Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
2.
Ann Oncol ; 27(5): 947-52, 2016 05.
Article En | MEDLINE | ID: mdl-26903311

BACKGROUND: Treatment options for sarcoma are limited. Histone deacetylase inhibitors increase the efficacy of topoisomerase II inhibitors by promoting access to chromatin and by down-regulating DNA repair. Thus, combined panobinostat and epirubicin therapy was evaluated to treat refractory sarcoma. PATIENTS AND METHODS: Patients with advanced solid tumors were enrolled in a 3 + 3 dose-escalation phase I trial of panobinostat given on days 1, 3, and 5 followed by 75 mg/m(2) of epirubicin on day 5 in 21-day cycles, with a dose expansion at maximum tolerated dose (MTD) in 20 sarcoma patients. Peripheral blood mononucleocyte histone acetylation was also evaluated. RESULTS: Forty patients received 20-60 mg panobinostat. Dose-limiting toxicities included thrombocytopenia, febrile neutropenia, and fatigue at 60 mg, defining a panobinostat MTD at 50 mg. Four responses were seen in 37 assessable patients, all after progression on prior topoisomerase II inhibitors. For those with sarcoma, 12 of 20 derived clinical benefit (1 partial response and 11 stable disease, median overall survival 8.3 months), including 8 of 14 previously progressed on topoisomerase II therapy. Treatment benefits correlated with increased histone acetylation and decreased neutrophil count on day 5. CONCLUSIONS: Panobinostat and epirubicin treatment is well tolerated and may reverse anthracycline resistance. Changes in histone acetylation and associated decrease in neutrophil count correlated with clinical benefit and warrant investigation as predictive biomarkers. CLINICAL TRIAL: This trial is registered at www.Clinicaltrials.gov, Identifier: NCT00878904.


Drug Therapy, Combination , Epirubicin/administration & dosage , Hydroxamic Acids/administration & dosage , Indoles/administration & dosage , Sarcoma/drug therapy , Adult , Aged , Chromatin/drug effects , DNA Repair/drug effects , Dose-Response Relationship, Drug , Epirubicin/adverse effects , Female , Histone Deacetylase Inhibitors/administration & dosage , Humans , Hydroxamic Acids/adverse effects , Indoles/adverse effects , Male , Maximum Tolerated Dose , Middle Aged , Panobinostat , Sarcoma/genetics , Sarcoma/pathology , Topoisomerase II Inhibitors/administration & dosage
3.
Eur J Cancer ; 51(14): 1865-73, 2015 Sep.
Article En | MEDLINE | ID: mdl-26199039

BACKGROUND: The phosphatidylinositol 3-kinase/protein kinase-B/mammalian target of rapamycin (PI3K-AKT-mTOR) signalling pathway is aberrantly activated in several cancers. Notch signalling maintains cell proliferation, growth and metabolism in part by driving the PI3K pathway. Combining the mTOR inhibitor ridaforolimus with the Notch inhibitor MK-0752 may increase blockade of the PI3K pathway. METHODS: This phase I dose-escalation study (NCT01295632) aimed to define the dose-limiting toxicities (DLTs) and maximum tolerated dose (MTD) of combination oral ridaforolimus (rising doses starting at 20 mg, 5 days/week) and oral MK-0752 (1800 mg once weekly) in patients with solid tumours. No intrapatient dose escalation was permitted. RESULTS: Twenty eight patients were treated on study. Ridaforolimus doses were escalated from 20 to 30 mg/day. Among 14 evaluable patients receiving ridaforolimus 20 mg, one DLT (grade 2 stomatitis, second episode) was reported. Among eight evaluable patients receiving ridaforolimus 30 mg, three DLTs were reported (one each grade 3 stomatitis, grade 3 diarrhoea, and grade 3 asthenia). The MTD was 20 mg daily ridaforolimus 5 days/week+1800 mg weekly MK-0752. The most common drug-related adverse events included stomatitis, diarrhoea, decreased appetite, hyperglycaemia, thrombocytopenia, asthenia and rash. Two of 15 (13%) patients with head and neck squamous cell carcinoma (HNSCC) had responses: one with complete response and one with partial response. In addition, one patient experienced stable disease ⩾6 months. CONCLUSIONS: Combined ridaforolimus and MK-0752 showed activity in HNSCC. However, a high number of adverse events were reported at the MTD, which would require careful management during future clinical development.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzene Derivatives/administration & dosage , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Drug Administration Schedule , Europe , Female , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/pathology , Humans , Male , Maximum Tolerated Dose , Middle Aged , Molecular Targeted Therapy , Multimodal Imaging/methods , Positron-Emission Tomography , Propionates/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Signal Transduction/drug effects , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Squamous Cell Carcinoma of Head and Neck , Sulfones/administration & dosage , Time Factors , Tomography, X-Ray Computed , Treatment Outcome , United States
4.
Ann Oncol ; 24(7): 1900-1907, 2013 Jul.
Article En | MEDLINE | ID: mdl-23519998

