Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 111
3.
Breast Cancer Res Treat ; 171(1): 21-31, 2018 Aug.
Article En | MEDLINE | ID: mdl-29736741

PURPOSE: Triple-negative breast cancer (TNBC) is associated with worse outcomes relative to other breast cancer subtypes. Chemotherapy remains the standard-of-care systemic therapy for patients with localized or metastatic disease, with few biomarkers to guide benefit. METHODS: We will discuss recent advances in our understanding of two key biological processes in TNBC, homologous recombination (HR) DNA repair deficiency and host anti-tumor immunity, and their intersection. RESULTS: Recent advances in our understanding of homologous recombination (HR) deficiency, including FDA approval of PARP inhibitor olaparib for BRCA1 or BRCA2 mutation carriers, and host anti-tumor immunity in TNBC offer potential for new and biomarker-driven approaches to treat TNBC. Assays interrogating HR DNA repair capacity may guide treatment with agents inducing or targeting DNA damage repair. Tumor infiltrating lymphocytes (TILs) are associated with improved prognosis in TNBC and recent efforts to characterize infiltrating immune cell subsets and activate host anti-tumor immunity offer promise, yet challenges remain particularly in tumors lacking pre-existing immune infiltrates. Advances in these fields provide potential biomarkers to stratify patients with TNBC and guide therapy: induction of DNA damage in HR-deficient tumors and activation of existing or recruitment of host anti-tumor immune cells. Importantly, these advances provide an opportunity to guide use of existing therapies and development of novel therapies for TNBC. Efforts to combine therapies that exploit HR deficiency to enhance the activity of immune-directed therapies offer promise. CONCLUSIONS: HR deficiency remains an important biomarker target and potentially effective adjunct to enhance immunogenicity of 'immune cold' TNBCs.


Homologous Recombination , Immunity/immunology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Biomarkers, Tumor , DNA Damage , DNA Repair , Disease Susceptibility , Female , Gene Expression Regulation, Neoplastic , Genes, BRCA1 , Genes, BRCA2 , Germ-Line Mutation , Humans , Immunomodulation , Molecular Targeted Therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism
4.
Ann Oncol ; 28(2): 208-217, 2017 02 01.
Article En | MEDLINE | ID: mdl-27831505

New research questions emerge as medical needs continue to evolve and as we improve our understanding of cancer biology and treatment of malignancies. Although significant advances have been made in some areas of breast cancer research resulting in improvements in therapies and outcomes over the last few decades, other areas have not benefited to the same degree and we continue to have many gaps in our knowledge. This article summarizes the 12 short and medium-term clinical research needs in breast cancer deemed as priorities in 2016 by a panel of experts, in an attempt to focus and accelerate future research in the most needed areas: (i) de-escalate breast cancer therapies in early breast cancer without sacrificing outcomes; (ii) explore optimal adjuvant treatment durations; (iii) develop better tools and strategies to identify patients with genetic predisposition; (iv) improve care in young patients with breast cancer; (v) develop tools to speed up drug development in biomarker-defined populations; (vi) identify and validate targets that mediate resistance to chemotherapy, endocrine therapy and anti-HER2 therapies; (vii) evaluate the efficacy of local-regional treatments for metastatic disease; (viii) better define the optimal sequence of treatments in the metastatic setting; (ix) evaluate the clinical impact of intra-patient heterogeneity (intra-tumor, inter-tumor and inter-lesion heterogeneity); (x) better understand the biology and identify new targets in triple-negative breast cancer; (xi) better understand immune surveillance in breast cancer and further develop immunotherapies; and (xii) increase survivorship research efforts including supportive care and quality of life.


