Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 198
Filter
1.
Oncogene ; 36(16): 2255-2264, 2017 04 20.
Article in English | MEDLINE | ID: mdl-27748766

ABSTRACT

Dysregulated activation of the CDK4/6 kinases is a hallmark of most mammary-derived carcinomas. ATP-competitive inhibitors against this complex have been recently advanced in the clinic and have shown significant activity, particularly against tumors driven by the estrogen receptor (ER). However, resistance to these compounds has begun to emerge often months to years after their initiation. We investigated potential mechanisms of resistance using cell line models that are highly sensitive to this class of drugs. After prolonged exposure to the selective and potent CDK4/6 inhibitor LY2835219, clones emerged and several were found to harbor amplification of the CDK6 kinase. Amplification of CDK6 resulted in a marked increase in CDK6 expression and reduced response of the CDK4/6 target, phospho-Rb (pRb), to CDK4/6 inhibitors. Knockdown of CDK6 restored drug sensitivity, while enforced overexpression of CDK6 was sufficient to mediate drug resistance. Not only did CDK6 overexpression mediate resistance to CDK4/6 inhibitors but it also led to reduced expression of the ER and progesterone receptor (PR), and diminished responsiveness to ER antagonism. The reduced ER/PR expression after CDK4/6 inhibitor resistance was additionally observed in tumor biopsy specimens from patients treated with these drugs. Alternative mechanisms of resistance to CDK4/6 inhibitors such as loss of pRb and cyclin E1 overexpression also exhibited decreased hormone responsiveness, suggesting that the clinical paradigm of sequential endocrine-based therapy may be ineffective in some settings of acquired CDK4/6 resistance.


Subject(s)
Breast Neoplasms/enzymology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Drug Resistance, Neoplasm , Protein Kinase Inhibitors/pharmacology , Receptors, Estrogen/metabolism , Aminopyridines/pharmacology , Benzimidazoles/pharmacology , Breast Neoplasms/diet therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase 6/genetics , Female , Gene Amplification , Humans , MCF-7 Cells , Signal Transduction/drug effects
2.
J Urol ; 190(5): 1846-51, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23727411

ABSTRACT

PURPOSE: Nephrectomy with lymph node sampling is the recommended treatment for children with unilateral Wilms tumor under the Children's Oncology Group protocols. Using radiological assessment, we determined the feasibility of performing partial nephrectomy in a select group of patients with very low risk unilateral Wilms tumor. MATERIALS AND METHODS: We reviewed imaging studies of 60 patients with a mean age of less than 2 years with very low risk unilateral Wilms tumor (mean weight less than 550 gm) to assess the feasibility of partial nephrectomy. We evaluated percentage of salvageable parenchyma, tumor location and anatomical features preventing a nephron sparing approach. RESULTS: A linear relationship exists between tumor weight and computerized tomography estimated tumor volume. Mean tumor weight in the study population was 315 gm. Partial nephrectomy was deemed feasible in only 5 of 60 patients (8%). CONCLUSIONS: When considering a select population with very low risk unilateral Wilms tumor (lower volume tumor), only a small percentage of nonpretreated patients are candidates for nephron sparing surgery.


Subject(s)
Kidney Neoplasms/diagnostic imaging , Kidney Neoplasms/surgery , Nephrectomy/methods , Wilms Tumor/diagnostic imaging , Wilms Tumor/surgery , Feasibility Studies , Female , Humans , Infant , Male , Organ Sparing Treatments , Radiography , Risk Assessment
5.
Oncogene ; 30(22): 2547-57, 2011 Jun 02.
Article in English | MEDLINE | ID: mdl-21278786