BACKGROUND: Based upon preclinical evidence for improved antitumor activity in combination, this phase I study investigated the maximum-tolerated dose (MTD), safety, activity, pharmacokinetics (PK), and biomarkers of the mammalian target of rapamycin inhibitor, temsirolimus, combined with sorafenib in hepatocellular carcinoma (HCC). PATIENTS AND METHODS: Patients with incurable HCC and Child Pugh score ≤B7 were treated with sorafenib plus temsirolimus by 3 + 3 design. The dose-limiting toxicity (DLT) interval was 28 days. The response was assessed every two cycles. PK of temsirolimus was measured in a cohort at MTD. RESULTS: Twenty-five patients were enrolled. The MTD was temsirolimus 10 mg weekly plus sorafenib 200 mg twice daily. Among 18 patients at MTD, DLT included grade 3 hand-foot skin reaction (HFSR) and grade 3 thrombocytopenia. Grade 3 or 4 related adverse events at MTD included hypophosphatemia (33%), infection (22%), thrombocytopenia (17%), HFSR (11%), and fatigue (11%). With sorafenib, temsirolimus clearance was more rapid (P < 0.05). Two patients (8%) had a confirmed partial response (PR); 15 (60%) had stable disease (SD). Alpha-fetoprotein (AFP) declined ≥50% in 60% assessable patients. CONCLUSION: The MTD of sorafenib plus temsirolimus in HCC was lower than in other tumor types. HCC-specific phase I studies are necessary. The observed efficacy warrants further study.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , alpha-Fetoproteins/metabolism , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Biomarkers/blood , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/pathology , Disease-Free Survival , Drug Administration Schedule , Female , Humans , Liver Neoplasms/blood , Liver Neoplasms/pathology , Male , Maximum Tolerated Dose , Middle Aged , Neoplastic Cells, Circulating , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Protein Precursors/blood , Prothrombin , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Sorafenib , Treatment Outcome
5.
Br J Cancer ; 106(1): 85-91, 2012 Jan 03.
Article En | MEDLINE | ID: mdl-22127285

BACKGROUND: Src inhibitors sensitise melanoma cells to chemotherapy in preclinical models. The combination of dasatinib and dacarbazine was tested in a phase I trial in melanoma. METHODS: Patients had ECOG performance status 0-2 and normal organ function. Dacarbazine was administered on day 1 and dasatinib on day 2 through 19 of each 21-day cycle. Both were escalated from 50 mg b.i.d. of dasatinib and 800 mg m(-2) of dacarbazine. Available pre-treatment biopsies were sequenced for BRAF, NRAS, and C-Kit mutations. RESULTS: Dose-limiting toxicity was reached at dasatinib 70 mg b.i.d./dacarbazine 1000 mg m(-2), and was predominantly haematological. In 29 patients receiving dasatinib 70 mg b.i.d., the objective response rate (ORR) was 13.8%, the clinical benefit rate (ORR+SD) was 72.4%, the 6-month progression-free survival (PFS) was 20.7%, and the 12-month overall survival (OS) was 34.5%. Two out of three patients who were wild type for BRAF, NRAS, and c-KIT mutations had confirmed partial responses, and one had a minor response. CONCLUSION: The recommended phase II dose is dasatinib 70 mg b.i.d with dacarbazine 800 mg m(-2). PFS and OS data for dasatinib at 70 mg b.i.d. with dacarbazine compared favourably with historical controls. Preliminary data support evaluating tumour mutation status further as a biomarker of response.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melanoma/drug therapy , Neoplasm Metastasis , src-Family Kinases/antagonists & inhibitors , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Dacarbazine/adverse effects , Dasatinib , Female , Humans , Male , Melanoma/pathology , Middle Aged , Pyrimidines/adverse effects , Survival Analysis , Thiazoles/adverse effects , Treatment Outcome
6.
Ann Oncol ; 23(3): 610-617, 2012 Mar.
Article En | MEDLINE | ID: mdl-21700731

