45 results on '"Sartor CI"'
Search Results
2. Phase 2 study of pre-excision single-dose intraoperative radiation therapy for early-stage breast cancers: six-year update with application of the ASTRO accelerated partial breast irradiation consensus statement criteria.
- Author
-
Vanderwalde NA, Jones EL, Kimple RJ, Moore DT, Klauber-Demore N, Sartor CI, and Ollila DW
- Subjects
- Aged, Aged, 80 and over, Combined Modality Therapy, Female, Humans, Intraoperative Care, Middle Aged, Breast Neoplasms radiotherapy, Breast Neoplasms surgery
- Abstract
Background: Intraoperative radiation therapy (IORT) allows delivery of high-dose radiation at the time of lumpectomy, potentially sparing adjuvant daily radiation. A phase 2 study of pre-excision IORT was performed for early-stage breast cancer., Methods: Patients ≥ 48 years of age with invasive ductal carcinoma, ≤ 3 cm, and clinically node-negative were eligible for this study, which was approved by institutional review board. Ultrasound was used to select electron energy and cone size to cover the tumor plus 1.5- to 2.0-cm lateral margins and 1-cm-deep margins (90% isodose). Fifteen Gy was delivered with a Mobetron irradiator, and immediate needle-localized partial mastectomy followed. Local event results were updated using the Kaplan-Meier method., Results: A total of 53 patients received IORT alone. Median age was 63 years, and median tumor size was 1.2 cm. Of these, 81% were positive for estrogen receptor or progesterone receptor, 11% were positive for human epidermal growth factor receptor 2, and 15% were triple-negative. Also, 42%, 49%, and 9% would have fallen into the Suitable, Cautionary, and Unsuitable groups, respectively, of the American Society of Therapeutic Radiation Oncology consensus statement for accelerated partial breast irradiation. Median follow-up was 69 months. Ipsilateral events occurred in 8 of 53 patients. The 6-year actuarial rate of ipsilateral events was 15% (95% confidence interval = 7%-29%). The crude event rate for Suitable and Cautionary groups was 1 of 22 (5%) and 7 of 26 (27%), respectively. Overall survival was 94.4%, and breast cancer-specific survival was 100%., Conclusions: The rate of local events in this study is a matter of concern, especially in the Cautionary group. On the basis of these findings, pre-excision IORT, as delivered in this study, may not provide adequate local control for less favorable early-stage breast cancers., (Copyright © 2013 American Cancer Society.)
- Published
- 2013
- Full Text
- View/download PDF
3. Phase I study and biomarker analysis of lapatinib and concurrent radiation for locally advanced breast cancer.
- Author
-
Kimple RJ, Horton JK, Livasy CA, Shields JM, Lawrence JA, Chiu WM, Ivanova A, Ollila DW, Carey LA, Halle JS, Sartor CI, and Dees EC
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor analysis, Biopsy methods, Cohort Studies, ErbB Receptors genetics, ErbB Receptors metabolism, Female, Humans, Immunohistochemistry, Lapatinib, Maximum Tolerated Dose, Middle Aged, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-akt metabolism, Receptor, ErbB-2 genetics, Receptor, ErbB-2 metabolism, Signal Transduction, Breast Neoplasms drug therapy, Breast Neoplasms radiotherapy, Quinazolines therapeutic use
- Abstract
Purpose: This phase I study assessed the toxicity and safety of combining daily lapatinib with radiation therapy. Sequential tumor biopsies were obtained to evaluate changes in biomarkers, such as epidermal growth factor receptor (EGFR) and human EGFR-2 (HER2) signaling pathways., Methods: Eligibility for this dose-escalation study included unresectable and locally recurrent or chemotherapy-refractory and locally advanced breast cancer, and adequate organ function. Patients underwent three serial biopsies: at baseline, after 1 week of lapatinib alone, and after 1 week of lapatinib and radiation. Endpoints included determination of toxicity, maximum tolerated dose, and analysis of the effect of lapatinib with or without radiation on EGFR and HER2 signaling pathways by immunohistochemistry., Results: Doses of lapatinib up to 1,500 mg/day were well tolerated. Toxicity of grade 3 or more was limited to radiation dermatitis and pain. Out of 19 patients treated, in field responses per response evaluation criteria in solid tumors criteria were complete in four patients and partial in six patients. Serial biopsies were obtained in 16 patients with no complications. Total Her2 was relatively unchanged while phospho-Her2, phospho-Akt, and phospho-ERK showed variable responses to both lapatinib alone and dual therapy with lapatinib and radiation., Conclusions: The combination of lapatinib and radiation was well tolerated in this patient cohort. Overall local response rates were comparable to those reported in other studies in this patient population. Biopsies were safely performed at all time points. Inhibition of HER2 and downstream signaling pathways was identified, although no strong correlation with response was seen.
- Published
- 2012
- Full Text
- View/download PDF
4. Local control following single-dose intraoperative radiotherapy prior to surgical excision of early-stage breast cancer.
- Author
-
Kimple RJ, Klauber-DeMore N, Kuzmiak CM, Pavic D, Lian J, Livasy CA, Chiu WM, Moore DT, Sartor CI, and Ollila DW
- Subjects
- Aged, Aged, 80 and over, Breast Neoplasms pathology, Female, Follow-Up Studies, Humans, Intraoperative Care, Middle Aged, Neoplasm Recurrence, Local pathology, Neoplasm Staging, Radiotherapy Dosage, Radiotherapy, Adjuvant, Survival Rate, Treatment Outcome, Breast Neoplasms radiotherapy, Breast Neoplasms surgery, Mastectomy, Neoplasm Recurrence, Local radiotherapy, Neoplasm Recurrence, Local surgery
- Abstract
Background: Multiple partial breast radiotherapy techniques are available. We have previously presented the technical details of our procedure of delivering partial breast irradiation with a single fraction of intraoperative radiotherapy (IORT) targeting the tumor in situ prior to partial mastectomy. This study details our completed, single-institution trial., Materials and Methods: An IRB-approved, DSMB-monitored phase II trial was performed with the following inclusion criteria: women age ≥48, ultrasound-visible invasive ductal cancers <3 cm, clinically negative axillary nodes. IORT was delivered using mobile electron irradiator, at least a 1.5-cm radial and 1-cm deep margin; patients received 15 Gy and immediately underwent partial mastectomy. Ipsilateral breast recurrence was classified as true/marginal, elsewhere in the breast or nodal basin. Kaplan-Meier methods were used to estimate survival functions and exact 95% confidence intervals are reported., Results: Between 2003 and 2007, 71 women underwent IORT (median follow-up: 3.5 years). For patients with tumor-involved or close margins, additional therapy was required: 7 patients, total mastectomy; 11, whole breast radiation. Four women experienced invasive ipsilateral breast failures (1 new primary, 3 margin recurrences) for a 3-year local control rate of 49 of 53 (94.8%; 95% confidence interval 92.4% [95% CI] 84.2–98.3%), actuarial three-year in breast recurrence was 8% (95% CI 2–18%), and breast cancer-specific survival was 100%., Conclusions: Intraoperative radiotherapy delivered to an in situ tumor is feasible, but our local control rate at 3.5 years is concerning. Possible changes to this technique to improve local control rates include better preoperative imaging (MRI), routine intraoperative ultrasound, and improved IORT delivery (larger cone, increased dose).