ABSTRACT

There is a strong rationale to therapeutically target the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway in breast cancer since it is highly deregulated in this disease and it also mediates resistance to anti-HER2 therapies. However, initial studies with rapalogs, allosteric inhibitors of mTORC1, have resulted in limited clinical efficacy probably due to the release of a negative regulatory feedback loop that triggers AKT and ERK signaling. Since activation of AKT occurs via PI3K, we decided to explore whether PI3K inhibitors prevent the activation of these compensatory pathways. Using HER2-overexpressing breast cancer cells as a model, we observed that PI3K inhibitors abolished AKT activation. However, PI3K inhibition resulted in a compensatory activation of the ERK signaling pathway. This enhanced ERK signaling occurred as a result of activation of HER family receptors as evidenced by induction of HER receptors dimerization and phosphorylation, increased expression of HER3 and binding of adaptor molecules to HER2 and HER3. The activation of ERK was prevented with either MEK inhibitors or anti-HER2 monoclonal antibodies and tyrosine kinase inhibitors. Combined administration of PI3K inhibitors with either HER2 or MEK inhibitors resulted in decreased proliferation, enhanced cell death and superior anti-tumor activity compared with single agent PI3K inhibitors. Our findings indicate that PI3K inhibition in HER2-overexpressing breast cancer activates a new compensatory pathway that results in ERK dependency. Combined anti-MEK or anti-HER2 therapy with PI3K inhibitors may be required in order to achieve optimal efficacy in HER2-overexpressing breast cancer. This approach warrants clinical evaluation.


Subject(s)
Breast Neoplasms/drug therapy , Extracellular Signal-Regulated MAP Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/therapeutic use , Receptor, ErbB-2/metabolism , Breast Neoplasms/enzymology , Cell Line, Tumor , Female , Humans , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-3/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors
6.
Skin Therapy Lett ; 15(5): 5-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20505896

ABSTRACT

An actinic keratosis (AK) is a pre-malignant cutaneous lesion that frequently manifests in sun-exposed areas of the skin as a small, rough, scaly erythematous papule. They are one of the most common presenting complaints for dermatologists. AKs should be treated due to their potential to progress into a squamous cell carcinoma (SCC). There are numerous treatments available for managing AKs including those broadly categorized as destructive, topical field, and procedural field therapies. The topical field therapies include 5-fluorouracil, imiquimod, and diclofenac gel. Recently, imiquimod 3.75% (Zyclara TM) has been approved for the treatment AKs on the face and scalp. It is a reasonable alternative to imiquimod 5%, as the approved indication includes a larger surface area for treatment, shorter duration course, and the potential for less severe local skin reactions. There is no widely accepted algorithm for the treatment of AKs, as comparative data is unavailable between all approaches. Therapy choices are guided by efficacy, adverse effects, cosmetic results, and patient compliance.


Subject(s)
Carcinoma, Squamous Cell/prevention & control , Keratosis, Actinic/therapy , Skin Neoplasms/prevention & control , Administration, Cutaneous , Aminoquinolines/administration & dosage , Aminoquinolines/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/etiology , Diclofenac/administration & dosage , Diclofenac/therapeutic use , Fluorouracil/administration & dosage , Fluorouracil/therapeutic use , Humans , Imiquimod , Keratosis, Actinic/complications , Keratosis, Actinic/physiopathology , Photochemotherapy/methods , Skin Neoplasms/etiology
7.
Oncogene ; 29(3): 325-34, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-19855434

ABSTRACT

The anti-HER2 antibody Trastuzumab (Herceptin) has been proven to be effective in the treatment of HER2-overexpressing breast cancer; resistance, however, invariably emerges in metastatic tumors. The expression of p95-HER2, a form of HER2 with a truncated extracellular domain that lacks the Trastuzumab binding epitope, has been implicated as a mechanism of resistance to the antibody. We utilized an in vivo tumor model that overexpresses p95-HER2 and showed it to be resistant to the signaling and antitumor effects of Trastuzumab. We find that both full-length and p95-HER2 interact with the HSP90 chaperone protein and are degraded in tumor cells exposed to HSP90 inhibitors in tissue culture and in vivo. Loss of expression of p95-HER2 is accompanied by downregulation of the phosphoinositide-3 kinase/AKT and extracellular signal-regulated kinase signaling pathways and inhibition of cell proliferation. Chronic administration of HSP90 inhibitors in vivo results in sustained loss of HER2 and p95-HER2 expression and inhibition of AKT activation, together with induction of apoptosis and complete inhibition of tumor growth in Trastuzumab-resistant, p95-HER2-overexpressing models. Thus, p95-HER2 is an HSP90 client protein, the expression and function of which can be effectively suppressed in vivo by HSP90 inhibitors. HSP90 inhibition is therefore a potentially effective therapeutic strategy for p95-HER2-mediated Trastuzumab-resistant breast cancer.