BACKGROUND: This phase II study evaluated single-agent bosutinib in pretreated patients with locally advanced or metastatic breast cancer. PATIENTS AND METHODS: Patients received oral bosutinib 400 mg/day. The primary end point was the progression-free survival (PFS) rate at 16 weeks. Secondary end points included objective response rate, clinical benefit rate, 2-year overall survival rate, safety, and changes in levels of bone resorption/formation biomarkers. RESULTS: Seventy-three patients were enrolled and treated. Median time from diagnosis of metastatic disease to initiation of bosutinib treatment was 24.5 months. For the intent-to-treat population, the PFS rate at 16 weeks was 39.6%. Unexpectedly, all responding patients (n = 4) were hormone receptor positive. The clinical benefit rate was 27.4%. The 2-year overall survival rate was 26.4%. The main toxic effects were diarrhea (66%), nausea (55%), and vomiting (47%). Grade 3-4 laboratory aminotransferase elevations occurred in 14 (19%) patients. Myelosuppression was minimal. No consistent changes in the levels of bone resorption/formation biomarkers were seen. CONCLUSIONS: Bosutinib showed promising efficacy in prolonging time to progression in chemotherapy-pretreated patients with locally advanced or metastatic breast cancer. Bosutinib was generally well tolerated, with a safety profile different from that of the Src/Abl tyrosine kinase inhibitor dasatinib in a similar patient population.


Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Nitriles/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Quinolines/therapeutic use , Salvage Therapy/methods , Adult , Aged , Bone Remodeling/drug effects , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Metastasis/drug therapy , Neoplasm Staging , Protein-Tyrosine Kinases/antagonists & inhibitors
7.
Br J Cancer ; 104(12): 1828-35, 2011 Jun 07.
Article En | MEDLINE | ID: mdl-21559012

BACKGROUND: Histone deacetylases (HDACs) are crucial components of the oestrogen receptor (ER) transcriptional complex. Preclinically, HDAC inhibitors can reverse tamoxifen/aromatase inhibitor resistance in hormone receptor-positive breast cancer. This concept was examined in a phase II combination trial with correlative end points. METHODS: Patients with ER-positive metastatic breast cancer progressing on endocrine therapy were treated with 400 mg of vorinostat daily for 3 of 4 weeks and 20 mg tamoxifen daily, continuously. Histone acetylation and HDAC2 expression in peripheral blood mononuclear cells were also evaluated. RESULTS: In all, 43 patients (median age 56 years (31-71)) were treated, 25 (58%) received prior adjuvant tamoxifen, 29 (67%) failed one prior chemotherapy regimen, 42 (98%) progressed after one, and 23 (54%) after two aromatase inhibitors. The objective response rate by Response Evaluation Criteria in Solid Tumours criteria was 19% and the clinical benefit rate (response or stable disease >24 weeks) was 40%. The median response duration was 10.3 months (confidence interval: 8.1-12.4). Histone hyperacetylation and higher baseline HDAC2 levels correlated with response. CONCLUSION: The combination of vorinostat and tamoxifen is well tolerated and exhibits encouraging activity in reversing hormone resistance. Correlative studies suggest that HDAC2 expression is a predictive marker and histone hyperacetylation is a useful pharmacodynamic marker for the efficacy of this combination.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Estrogen Antagonists/administration & dosage , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Tamoxifen/therapeutic use , Acetylation , Adult , Aged , Breast Neoplasms/mortality , Female , Histone Deacetylase 2/analysis , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/adverse effects , Histones/metabolism , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/adverse effects , Middle Aged , Tamoxifen/administration & dosage , Tamoxifen/adverse effects , Vorinostat
8.
Br J Cancer ; 101(7): 1044-50, 2009 Oct 06.
Article En | MEDLINE | ID: mdl-19738609