- Published
- 2011
- Full Text
- View/download PDF
5. Cosmetic outcomes for accelerated partial breast irradiation before surgical excision of early-stage breast cancer using single-dose intraoperative radiotherapy.
- Author
-
Kimple RJ, Klauber-DeMore N, Kuzmiak CM, Pavic D, Lian J, Livasy CA, Esler L, Moore DT, Sartor CI, and Ollila DW
- Subjects
- Aged, Aged, 80 and over, Breast Neoplasms diagnostic imaging, Breast Neoplasms pathology, Breast Neoplasms surgery, Carcinoma, Ductal, Breast diagnostic imaging, Carcinoma, Ductal, Breast pathology, Carcinoma, Ductal, Breast surgery, Combined Modality Therapy methods, Female, Follow-Up Studies, Humans, Intraoperative Period, Mastectomy, Segmental methods, Middle Aged, Patient Satisfaction, Prospective Studies, Radiotherapy Dosage, Treatment Outcome, Tumor Burden, Ultrasonography, Breast Neoplasms radiotherapy, Carcinoma, Ductal, Breast radiotherapy
- Abstract
Purpose: Determine cosmetic outcome and toxicity profile of intraoperative radiation delivered before tumor excision for patients with early-stage breast cancer., Methods and Materials: Patients age 48 or older with ultrasound-visible invasive ductal cancers <3 cm and clinically negative lymph nodes were eligible for treatment on this institutional review board-approved Phase II clinical trial. Treatment planning ultrasound was used to select an electron energy and cone size sufficient to cover the tumor plus a 1.5- to 2.0-cm circumferential margin laterally and a 1-cm-deep margin with the 90% isodose line. The dose was prescribed to a nominal 15 Gy and delivered using a Mobetron electron irradiator before tumor excision by segmental mastectomy. Physician- and patient-assessed cosmetic outcome and patient satisfaction were determined by questionnaire., Results: From March 2003 to July 2007, 71 patients were treated with intraoperative radiation therapy. Of those, 56 patients were evaluable, with a median follow-up of 3.1 years (minimum 1 year). Physician and patient assessment of cosmesis was "good or excellent" (Radiation Therapy Oncology Group cosmesis scale) in 45/56 (80%) and 32/42 (76%) of all patients, respectively. Eleven patients who received additional whole breast radiation had similar rates of good or excellent cosmesis: 40/48 (83%) and 29/36 (81%), respectively). Grade 1 or 2 acute toxicities were seen in 4/71 (6%) patients. No Grade 3 or 4 toxicities or serious adverse events have been seen., Conclusion: Intraoperative radiotherapy delivered to an in situ tumor is feasible with acceptable acute tolerance. Patient and physician assessment of the cosmetic outcome is good to excellent., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
6. Lapatinib in combination with radiation diminishes tumor regrowth in HER2+ and basal-like/EGFR+ breast tumor xenografts.
- Author
-
Sambade MJ, Kimple RJ, Camp JT, Peters E, Livasy CA, Sartor CI, and Shields JM
- Subjects
- Animals, Breast Neoplasms metabolism, ErbB Receptors metabolism, Female, Humans, Lapatinib, Mice, Mitogen-Activated Protein Kinase 3 antagonists & inhibitors, Mitogen-Activated Protein Kinase 3 metabolism, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins metabolism, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Proto-Oncogene Proteins c-akt metabolism, Quinazolines, Receptor, ErbB-2 metabolism, Transplantation, Heterologous, Breast Neoplasms radiotherapy, ErbB Receptors antagonists & inhibitors, Protein Kinase Inhibitors therapeutic use, Radiation Tolerance, Radiation-Sensitizing Agents therapeutic use, Receptor, ErbB-2 antagonists & inhibitors
- Abstract
Purpose: To determine whether lapatinib, a dual epidermal growth factor receptor (EGFR)/HER2 kinase inhibitor, can radiosensitize EGFR+ or HER2+ breast cancer xenografts., Methods and Materials: Mice bearing xenografts of basal-like/EGFR+ SUM149 and HER2+ SUM225 breast cancer cells were treated with lapatinib and fractionated radiotherapy and tumor growth inhibition correlated with alterations in ERK1 and AKT activation by immunohistochemistry., Results: Basal-like/EGFR+ SUM149 breast cancer tumors were completely resistant to treatment with lapatinib alone but highly growth impaired with lapatinib plus radiotherapy, exhibiting an enhancement ratio average of 2.75 and a fractional tumor product ratio average of 2.20 during the study period. In contrast, HER2+ SUM225 breast cancer tumors were highly responsive to treatment with lapatinib alone and yielded a relatively lower enhancement ratio average of 1.25 during the study period with lapatinib plus radiotherapy. Durable tumor control in the HER2+ SUM225 model was more effective with the combination treatment than either lapatinib or radiotherapy alone. Immunohistochemical analyses demonstrated that radiosensitization by lapatinib correlated with ERK1/2 inhibition in the EGFR+ SUM149 model and with AKT inhibition in the HER2+ SUM225 model., Conclusion: Our data suggest that lapatinib combined with fractionated radiotherapy may be useful against EGFR+ and HER2+ breast cancers and that inhibition of downstream signaling to ERK1/2 and AKT correlates with sensitization in EGFR+ and HER2+ cells, respectively., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
7. Radiosensitization of chemotherapy-refractory, locally advanced or locally recurrent breast cancer with trastuzumab: a phase II trial.