Subject(s)
Antibodies, Monoclonal/pharmacology , HSP90 Heat-Shock Proteins/metabolism , Neoplasms/drug therapy , Receptor, ErbB-2/metabolism , 3T3 Cells , Animals , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Binding Sites/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Immunoblotting , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Neoplasms/metabolism , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/genetics , Signal Transduction/drug effects , Trastuzumab , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
8.
Curr Cancer Drug Targets ; 8(6): 522-32, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18781898

ABSTRACT

Hsp90 (heat shock protein 90) is a molecular chaperone that modulates the stability and/or transport of a diverse set of critical cellular regulatory, metabolism, organization, and signaling proteins. Binding to Hsp90 is required for normal function of many proteins. In addition, Hsp90 has an extra-cellular function. It is found in two isotypes: alpha which is inducible and beta which is constitutive. Tumor cells frequently over express Hsp90alpha, and Hsp90 is implicated in cancer progression. Hence Hsp90 has emerged as a potential target for cancer treatment. A variety of agents have been found to interfere with Hsp function, mainly by binding to an ATP binding site on the molecule. More recent agents interfere with protein binding or the dimerization of Hsp90 needed for function. Preclinical studies have demonstrated that disruption of the many client proteins chaperoned by Hsp90 is achievable and associated with significant growth inhibition, both in vitro and in tumor xenografts. As a result, agents which interfere with this protein's function are being tested in the clinic as a targeted method of interfering with malignant growth. We review the current clinical status of therapeutic efforts to perturb this pathway and discuss future directions.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Animals , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/chemistry , Humans , Immunotherapy , Neoplasms/metabolism , Neoplasms/therapy , Protein Binding , Protein Conformation
10.
Proc Natl Acad Sci U S A ; 103(1): 57-62, 2006 Jan 03.
Article in English | MEDLINE | ID: mdl-16371460

ABSTRACT

The Raf family includes three members, of which B-Raf is frequently mutated in melanoma and other tumors. We show that Raf-1 and A-Raf require Hsp90 for stability, whereas B-Raf does not. In contrast, mutated, activated B-Raf binds to an Hsp90-cdc37 complex, which is required for its stability and function. Exposure of melanoma cells and tumors to the Hsp90 inhibitor 17-allylamino-17-demethoxygeldanamycin results in the degradation of mutant B-Raf, inhibition of mitogen-activated protein kinase activation and cell proliferation, induction of apoptosis, and antitumor activity. These data suggest that activated mutated B-Raf proteins are incompetent for folding in the absence of Hsp90, thus suggesting that the chaperone is required for the clonal evolution of melanomas and other tumors that depend on this mutation. Hsp90 inhibition represents a therapeutic strategy for the treatment of melanoma.


Subject(s)
Cell Cycle Proteins/metabolism , Chaperonins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Melanoma/metabolism , Mutation, Missense/genetics , Proto-Oncogene Proteins B-raf/metabolism , Animals , Apoptosis/drug effects , Benzoquinones , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Chaperonins/antagonists & inhibitors , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/drug effects , Female , Humans , Lactams, Macrocyclic , Melanoma/genetics , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins A-raf/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Rifabutin/analogs & derivatives , Rifabutin/pharmacology
11.
Cytotherapy ; 7(4): 374-84, 2005.
Article in English | MEDLINE | ID: mdl-16162460