BACKGROUND: Histone deacetylase inhibitors (HDACi) can sensitise cancer cells to topoisomerase inhibitors by increasing their access and binding to DNA. METHODS: This phase I trial was designed to determine the toxicity profile, tolerability, and recommended phase II dose of escalating doses of the HDACi vorinostat, with weekly doxorubicin. RESULTS: In total, 32 patients were treated; vorinostat was dosed at 400, 600, 800, or 1000 mg day(-1) on days 1-3, followed by doxorubicin (20 mg m(-2)) on day 3 for 3 of 4 weeks. Maximal tolerated dose was determined to be 800 mg day(-1) of vorinostat. Dose-limiting toxicities were grade 3 nausea/vomiting (two out of six) and fatigue (one out of six) at 1000 mg day(-1). Non-dose-limiting grade 3/4 toxicities included haematological toxicity and venous thromboembolism. Antitumor activity in 24 evaluable patients included two partial responses (breast and prostate cancer). Two patients with melanoma had stable disease for > or =8 months. Histone hyperacetylation changes in peripheral blood mononuclear and tumour cells were comparable. Histone hyperacetylation seemed to correlate with pre-treatment HDAC2 expression. CONCLUSION: These findings suggest that vorinostat can be combined with weekly doxorubicin in this schedule at a dose of 800 mg day(-1). The HDAC2 expression may be a marker predictive of HDAC inhibition. Antitumor activity of this regimen in breast cancer, prostate cancer, and melanoma seems interesting.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Histone Deacetylase Inhibitors , Neoplasms/drug therapy , Acetylation , Adult , Aged , Aged, 80 and over , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Female , Heart/drug effects , Histone Deacetylases/analysis , Histones/metabolism , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/adverse effects , Male , Middle Aged , Vorinostat
9.
Cancer Res ; 61(23): 8492-7, 2001 Dec 01.
Article En | MEDLINE | ID: mdl-11731433

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.


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
10.
Breast Cancer Res ; 3(6): 361-4, 2001.
Article En | MEDLINE | ID: mdl-11737886

One of the major challenges of early-stage breast cancer is to select the adjuvant therapy that ensures the most benefits and the least harm for the patient. The definition of accurate predictive factors is therefore of paramount importance. So far the choice of adjuvant therapy has been based on the number of affected lymph nodes and the hormone receptor status of the patient. This paper evaluates the use of other tumor-related markers as predictive factors for adjuvant therapy. These include HER2, p53 and Bcl-2, cathepsin B, p27, proliferating cell nuclear antigen (PCNA), cyclin D, Ki-67, and vascular endothelial growth factor (VEGF).


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Chemotherapy, Adjuvant , Female , Humans , Predictive Value of Tests , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor, ErbB-2/metabolism , Tumor Suppressor Protein p53/metabolism
11.
Clin Cancer Res ; 7(8): 2228-36, 2001 Aug.
Article En | MEDLINE | ID: mdl-11489796

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.


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
12.
J Clin Oncol ; 19(7): 2002-9, 2001 Apr 01.
Article En | MEDLINE | ID: mdl-11283133

PURPOSE: We conducted this phase I trial to determine the safety and toxicity profile of LY353381.HCl-a novel, potent, third-generation selective estrogen receptor modulator (SERM)-because this benzothiophene derivative demonstrated an SERM profile in preclinical studies. PATIENTS AND METHODS: We studied 32 patients with recurrent or metastatic breast cancer. Patients were treated in four cohorts with oral daily doses of 10, 20, 50, and 100 mg. Pharmacokinetic sampling was performed during the first 72 hours following the first dose on day 1 and during the 24 hours after the day 57 dose. Eligibility criteria included Eastern Cooperative Oncology Group performance status of 0 to 2; no significant major organ dysfunction; and at least 3 weeks elapsed since most recent hormonal therapy, chemotherapy, and estrogen replacement therapy. RESULTS: The median patient age was 56 years (range, 30 years to 76 years). The median number of prior chemotherapies for metastatic disease was one (range, zero to four), while the median number of prior hormone regimens for metastatic disease was two (range, zero to five). Receptor status was estrogen receptor (ER) positive and progesterone receptor (PR) positive, 19 patients; ER positive and PR negative, eight patients; ER positive and PR unknown, two patients; and ER and PR unknown, three patients. Dose-limiting toxicity was not observed. Treatment was well tolerated with mild to moderate hot flashes in 18 of 32 patients (56%) at all dose levels. Transvaginal ultrasound performed at baseline and after 12 weeks of treatment showed no endometrial thickening. Of the 32 patients evaluable for response, six patients had stable disease for at least 6 months with a median duration of 7.7 months (range, 6.2 months to 33.8 months). The pharmacokinetics of LY353381.HCl were generally linear with respect to time and studied dose range. CONCLUSION: As predicted in preclinical testing, daily oral LY353381.HCl is safe, is well tolerated at all tested dose levels, and may be clinically beneficial in patients with extensively pretreated metastatic breast cancer. Further studies with LY353381 to evaluate the efficacy in patients with or without prior exposure to tamoxifen and fewer overall prior regimens are under way.