- Author
-
Horton JK, Halle J, Ferraro M, Carey L, Moore DT, Ollila D, and Sartor CI
- Subjects
- Adult, Aged, Antibodies, Monoclonal, Humanized, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms metabolism, Breast Neoplasms pathology, Combined Modality Therapy methods, Female, Humans, Lymphatic Irradiation, Middle Aged, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins metabolism, Prospective Studies, Radiotherapy Dosage, Receptor, ErbB-2 metabolism, Remission Induction, Trastuzumab, Antibodies, Monoclonal therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms radiotherapy, Radiation-Sensitizing Agents therapeutic use
- Abstract
Purpose: Trastuzumab (Herceptin), an anti-human epidermal growth factor receptor 2 (HER2) antibody, has been shown to be an effective radiosensitizer in preclinical studies. The present Phase II trial evaluated trastuzumab plus radiotherapy in patients with HER2-positive, chemotherapy-refractory, locally advanced or locoregionally recurrent breast cancer., Methods and Materials: Eligible patients had measurable disease, normal cardiac function, and biopsy-confirmed residual HER2-positive disease. Patients received weekly trastuzumab (2 mg/kg intravenously), concurrent with radiotherapy (50 Gy) to the breast and regional lymph nodes for 5 weeks. If feasible, surgery followed radiotherapy. The primary endpoint was safety, and the secondary endpoint was efficacy (pathologic response and interval to symptomatic local progression)., Results: Of the 19 patients enrolled, 7 were ineligible and received radiotherapy alone and 12 received therapy per protocol. Of these 12 patients, 11 had a Stage T4 diagnosis. Grade 3 toxicities included skin (n = 2) and lymphopenia (n = 1). One patient experienced delayed wound healing after surgery. No patients developed symptomatic cardiac dysfunction. Of the 7 patients who had undergone mastectomy, 3 (43%) had a substantial pathologic response (complete response or microscopic residual disease), significantly more than a comparison cohort (2 of 38 or 5%, p = .02). The median interval to symptomatic local progression was not reached. The median overall survival was 39 months., Conclusion: This is the first prospective trial providing evidence for a radiosensitizing effect of trastuzumab in breast cancer. The combination of trastuzumab and radiotherapy was well tolerated., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
8. Radiosensitization of epidermal growth factor receptor/HER2-positive pancreatic cancer is mediated by inhibition of Akt independent of ras mutational status.
- Author
-
Kimple RJ, Vaseva AV, Cox AD, Baerman KM, Calvo BF, Tepper JE, Shields JM, and Sartor CI
- Subjects
- Animals, Cell Line, Tumor, Female, Humans, Lapatinib, Mice, Mice, Inbred BALB C, Mutation, Nelfinavir pharmacology, Oncogene Protein v-akt metabolism, Phosphatidylinositol 3-Kinases metabolism, Radiation Tolerance, Radiation-Sensitizing Agents pharmacology, Receptor, ErbB-2 metabolism, Signal Transduction drug effects, Xenograft Model Antitumor Assays, ErbB Receptors metabolism, Genes, ras, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Proto-Oncogene Proteins c-akt metabolism, Quinazolines pharmacology
- Abstract
Purpose: Epidermal growth factor receptor (EGFR) family members (e.g., EGFR, HER2, HER3, and HER4) are commonly overexpressed in pancreatic cancer. We investigated the effects of inhibition of EGFR/HER2 signaling on pancreatic cancer to elucidate the role(s) of EGFR/HER2 in radiosensitization and to provide evidence in support of further clinical investigations., Experimental Design: Expression of EGFR family members in pancreatic cancer lines was assessed by quantitative reverse transcription-PCR. Cell growth inhibition was determined by MTS assay. The effects of inhibition of EGFR family receptors and downstream signaling pathways on in vitro radiosensitivity were evaluated using clonogenic assays. Growth delay was used to evaluate the effects of nelfinavir on in vivo tumor radiosensitivity., Results: Lapatinib inhibited cell growth in four pancreatic cancer cell lines, but radiosensitized only wild-type K-ras-expressing T3M4 cells. Akt activation was blocked in a wild-type K-ras cell line, whereas constitutive phosphorylation of Akt and extracellular signal-regulated kinase (ERK) was seen in lines expressing mutant K-ras. Overexpression of constitutively active K-ras (G12V) abrogated lapatinib-mediated inhibition of both Akt phosphorylation and radiosensitization. Inhibition of MAP/ERK kinase/ERK signaling with U0126 had no effect on radiosensitization, whereas inhibition of activated Akt with LY294002 (enhancement ratio, 1.2-1.8) or nelfinavir (enhancement ratio, 1.2-1.4) radiosensitized cells regardless of K-ras mutation status. Oral nelfinavir administration to mice bearing mutant K-ras-containing Capan-2 xenografts resulted in a greater than additive increase in radiation-mediated tumor growth delay (synergy assessment ratio of 1.5)., Conclusions: Inhibition of EGFR/HER2 enhances radiosensitivity in wild-type K-ras pancreatic cancer. Nelfinavir, and other phosphoinositide 3-kinase/Akt inhibitors, are effective pancreatic radiosensitizers regardless of K-ras mutation status.
- Published
- 2010
- Full Text
- View/download PDF
9. Mechanism of lapatinib-mediated radiosensitization of breast cancer cells is primarily by inhibition of the Raf>MEK>ERK mitogen-activated protein kinase cascade and radiosensitization of lapatinib-resistant cells restored by direct inhibition of MEK.
- Author
-
Sambade MJ, Camp JT, Kimple RJ, Sartor CI, and Shields JM
- Subjects
- Anthracenes pharmacology, Cell Line, Tumor, Enzyme Activation drug effects, Extracellular Signal-Regulated MAP Kinases metabolism, Female, Humans, Lapatinib, Mitogen-Activated Protein Kinase 9 antagonists & inhibitors, Mitogen-Activated Protein Kinase Kinases metabolism, Proto-Oncogene Proteins c-raf metabolism, Receptor, ErbB-2 antagonists & inhibitors, Tumor Cells, Cultured, Breast Neoplasms metabolism, Breast Neoplasms radiotherapy, ErbB Receptors antagonists & inhibitors, MAP Kinase Kinase 1 antagonists & inhibitors, MAP Kinase Signaling System drug effects, Quinazolines pharmacology, Radiation Tolerance drug effects
- Abstract
Background and Purpose: We recently showed that lapatinib, an EGFR/HER2 inhibitor, radiosensitized breast cancer cells of the basal and HER2+ subtypes. The purpose of this study was to identify the downstream signaling pathways responsible for lapatinib-mediated radiosensitization in breast cancer., Materials and Methods: Response of EGFR downstream signaling pathways was assessed by Western blot and clonogenic cell survival assays in breast tumor cells after irradiation (5Gy), lapatinib, CI-1040, or combined treatment., Results: In SUM102 cells, an EGFR+ basal breast cancer cell line, exposure to ionizing radiation elicited strong activation of ERK1/2 and JNK, which was blocked by lapatinib, and weak/no activation of p38, AKT or STAT3. Direct inhibition of MEK1 with CI-1040 resulted in 95% inhibition of surviving colonies when combined with radiation while inhibition of JNK with SP600125 had no effect. Lapatinib-mediated radiosensitization of SUM102 cells was completely abrogated with expression of constitutively active Raf. Treatment of lapatinib-resistant SUM185 cells with CI-1040 restored radiosensitization with 45% fewer surviving colonies when combined with radiation., Conclusions: These data suggest that radiosensitization by lapatinib is mediated largely through inhibition of MEK/ERK and that direct inhibition of this pathway may provide an additional avenue of radiosensitization in EGFR+ or HER2+ breast cancers.
