Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Cell Death Differ ; 21(2): 234-46, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24037089

ABSTRACT

We previously reported that STAT1 expression is frequently abrogated in human estrogen receptor-α-positive (ERα(+)) breast cancers and mice lacking STAT1 spontaneously develop ERα(+) mammary tumors. However, the precise mechanism by which STAT1 suppresses mammary gland tumorigenesis has not been fully elucidated. Here we show that STAT1-deficient mammary epithelial cells (MECs) display persistent prolactin receptor (PrlR) signaling, resulting in activation of JAK2, STAT3 and STAT5A/5B, expansion of CD61(+) luminal progenitor cells and development of ERα(+) mammary tumors. A failure to upregulate SOCS1, a STAT1-induced inhibitor of JAK2, leads to unopposed oncogenic PrlR signaling in STAT1(-/-) MECs. Prophylactic use of a pharmacological JAK2 inhibitor restrains the proportion of luminal progenitors and prevents disease induction. Systemic inhibition of activated JAK2 induces tumor cell death and produces therapeutic regression of pre-existing endocrine-sensitive and refractory mammary tumors. Thus, STAT1 suppresses tumor formation in mammary glands by preventing the natural developmental function of a growth factor signaling pathway from becoming pro-oncogenic. In addition, targeted inhibition of JAK2 may have significant therapeutic potential in controlling ERα(+) breast cancer in humans.


Subject(s)
Estrogen Receptor alpha/metabolism , Janus Kinase 2/metabolism , Mammary Neoplasms, Animal/metabolism , Neoplastic Stem Cells/metabolism , STAT1 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Female , Heterocyclic Compounds, 3-Ring/pharmacology , Janus Kinase 2/antagonists & inhibitors , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/genetics , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , STAT1 Transcription Factor/deficiency , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 1 Protein
2.
Endocr Relat Cancer ; 12 Suppl 1: S113-7, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16113087

ABSTRACT

De novo resistance to endocrine therapy is a near-universal feature of oestrogen receptor (ER)- negative breast cancer. Although many ER-positive breast cancers also show no response to tamoxifen or aromatase inhibitors on objective clinical grounds the large majority show reduced proliferation indicating that some oestrogen dependence is present in almost all ER-positive breast cancer. In neoadjuvant studies HER2 positivity is associated with poor response rates to tamoxifen but not aromatase inhibitors, consistent with preclinical models. Acquired resistance to tamoxifen is associated with decreases in ER positivity but most recurrent lesions remain ER-positive. A small proportion of these show increased HER2 expression and in these patients increased phospho-p38 may contribute to the tamoxifen-resistant phenotype. There is an unfortunate paucity of clinical and biological data on acquired resistance to aromatase inhibitors.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Neoplasms, Hormone-Dependent/drug therapy , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Tamoxifen/therapeutic use , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Estrogens/metabolism , Female , Humans , Neoplasms, Hormone-Dependent/metabolism , Signal Transduction
3.
Breast Cancer Res Treat ; 92(1): 69-75, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15980993