Breast Neoplasms/drug therapy , Estrogen Antagonists/pharmacology , Piperidines/pharmacology , Thiophenes/pharmacology , Actuarial Analysis , Adult , Aged , Breast Neoplasms/pathology , Dose-Response Relationship, Drug , Endometrium/drug effects , Estrogen Antagonists/therapeutic use , Female , Humans , Middle Aged , Neoplasm Metastasis , Neoplasm Recurrence, Local/drug therapy , Piperidines/therapeutic use , Thiophenes/therapeutic use
13.
Chem Biol ; 8(3): 289-99, 2001 Mar.
Article En | MEDLINE | ID: mdl-11306353

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.


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
14.
Cancer Res ; 61(7): 2945-52, 2001 Apr 01.
Article En | MEDLINE | ID: mdl-11306472

17-(Allylamino)-17-demethoxygeldanamycin (17-AAG) is an ansamycin antibiotic that binds to a conserved pocket in Hsp90 and induces the degradation of proteins that require this chaperone for conformational maturation. 17-AAG causes a retinoblastoma (RB)-dependent G1 block in cancer cells and is now in clinical trial. In breast cancer cells, G1 block is accompanied by differentiation and followed by apoptosis. The differentiation is characterized by specific changes in morphology and induction of milk fat proteins and lipid droplets. In cells lacking RB, neither G1 arrest nor differentiation occurs; instead, they undergo apoptosis in mitosis. Introduction of RB into these cells restores the differentiation response to 17-AAG. Inhibitors of the ras, mitogen-activated protein kinase, and phosphatidylinositol 3-kinase pathways cause accumulation of milk fat proteins and induction of lipid droplets when associated with G1 arrest but do not cause morphological changes. Thus, regulation of Hsp90 function by 17-AAG in breast cancer cells induces RB-dependent morphological and functional mammary differentiation. G1 arrest is sufficient for some but not all aspects of the phenotype. Induction of differentiation may be responsible for some of the antitumor effects of this drug.


Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/pathology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Quinones/pharmacology , Rifabutin/pharmacology , Apoptosis/drug effects , Benzoquinones , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , G1 Phase/drug effects , Glycolipids/metabolism , Glycoproteins/metabolism , HSP90 Heat-Shock Proteins/physiology , Lactams, Macrocyclic , Lipid Droplets , Milk Proteins/metabolism , Phenotype , Phosphorylation , Retinoblastoma Protein/metabolism , Retinoblastoma Protein/physiology , Rifabutin/analogs & derivatives , Tumor Cells, Cultured
16.
Cancer Res ; 60(8): 2090-4, 2000 Apr 15.
Article En | MEDLINE | ID: mdl-10786665

Geldanamycin (GM) is a natural antibiotic that binds Hsp90 and induces the degradation of receptor tyrosine kinases, steroid receptors, and Raf. It is a potent inhibitor of cancer cells that overexpress HER-kinases, but its effects on other important proteins may cause significant toxicity and limit its clinical use. We report the synthesis and identification of a GM dimer, GMD-4c, which had selective activity against HER-kinases. Selectivity was a function of linker length and required two intact GM moieties. GMD-4c is a potent inducer of G1 block and apoptosis of breast cancer cell lines that overexpress HER2, but does not appreciably inhibit the growth of 32D cells that lack HER-kinases. GMD-4c could be useful in the treatment of carcinomas dependent on HER-kinases.


Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/enzymology , Quinones/pharmacology , Receptor, ErbB-2/metabolism , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/therapeutic use , Benzoquinones , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Division/drug effects , Dimerization , Down-Regulation/drug effects , Humans , Immunohistochemistry , Inhibitory Concentration 50 , Lactams, Macrocyclic , Proto-Oncogene Proteins c-raf/metabolism , Quinones/chemistry , Quinones/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/biosynthesis , Receptor, IGF Type 1/metabolism , Receptors, Estrogen/metabolism , Substrate Specificity , Tumor Cells, Cultured
18.
Cell Biol Int ; 23(4): 299-306, 1999.
Article En | MEDLINE | ID: mdl-10600238

A 35-kDa protein (designated p35) showing antigenic homology with an N-terminal epitope on the SV-40 large T-antigen oncoprotein was purified from transformed cardiomyocytes. Sequence analysis of several tryptic peptides indicated that p35 was not homologous to previously described sequences. Polyclonal antibody raised against synthetic peptide containing one of the tryptic fragments was used in Western blot analyses to ascertain the tissue-specific pattern of p35 expression. p35 was expressed ubiquitously in adult mouse tissues, and was detected in both embryonic and transformed cardiomyocyte preparations. Subcellular fractionation studies indicated that p35 is an integral membrane protein. Expression of p35 appeared to be regulated by growth conditions as evidenced by a transient decrease in protein levels following the addition of serum to quiescent NIH 3T3 cells.


Membrane Proteins/isolation & purification , Myocardium/chemistry , 3T3 Cells , Animals , Antigens, Viral, Tumor , Blotting, Western , Cell Transformation, Neoplastic , Cells, Cultured , Databases, Factual , Dogs , Epitopes/chemistry , Membrane Proteins/chemistry , Mice , Mice, Transgenic , Myocardium/cytology , Myocardium/metabolism , Peptide Mapping/methods , Sequence Homology, Amino Acid , Simian virus 40
19.
Hematol Oncol Clin North Am ; 13(2): 391-413, vi, 1999 Apr.
Article En | MEDLINE | ID: mdl-10363137

The breast cancer mortality rate is falling, most likely because of a combination of early detection, refined surgical and radiation therapy techniques, and improved systemic therapy efficacy. The proper integration and application of these treatment modalities present evolving challenges for clinicians. Systemic therapy, in particular, is changing rapidly with the advent of new chemotherapy drugs, new classes of agents, and new therapeutic regimens. The most recent studies suggest that optimal outcomes are possible through the broad but appropriate use of hormone therapy and chemotherapy to prevent relapse and possibly prevent second primary tumors. The choice of therapy for patients remains a matter for careful consideration and discussion in each individual case.


Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Female , Humans , Neoplasm Metastasis , Randomized Controlled Trials as Topic , Recurrence , Remission Induction , Treatment Outcome
20.
Semin Oncol ; 26(1): 35-47, 1999 Feb.
Article En | MEDLINE | ID: mdl-10073560

In recent years, the most frequent indication for high-dose autologous stem-cell-supported chemotherapy in the United States has been breast cancer. This approach is applied in "high-risk," early-stage disease as adjuvant treatment, and with either curative or palliative intent in metastatic disease. Among both lay persons and medical professionals, high-dose therapy is broadly viewed as standard and appropriate, even though the results of the largest prospective randomized studies are not yet available. This view is based on extrapolation from preclinical data, a fundamental belief that "more is better," a faith in high technology, and the results of numerous pilot and phase I and II trials. Because high-dose therapy is promising, but also more difficult to administer than standard therapy, controversy rages in all circles concerning its use. Unless and until we have the results of properly randomized, prospective studies, this controversy will likely continue. Against this background, we will review the theoretical basis for high-dose treatment in breast cancer, the translation of this laboratory science into the clinic, usual treatment approaches, the available data, ongoing clinical trials, and future research directions.


Antineoplastic Agents/administration & dosage , Bone Marrow Transplantation , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Chemotherapy, Adjuvant , Clinical Trials as Topic , Drug Administration Schedule , Female , Humans , Multicenter Studies as Topic , Randomized Controlled Trials as Topic , Registries , Treatment Outcome , United States
...