Breast Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Biomedical Research , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Female , Humans , Molecular Targeted Therapy , Quality Improvement , Treatment Outcome
5.
Br J Cancer ; 111(6): 1241-8, 2014 Sep 09.
Article En | MEDLINE | ID: mdl-25117820

BACKGROUND: Bevacizumab has broad anti-tumour activity, but substantial risk of hypertension. No reliable markers are available for predicting bevacizumab-induced hypertension. METHODS: A genome-wide association study (GWAS) was performed in the phase III bevacizumab-based adjuvant breast cancer trial, ECOG-5103, to evaluate for an association between genotypes and hypertension. GWAS was conducted in those who had experienced systolic blood pressure (SBP) >160 mm Hg during therapy using binary analysis and a cumulative dose model for the total exposure of bevacizumab. Common toxicity criteria (CTC) grade 3-5 hypertension was also assessed. Candidate SNP validation was performed in the randomised phase III trial, ECOG-2100. RESULTS: When using the phenotype of SBP>160 mm Hg, the most significant association in SV2C (rs6453204) approached and met genome-wide significance in the binary model (P=6.0 × 10(-8); OR=3.3) and in the cumulative dose model (P=4.7 × 10(-8); HR=2.2), respectively. Similar associations with rs6453204 were seen for CTC grade 3-5 hypertension but did not meet genome-wide significance. Validation study from ECOG-2100 demonstrated a statistically significant association between this SNP and grade 3/4 hypertension using the binary model (P-value=0.037; OR=2.4). CONCLUSIONS: A genetic variant in SV2C predicted clinically relevant bevacizumab-induced hypertension in two independent, randomised phase III trials.


Angiogenesis Inhibitors/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Breast Neoplasms/drug therapy , Hypertension/chemically induced , Hypertension/genetics , Membrane Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Bevacizumab , Biomarkers , Blood Pressure , Female , Genome-Wide Association Study , Genotype , Humans , Polymorphism, Single Nucleotide
6.
Ann Oncol ; 23(2): 331-7, 2012 Feb.
Article En | MEDLINE | ID: mdl-21821545

BACKGROUND: E2104 was designed to evaluate the safety of two different strategies incorporating bevacizumab into anthracycline-containing adjuvant therapy as a precursor to a definitive randomized phase III trial. PATIENTS AND METHODS: Patients were sequentially assigned to one of two treatment arms. In addition to dose-dense doxorubicin and cyclophosphamide followed by paclitaxel (Taxol) (ddAC→T), all patients received bevacizumab (10 mg/kg every 2 weeks × 26) initiated either concurrently with AC (Arm A: ddBAC→BT→B) or with paclitaxel (Arm B: ddAC→BT→B). The primary end point was incidence of clinically apparent cardiac dysfunction (CHF). RESULTS: Patients enrolled were 226 in number (Arm A 104, Arm B 122). Grade 3 hypertension, thrombosis, proteinuria and hemorrhage were reported for 12, 2, 2 and <1% of patients, respectively. Two patients developed grade 3 or more cerebrovascular ischemia. Three patients in each arm developed CHF. There was no significant difference between arms in the proportion of patients with an absolute decrease in left ventricular ejection fraction of >15% or >10% to below the lower limit of normal post AC or post bevacizumab. CONCLUSIONS: Incorporation of bevacizumab into anthracycline-containing adjuvant therapy does not result in prohibitive cardiac toxicity. The definitive phase III trial (E5103) was activated with systematic and extensive cardiac monitoring to define the true impact of bevacizumab on cardiac function.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Heart Diseases/chemically induced , Adult , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Female , Humans , Middle Aged , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Pilot Projects
7.
Ann Oncol ; 16(9): 1545-50, 2005 Sep.
Article En | MEDLINE | ID: mdl-15919683

BACKGROUND: To understand the pathogenesis of fatigue in cancer, we conducted a cross-sectional study using Brief Fatigue Inventory (BFI) and Functional Assessment of Cancer Therapy-Fatigue (FACT-F) instruments to measure fatigue and assessed laboratory studies. PATIENTS AND METHODS: 174 patients with cancer, who had undergone treatment within the last six months, answered the questionnaires and the Brief Version Zung Self-Rating Depression Scale (BZSDS). Blood samples were drawn for hemoglobin, albumin, thyroid stimulating hormone (TSH), dehydroepiandrosterone-sulfate (DHEAS) and tumor necrosis factor-alpha (TNF- alpha). Testosterone levels were checked in male patients. RESULTS: Clinically significant fatigue with BFI > or =4 was present in 52.0% of patients. Measurement of laboratory parameters revealed the following: DHEAS levels <80 mcg/dl in males and <36 mcg/dl in females=54.1%; BZSDS scores > or =27=20.1%; testosterone levels <200 ng/dl=26.4% of male patients. Significant correlations were noted between BFI and FACT-F, albumin levels, hemoglobin levels and BZSDS scores. In addition, for male patients BFI correlated with DHEAS and testosterone levels. In multiple linear regression, hemoglobin, BZSDS scores and current opioid use were associated with response BFI. For male patients, DHEAS <80 mcg/dl, increased BZSDS and testosterone <200 ng/dl were associated with increased BFI. CONCLUSION: Fatigue is common in this population and BFI correlates with more extensive measurements. Abnormalities such as decreased testosterone and DHEAS may lead to interventions that can be therapeutically exploited.