ABSTRACT

PURPOSE: Laboratory evidence indicates that tumor growth depends on the balance between cell proliferation and cell death, and many anticancer agents may exert their therapeutic effect by decreasing proliferation and increasing apoptosis. Additionally, clinical observations indicate that overexpression of HER-2 or topoisomerase IIalpha (topo IIalpha) may be predictors of better response to anthracyclines in breast cancer. The objective of this study was to determine if proliferation (Ki-67), apoptosis (TUNEL), and expression of HER-2 and topo IIalpha are affected by anthracycline treatment, and if these molecular markers predict anthracycline responsiveness. EXPERIMENTAL DESIGN: Thirty-three women with primary breast tumors > or =3 cm received either doxorubicin (75 mg/m(2)) or epirubicin (120 mg/m(2)) for 4 cycles before surgery. Clinical response was evaluated after 4 cycles of treatment. Changes in molecular markers were assessed from core needle taken before treatment (D0), at 24-48 h (Dl) and on day 7 (D7) while on treatment, and from the surgical specimen excised on day 84 (D84) after the fourth cycle of chemotherapy. RESULTS: The overall response rate was 51% (17 of 33 patients), with a 12% complete clinical response rate (4 of 33 patients). There were trends for tumors with higher apoptosis and topo IIalpha at baseline (D0) to be more responsive to anthracyclines, p = 0.1 and p = 0.08, respectively. Median apoptosis increased from D0 to Dl (p = 0.06) while median Ki-67 decreased (p = 0.07). Overall, expression of HER-2 remained stable throughout the chemotherapy administration. By Day 84, topo IIalpha had significantly decreased from baseline in responders, while it increased in non-responders, p = 0.03. CONCLUSIONS: In human primary breast cancer, anthracycline treatment causes an early increase in apoptosis and a decrease in proliferation. In this pilot study, higher apoptosis and topo IIalphaa levels in primary tumors were associated with greater responsiveness to anthracyclines, and topo IIalpha levels declined in responsive tumors.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/biosynthesis , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Adult , Aged , Antibiotics, Antineoplastic/therapeutic use , Antigens, Neoplasm/biosynthesis , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/physiopathology , Carcinoma, Lobular/drug therapy , Carcinoma, Lobular/pathology , Carcinoma, Lobular/physiopathology , DNA Topoisomerases, Type II/biosynthesis , DNA-Binding Proteins/biosynthesis , Doxorubicin/therapeutic use , Epirubicin/pharmacology , Epirubicin/therapeutic use , Female , Genes, erbB-2/physiology , Humans , Middle Aged , Neoplasm Staging , Pilot Projects , Predictive Value of Tests
4.
Endocrinology ; 144(6): 2683-94, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12746333

ABSTRACT

Insulin receptor substrates (IRS) are central integrators of hormone, cytokine, and growth factor signaling. IRS proteins can be phosphorylated by a number of signaling pathways critical to normal mammary gland development. Studies in transgenic mice that overexpress IGF-I in the mammary gland suggested that IRS expression is important in the regulation of normal postlactational mammary involution. The goal of these studies was to examine IRS expression in the mouse mammary gland and determine the importance of IRS-1 to mammary development in the virgin mouse. IRS-1 and -2 show distinct patterns of protein expression in the virgin mouse mammary gland, and protein abundance is dramatically increased during pregnancy and lactation, but rapidly lost during involution. Consistent with hormone regulation, IRS-1 protein levels are reduced by ovariectomy, induced by combined treatment with estrogen and progesterone, and vary considerably throughout the estrous cycle. These changes occur without similar changes in mRNA levels, suggesting posttranscriptional control. Mammary glands from IRS-1 null mice have smaller fat pads than wild-type controls, but this reduction is proportional to the overall reduction in body size. Development of the mammary duct (terminal endbuds and branch points) is not altered by the loss of IRS-1, and pregnancy-induced proliferation is not changed. These data indicate that IRS undergo complex developmental and hormonal regulation in the mammary gland, and that IRS-1 is more likely to regulate mammary function in lactating mice than in virgin or pregnant mice.


Subject(s)
Estrogens/pharmacology , Mammary Glands, Animal/physiology , Phosphoproteins/genetics , Progesterone/pharmacology , Signal Transduction/physiology , Adipose Tissue/chemistry , Adipose Tissue/growth & development , Adipose Tissue/physiology , Animals , Estrous Cycle/physiology , Female , Gene Expression/drug effects , Gene Expression/physiology , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Mammary Glands, Animal/chemistry , Mammary Glands, Animal/growth & development , Mice , Mice, Inbred Strains , Ovariectomy , Phosphoproteins/analysis , Pregnancy , Signal Transduction/drug effects
5.
Arch Pathol Lab Med ; 125(10): 1316-20, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11570906

