ABSTRACT

Bispecific antibodies, which simultaneously target CD3 on T cells and tumor-associated antigens to recruit cytotoxic T cells to cancer cells, are a promising new approach to the treatment of hormone-refractory prostate cancer. Here we report a site-specific, semisynthetic method for the production of bispecific antibody-like therapeutics in which a derivative of the prostate-specific membrane antigen-binding small molecule DUPA was selectively conjugated to a mutant αCD3 Fab containing the unnatural amino acid, p-acetylphenylalanine, at a defined site. Homogeneous conjugates were generated in excellent yields and had good solubility. The efficacy of the conjugate was optimized by modifying the linker structure, relative binding orientation, and stoichiometry of the ligand. The optimized conjugate showed potent and selective in vitro activity (EC50 ~ 100 pM), good serum half-life, and potent in vivo activity in prophylactic and treatment xenograft mouse models. This semisynthetic approach is likely to be applicable to the generation of additional bispecific agents using drug-like ligands selective for other cell-surface receptors.


Subject(s)
Drug Discovery/methods , Immunoglobulin Fab Fragments/therapeutic use , Prostatic Neoplasms/drug therapy , Animals , CD3 Complex/immunology , Heterografts/immunology , Humans , Immunoglobulin Fab Fragments/immunology , Immunotherapy/methods , Leukocytes, Mononuclear , Male , Mice , Prostatic Neoplasms/immunology , Protein Engineering
6.
Proc Natl Acad Sci U S A ; 109(40): 16101-6, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-22988081

ABSTRACT

Antibody-drug conjugates (ADCs) allow selective targeting of cytotoxic drugs to cancer cells presenting tumor-associated surface markers, thereby minimizing systemic toxicity. Traditionally, the drug is conjugated nonselectively to cysteine or lysine residues in the antibody. However, these strategies often lead to heterogeneous products, which make optimization of the biological, physical, and pharmacological properties of an ADC challenging. Here we demonstrate the use of genetically encoded unnatural amino acids with orthogonal chemical reactivity to synthesize homogeneous ADCs with precise control of conjugation site and stoichiometry. p-Acetylphenylalanine was site-specifically incorporated into an anti-Her2 antibody Fab fragment and full-length IgG in Escherichia coli and mammalian cells, respectively. The mutant protein was selectively and efficiently conjugated to an auristatin derivative through a stable oxime linkage. The resulting conjugates demonstrated excellent pharmacokinetics, potent in vitro cytotoxic activity against Her2(+) cancer cells, and complete tumor regression in rodent xenograft treatment models. The synthesis and characterization of homogeneous ADCs with medicinal chemistry-like control over macromolecular structure should facilitate the optimization of ADCs for a host of therapeutic uses.


Subject(s)
Amino Acids/chemistry , Antibodies, Monoclonal, Humanized/chemistry , Breast Neoplasms/drug therapy , Immunoconjugates/chemistry , Protein Engineering/methods , Aminobenzoates/chemistry , Animals , Cell Line, Tumor , Drug Discovery/methods , Enzyme-Linked Immunosorbent Assay , Escherichia coli , Female , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Immunoglobulin G/chemistry , Mice , Mice, SCID , Oligopeptides/chemistry , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/immunology , Trastuzumab
7.
Chem Biol Drug Des ; 74(1): 43-50, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19519743

ABSTRACT

Heat-shock protein-90 is an attractive target for anticancer drugs, as heat-shock protein-90 blockers such as the ansamycin 17-(allylamino)-17-demethoxygeldanamycin greatly reduce the expression of many signaling molecules that are disregulated in cancer cells and are key drivers of tumor growth and metastasis. While 17-(allylamino)-17-demethoxygeldanamycin has shown promise in clinical trials, this compound class has significant template-related drawbacks. In this paper, we describe a new, potent non-ansamycin small-molecule inhibitor of heat-shock protein-90, BX-2819, containing resorcinol and triazolothione rings. Structural studies demonstrate binding of BX-2819 to the ADP/ATP-binding pocket of heat-shock protein-90. The compound blocked expression of heat-shock protein-90 client proteins in cancer cell lines and inhibited cell growth with a potency similar to 17-(allylamino)-17-demethoxygeldanamycin. In a panel of four cancer cell lines, BX-2819 blocked growth with an average IC(50) value of 32 nM (range of 7-72 nM). Efficacy studies demonstrated that treatment with BX-2819 significantly inhibited the growth of NCI-N87 and HT-29 tumors in nude mice, consistent with pharmacodynamic studies showing inhibition of heat-shock protein-90 client protein expression in tumors for greater than 16 h after dosing. These data support further studies to assess the potential of BX-2819 and related analogs for the treatment of cancer.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Triazoles/pharmacology , Animals , Benzoquinones/chemistry , Benzoquinones/pharmacology , Cell Line, Tumor , Computer Simulation , Crystallography, X-Ray , Drug Screening Assays, Antitumor , HSP90 Heat-Shock Proteins/metabolism , HT29 Cells , Humans , Lactams, Macrocyclic/chemistry , Lactams, Macrocyclic/pharmacology , Mice , Mice, Nude , Transplantation, Heterologous , Triazoles/chemistry , Xenograft Model Antitumor Assays
8.
Mol Cancer Ther ; 5(9): 2158-64, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16985048