Fatigue/diagnosis , Neoplasms/complications , Cross-Sectional Studies , Dehydroepiandrosterone Sulfate/blood , Fatigue/blood , Fatigue/complications , Female , Hemoglobins/analysis , Humans , Male , Neoplasms/blood , Neoplasms/therapy , Serum Albumin/analysis , Surveys and Questionnaires , Testosterone/blood , Thyrotropin/analysis , Tumor Necrosis Factor-alpha/analysis
8.
Eur J Cancer ; 40(8): 1193-8, 2004 May.
Article En | MEDLINE | ID: mdl-15110883

Cancer patients, especially the elderly, present with a highly variable susceptibility to toxicity from chemotherapy. To estimate correctly a patient's risk for toxicity, both the average toxicity of a chemotherapy regimen and patient-related variables need to be assessed. However, treatment toxicities are typically reported item by item, not summarised per patient. We tested an index derived from a pilot study, the MAX2, on the ECOG database. Studies including 20 or more patients aged 70 years and older per arm were selected. Four studies were identified, representing 2526 patients, 410 (16%) being elderly. The association of the MAX2 index with the per patient incidence of grade 4 haematological and/or grade 3 or 4 non-haematological toxicity was highly significant, both for the overall group and for the elderly subgroup. The MAX2 index is a convenient and reproducible way of comparing the average per patient risk for toxicity from chemotherapy across several regimens.


Antineoplastic Combined Chemotherapy Protocols/adverse effects , Health Status Indicators , Neoplasms/drug therapy , Adult , Age Factors , Aged , Aged, 80 and over , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Humans , Middle Aged , Regression Analysis , Risk Factors , Sensitivity and Specificity
9.
J Clin Oncol ; 22(6): 1071-7, 2004 Mar 15.
Article En | MEDLINE | ID: mdl-15020608

PURPOSE: To determine the efficacy and safety of weekly docetaxel and trastuzumab as first- or second-line therapy in women with HER-2-overexpressing metastatic breast cancer and to correlate the efficacy of trastuzumab with HER-2 status as determined by immunohistochemistry assay and fluorescent in situ hybridization (FISH). PATIENTS AND METHODS: Twenty-six women with HER-2-positive (HercepTest [Dako Corp, Carpenteria, CA]2 to 3+) metastatic breast cancer were enrolled onto this study of trastuzumab (4 mg/kg load; 2 mg/kg/wk administered intravenously) and docetaxel (35 mg/m2/wk for 6 weeks). RESULTS: Using an intent-to-treat analysis, the overall response rate was 50% (13 of 26 patients). Eight patients (31%) had a period of stable disease posttherapy. Among HER-2 3+ patients, the overall response rate was 63% (12 of 19 patients) compared with a 14% response rate (one of seven patients) for HER-2 2+ patients (P=.07). Patients with FISH-positive tumors experienced an overall response rate of 64%. Median time to progression was 12.4 months for the entire cohort (HER-2 3+ tumors, 12.3 months; HER-2 2+ lesions, 9.5 months) and median survival was 22.1 months. All HER-2 3+ patients were FISH-positive; the only HER-2 2+ patient responding to treatment was also FISH-positive. Grade 4 toxicities occurred in four patients; most toxicities were mild. CONCLUSION: Trastuzumab plus docetaxel is an active and well-tolerated regimen in women with HER-2 3+ overexpressing or FISH-positive metastatic breast cancer.


Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-3/antagonists & inhibitors , Adult , Aged , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/pathology , Disease-Free Survival , Docetaxel , Female , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Middle Aged , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Survival Analysis , Taxoids/administration & dosage , Trastuzumab , United States , Up-Regulation
10.
Ann Oncol ; 14(7): 1072-7, 2003 Jul.
Article En | MEDLINE | ID: mdl-12853349

BACKGROUND: As screening central nervous system (CNS) imaging is not routinely performed, the incidence and clinical relevance of occult CNS metastases in advanced breast cancer is unknown. PATIENTS AND METHODS: All patients screened for participation in one of four clinical trials were included; each of the trials excluded patients with known CNS involvement and required screening CNS imaging. A cohort of breast cancer patients with symptomatic CNS metastases was identified from the IU Cancer Center Tumor Registry for comparison. RESULTS: From November 1998 to August 2001, 155 screening imaging studies were performed. Twenty-three patients (14.8%) had occult CNS metastases. HER-2 overexpression (P = 0.02) and number of metastatic sites (P = 0.03) were predictive of CNS involvement by multivariate analysis. Median survival from time of metastasis (1.78 versus 2.76 years; P <0.0001) and from screening (4.67 versus 10.4 months; P = 0.0013) was shorter in patients with than without occult CNS metastasis. Survival among patients with occult CNS metastasis was similar to patients with symptomatic CNS disease. CONCLUSIONS: Patients with CNS involvement, whether occult or symptomatic, have an impaired survival. Occult CNS metastasis is relatively common, but impact on survival of treating occult CNS disease in patients with progressive systemic metastases is questionable.


Breast Neoplasms/pathology , Central Nervous System Neoplasms/secondary , Mass Screening , Registries/statistics & numerical data , Adult , Aged , Biomarkers, Tumor/analysis , Clinical Trials as Topic , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Predictive Value of Tests , Prevalence , Prognosis , Prospective Studies , Receptor, ErbB-2/biosynthesis , Survival Analysis
11.
Eur J Cancer ; 39(12): 1668-75, 2003 Aug.
Article En | MEDLINE | ID: mdl-12888360

What separates a malignant from a normal cell? This question has occupied scientists for decades. Although a simple answer remains elusive, several hallmarks of malignancy have been identified. These critical features include uncontrolled proliferation, insensitivity to negative growth regulation, evasion of apoptosis, lack of senescence, invasion and metastasis, angiogenesis and genomic elasticity. Existing therapies predominantly target proliferation either with cytotoxic agents, ionising radiation or more targeted attacks on growth factor signalling pathways. Our most successful therapies to date inhibit proliferation via the oestrogen receptor (ER) and HER2 pathways. Further improvements in therapy must attack the other hallmarks of malignancy and will undoubtedly be accompanied by a better means of individual patient selection for such therapies. Indeed, each of these hallmarks presents a therapeutic opportunity. To believe otherwise would be to assume that a feature is both biologically crucial, yet therapeutically unimportant, an unlikely paradox. Here, we suggest the hallmarks of malignancy as a conceptual framework for understanding novel breast cancer therapies.


Breast Neoplasms/prevention & control , Apoptosis , Breast Neoplasms/pathology , Cell Transformation, Neoplastic , Female , Forecasting , Genome, Human , Growth Substances/physiology , Humans , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/prevention & control , Neovascularization, Pathologic/prevention & control , Receptor, ErbB-2/antagonists & inhibitors , Receptors, Estrogen/antagonists & inhibitors
12.
Ann Oncol ; 14(1): 20-8, 2003 Jan.
Article En | MEDLINE | ID: mdl-12488288

If your Snark be a Snark, that is right: Fetch it home by all means-you may serve it with greens, And it's handy for striking a light. "But oh, beamish nephew, beware of the day, If your Snark be a Boojum! For then You will softly and suddenly vanish away, And never be met with again!" Lewis Carroll The Hunting of the Snark


Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Humans , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/etiology
13.
Ann Oncol ; 13(10): 1612-5, 2002 Oct.
Article En | MEDLINE | ID: mdl-12377650

BACKGROUND: Docetaxel and estramustine exert anti-tumor effects by inhibiting microtubule function. In vitro data suggest synergism with this combination. This phase II study evaluated the response rate and toxicity of docetaxel and estramustine in patients with metastatic breast cancer (MBC). PATIENTS AND METHODS: Patients were treated with docetaxel 35 mg/m(2) on day 2 and estramustine phosphate 280 mg p.o. tds days 1-3 weekly for 3 of 4 weeks, for a maximum of six treatment cycles. RESULTS: Thirty-nine patients were enrolled between August 1999 and March 2001; 36 were eligible. Of 31 evaluable patients, responses were observed in 15 patients (47%); two patients (6%) obtained a complete response. Median time to treatment failure was 6 months; median survival was 1 year. Thromboembolic toxicity occurred in 11% of patients: three experienced deep venous thromboses and one had a fatal pulmonary embolism. Myelosuppression was minimal with this regimen. CONCLUSIONS: Despite modest activity in metastatic breast cancer, the toxicity observed with the combination of estramustine and docetaxel precludes the routine use of this combination in the treatment of breast cancer. Further studies using this compound in metastatic breast cancer are not warranted.


Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Paclitaxel/analogs & derivatives , Taxoids , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Docetaxel , Drug Administration Schedule , Estramustine/administration & dosage , Female , Humans , Microtubules/physiology , Middle Aged , Neoplasm Metastasis , Paclitaxel/administration & dosage , Survival , Treatment Outcome , Venous Thrombosis/chemically induced
14.
Ann Oncol ; 13(8): 1220-4, 2002 Aug.
Article En | MEDLINE | ID: mdl-12181245

BACKGROUND: This pilot trial was performed to evaluate the safety, toxicity and pharmacokinetics of chronic therapy with the matrix metalloproteinase inhibitor marimastat in the adjuvant treatment of breast cancer. PATIENTS AND METHODS: Patients with high-risk node negative or node positive breast cancer received marimastat either 5 or 10 mg p.o. b.i.d. for 12 months. Marimastat was given either as a single agent following completion of adjuvant chemotherapy or concurrently with tamoxifen. RESULTS: Sixty-three patients were enrolled from June 1997 to May 1998. All patients have completed 12 months of treatment or have discontinued therapy due to toxicity, relapse or intercurrent illness. Moderate (WHO criteria) arthralgia/arthritis was reported by 34% of patients receiving 5 mg b.i.d. and 45% of patients receiving 10 mg b.i.d.; severe arthralgia/arthritis was reported by 6% and 23% of patients, respectively. Six patients (19%) receiving 5 mg b.i.d. and 11 (35%) receiving 10 mg b.i.d. discontinued marimastat therapy due to toxicity. Trough plasma levels were rarely within the target range for biological activity (40-200 ng/ml) with mean concentration for patients receiving: 5 mg b.i.d. = 7.5; 5 mg b.i.d. plus tamoxifen = 6.9; 10 mg b.i.d. = 11.9; 10 mg b.i.d. plus tamoxifen = 12.8. CONCLUSIONS: A randomized adjuvant trial with marimastat is not warranted as chronic administration cannot maintain plasma levels with the target range.


Breast Neoplasms/metabolism , Enzyme Inhibitors/pharmacokinetics , Hydroxamic Acids/pharmacokinetics , Adult , Aged , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/therapeutic use , Female , Humans , Hydroxamic Acids/adverse effects , Hydroxamic Acids/therapeutic use , Matrix Metalloproteinase Inhibitors , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Pilot Projects , Safety , Survival Rate , Treatment Outcome
16.
Semin Oncol ; 28(4 Suppl 12): 24-31, 2001 Aug.
Article En | MEDLINE | ID: mdl-11552227