ABSTRACT

CONTEXT: Previous studies have indicated certain immunohistochemical markers, including WT1, may be helpful in distinguishing adenocarcinomas from mesotheliomas, but to date there are no reliable, widely accepted, commercially available antibodies positive in mesotheliomas and negative in adenocarcinomas. We compared the nuclear and cytoplasmic staining patterns of WT1 in these 2 malignancies using a commercially available antibody and examined the expression of 2 other previously reported positive markers, calretinin and thrombomodulin. METHODS: Sixty-seven mesotheliomas and 51 adenocarcinomas, all paraffin embedded, were retrieved from recent case files. The diagnosis of mesothelioma was based on typical clinical and morphologic features, as well as immunohistochemistry; electron microscopy had been performed on 16 cases. The diagnosis of adenocarcinoma was based on typical light microscopic findings and a positive stain for mucin. Commercially available antibodies to WT1, thrombomodulin, and calretinin were applied. Because of the conflict surrounding calretinin, 2 anticalretinin antibodies (from Chemicon Inc and Zymed Laboratories) were utilized. RESULTS: Fifty of 67 mesotheliomas showed strong nuclear staining with WT1. No adenocarcinomas (0/51) showed nuclear staining. Twenty-three of 67 mesotheliomas were positive for thrombomodulin, and 35 of 67 mesotheliomas were positive for calretinin with the Chemicon antibody. Nine of 15 mesotheliomas were positive for calretinin with the Zymed antibody. CONCLUSIONS: Thrombomodulin and calretinin did not prove useful in discriminating between mesotheliomas and adenocarcinomas. The degree of positivity with calretinin may be dependent on the specific antibody utilized. Nuclear staining for WT1 is highly specific for mesothelioma and, in the appropriate clinical setting, can be a helpful adjunct in the distinction between adenocarcinomas and mesotheliomas.


Subject(s)
Adenocarcinoma/ultrastructure , Cell Nucleus/chemistry , Cytoplasm/chemistry , Lung Neoplasms/ultrastructure , Mesothelioma/ultrastructure , WT1 Proteins/analysis , Adenocarcinoma/chemistry , Humans , Immunohistochemistry , Lung Neoplasms/chemistry , Mesothelioma/chemistry , Staining and Labeling
6.
Breast Cancer Res Treat ; 66(3): 217-24, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11510693

ABSTRACT

Heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNP-A2/B1) is highly expressed during critical stages of lung development and carcinogenesis. To determine if the expression of hnRNP-A2/B1 is an informative biomarker in breast carcinogenesis, we analyzed hnRNP-A2/B1 overexpression by immunohistochemistry in archived specimens. Expression was detected in 48/85 (56.5%) primary invasive breast cancers and 7/72 (9.7%) specimens of normal breast tissue. Northern analysis of breast cancer cells also demonstrated higher level of hnRNP-A2/B1 expression compared to normal or transformed breast cells. Expression of hnRNP-A2/B1 in breast cancer cells was decreased by exposure to retinoids coordinately with decreased cell growth. These results warrant further evaluation of hnRNP-A2/B1 as a marker of breast carcinogenesis.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/genetics , Cell Transformation, Neoplastic , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Breast Neoplasms/pathology , Cell Division , Female , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , Humans , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Invasiveness
7.
Endocr Relat Cancer ; 8(1): 47-61, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11350726

ABSTRACT

Most human invasive breast cancers (IBCs) appear to develop over long periods of time from certain pre-existing benign lesions. Of the many types of benign lesions in the human breast, only a few appear to have significant premalignant potential. The best characterized of these include atypical hyperplasias and in situ carcinomas and both categories are probably well on along the evolutionary pathway to IBC. Very little is known about earlier premalignant alterations. All types of premalignant breast lesions are relatively common but only a small proportion appear to progress to IBC. They are currently defined by their histological features and their prognosis is imprecisely estimated from indirect epidemiological evidence. Although lesions within specific categories look alike, they must possess underlying biological differences causing some to remain stable and others to progress. Recent studies suggest that they evolve by highly diverse genetic mechanisms and research into these altered pathways may identify specific early defects that can be targeted to prevent premalignant lesions from developing or becoming cancerous. It is far more rational to think that breast cancer can be prevented than cured once it has developed fully. This review discusses histological models of human premalignant breast disease that provide the framework for scientific investigations into the biological alterations behind them and examples of specific biological alterations that appear to be particularly important.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Carcinoma, Lobular/pathology , Precancerous Conditions/pathology , Breast/pathology , Breast Neoplasms/etiology , Breast Neoplasms/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Lobular/genetics , Female , Humans , Hyperplasia/pathology , Models, Biological , Precancerous Conditions/etiology , Precancerous Conditions/genetics
8.
Cancer Res ; 61(9): 3578-80, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11325822