ABSTRACT

The ability of cancer cells to undergo invasion and migration is a prerequisite for tumor metastasis. Rho, a Ras-related small GTPase, and the Rho-associated coiled coil-containing protein kinases (Rho kinases, ROCK1 and ROCK2) are key regulators of focal adhesion, actomyosin contraction, and thus cell motility. Inhibitors of this pathway have been shown to inhibit tumor cell motility and metastasis. Here, we show that fasudil [1-(5-isoquinolinesulfonyl)-homopiperazine], an orally available inhibitor of Rho kinases, and its metabolite 1-(hydroxy-5-isoquinoline sulfonyl-homopiperazine) (fasudil-OH) modify tumor cell morphology and inhibit tumor cell migration and anchorage-independent growth. In addition, we show that fasudil inhibited tumor progression in three independent animal models. In the MM1 peritoneal dissemination model, tumor burden and ascites production were reduced by > 50% (P < 0.05). In the HT1080 experimental lung metastasis model, fasudil decreased lung nodules by approximately 40% (P < 0.05). In the orthotopic breast cancer model with MDA-MB-231, there were 3-fold more tumor-free mice in the fasudil-treated group versus saline control group (P < 0.01). Fasudil has been approved for the treatment of cerebral vasospasm and associated cerebral ischemic symptoms. In patients, fasudil is well tolerated without any serious adverse reactions. Therefore, the concept of Rho kinase inhibition as an antimetastatic therapy for cancer can now be clinically explored.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Breast Neoplasms/drug therapy , Fibrosarcoma/drug therapy , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , rho-Associated Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Disease Progression , Female , Fibrosarcoma/enzymology , Fibrosarcoma/pathology , Humans , Male , Mice , Mice, Nude , Rats , Xenograft Model Antitumor Assays
9.
Neoplasia ; 8(1): 18-30, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16533422

ABSTRACT

The EphA2 receptor tyrosine kinase has been shown to be over-expressed in cancer and a monoclonal antibody (mAb) that activates and down-modulates EphA2 was reported to inhibit the growth of human breast and lung tumor xenografts in nude mice. Reduction of EphA2 levels by treatment with anti-EphA2 siRNA also inhibited tumor growth, suggesting that the anti-tumor effects of these agents are mediated by decreasing the levels of EphA2. As these studies employed human tumor xenograft models in nude mice with reagents whose cross reactivity with murine EphA2 is unknown, we generated a mAb (Ab20) that preferentially binds, activates, and induces the degradation of murine EphA2. Treatment of established murine CT26 colorectal tumors with Ab20 reduced EphA2 protein levels to approximately 12% of control tumor levels, yet had no effect on tumor growth. CT26 tumor cell colonization of the lung was also not affected by Ab20 administration despite having barely detectable levels of EphA2. We also generated and tested a potent agonistic mAb against human EphA2 (1G9-H7). No inhibition of humanMDA-231 breast tumor xenograft growth was observed despite evidence for >85% reduction of EphA2 protein levels in the tumors. These results suggest that molecular characteristics of the tumors in addition to EphA2 over-expression may be important for predicting responsiveness to EphA2-directed therapies.