ABSTRACT

BACKGROUND: The use of myeloma Ag-loaded mature DC vaccines, cryopreserved in single-use aliquots, is an attractive immunotherapeutic strategy. In this study we investigated the retention of phenotype, viability and potency of DC vaccines after freezing and thawing. METHODS: Plastic-adherent monocytes, derived from a steady-state leukapheresis, were cultured in serum-free media containing GM-CSF and IL-4. DC were loaded on day 6 with myeloma lysate (ML) or idiotype (Id) Ag and keyhole limpet hemocyanin (KLH), induced to mature on day 7 with CD40-ligand and cryopreserved on day 9. Seventeen clinical-scale cultures were evaluated for DC yield, recovery and immunophenotype after potency was validated with allogeneic mixed lymphocyte culture and Ag presentation assays. RESULTS: We produced 88 individual vaccines from 17 clinical-scale cultures. Median DC yield at harvest was 131 x 10(6) (range 37-375 x 10(6)) and median recovery of viable DC after thawing was 69% (range 11-100%). We confirmed viability (7AAD-), phenotype (CD14-, CD83+/CD40+, CD83+/CD80+, CD83+/CD86+, CD83+/CD54+, HLA-DR++) and the ability of the DC to present Ag and stimulate allogeneic T cells post-thawing. DISCUSSION: We have validated a serum-free culture system for the production of DC. Cryopreservation did not interfere with DC activity, allowed time for rigorous quality control (QC) and flexible scheduling of intranodal vaccination, and reduced the time to prepare multiple vaccines.


Subject(s)
Antigens, Neoplasm , Cancer Vaccines , Cryopreservation , Dendritic Cells , Multiple Myeloma/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Antigens, Neoplasm/therapeutic use , Cancer Vaccines/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Leukapheresis , Multiple Myeloma/drug therapy , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Neoplasm Proteins/therapeutic use , Quality Control
12.
Article in English | MEDLINE | ID: mdl-16869774

ABSTRACT

Over the past decade, mouse models of cancer have come to resemble human disease much more closely than simple subcutaneous or orthotopic systems. Intervention strategies that work on these new model systems are more likely to have an impact clinically. We have shown recently that antiangiogenic stress imposed by loss of Id protein in endothelial progenitor cells results in dramatic central necrosis in breast tumors initiated in mice by overexpression of the her2/neu oncogene. Tumor cells remain viable at the periphery, perhaps via the hypoxic response pathway which allows the lesions to expand. Inhibition of this pathway by the inactivation of the Hif-1alpha chaperone Hsp90 in combination with antiangiogenic stress leads to the first reported complete regression of these aggressive breast tumors.


Subject(s)
Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/therapy , Angiogenesis Inhibitors/therapeutic use , Animals , Benzoquinones/therapeutic use , Cell Hypoxia , Female , Genes, erbB-2 , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inhibitor of Differentiation Proteins/deficiency , Inhibitor of Differentiation Proteins/genetics , Lactams, Macrocyclic/therapeutic use , Mammary Neoplasms, Experimental/blood supply , Mice , Mice, Knockout , Mice, Transgenic
13.
Hum Gene Ther ; 14(14): 1333-45, 2003 Sep 20.
Article in English | MEDLINE | ID: mdl-14503968

ABSTRACT

Tumor-specific genes delivered to dendritic cells (DCs) have been used for the generation of cytotoxic T cells (CTLs), but their application has been limited on the one hand by low viral titers resulting in low transduction efficiency and poor protein production, and on the other hand by immunogenicity of the selectable marker and poor viability of the DCs. We addressed these limitations by creating a multipurpose master vector (pMV) and cloning the tumor gene NY-ESO-1, which is highly expressed in more than 50% of advanced myeloma patients. pMV was constructed from a Moloney murine leukemia virus (Mo-MuLV)-based retroviral backbone with the following features: (1) an extended packaging signal to achieve high viral titers, (2) a splice acceptor region to facilitate protein production, (3) a nonimmunogenic selectable marker, dihydrofolate reductase-L22Y (DHFR(L22Y)), to exclude the generation of CTLs against the selectable marker, (4) an internal ribosomal entry site between the tumor-specific gene (NY-ESO-1) and the selectable marker DHFR(L22Y) for coexpression of two heterologous gene products from a single bicistronic mRNA, minimizing the possibility of differential expression of these two genes, and (5) human granulocyte-macrophage colony-stimulating factor (hGM-CSF) cDNA driven by the human T-lymphotropic virus promoter to enhance DC function and viability. Recombinant virus of pMV-NY-ESO-1 was generated with vesicular stomatitis virus G envelope protein (VSV-G) in the GP2-293 cell line for efficient transduction. We present evidence that the DC phenotype is unaltered after transduction and that more than 85% of DCs express NY-ESO-1, which secrete approximately 40 ng of GM-CSF per 10(6) DCs.