ABSTRACT

Breast cancer mortality is seldom attributable to the primary tumor, but rather to the presence of systemic (metastatic) disease. Axillary lymph node dissection can identify the presence of metastatic breast cancer cells and serves as a marker for systemic disease. Previous work in our laboratory determined that rates of loss of heterozygosity (LOH) of a 1.6-Mb region of chromosome 14q 31.2 is much higher in axillary lymph node-negative primary breast tumors than in axillary lymph node-positive primary breast tumors (P. O'Connell et al., J. NATL: Cancer INST:, 91: 1391-1397, 1999.). This unusual observation suggests that, whereas the LOH of this region promotes primary breast cancer formation, some gene(s) mapping to this 1.6-Mb region is rate-limiting for breast cancer metastasis. Thus, if primary breast cancers delete this region, their ability to metastasize decreases. To identify this gene(s), we have physically mapped this area of chromosome 14q, confirmed the position of two known genes and 13 other expressed sequence tags into this 1.6-Mb region. One of these, the metastasis-associated 1 (MTA1) gene, previously identified as a metastasis-promoting gene (Y. Toh et al., J. BIOL: CHEM:, 269: 22958-22963, 1994.), mapped to the center of our 1.6-Mb target region. Thus, MTA1 represents a strong candidate for this breast cancer metastasis-promoting gene.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Histone Deacetylases , Loss of Heterozygosity , Proteins/genetics , Repressor Proteins , Chromosomes, Artificial, Bacterial , Chromosomes, Artificial, Yeast , Chromosomes, Human, Pair 14 , Expressed Sequence Tags , Humans , Neoplasm Metastasis , Physical Chromosome Mapping , Trans-Activators
9.
Am J Clin Oncol ; 24(1): 10-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11232942

ABSTRACT

Histologic evaluation and reporting of invasive breast cancer has effectively used Nottingham combined histologic grade (NCHG). This approach to predict outcome in invasive breast cancer has not been tested in multicenter cooperative trials. Histologic slides from selected breast cancer cases entered on node-negative Eastern Cooperative Oncology Group trials were assigned grades. Two pathologists evaluated cases for NCHG defined from differentiation, mitotic index, and nuclear grade. The study population consisted of separate samples from low- and high-risk strata, where low risk was estrogen receptor positive with a tumor size of less than 3 cm and high risk was estrogen receptor negative or tumor size greater than or equal to 3 cm. The rate of agreement was generally good, with 80% of cases classified the same for mitotic count and 76% of the cases classified the same for combined grade. There were no cases disagreeing from the lowest to the highest of the three categories. The median follow-up is 11.6 years, but for analysis of survival, this was truncated at 5 years. Mitotic index and combined grade as assessed by both pathologists showed significant associations with survival. High combined histologic grade was predictive for response to cyclophosphamide/methotrexate/5-fluorouracil (CMF) with survival differences at 5 years of 30% in the treated high-grade patients over the untreated patients. Overall, it is clear that pathologists can have close agreement in assignment of combined histologic grades, with highly significant prediction in univariate and borderline significance in multivariate analysis in prognostication of time to recurrence as well as survival. Thus, stratification used in these trials was highly prognostic as hoped, leaving a role for histologic grading in these relatively large tumors, more powerful than S-phase analysis in this series. In the subgroups of high-risk patients randomized between CMF and observation, there was a suggestion that the high-combined-grade group was predictive of treatment efficacy. We conclude that a combined histologic grade with defined criteria may be reliably assigned by practiced pathologists using readily available criteria, and that the measure may be of use in prognostication and prediction of therapeutic responsiveness when done in a technically ideal fashion.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Lymph Nodes/pathology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/mortality , Chemotherapy, Adjuvant , Cyclophosphamide/administration & dosage , Disease-Free Survival , Female , Flow Cytometry , Fluorouracil/administration & dosage , Humans , Lymphatic Metastasis , Medical Futility , Methotrexate/administration & dosage , Multivariate Analysis , Predictive Value of Tests , Prednisone/administration & dosage , Proportional Hazards Models , Reproducibility of Results , S Phase/physiology , Survival Rate
10.
Cancer Res ; 60(15): 4026-9, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10945602