Subject(s)
Breast Neoplasms/metabolism , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Immunotherapy/methods , Mammary Neoplasms, Animal/metabolism , Receptor, EphA2/chemistry , Animals , Antibodies, Monoclonal/chemistry , Breast Neoplasms/therapy , Cell Line, Tumor , Colorectal Neoplasms/therapy , Female , Humans , Mammary Neoplasms, Animal/therapy , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Receptor, EphA2/immunology
10.
Am J Pathol ; 168(2): 608-20, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16436674

ABSTRACT

The mammary gland microenvironment during postlactational involution shares similarities with inflammation, including high matrix metalloproteinase activity, fibrillar collagen deposition, and release of bioactive fragments of fibronectin and laminin. Because inflammation can promote tumorigenesis, we evaluated whether the tissue microenvironment of the involuting gland is also promotional. Extracellular matrix was isolated from mammary glands of nulliparous rats or rats with mammary glands undergoing weaning-induced involution. Using these matrices as substratum, nulliparous matrix was found to promote ductal organization of normal mammary epithelial MCF-12A cells in three-dimensional culture and to suppress invasion of mammary tumor MDA-MB-231 cells in transwell filter assays. Conversely, involution matrix failed to support ductal development in normal cells and promoted invasiveness in tumor cells. To evaluate the effects of these matrices on metastasis in vivo, MDA-MB-231 cells, premixed with Matrigel, nulliparous matrix, or involution matrix, were injected into mammary fat pads of nude mice. Metastases to lung, liver, and kidney were increased in the involution matrix group, and correlated with a twofold increase in tumor vascular endothelial growth factor expression and increased angiogenesis. These data suggest that the mammary gland microenvironment becomes promotional for tumor cell dissemination during involution, thus providing a plausible mechanism to explain the high rate of metastases that occur with pregnancy-associated breast cancer.


Subject(s)
Breast Neoplasms/pathology , Kidney Neoplasms/secondary , Lactation/physiology , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Mammary Glands, Animal/physiology , Animals , Collagen/metabolism , Drug Combinations , Extracellular Matrix/pathology , Female , Humans , Kidney Neoplasms/pathology , Laminin/metabolism , Liver Neoplasms/pathology , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasm Invasiveness/pathology , Neovascularization, Pathologic , Pregnancy , Pregnancy, Animal , Proteoglycans/metabolism , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/metabolism
11.
Cancer Res ; 65(18): 8397-405, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16166318

ABSTRACT

Gene expression analysis showed that a human mindin homologue, mindin/RG-1, is expressed selectively in prostate tissues and that its expression level is elevated in some prostate tumors. Mindin/RG-1 protein expression is maintained in >80% of prostate cancers metastatic to bone or lymph nodes as well as in locally recurrent tumors in androgen-unresponsive patients. In contrast, mindin/RG-1 expression in other normal tissues is significantly lower than that seen in the prostate. A fully human antibody, 19G9, was generated against mindin/RG-1 protein and was shown to accumulate at high abundance in LNCaP tumor xenografts. Conjugates of this antibody with the chelator CHX-A''-DTPA were generated and radiolabeled with either 111In, 90Y, or 86Y. Small animal positron emission tomography imaging with the 86Y-radiolabeled conjugate showed very specific accumulation of the antibody in LNCaP tumor xenografts with clear tumor delineation apparent at 4 hours. The therapeutic efficacy of [90Y]-CHX-A''-DTPA-19G9 was evaluated in mice bearing LNCaP xenografts. A dose-finding study identified a nontoxic therapeutic dose to be approximately 75 microCi. Significant antitumor effects were seen with a single administration of radiolabeled antibody to animals bearing 200 to 400 mm3 tumors. Inhibition of tumor growth was observed in all treated animals over a 49-day period. At 49 days posttreatment, slow tumor growth recurred but this could be prevented for an additional 40-day period by a second administration of a 75 microCi dose at day 49. We conclude that [90Y]-CHX-A''-DTPA-19G9 is a novel antibody conjugate that has considerable promise for therapy of metastatic prostate cancer in androgen-unresponsive patients.