- Published
- 2009
- Full Text
- View/download PDF
10. The HER4 cytoplasmic domain, but not its C terminus, inhibits mammary cell proliferation.
- Author
-
Feng SM, Sartor CI, Hunter D, Zhou H, Yang X, Caskey LS, Dy R, Muraoka-Cook RS, and Earp HS 3rd
- Subjects
- Amyloid Precursor Protein Secretases antagonists & inhibitors, Cell Line, Tumor, Female, Humans, Mammary Glands, Human enzymology, Neuregulin-1 antagonists & inhibitors, Neuregulin-1 physiology, Protein Structure, Tertiary, Receptor, ErbB-4, Cell Proliferation, Cytoplasm physiology, ErbB Receptors physiology, Growth Inhibitors physiology, Mammary Glands, Human cytology, Mammary Glands, Human metabolism, Peptide Fragments physiology
- Abstract
Unlike the proliferative action of other epidermal growth factor (EGF) receptor family members, HER4/ErbB4 is often associated with growth-inhibitory and differentiation signaling. These actions may involve HER4 two-step proteolytic processing by intramembraneous gamma-secretase, releasing the soluble, intracellular 80-kDa HER4 cytoplasmic domain, s80HER4. We demonstrate that pharmacological inhibition of either gamma-secretase activity or HER4 tyrosine kinase activity blocked heregulin-dependent growth inhibition of SUM44 breast cancer cells. We next generated breast cell lines stably expressing GFP-s80HER4 [green fluorescent protein (GFP) fused to the N terminus of the HER4 cytoplasmic domain, residues 676-1308], GFP-CT(HER4) (GFP fused to N terminus of the HER4 C-terminus distal to the tyrosine kinase domain, residues 989-1308), or GFP alone. Both GFP-s80HER4 and GFP-CTHER4 were found in the nucleus, but GFP-s80HER4 accumulated to a greater extent and sustained its nuclear localization. s80HER4 was constitutively tyrosine phosphorylated, and treatment of cells with a specific HER family tyrosine kinase inhibitor 1) blocked tyrosine phosphorylation; 2) markedly diminished GFP-s80HER4 nuclear localization; and 3) reduced signal transducer and activator of transcription (STAT)5A tyrosine phosphorylation and nuclear localization as well as GFP-s80HER4:STAT5A interaction. Multiple normal mammary and breast cancer cell lines, stably expressing GFP-s80HER4 (SUM44, MDA-MB-453, MCF10A, SUM102, and HC11) were growth inhibited compared with the same cell line expressing GFP-CTHER4 or GFP alone. The s80HER4-induced cell number reduction was due to slower growth because rates of apoptosis were equivalent in GFP-, GFP-CTHER4-, and GFP-s80HER4-expressing cells. Lastly, GFP-s80HER4 enhanced differentiation signaling as indicated by increased basal and prolactin-dependent beta-casein expression. These results indicate that surface HER4 tyrosine phosphorylation and ligand-dependent release of s80HER4 are necessary, and s80HER4 signaling is sufficient for HER4-dependent growth inhibition.
- Published
- 2007
- Full Text
- View/download PDF
11. EGFR associated expression profiles vary with breast tumor subtype.
- Author
-
Hoadley KA, Weigman VJ, Fan C, Sawyer LR, He X, Troester MA, Sartor CI, Rieger-House T, Bernard PS, Carey LA, and Perou CM
- Subjects
- Breast Neoplasms pathology, Cell Line, Tumor, Drug Screening Assays, Antitumor, Enzyme Inhibitors pharmacology, ErbB Receptors antagonists & inhibitors, Humans, MAP Kinase Kinase Kinases metabolism, Phosphatidylinositol 3-Kinases metabolism, Breast Neoplasms genetics, ErbB Receptors physiology, Gene Expression Profiling
- Abstract
Background: The epidermal growth factor receptor (EGFR/HER1) and its downstream signaling events are important for regulating cell growth and behavior in many epithelial tumors types. In breast cancer, the role of EGFR is complex and appears to vary relative to important clinical features including estrogen receptor (ER) status. To investigate EGFR-signaling using a genomics approach, several breast basal-like and luminal epithelial cell lines were examined for sensitivity to EGFR inhibitors. An EGFR-associated gene expression signature was identified in the basal-like SUM102 cell line and was used to classify a diverse set of sporadic breast tumors., Results: In vitro, breast basal-like cell lines were more sensitive to EGFR inhibitors compared to luminal cell lines. The basal-like tumor derived lines were also the most sensitive to carboplatin, which acted synergistically with cetuximab. An EGFR-associated signature was developed in vitro, evaluated on 241 primary breast tumors; three distinct clusters of genes were evident in vivo, two of which were predictive of poor patient outcomes. These EGFR-associated poor prognostic signatures were highly expressed in almost all basal-like tumors and many of the HER2+/ER- and Luminal B tumors., Conclusion: These results suggest that breast basal-like cell lines are sensitive to EGFR inhibitors and carboplatin, and this combination may also be synergistic. In vivo, the EGFR-signatures were of prognostic value, were associated with tumor subtype, and were uniquely associated with the high expression of distinct EGFR-RAS-MEK pathway genes.
- Published
- 2007
- Full Text
- View/download PDF
12. The triple negative paradox: primary tumor chemosensitivity of breast cancer subtypes.
- Author
-
Carey LA, Dees EC, Sawyer L, Gatti L, Moore DT, Collichio F, Ollila DW, Sartor CI, Graham ML, and Perou CM
- Subjects
- Adult, Aged, Breast Neoplasms mortality, Female, Humans, Immunohistochemistry, Middle Aged, Neoplasm Staging, Receptor, ErbB-2 analysis, Receptors, Estrogen analysis, Receptors, Progesterone analysis, Survival Analysis, Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms pathology
- Abstract
Purpose: Gene expression analysis identifies several breast cancer subtypes. We examined the relationship of neoadjuvant chemotherapy response to outcome among these breast cancer subtypes., Experimental Design: We used immunohistochemical profiles [human epidermal growth factor receptor 2-positive (HER2+)/hormone receptor-negative for HER2+/estrogen receptor-negative (ER-), hormone receptor and HER2- for basal-like, hormone receptor-positive for luminal] to subtype a prospectively maintained data set of patients with breast cancer treated with neoadjuvant anthracycline-based (doxorubicin plus cyclophosphamide, AC) chemotherapy. We analyzed each subtype for clinical and pathologic response to neoadjuvant chemotherapy and examined the relationship of response to distant disease-free survival and overall survival., Results: Of the 107 patients tested, 34 (32%) were basal-like, 11 (10%) were HER2+/ER-, and 62 (58%) were luminal. After neoadjuvant AC, 75% received subsequent chemotherapy and all received endocrine therapy if hormone receptor-positive. The chemotherapy regimen and pretreatment stage did not differ by subtype. Clinical response to AC was higher among the HER2+/ER- (70%) and basal-like (85%) than the luminal subtypes (47%; P < 0.0001). Pathologic complete response occurred in 36% of HER2+/ER-, 27% of basal-like, and 7% of luminal subtypes (P = 0.01). Despite initial chemosensitivity, patients with the basal-like and HER2+/ER- subtypes had worse distant disease-free survival (P = 0.04) and overall survival (P = 0.02) than those with the luminal subtypes. Regardless of subtype, only 2 of 17 patients with pathologic complete response relapsed. The worse outcome among basal-like and HER+/ER- subtypes was due to higher relapse among those with residual disease (P = 0.003)., Conclusions: Basal-like and HER2+/ER- subtypes are more sensitive to anthracycline-based neoadjuvant chemotherapy than luminal breast cancers. Patients that had pathologic complete response to chemotherapy had a good prognosis regardless of subtype. The poorer prognosis of basal-like and HER2+/ER- breast cancers could be explained by a higher likelihood of relapse in those patients in whom pathologic complete response was not achieved.