ABSTRACT

The best current model of breast cancer evolution suggests that most cancers arise from certain premalignant lesions. We have identified a common (34%) somatic mutation in the estrogen receptor (ER)-alpha gene in a series of 59 typical hyperplasias, a type of early premalignant breast lesion. The mutation, which affects the border of the hinge and hormone binding domains of ER-alpha, showed increased sensitivity to estrogen as compared with wild-type ER-alpha in stably transfected breast cancer cells, including markedly increased proliferation at subphysiological levels of estrogen. The mutated ER-alpha exhibits enhanced binding to the TIF-2 coactivator at low levels of hormone, which may partially explain its increased estrogen responsiveness. These data suggest that this mutation may promote or accelerate the development of cancer from premalignant breast lesions.


Subject(s)
Breast Neoplasms/genetics , Breast/pathology , Mutation , Precancerous Conditions/genetics , Receptors, Estrogen/genetics , Breast/physiology , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Division/physiology , DNA/analysis , DNA/genetics , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , Dose-Response Relationship, Drug , Estradiol/pharmacology , Estrogen Receptor alpha , Humans , Hyperplasia/genetics , Hyperplasia/pathology , Precancerous Conditions/pathology , Receptors, Estrogen/drug effects , Receptors, Estrogen/physiology , Reverse Transcriptase Polymerase Chain Reaction , Transfection
11.
Arch Pathol Lab Med ; 124(7): 966-78, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10888772

ABSTRACT

BACKGROUND: Under the auspices of the College of American Pathologists, a multidisciplinary group of clinicians, pathologists, and statisticians considered prognostic and predictive factors in breast cancer and stratified them into categories reflecting the strength of published evidence. MATERIALS AND METHODS: Factors were ranked according to previously established College of American Pathologists categorical rankings: category I, factors proven to be of prognostic import and useful in clinical patient management; category II, factors that had been extensively studied biologically and clinically, but whose import remains to be validated in statistically robust studies; and category III, all other factors not sufficiently studied to demonstrate their prognostic value. Factors in categories I and II were considered with respect to variations in methods of analysis, interpretation of findings, reporting of data, and statistical evaluation. For each factor, detailed recommendations for improvement were made. Recommendations were based on the following aims: (1) increasing uniformity and completeness of pathologic evaluation of tumor specimens, (2) enhancing the quality of data collected about existing prognostic factors, and (3) improving patient care. RESULTS AND CONCLUSIONS: Factors ranked in category I included TNM staging information, histologic grade, histologic type, mitotic figure counts, and hormone receptor status. Category II factors included c-erbB-2 (Her2-neu), proliferation markers, lymphatic and vascular channel invasion, and p53. Factors in category III included DNA ploidy analysis, microvessel density, epidermal growth factor receptor, transforming growth factor-alpha, bcl-2, pS2, and cathepsin D. This report constitutes a detailed outline of the findings and recommendations of the consensus conference group, organized according to structural guidelines as defined.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , Female , Genes, erbB , Genes, p53 , Humans , Lymph Node Excision , Lymphatic Metastasis , Mitosis , Pathology, Clinical , Ploidies , Prognosis , Receptors, Cell Surface/metabolism , Societies, Medical , United States
12.
J Clin Oncol ; 18(9): 1906-13, 2000 May.
Article in English | MEDLINE | ID: mdl-10784631