Subject(s)
Antibodies, Monoclonal/immunology , Extracellular Matrix Proteins/immunology , Immunotoxins/immunology , Prostatic Neoplasms/radiotherapy , Radioimmunotherapy/methods , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antibody Specificity , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , CHO Cells , Cricetinae , Dose-Response Relationship, Immunologic , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Humans , Immunotoxins/pharmacokinetics , Immunotoxins/pharmacology , Isothiocyanates/immunology , Isothiocyanates/pharmacokinetics , Isothiocyanates/pharmacology , Male , Molecular Sequence Data , Pentetic Acid/analogs & derivatives , Pentetic Acid/immunology , Pentetic Acid/pharmacokinetics , Pentetic Acid/pharmacology , Positron-Emission Tomography , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tissue Distribution , Xenograft Model Antitumor Assays , Yttrium Radioisotopes/administration & dosage , Yttrium Radioisotopes/pharmacokinetics , Yttrium Radioisotopes/pharmacology
12.
Cancer Res ; 65(7): 2846-53, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15805286

ABSTRACT

Radiotherapy is an effective approach for the treatment of local prostate cancer. However, once prostate cancer metastasizes, radiotherapy cannot be used due to the distribution of multiple metastases to lymph nodes and bones. In contrast, radioimmunotherapy should still be efficacious in metastatic prostate cancer as radioisotopes are brought to tumor cells by targeting antibodies. Here we identify and validate a prostate-expressed molecule, tomoregulin, as a target for radioimmunotherapy of prostate cancer. Tomoregulin is a transmembrane protein selectively expressed in the brain, prostate, and prostate cancer, but not expressed in other normal tissues. Immunohistochemical studies of tomoregulin protein in clinical samples show its location in the luminal epithelium of normal prostate, benign prostatic hyperplasia, and prostatic intraepithelial neoplasia. More importantly, the tomoregulin protein is expressed in primary prostate tumors and in their lymph node and bone metastases. The nature of tomoregulin as a transmembrane protein and its tissue-specific expression make tomoregulin an attractive target for radioimmunotherapy, in which tomoregulin-specific antibodies will deliver a radioisotope to prostate tumor cells and metastases. Indeed, biodistribution studies using a prostate tumor xenograft model showed that the (111)In-labeled anti-tomoregulin antibody 2H8 specifically recognizes tomoregulin protein in vivo, leading to a strong tumor-specific accumulation of the antibody. In efficacy studies, a single i.p. dose of 150 microCi (163 microg) (90)Y-labeled 2H8 substantially inhibits the growth rate of established LNCaP human prostate tumor xenograft in nude mice but produces no overt toxicity despite cross-reactivity of 2H8 with mouse tomoregulin. Our data clearly validate tomoregulin as a target for radioimmunotherapy of prostate cancer.


Subject(s)
Immunotoxins/therapeutic use , Indium Radioisotopes/therapeutic use , Membrane Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms/radiotherapy , Radioisotopes/therapeutic use , Ytterbium/therapeutic use , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Brain/metabolism , Cell Line, Tumor , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Prostate/metabolism , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Radioimmunotherapy , Radiopharmaceuticals/immunology , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Tissue Distribution , Xenograft Model Antitumor Assays
13.
J Biol Chem ; 280(20): 19867-74, 2005 May 20.
Article in English | MEDLINE | ID: mdl-15772071

ABSTRACT

The phosphoinositide 3-kinase/3-phosphoinositide-dependent kinase 1 (PDK1)/Akt signaling pathway plays a key role in cancer cell growth, survival, and tumor angiogenesis and represents a promising target for anticancer drugs. Here, we describe three potent PDK1 inhibitors, BX-795, BX-912, and BX-320 (IC(50) = 11-30 nm) and their initial biological characterization. The inhibitors blocked PDK1/Akt signaling in tumor cells and inhibited the anchorage-dependent growth of a variety of tumor cell lines in culture or induced apoptosis. A number of cancer cell lines with elevated Akt activity were >30-fold more sensitive to growth inhibition by PDK1 inhibitors in soft agar than on tissue culture plastic, consistent with the cell survival function of the PDK1/Akt signaling pathway, which is particularly important for unattached cells. BX-320 inhibited the growth of LOX melanoma tumors in the lungs of nude mice after injection of tumor cells into the tail vein. The effect of BX-320 on cancer cell growth in vitro and in vivo indicates that PDK1 inhibitors may have clinical utility as anticancer agents.