- Published
- 2007
- Full Text
- View/download PDF
13. In vivo intraoperative radiotherapy: a novel approach to radiotherapy for early stage breast cancer.
- Author
-
Stitzenberg KB, Klauber-Demore N, Chang XS, Calvo BF, Ollila DW, Goyal LK, Meyers MO, Kim HJ, Tepper JE, and Sartor CI
- Subjects
- Breast Neoplasms pathology, Feasibility Studies, Female, Humans, Intraoperative Care, Lymph Node Excision, Mastectomy, Segmental, Middle Aged, Neoplasm Recurrence, Local prevention & control, Neoplasm Staging, Radiotherapy Dosage, Treatment Outcome, Breast Neoplasms radiotherapy
- Abstract
Introduction: Intraoperative radiotherapy (IORT) has the potential to eliminate the access problems associated with standard 6-week post-operative external beam radiotherapy for patients with breast cancer. However, accurate delivery of the IORT dose for breast cancer has been problematic due to difficulty estimating the tumor bed after tumor removal and tissue re-approximation. We are investigating the feasibility of partial breast irradiation using a single fraction of IORT delivered to the tumor in vivo prior to surgical resection., Methods: In a trial, approved by the University of North Carolina School of Medicine Institutional Review Board, patients > or =55 years old with infiltrating ductal carcinoma without an extensive intraductal component with an overall tumor size < or =3.0 cm receive a single dose of IORT in place of standard post-operative radiotherapy., Results: All patients undergo preoperative ultrasonography to define the target volume. In a standard operating room, the tumor is exposed through a standard partial mastectomy incision. IORT is then delivered using a mobile, self-shielded, magnetron-driven X-band linear accelerator (Intraop Corp, Santa Clara, CA, USA). 15 Gy is delivered to the 90% isodose line covering the tumor with a 1 cm margin anterior-posterior and 2 cm margins laterally. After IORT, partial mastectomy is performed in the usual manner., Conclusions: IORT for breast cancer, delivered to the exposed tumor in vivo, is feasible and allows accurate estimation of the tumor bed. Further follow-up is ongoing to determine the efficacy of this approach.
- Published
- 2007
- Full Text
- View/download PDF
14. Feasibility of breast preserving therapy with single fraction in situ radiotherapy delivered intraoperatively.
- Author
-
Ollila DW, Klauber-DeMore N, Tesche LJ, Kuzmiak CM, Pavic D, Goyal LK, Lian J, Chang S, Livasy CA, Sherron RF, and Sartor CI
- Subjects
- Aged, Aged, 80 and over, Breast Neoplasms pathology, Carcinoma, Ductal, Breast pathology, Feasibility Studies, Female, Humans, Intraoperative Period, Mastectomy, Segmental, Middle Aged, Sentinel Lymph Node Biopsy, Breast Neoplasms radiotherapy, Breast Neoplasms surgery, Carcinoma, Ductal, Breast radiotherapy, Carcinoma, Ductal, Breast surgery, Dose Fractionation, Radiation
- Abstract
Background: Accelerated partial breast irradiation (APBI) has gained widespread interest as a means of improving the convenience and availability of breast conserving radiotherapy. Intraoperative radiation therapy (IORT) is an APBI technique that delivers breast radiotherapy as a single dose at the time of partial mastectomy. We adapted the technique of Veronesi to deliver IORT prior to tumor excision to improve delivery to the region at risk and reduce the volume of normal tissue irradiated., Methods: Patients age >or=55 with ultrasonographically defined tumors
- Published
- 2007
- Full Text
- View/download PDF
15. Radiation therapy and biologics: a ripe opportunity?
- Author
-
Tepper JE and Sartor CI
- Subjects
- Biological Therapy, Biotechnology, Humans, Neoplasms drug therapy, Neoplasms radiotherapy
- Published
- 2006
- Full Text
- View/download PDF
16. Heregulin-dependent delay in mitotic progression requires HER4 and BRCA1.
- Author
-
Muraoka-Cook RS, Caskey LS, Sandahl MA, Hunter DM, Husted C, Strunk KE, Sartor CI, Rearick WA Jr, McCall W, Sgagias MK, Cowan KH, and Earp HS 3rd
- Subjects
- Animals, BRCA1 Protein deficiency, BRCA1 Protein genetics, Breast Neoplasms pathology, Epithelial Cells cytology, Epithelial Cells drug effects, ErbB Receptors genetics, Exons genetics, G2 Phase drug effects, Gene Expression Regulation, Neoplastic drug effects, HeLa Cells, Humans, Mammary Glands, Animal cytology, Mice, Mitogen-Activated Protein Kinase 8 metabolism, Mitogen-Activated Protein Kinase 9 metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Receptor, ErbB-2 metabolism, Receptor, ErbB-4, Tumor Cells, Cultured, BRCA1 Protein metabolism, ErbB Receptors metabolism, Mitosis drug effects, Neuregulin-1 pharmacology
- Abstract
HER4 expression in human breast cancers correlates with a positive prognosis. While heregulin inhibits the growth of HER4-positive breast cancer cells, it does so by undefined mechanisms. We demonstrate that heregulin-induced HER4 activity inhibits cell proliferation and delays G(2)/M progression of breast cancer cells. While investigating pathways of G(2)/M delay, we noted that heregulin increased the expression of BRCA1 in a HER4-dependent, HER2-independent manner. Induction of BRCA1 by HER4 occurred independently of the cell cycle. Moreover, BRCA1 expression was elevated in HER4-postive human breast cancer specimens. Heregulin stimulated c-Jun N-terminal kinase (JNK), and pharmacologic inhibition of JNK impaired heregulin-enhanced expression of BRCA1 and mitotic delay; inhibition of Erk1/2 did not. Knockdown of BRCA1 with small interfering RNA in a human breast cancer cell line interfered with HER4-mediated mitotic delay. Heregulin/HER4-dependent mitotic delay was examined further with an isogenic pair of mouse mammary epithelial cells (MECs) derived from mice harboring homozygous LoxP sites flanking exon 11 of BRCA1, such that one cell line expressed BRCA1 while the other cell line, after Cre-mediated excision, did not. BRCA1-positive MECs displayed heregulin-dependent mitotic delay; however, the isogenic BRCA1-negative MECs did not. These results suggest that heregulin-mediated growth inhibition in HER4-postive breast cancer cells requires BRCA1.