ABSTRACT

PURPOSE: The loss of p53 function is a recognized adverse prognostic factor in invasive breast cancer. Several studies have shown a relationship between the nuclear accumulation of p53 protein (a surrogate marker of p53 inactivation) and poor disease-free and overall survival. In general, however, these studies did not report the prognostic value of p53 for local failure, which we have therefore assessed retrospectively here. MATERIALS AND METHODS: Accumulation of p53 protein was evaluated by immunohistochemistry in 1,530 mastectomy-treated breast cancer patients (259 radiation therapy [RT]- and 1,271 mastectomy only [No RT]-treated patients). Statistical comparisons were made between p53 protein accumulation, estrogen/progesterone receptors, nodal status, tumor size, and local failure rate (LFR). Local failure was defined as tumor recurrence involving the chest wall and/or the ipsilateral supraclavicular/axillary lymph nodes. The median follow-up period was 62 months. RESULTS: In the No RT group, the LFR was 9.1% and 16. 5% in p53-negative and p53-positive patients, respectively (P<.001). Multivariate analysis revealed that p53 protein accumulation was significantly associated with an increased risk of local relapse (relative risk [RR], 1.7; 95% confidence interval [CI], 1.2 to 2.4). Nodal status and tumor size were also significant factors. In the RT group, the LFR was 9.3% and 21.5% in p53-negative and p53-positive patients, respectively (P = .009). Multivariate analysis revealed that p53 protein accumulation was significantly associated with an increased risk of local relapse (RR, 2.5; 95% CI, 1.1 to 5.7), as was nodal status. CONCLUSION: Nuclear accumulation of p53 protein is independently associated with a significantly increased local failure rate in breast cancer patients treated with mastectomy, with or without radiation.


Subject(s)
Breast Neoplasms/genetics , Genes, p53/genetics , Neoplasm Recurrence, Local , Tumor Suppressor Protein p53/analysis , Adult , Aged , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Disease-Free Survival , Female , Humans , Immunohistochemistry , Mastectomy, Radical , Middle Aged , Predictive Value of Tests , Prognosis , Retrospective Studies
14.
Clin Cancer Res ; 6(2): 616-21, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10690547

ABSTRACT

The aim of this study was to evaluate pretreatment clinical features and biological markers together with changes in these factors as predictors of response and relapse in patients receiving tamoxifen for primary breast cancer. Fine-needle aspiration cytology of the primary breast cancer was performed before tamoxifen treatment in 54 patients and repeated after therapy on day 14, day 60, or on both days in a subset of 35 patients. These samples were evaluated for estrogen receptor (ER), progesterone receptor (PgR), Ki67, S-phase fraction and ploidy. The overall response to tamoxifen was 57% (31 of 54 patients). Pretreatment ER and PgR significantly predicted for response by univariate analysis (P < 0.0001 and P < 0.003, respectively). By multivariate analysis, ER expression was the only independent predictor of response, and it was associated with 27 times the likelihood of response (95% confidence interval, 6-136). Increase in PgR and decrease in Ki67 on day 14 significantly predicted for response to tamoxifen (P < 0.03 and P < 0.04, respectively). Lack of ER, clinical node-positive disease, and failure to decrease Ki67 on day 14 were significantly associated with increased risk of relapse (P < 0.05). By multivariate analysis, ER expression was the only independent predictor of relapse (P < 0.005). Pretreatment and early changes in molecular marker expression may assist in the prediction of response and clinical outcome in primary breast cancer patients receiving tamoxifen.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Biomarkers, Tumor/analysis , Breast Neoplasms/drug therapy , Tamoxifen/therapeutic use , Aged , Aged, 80 and over , Biopsy, Needle , Breast Neoplasms/pathology , Disease Progression , Disease-Free Survival , Female , Humans , Ki-67 Antigen/analysis , Ploidies , Predictive Value of Tests , Prognosis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Recurrence , S Phase , Treatment Outcome
15.
Cancer ; 89(11): 2145-52, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11147583