Subject(s)
Antigens, Neoplasm/metabolism , Dendritic Cells/metabolism , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Membrane Proteins/metabolism , Testis/metabolism , Antigens, CD34/metabolism , Antigens, Neoplasm/analysis , Antigens, Neoplasm/genetics , Gene Transfer Techniques , Genetic Markers , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , HeLa Cells , Humans , Immunohistochemistry , Male , Moloney murine leukemia virus/genetics , Multiple Myeloma/metabolism , Promoter Regions, Genetic , RNA, Messenger/analysis , Retroviridae/genetics , Transduction, Genetic
14.
Cancer Res ; 61(23): 8492-7, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11731433

ABSTRACT

Histone deacetylase (HDACs) regulate histone acetylation by catalyzing the removal of acetyl groups on the NH(2)-terminal lysine residues of the core nucleosomal histones. Modulation of the acetylation status of core histones is involved in the regulation of the transcriptional activity of certain genes. HDAC activity is generally associated with transcriptional repression. Aberrant recruitment of HDAC activity has been associated with the development of certain human cancers. We have developed a class of HDAC inhibitors, such as suberoylanilide hydroxamic acid (SAHA), that were initially identified based on their ability to induce differentiation of cultured murine erythroleukemia cells. Additional studies have demonstrated that SAHA inhibits the growth of tumors in rodents. In this study we have examined the effects of SAHA on MCF-7 human breast cancer cells. We found that SAHA causes the inhibition of proliferation, accumulation of cells in a dose-dependent manner in G(1) then G(2)-M phase of the cell cycle, and induction of milk fat globule protein, milk fat membrane globule protein, and lipid droplets. Growth inhibition was associated with morphological changes including the flattening and enlargement of the cytoplasm, and a decrease in the nuclear:cytoplasmic ratio. Withdrawal of SAHA led to reentry of cells into the cell cycle and reversal to a less differentiated phenotype. SAHA induced differentiation in the estrogen receptor-negative cell line SKBr-3 and the retinoblastoma-negative cell line MDA-468. We propose that SAHA has profound antiproliferative activity by causing these cells to undergo cell cycle arrest and differentiation that is dependent on the presence of SAHA. SAHA and other HDAC inhibitors are currently in Phase I clinical trials. These findings may impact the clinical use of these drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Breast Neoplasms/metabolism , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line, Transformed , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Histone Deacetylase Inhibitors , Humans , Lipids/biosynthesis , Milk Proteins/biosynthesis , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/physiology , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/physiology , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/physiology , Vorinostat
16.
Cancer Res ; 61(19): 7184-8, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11585753

ABSTRACT

The epidermal growth factor receptor (EGFR) is commonly overexpressed in many human tumors and provides a new target for anticancer drug development. ZD1839 ("Iressa"), a quinazoline tyrosine kinase inhibitor selective for the EGFR, has shown good activity in preclinical studies and in the early phase of clinical trials. However, because it remains unclear which tumor types are the best targets for treatment with this agent, the molecular characteristics that correlate with tumor sensitivity to ZD1839 have been studied. In a panel of human breast cancer and other epithelial tumor cell lines, HER2-overexpressing tumors were particularly sensitive to ZD1839. Growth inhibition of these tumor cell lines was associated with the dephosphorylation of EGFR, HER2, and HER3, accompanied by the loss of association of HER3 with phosphatidylinositol 3-kinase, and down-regulation of Akt activity. These studies suggest that HER2-overexpressing tumors are particularly susceptible to the inhibition of HER family tyrosine kinase signaling and suggest novel strategies to treat these particularly aggressive tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Serine-Threonine Kinases , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Division/physiology , Down-Regulation/drug effects , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/biosynthesis , Gefitinib , Humans , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt , Quinazolines/administration & dosage , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Trastuzumab , Tumor Cells, Cultured
18.
Clin Cancer Res ; 7(8): 2228-36, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489796