- Published
- 2006
- Full Text
- View/download PDF
17. The ERBB4/HER4 intracellular domain 4ICD is a BH3-only protein promoting apoptosis of breast cancer cells.
- Author
-
Naresh A, Long W, Vidal GA, Wimley WC, Marrero L, Sartor CI, Tovey S, Cooke TG, Bartlett JM, and Jones FE
- Subjects
- Amino Acid Sequence, Breast Neoplasms metabolism, Cell Line, Tumor, Cytochromes c metabolism, ErbB Receptors biosynthesis, Humans, Mitochondria drug effects, Mitochondria metabolism, Models, Molecular, Molecular Sequence Data, Neuregulin-1 pharmacology, Peptide Fragments metabolism, Protein Structure, Tertiary, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-bcl-2 metabolism, Receptor, ErbB-4, Sequence Homology, Amino Acid, bcl-2 Homologous Antagonist-Killer Protein metabolism, Apoptosis physiology, Breast Neoplasms pathology, ErbB Receptors metabolism
- Abstract
ERBB4/HER4 (referred to here as ERBB4) is a unique member of the epidermal growth factor receptor (EGFR) family of receptor tyrosine kinases. In contrast to the other three members of the EGFR family (i.e., EGFR, ERBB2/HER2/NEU, and ERBB3), which are associated with aggressive forms of human cancers, ERBB4 expression seems to be selectively lost in tumors with aggressive phenotypes. Consistent with this observation, we show that ERBB4 induces apoptosis when reintroduced into breast cancer cell lines or when endogenous ERBB4 is activated by a ligand. We further show that ligand activation and subsequent proteolytic processing of endogenous ERBB4 results in mitochondrial accumulation of the ERBB4 intracellular domain (4ICD) and cytochrome c efflux, the essential and committed step of mitochondrial regulated apoptosis. Our results indicate that 4ICD is functionally similar to BH3-only proteins, proapoptotic members of the BCL-2 family required for initiation of mitochondrial dysfunction through activation of the proapoptotic multi-BH domain proteins BAX/BAK. Similar to other BH3-only proteins, 4ICD cell-killing activity requires an intact BH3 domain and 4ICD interaction with the antiapoptotic protein BCL-2, suppressed 4ICD-induced apoptosis. Unique among BH3-only proteins, however, is the essential requirement of BAK but not BAX to transmit the 4ICD apoptotic signal. Clinically, cytosolic but not membrane ERBB4/4ICD expression in primary human breast tumors was associated with tumor apoptosis, providing a mechanistic explanation for the loss of ERBB4 expression during tumor progression. Thus, we propose that ligand-induced mitochondrial accumulation of 4ICD represents a unique mechanism of action for transmembrane receptors, directly coupling a cell surface signal to the tumor cell mitochondrial apoptotic pathway.
- Published
- 2006
- Full Text
- View/download PDF
18. Axillary lymph node count is lower after neoadjuvant chemotherapy.
- Author
-
Neuman H, Carey LA, Ollila DW, Livasy C, Calvo BF, Meyer AA, Kim HJ, Meyers MO, Dees EC, Collichio FA, Sartor CI, Moore DT, Sawyer LR, Frank J, and Klauber-DeMore N
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents therapeutic use, Axilla, Breast Neoplasms surgery, Female, Humans, Lymph Node Excision methods, Lymph Nodes surgery, Lymphatic Metastasis pathology, Lymphatic Metastasis prevention & control, Mastectomy methods, Middle Aged, Neoplasm Staging, Probability, Prognosis, Prospective Studies, Sentinel Lymph Node Biopsy, Statistics, Nonparametric, Treatment Outcome, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Lymph Nodes pathology, Neoadjuvant Therapy, Neoplasm Invasiveness pathology
- Abstract
Background: Retrieval of fewer than 10 lymph nodes at axillary dissection (ALND) for breast cancer can represent anatomic variation or inadequate dissection. We postulated that despite aggressive ALND, a lower lymph node count is more frequent after neoadjuvant chemotherapy., Methods: Patients who received neoadjuvant chemotherapy followed by ALND were compared with patients who received surgery first. All patients received a level I and II ALND at a single institution by one of the breast surgeons. The number of nodes retrieved at ALND was dichotomized into categories (< 10 and > or = 10), and compared using Fisher exact test., Results: A total of 143 neoadjuvant and 170 surgery-first patients were studied. Patients treated with neoadjuvant chemotherapy were significantly more likely to have fewer than 10 lymph nodes retrieved at ALND than were the surgery-first patients (19/143 or 13% vs. 6/170 or 4%, P = .003)., Conclusions: A low lymph node count is more common in patients after treatment with neoadjuvant chemotherapy and should not be assumed to represent an incomplete ALND.
- Published
- 2006
- Full Text
- View/download PDF
19. Size of residual lymph node metastasis after neoadjuvant chemotherapy in locally advanced breast cancer patients is prognostic.
- Author
-
Klauber-DeMore N, Ollila DW, Moore DT, Livasy C, Calvo BF, Kim HJ, Dees EC, Sartor CI, Sawyer LR, Graham M 2nd, and Carey LA
- Subjects
- Adult, Breast Neoplasms pathology, Chi-Square Distribution, Female, Humans, Neoadjuvant Therapy, Prognosis, Proportional Hazards Models, Survival Analysis, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms drug therapy, Lymphatic Metastasis pathology
- Abstract
Background: The prognostic significance of micrometastasis after neoadjuvant chemotherapy for locally advanced breast cancer is unknown. We examined the residual lymph node metastasis size in patients after treatment with neoadjuvant chemotherapy to determine the relevance of metastasis size on outcome., Methods: Stage II/III breast cancer patients treated with neoadjuvant chemotherapy at our institution from 1991 to 2002 were included. We examined the relationship of postneoadjuvant chemotherapy lymph node metastasis size and number with distant disease-free survival (DDFS) and overall survival (OS)., Results: In 122 patients with a median follow-up of 5.4 years, we found not only that patients with an increasing number of residual positive nodes had progressively worse DDFS and OS (P < .0001 for both) compared with patients with negative nodes, but also that the size of the largest lymph node metastasis was associated with worse DDFS and OS (P < .0001 for both) in both univariate and multivariate analysis. Compared with negative nodes, even lymph node micrometastasis (<2 mm) was associated with worsened DDFS and OS (adjusted P = .02 and P = .005, respectively)., Conclusions: Residual micrometastatic disease in the axillary lymph nodes after neoadjuvant chemotherapy is predictive of worse prognosis than negative nodes. In this study, the lymph node metastasis size and the number of involved lymph nodes were independent powerful predictors of DDFS and OS.