ABSTRACT

BACKGROUND: Laboratory evidence suggests that many anticancer agents exert their effect by altering the ratio between apoptosis and cellular proliferation. The objective of this clinical study was to examine in vivo changes in apoptotic index (AI), Bcl-2 expression, proliferation (Ki-67 and S-phase fraction [SPF]), and ploidy as potential indicators of chemoresponsiveness. METHODS: Twenty-eight women with primary operable breast carcinoma received 2M (mitoxantrone 11 mg/m2, methotrexate 35 mg/m2 every 3 weeks) for 4 cycles before surgery/radiotherapy, and an additional 2 cycles were given after surgery. Clinical response was assessed after four cycles of treatment according to World Health Organization criteria. Changes in molecular markers were assessed from biopsies obtained by fine-needle aspiration performed before treatment, repeated after 24 and/or 72 hours (T1), and on Days 7 and 21 after the first cycle of chemotherapy. Flow cytometric analysis was used to assess SPF, ploidy, and AI (in situ DNA nick end labeling assay) whereas Ki-67 and Bcl-2 were evaluated by immunocytochemical analysis. RESULTS: The overall response rate was 61% (17 of 28 patients), with a 14% (4 of 28 patients) complete response rate. Patients with diploid carcinomas (P = 0.04) with high Ki-67 (P = 0.0001) and SPF (P = 0.09) were more likely to respond to treatment. Median AI increased by 3.4% with interquartile (IQ) range of 3.2 in responders, compared with only -0.1% (IQ range, 2.2) in nonresponders at T1 (P = 0.03). Median Ki-67 decreased by -12.0% (IQ range, 22.9) in responders and increased by 18.5% (IQ range, 15.1) in nonresponders on Day 21 (P = 0.003). Median Bcl-2 scores increased by 1.0 (IQ range, 4.0) in responders and were unchanged at 0.0 (IQ range, 0.5) in nonresponders (P = 0.08). Changes in SPF or ploidy were not significantly predictive of response. CONCLUSIONS: The results of this preliminary study support evidence that chemotherapy may increase apoptosis, decrease Ki-67, and increase Bcl-2 expression in primary breast carcinomas that subsequently respond to therapy. Methodology allowing morphological confirmation of apoptosis would be advantageous.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma/drug therapy , Carcinoma/pathology , Apoptosis/physiology , Biomarkers, Tumor/metabolism , Biopsy, Needle , Breast Neoplasms/surgery , Carcinoma/surgery , Cell Division/drug effects , Cell Division/physiology , Female , Flow Cytometry , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Methotrexate/administration & dosage , Middle Aged , Mitoxantrone/administration & dosage , Neoadjuvant Therapy , Ploidies , Proto-Oncogene Proteins c-bcl-2/metabolism , S Phase/drug effects , S Phase/physiology
16.
J Mammary Gland Biol Neoplasia ; 5(4): 351-64, 2000 Oct.
Article in English | MEDLINE | ID: mdl-14973381

ABSTRACT

Most human invasive breast cancers (IBCs) arise from preexisting benign lesions. There are many types of benign lesions in the human breast and only a few appear to have significant premalignant potential (atypical hyperplasias and in situ carcinomas). These lesions are relatively common and only a small proportion progress to IBC. They are currently defined by their histological features and their prognosis is imprecisely estimated from indirect evidence based on epidemiological studies. Although lesions within specific categories look alike, they must possess morphologically silent biological differences motivating some to remain stable and others to progress. Understanding the biological changes responsible for the development and progression of premalignant disease is a very active area of medical research. Progress in this area may provide new opportunities for breast cancer prevention by providing strategies to treat premalignant lesions before they develop or become cancerous. A large number of biological features have been evaluated in this setting during the past decade. This review discusses a few features that appear to be particularly important and have been studied in a relatively comprehensive manner.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Breast/pathology , Precancerous Conditions , Alleles , Breast Neoplasms/genetics , Disease Progression , Genes, Tumor Suppressor , Humans , Loss of Heterozygosity , Models, Biological , Neoplasm Invasiveness , Prognosis
18.
Int J Cancer ; 89(2): 111-7, 2000 Mar 20.
Article in English | MEDLINE | ID: mdl-10754487