ABSTRACT

17-allyl-aminogeldanamycin (17-AAG) is an ansamycin antibiotic that binds to a highly conserved pocket in the Hsp90 chaperone protein and inhibits its function. Hsp90 is required for the refolding of proteins during cellular stress and the conformational maturation of certain signaling proteins. 17-AAG has antitumor activity in cell culture and animal xenograft models and is currently in clinical trial. It causes an RB-dependent G(1) arrest, differentiation, and apoptosis. RB-negative cells arrest in mitosis and undergo apoptosis. Hsp90 plays an important role in the cellular response to environmental stress. Therefore, we tested whether the regulation of Hsp90 function by 17-AAG could sensitize cells to cytotoxic agents. 17-AAG sensitized tumor cells to Taxol and doxorubicin. Taxanes cause growth arrest in mitosis and apoptosis. The addition of 17-AAG to cells after exposure to Taxol significantly increased both the activation of caspases 9 and 3 and apoptosis. In cells with intact RB, exposure to 17-AAG before Taxol resulted in G(1) arrest and abrogated apoptosis. Schedule dependence was not seen in cells with mutated RB, because both agents blocked cells in mitosis. Schedule- or RB-dependence was also not observed when cells were treated with 17-AAG and doxorubicin, a DNA-intercalating agent that acts on different phases of the cell cycle. These findings suggest that inhibition of Hsp90 function by 17-AAG enhances the apoptotic effects of cytotoxic agents. The sequence of drug administration and the RB status significantly influence efficacy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , HSP90 Heat-Shock Proteins/drug effects , Retinoblastoma Protein/physiology , Antineoplastic Agents, Phytogenic/pharmacology , Benzoquinones , Breast Neoplasms/drug therapy , Breast Neoplasms/physiopathology , Caspase 3 , Caspase 9 , Caspases/drug effects , Caspases/metabolism , Cell Division/drug effects , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Drug Synergism , Enzyme Activation/drug effects , HSP90 Heat-Shock Proteins/physiology , Humans , Lactams, Macrocyclic , Mitosis/physiology , Paclitaxel/pharmacology , Retinoblastoma Protein/genetics , Rifabutin/analogs & derivatives , Rifabutin/pharmacology , Time Factors , Tumor Cells, Cultured/drug effects
19.
Chem Biol ; 8(3): 289-99, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11306353

ABSTRACT

BACKGROUND: The Hsp90s contain a conserved pocket that binds ATP/ADP and plays an important role in the regulation of chaperone function. Occupancy of this pocket by several natural products (geldanamycin (GM) and radicicol) alters Hsp90 function and results in the degradation of a subset of proteins (i.e. steroid receptors, Her2, Raf). We have used the structural features of this pocket to design a small molecule inhibitor of Hsp90. RESULTS: The designed small molecule PU3 competes with GM for Hsp90 binding with a relative affinity of 15-20 microM. PU3 induces degradation of proteins, including Her2, in a manner similar to GM. Furthermore, PU3 inhibits the growth of breast cancer cells causing retinoblastoma protein hypophosphorylation, G1 arrest and differentiation. CONCLUSIONS: PU3 is representative of a novel class of synthetic compounds that binds to Hsp90 and inhibits the proliferation of cancer cells. These reagents could provide a new strategy for the treatment of cancers.


Subject(s)
Adenine Nucleotides/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Drug Design , HSP90 Heat-Shock Proteins/metabolism , Receptor, ErbB-2/drug effects , Benzoquinones , Binding, Competitive , Cell Differentiation/drug effects , Female , Humans , Lactams, Macrocyclic , Protein Binding , Quinones/metabolism , Receptor, ErbB-2/metabolism , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...