- Published
- 2006
- Full Text
- View/download PDF
20. Comparison of three concomitant boost techniques for early-stage breast cancer.
- Author
-
Horton JK, Halle JS, Chang SX, and Sartor CI
- Subjects
- Adult, Aged, Breast Neoplasms diagnostic imaging, Evaluation Studies as Topic, Female, Humans, Male, Middle Aged, Radiotherapy Dosage, Retrospective Studies, Tomography, X-Ray Computed, Breast Neoplasms radiotherapy, Radiotherapy, Intensity-Modulated methods
- Abstract
Purpose: Whole breast radiotherapy (RT) followed by a tumor bed boost typically spans 5-6 weeks of treatment. Interest is growing in RT regimens, such as concomitant boost, that decrease overall treatment time, lessening the time/cost burden to patients and facilities., Methods and Materials: Computed tomography (CT) scans from 20 cases were selected for this retrospective, dosimetric study to compare three different techniques of concomitant boost delivery: (1) standard tangents plus an electron boost, (2) intensity-modulated RT (IMRT) tangents using custom compensators plus an electron boost, and (3) IMRT tangents plus a conformal photon boost. The equivalent uniform dose model was used to compare the plans., Results: The average breast equivalent uniform dose value for the three techniques (standard, IMRT plus electrons, and IMRT plus photons) was 48.6, 47.9, and 48.3, respectively. The plans using IMRT more closely approximated the prescribed dose of 46 Gy to the whole breast. The breast volume receiving >110% of the dose was less with the IMRT tangents than with standard RT (p = 0.037), but no significant difference in the maximal dose or other evaluated parameters was noted., Conclusion: Although the IMRT techniques delivered the prescribed dose with better dose uniformity, the small improvement seen did not support a goal of improved resource use.
- Published
- 2006
- Full Text
- View/download PDF
21. Postmastectomy radiotherapy in intermediate-risk patients: the gray zone.
- Author
-
Sartor CI
- Subjects
- Clinical Trials, Phase III as Topic, Combined Modality Therapy, Female, Humans, Mastectomy adverse effects, Radiotherapy, Adjuvant adverse effects, Risk Factors, Breast Neoplasms radiotherapy, Breast Neoplasms surgery
- Abstract
Postmastectomy radiotherapy continues to be one of the most controversial issues in breast radiotherapy. At the crux of the controversy lies the lack of conclusive studies that specifically address the risk-benefit ratio of postmastectomy radiotherapy for patients at intermediate risk of developing locoregional recurrence. A well-designed phase III trial was initiated, but the trial failed to accrue and was closed prematurely, leaving the issue unresolved. Recent data confirm that postmastectomy radiotherapy yields a clear benefit in breast cancer-specific survival. Furthermore, the risk of cardiac morbidity that historically has offset the benefit of postmastectomy radiotherapy appears to be lessening with modern radiotherapy approaches. However, newer, more efficacious systemic therapy regimens may decrease the risk of locoregional recurrence and increase the risk of toxicity from combined-modality therapy. Recent studies attempt to better stratify patients into risk categories based on disease factors and to estimate the benefit of postmastectomy radiotherapy when traditional risk estimates, such as nodal status, are obscured by neoadjuvant systemic therapy. Nonetheless, a clear consensus on the role of postmastectomy radiotherapy remains elusive for patients who are at intermediate risk of locoregional recurrence after mastectomy.
- Published
- 2006
- Full Text
- View/download PDF
22. Staged sentinel lymph node biopsy before mastectomy facilitates surgical planning for breast cancer patients.
- Author
-
Klauber-Demore N, Calvo BF, Hultman CS, Kim HJ, Meyers MO, Damitz L, Frank JS, Stitzenberg KB, Sartor CI, and Ollila DW
- Subjects
- Adult, Aged, Axilla, Breast Neoplasms therapy, Carcinoma, Ductal, Breast therapy, Carcinoma, Intraductal, Noninfiltrating therapy, Carcinoma, Lobular therapy, Female, Humans, Lymph Node Excision, Lymphatic Metastasis, Mammaplasty, Mastectomy, Middle Aged, Neoplasm Staging, Radiotherapy, Adjuvant, Breast Neoplasms pathology, Carcinoma, Ductal, Breast pathology, Carcinoma, Intraductal, Noninfiltrating pathology, Carcinoma, Lobular pathology, Sentinel Lymph Node Biopsy methods
- Abstract
Background: In patients with breast cancer who choose mastectomy with immediate reconstruction, the sentinel lymph node (SLN) status on permanent histology may complicate treatment if a metastasis is found. The purpose of this study was to determine how performing an SLN biopsy (SLNB) before the definitive operation would influence subsequent surgical procedures., Methods: Our SLN database was searched for patients who underwent staged SLNB with subsequent mastectomy between 2001 and 2004., Results: Twenty-five patients with 27 breast cancers underwent SLNB before mastectomy. Of them, 9 of 27 (33%) were node positive. All 9 patients underwent modified radical mastectomy. Three node-positive patients did not undergo immediate reconstruction. Of the remaining 6 node-positive patients, 5 underwent reconstruction with autologous tissue rather than a tissue expander. In contrast, 6 of 16 (37%) node-negative patients underwent reconstruction with a tissue expander., Conclusions: Staged SLNB assists in selecting the appropriate operation in patients who are considering immediate reconstruction.
- Published
- 2005
- Full Text
- View/download PDF
23. American Joint Committee on Cancer tumor-node-metastasis stage after neoadjuvant chemotherapy and breast cancer outcome.
- Author
-
Carey LA, Metzger R, Dees EC, Collichio F, Sartor CI, Ollila DW, Klauber-DeMore N, Halle J, Sawyer L, Moore DT, and Graham ML
- Subjects
- Adult, Aged, Breast Neoplasms pathology, Chemotherapy, Adjuvant, Disease-Free Survival, Female, Humans, Lymph Node Excision, Lymphatic Metastasis, Mastectomy methods, Middle Aged, Neoplasm, Residual, Prognosis, Proportional Hazards Models, Survival Analysis, United States, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms classification, Breast Neoplasms drug therapy, Lymph Nodes pathology, Neoadjuvant Therapy methods, Neoplasm Staging methods
- Abstract
Background: Response to neoadjuvant chemotherapy is used as an intermediate endpoint for breast cancer relapse and survival. Most breast cancer response classification systems use pathologic complete response, either alone or in conjunction with clinical assessments, to categorize response. We examined the ability of the revised 2003 American Joint Committee on Cancer (AJCC) tumor-node-metastasis (TNM) staging system, which considers both the number of involved axillary lymph nodes and the extent of tumor in the breast to predict patient survival after neoadjuvant chemotherapy for breast cancer., Methods: We assessed the pathologic stage of residual tumor in 132 patients with nonmetastatic breast cancer after they had undergone neoadjuvant chemotherapy and examined the association between AJCC TNM stage and subsequent distant disease-free survival and overall survival. All statistical tests were two-sided., Results: At a median follow-up of 5 years, pathologic stage in the surgical specimens after neoadjuvant chemotherapy using the revised AJCC system was strongly associated with both distant disease-free survival and overall survival. A higher pathologic stage of residual tumor after neoadjuvant chemotherapy was associated with a statistically significant lower rate of distant disease-free survival (stage 0: 95%, stage I: 84%, stage II: 72%, and stage III: 47%; Ptrend < .001). The 5-year distant disease-free survival for patients with residual stage IIIC tumors was only 18% (95% CI = 0% to 36%)., Conclusion: Classification of residual tumor in the breast and axillary surgical specimens after neoadjuvant chemotherapy using the revised AJCC TNM system is useful for predicting distant relapse and survival.