Within the past decade there has been enormous interest in integrating the taxanes into the adjuvant breast cancer setting. Adjuvant trial designs in the early 1990s were absent of taxanes. By the mid 1990s, the taxanes were included in adjuvant trials, but were mainly limited to trials conducted in node-positive patients. Currently, taxanes are a chemotherapeutic modality in the majority of ongoing adjuvant trials in both node-negative and node-positive patients. These trials are being conducted in thousands of patients worldwide by several of the cooperative research organizations. Most of the adjuvant trials have focused on defining the clinical efficacy and toxicity of the concurrent or sequential use of taxanes with anthracyclines. The collective experience with taxanes over the next 3 to 5 years will make them one of the most intensely studied treatments in the history of patients with breast cancer. The outcome of these trials is greatly anticipated because they have the potential of changing the current standards of care in the adjuvant treatment of patients with breast cancer.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Antibiotics, Antineoplastic/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Clinical Trials as Topic , Humans , Paclitaxel/administration & dosage
17.
Oncogene ; 20(17): 2178-85, 2001 04 19.
Article En | MEDLINE | ID: mdl-11360202

Exposure of mammalian cells to ultraviolet light, nutrient deprived culture media, hypoxia, environmental toxicants such as methyl mercury, methyl methanesulfonate, crocodilite asbestos or the agents that disrupt the function of endoplasmic reticulum (ER) leads to activation of the pro-apoptotic transcription factor GADD153/CHOP. Paradoxically, several of these agents also induce the anti-apoptotic transcription factor NF-kappaB. In this report, we demonstrate that NF-kappaB inhibits GADD153 activation in breast cancer cells exposed to nutrient deprived media, tunicamycin (which blocks protein folding in ER) or calcium ionopore (which depletes calcium stores in ER). Basal and calcium ionopore-induced GADD153 expression was more pronounced in fibroblasts obtained from mouse embryos lacking in p65 subunit of NF-kappaB compared to fibroblasts from wild type littermate embryos. Moreover, p65-/- fibroblasts were killed more efficiently by calcium ionopore and tunicamycin but not hydrogen peroxide compared to wild type fibroblasts. We also show that parthenolide, a NF-kappaB inhibitor, sensitizes breast cancer cells to tunicamycin. Transient transfection assay revealed that the p65 subunit but not the p50 subunit of NF-kappaB represses GADD153 promoter activity. These results establish a correlation between repression of pro-apoptotic genes by NF-kappaB and increased cell survival during ER stress as well as identify a distinct NF-kappaB regulated cell survival pathway.


Apoptosis/physiology , CCAAT-Enhancer-Binding Proteins/genetics , Endoplasmic Reticulum/physiology , NF-kappa B/physiology , Transcription Factors/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CCAAT-Enhancer-Binding Proteins/biosynthesis , Calcimycin/pharmacology , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans , Ionophores/pharmacology , Mice , NF-kappa B/antagonists & inhibitors , Sesquiterpenes/pharmacology , Stress, Physiological/genetics , Transcription Factor CHOP , Transcription Factors/biosynthesis , Transfection , Tumor Cells, Cultured , Tunicamycin/pharmacology
18.
Cancer Res ; 61(8): 3369-72, 2001 Apr 15.
Article En | MEDLINE | ID: mdl-11309294

Numerous chemotherapeutic agents have been shown to have an inhibitory effect on endothelial cell proliferation and migration, and tubule formation. In this study, we examined the antiangiogenic activity of docetaxel. Docetaxel inhibited endothelial cell proliferation and tubule formation in vitro in a dose-dependent fashion. Docetaxel treatment also inhibited angiogenesis in an in vivo Matrigel plug assay. The endothelial stimulating factors, vascular endothelial cell growth factor (VEGF) and basic fibroblast growth factor are able to protect endothelial cells from the antiangiogenic properties of docetaxel. This protective effect can be overcome by a recombinant humanized monoclonal antibody directed against VEGF in both in vitro and in vivo models. Similarly, combination of docetaxel with the antiangiogenic agent 2-methoxyestradiol also overcomes the protective effect of VEGF in both in vitro and in vivo models. These data suggest that microenvironmental factors (e.g., local release of VEGF and basic fibroblast growth factor) could play a role in decreasing the antiangiogenic effects of docetaxel, whereas agents such as 2- methoxyestradiol and recombinant humanized monoclonal antibody directed against VEGF may reverse this protective effect.


Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Endothelial Growth Factors/immunology , Estradiol/pharmacology , Lymphokines/immunology , Paclitaxel/analogs & derivatives , Paclitaxel/pharmacology , Taxoids , 2-Methoxyestradiol , Angiogenesis Inhibitors/antagonists & inhibitors , Animals , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Apoptosis/drug effects , Cell Division/drug effects , Docetaxel , Drug Synergism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Estradiol/analogs & derivatives , Fibroblast Growth Factor 2/physiology , Humans , Mice , Mice, Nude , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Paclitaxel/antagonists & inhibitors , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
19.
Oncology (Williston Park) ; 15(2 Suppl 3): 38-40, 2001 Feb.
Article En | MEDLINE | ID: mdl-11252888

A phase II trial evaluated the effectiveness and toxicity of combination paclitaxel (Taxol), gemcitabine (Gemzar), and trastuzumab (Herceptin) as first-line therapy for patients with newly diagnosed HER2-overexpressing metastatic breast cancer. To date, 27 patients have received paclitaxel at 175 mg/m2 over 3 hours on day 1, plus gemcitabine at 1,200 mg/m2 on days 1 and 8, plus trastuzumab at a 4-mg/kg loading dose on day 1, followed by 2 mg/kg weekly. Treatment cycles were repeated every 21 days. Responding or stable patients who had received six cycles of combination therapy continued single-agent trastuzumab weekly until disease progression. Treatment was generally well tolerated with grade 4 toxicity limited to myelosuppression. In all, 12 patients have achieved a partial remission and 1 patient had progressive disease; 14 patients continue treatment and have not yet been evaluated for response. Combination treatment with paclitaxel, gemcitabine, and trastuzumab is well tolerated and appears to be highly active. Accrual will continue to a total enrollment of 46 patients.


Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Deoxycytidine/therapeutic use , Paclitaxel/therapeutic use , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Clinical Trials, Phase II as Topic , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Female , Humans , Middle Aged , Neoplasm Metastasis , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Trastuzumab , Gemcitabine
20.
J Clin Oncol ; 19(5): 1539-69, 2001 Mar 01.
Article En | MEDLINE | ID: mdl-11230499

OBJECTIVE: To determine indications for the use of postmastectomy radiotherapy (PMRT) for patients with invasive breast cancer with involved axillary lymph nodes or locally advanced disease who receive systemic therapy. These guidelines are intended for use in the care of patients outside of clinical trials. POTENTIAL INTERVENTION: The benefits and risks of PMRT in such patients, as well as subgroups of these patients, were considered. The details of the PMRT technique were also evaluated. OUTCOMES: The outcomes considered included freedom from local-regional recurrence, survival (disease-free and overall), and long-term toxicity. EVIDENCE: An expert multidisciplinary panel reviewed pertinent information from the published literature through July 2000; certain investigators were contacted for more recent and, in some cases, unpublished information. A computerized search was performed of MEDLINE data; directed searches based on the bibliographies of primary articles were also performed. VALUES: Levels of evidence and guideline grades were assigned by the Panel using standard criteria. A "recommendation" was made when level I or II evidence was available and there was consensus as to its meaning. A "suggestion" was made based on level III, IV, or V evidence and there was consensus as to its meaning. Areas of clinical importance were pointed out where guidelines could not be formulated due to insufficient evidence or lack of consensus. RECOMMENDATIONS: The recommendations, suggestions, and expert opinions of the Panel are described in this article. VALIDATION: Seven outside reviewers, the American Society of Clinical Oncology (ASCO) Health Services Research Committee members, and the ASCO Board of Directors reviewed this document.


Breast Neoplasms/radiotherapy , Breast Neoplasms/surgery , Mastectomy , Radiotherapy, Adjuvant , Axilla/pathology , Breast Neoplasms/pathology , Cost-Benefit Analysis , Female , Humans , Lymphatic Metastasis , Neoplasm Invasiveness , Prognosis , Radiotherapy, Adjuvant/adverse effects , Radiotherapy, Adjuvant/economics , Survival Analysis
...