ABSTRACT

Results of estrogen receptor (ER) and progesterone receptor (PgR) ligand-binding assays (LBAs) are strongly correlated with ER and PgR by immuno-histochemistry (IHC). To investigate whether ER and PgR by IHC are also strongly correlated with tamoxifen response, time to treatment failure (TTF) and overall survival (OS), the results of the 2 methods were directly compared in 205 patients with ER(+) metastatic breast cancer treated with daily tamoxifen (Southwest Oncology Group protocol 8228) with 9 years median follow-up. pS2, another estrogen-regulated molecule, was also analyzed. Tumors were scored for IHC from 0 to 5, according to the proportion of positively stained cells. These IHC scores for both ER and PgR were significantly associated with LBA levels (p < 0.001). There was a significant direct relationship between higher IHC ER, PgR and pS2 and increasing response to tamoxifen. TTF and OS were also significantly longer for patients with higher ER or PgR, but not pS2, IHC scores. Low, intermediate and high ER or PgR categories showed similar differences in response rates whether defined by LBA or IHC. In logistic regression models which included ER, PgR and pS2 by IHC; ER and PgR by LBA; and menopausal status, only ER (IHC) and pS2 (IHC) retained significance for predicting tamoxifen response (p = 0. 02 and p = 0.005, respectively), along with menopausal status (for PgR by IHC, p = 0.09). Increasing ER and PgR by IHC, as by LBA, are thus significantly associated with a progressively better response and longer survival in ER(+) metastatic breast cancer. pS2 is also predictive in this setting.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Breast Neoplasms/drug therapy , Estrogen Receptor Modulators/therapeutic use , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Tamoxifen/therapeutic use , Binding, Competitive , Breast Neoplasms/pathology , Female , Follow-Up Studies , Humans , Immunohistochemistry , Ligands , Logistic Models , Predictive Value of Tests , Southwestern United States , Survival Analysis , Treatment Outcome
19.
J Clin Oncol ; 17(10): 3058-63, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10506600

ABSTRACT

PURPOSE: To determine whether pretreatment clinical features and molecular markers, together with changes in these factors, can predict treatment response and survival in patients with primary operable breast cancer who receive neoadjuvant therapy. PATIENTS AND METHODS: Mitoxantrone, methotrexate (with or without mitomycin), and tamoxifen chemoendocrine therapy was administered to 158 patients before surgery. Clinical response was assessed after four cycles of treatment. Fine-needle aspiration cytology for estrogen receptor (ER), progesterone receptor (PgR), c-erbB-2, p53, bcl-2, Ki67, S-phase fraction (SPF), and ploidy were performed pretreatment and repeated on day 10 or day 21 after the first cycle of treatment. RESULTS: Good clinical response (GCR, defined as complete response or minimal residual disease) was achieved in 31% of patients (49 of 158). Tumor size, nodal disease, response, ER, PgR, c-erbB-2, p53, bcl-2, Ki67, SPF, and ploidy were analyzed as predictors of survival. By univariate analysis, node-positive disease (P =.05), lack of ER (P <.05) and PgR (P <.05), and failure to attain GCR (P =.008) were associated with a significantly increased risk of relapse. A significantly increased risk of death was associated with node-positive disease (P =.02), lack of ER expression (P =.04), and failure to attain GCR. By multivariate analysis, GCR was an independent predictor for survival (P =.05). ER expression (P =.03), absence of c-erbB-2 (P =.03), and a decrease in Ki67 on day 10 or day 21 of the first cycle (P <.05) significantly predicted for subsequent GCR. CONCLUSION: Molecular markers may be used to predict the likelihood of achieving GCR, which seems to be a valid surrogate marker for survival.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Breast Neoplasms/surgery , Adult , Aged , Breast Neoplasms/drug therapy , DNA, Neoplasm/analysis , Female , Genes, Tumor Suppressor/genetics , Humans , Lymphatic Metastasis , Methotrexate/administration & dosage , Middle Aged , Mitoxantrone/administration & dosage , Neoadjuvant Therapy , Ploidies , Predictive Value of Tests , Prognosis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Tamoxifen/administration & dosage , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...