Subject(s)
Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Catalytic Domain , Cell Division/drug effects , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , HeLa Cells , Humans , In Vitro Techniques , Kinetics , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Mice , Mice, Nude , Models, Molecular , Molecular Structure , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Pyrimidines/chemistry , Pyrimidines/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
14.
Int J Cancer ; 114(6): 1002-9, 2005 May 10.
Article in English | MEDLINE | ID: mdl-15645422

ABSTRACT

1-(2-deoxy-2-fluoro-4-thio-beta-D-arabinofuranosyl) cytosine (4'-thio-FAC) is a deoxycytidine analog that has been shown previously to have impressive anti-proliferative and cytotoxic effects in vitro and in vivo toward colorectal and gastric tumors. In our present studies, the pharmacokinetic behavior in nude mice and the effectiveness of 4'-thio-FAC against human pancreatic and ovarian tumor growth were assessed in comparison with standard chemotherapeutic agents. Potent in vitro anti-proliferative effects were observed against pancreatic (Capan-1, MIA-PaCa-2, BxPC-3) and ovarian (SK-OV-3, OVCAR-3, ES-2) cancer cell lines with IC(50) of 0.01-0.2 microM. In vivo anti-tumor activity was evaluated in nude mice bearing subcutaneously (s.c.) implanted human pancreatic tumor xenografts or intraperitoneally (i.p.) disseminated human ovarian xenografted tumors. Oral daily administration of 4'-thio-FAC for 8-10 days significantly inhibited the growth of gemcitabine-resistant BxPC-3 pancreatic tumors and induced regression of gemcitabine-refractory Capan-1 tumors. 4'-Thio-FAC was also a highly effective inhibitor of ovarian peritoneal carcinomatosis. In the SK-OV-3 and ES-2 ovarian cancer models, 4'-thio-FAC prolonged survival to a greater extent than that observed with gemcitabine. Furthermore, the superiority of 4'-thio-FAC to carboplatin and paclitaxel was demonstrated in the ES-2 clear cell ovarian carcinoma model. Studies provide evidence that 4'-thio-FAC is a promising new alternative to gemcitabine and other chemotherapeutic drugs in the treatment of a variety of tumor indications, including pancreatic and ovarian carcinoma.


Subject(s)
Carcinoma/drug therapy , Cytarabine/analogs & derivatives , Cytarabine/pharmacology , Ovarian Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Animals , Carcinoma/pathology , Carcinoma/veterinary , Disease Models, Animal , Female , Mice , Mice, Nude , Ovarian Neoplasms/pathology , Ovarian Neoplasms/veterinary , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/veterinary , Transplantation, Heterologous , Tumor Cells, Cultured
15.
Int J Cancer ; 111(1): 32-42, 2004 Aug 10.
Article in English | MEDLINE | ID: mdl-15185340

ABSTRACT

In addition to antiviral effects, Type I interferons (IFN) have potent antiproliferative and immunomodulatory activities. Because of these properties IFNs have been evaluated as therapeutics for the treatment of a number of human diseases, including cancer. Currently, IFNs have been shown to be efficacious for the treatment of only a select number of cancers. The reason for this is unclear. Recent evidence has demonstrated that some cancer cell types seem to be defective in their ability to respond to IFN. It has been suggested that defects in IFN signaling is one mechanism by which cancer cells escape responsiveness to Type I IFNs and growth control in general. We report that transfection and enhanced expression of the Type I IFN receptor chain (IFNAR2c) in 3 different human cancer cell lines markedly increases the sensitivity of these cells to the antiproliferative effects of IFNs. In cancer cells transfected with IFNAR2c, dose response curves demonstrate a significant decrease in the concentrations of IFN required to achieve maximum cell death. Furthermore, in these transfected cells, we observe a significant increase in the number of cells undergoing apoptosis, as measured by DNA fragmentation and Caspase 3 activation. In addition, using an in vivo xenograft tumor model we show an increase in the effectiveness of systemically delivered Betaseron in decreasing tumor burden in animals in which solid tumors were generated from IFNAR2c transfected cells. These data show that specific regulation of IFN receptor expression can play a major role in determining the clinical outcome of IFN-based cancer therapeutics by regulating the relative sensitivity of cancer cells to IFN-dependent growth control.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Interferons/pharmacology , Receptors, Interferon/biosynthesis , Animals , Apoptosis , Caspase 3 , Caspases/pharmacology , Cell Line, Tumor , DNA Damage , Female , Genetic Therapy/methods , Humans , Mice , Mice, Nude , Receptor, Interferon alpha-beta , Receptors, Interferon/physiology , Signal Transduction , Transfection , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...