- Published
- 2005
- Full Text
- View/download PDF
24. Inhibition of HER-2/neu kinase impairs androgen receptor recruitment to the androgen responsive enhancer.
- Author
-
Liu Y, Majumder S, McCall W, Sartor CI, Mohler JL, Gregory CW, Earp HS, and Whang YE
- Subjects
- Acetylation drug effects, Androgens physiology, Cell Line, Tumor, Chromatin drug effects, Chromatin genetics, Chromatin Immunoprecipitation, Dihydrotestosterone pharmacology, ErbB Receptors antagonists & inhibitors, Gefitinib, Histones metabolism, Humans, Immunoglobulin Fragments biosynthesis, Immunoglobulin Fragments genetics, Immunoglobulin Fragments immunology, Lapatinib, Male, Prostate-Specific Antigen antagonists & inhibitors, Prostate-Specific Antigen biosynthesis, Prostate-Specific Antigen genetics, Prostatic Neoplasms drug therapy, Prostatic Neoplasms enzymology, Prostatic Neoplasms genetics, Quinazolines pharmacology, Receptor, ErbB-2 immunology, Receptors, Androgen genetics, Receptors, Androgen metabolism, Receptors, Androgen physiology, Response Elements, Transcription, Genetic drug effects, Transcription, Genetic physiology, Androgen Receptor Antagonists, Prostatic Neoplasms metabolism, Receptor, ErbB-2 antagonists & inhibitors
- Abstract
Advanced prostate cancer invariably recurs despite androgen deprivation therapy. The androgen receptor (AR) likely plays a key role in this progression and in the continued survival and proliferation of prostate cancer cells in the low androgen environment. Cross-talk with growth factor receptors, such as epidermal growth factor receptor (EGFR) family, has been postulated as a potential mechanism to activate AR in recurrent prostate cancer. We have investigated the role of HER-2/neu (ErbB-2) tyrosine kinase in AR function by characterizing the effect of inhibiting endogenous HER-2 activity in LNCaP cells. We used two independent methods, expression of intracellular single-chain antibody against HER-2 and treatment with a novel dual EGFR/HER-2 kinase inhibitor GW572016 (lapatinib). Expression of intracellular HER-2 antibody scFv-5R and treatment with GW572016 inhibited HER-2 signaling. This HER-2 inhibition led to impairment of AR-mediated functions, such as androgen-stimulated growth and the induction of endogenous prostate-specific antigen (PSA) mRNA and protein. Androgen-stimulated recruitment of AR and histone acetylation at the androgen responsive enhancer of the PSA gene, detected by chromatin immunoprecipitation analysis, were impaired by HER-2 inhibition. GW572016 was more potent in its ability to inhibit PSA expression and AR recruitment and histone acetylation than the EGFR-selective kinase inhibitor ZD1839 (gefitinib), consistent with the HER-2 kinase playing the major role in AR regulation. These results show that HER-2 signaling is required for optimal transcriptional activity of AR in prostate cancer cells and suggest that HER-2 inhibition may provide a novel strategy to disrupt AR function in prostate cancer.
- Published
- 2005
- Full Text
- View/download PDF
25. Effect of addition of adjuvant paclitaxel on radiotherapy delivery and locoregional control of node-positive breast cancer: cancer and leukemia group B 9344.
- Author
-
Sartor CI, Peterson BL, Woolf S, Fitzgerald TJ, Laurie F, Turrisi AJ, Bogart J, Henderson IC, and Norton L
- Subjects
- Aged, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Chemotherapy, Adjuvant, Combined Modality Therapy, Cyclophosphamide administration & dosage, Doxorubicin administration & dosage, Elective Surgical Procedures, Female, Humans, Mastectomy, Mastectomy, Segmental, Middle Aged, Neoplasm Recurrence, Local, Radiotherapy methods, Retrospective Studies, Antineoplastic Agents, Phytogenic administration & dosage, Breast Neoplasms therapy, Paclitaxel administration & dosage
- Abstract
Purpose: We compared radiotherapy (RT) delivery and locoregional control in patients with node-positive breast cancer randomly assigned on Cancer and Leukemia Group B 9344 to receive adjuvant doxorubicin/cyclophosphamide (AC) with patients assigned to receive AC followed by paclitaxel (AC+T)., Methods: Eligible patients were randomly assigned to receive adjuvant AC versus AC+T chemotherapy. RT was required if breast-conserving surgery was performed but was elective after mastectomy. Information about RT delivery was retrospectively collected. Cumulative incidence of locoregional recurrence (LRR), use of elective RT, and RT delivery were compared between treatment arms., Results: For patients treated with breast-conserving surgery and RT, the 5-year cumulative incidence of isolated LRR was 9.7% in the AC arm and 3.7% in the AC+T arm (P = .04) and of LRR as any component of failure was 12.9% versus 6.1%, respectively (P = .04). Although LRR rates in patients who did not receive postmastectomy RT were lower in the AC+T arm, the difference was not statistically significant. Despite the lack of protocol guidelines, RT use did not differ between arms, nor did RT dose, treatment interruption, or completion., Conclusion: Despite the delay to RT during additional chemotherapy, adjuvant AC+T afforded better local control than AC alone in patients treated with breast-conserving therapy. Addition of paclitaxel did not adversely affect delivery or ability to tolerate RT, as indicated by similar rates of completion of timely, full-dose RT between arms.
- Published
- 2005
- Full Text
- View/download PDF
26. Central nervous system metastases in women after multimodality therapy for high risk breast cancer.
- Author
-
Carey LA, Ewend MG, Metzger R, Sawyer L, Dees EC, Sartor CI, Moore DT, and Graham ML
- Subjects
- Adult, Aged, Breast Neoplasms pathology, Carcinoma, Ductal, Breast epidemiology, Carcinoma, Ductal, Breast secondary, Carcinoma, Lobular epidemiology, Carcinoma, Lobular secondary, Central Nervous System Neoplasms epidemiology, Central Nervous System Neoplasms secondary, Cohort Studies, Combined Modality Therapy, Female, Humans, Middle Aged, Prevalence, Retrospective Studies, Risk Factors, Antineoplastic Agents therapeutic use, Breast Neoplasms drug therapy, Carcinoma, Ductal, Breast drug therapy, Carcinoma, Lobular drug therapy, Central Nervous System Neoplasms drug therapy
- Abstract
Background: Central nervous system (CNS) relapse is increasing in breast cancer. This increase may reflect altered failure patterns from adjuvant therapy, more effective systemic therapy with improved control in non-CNS sites, or a resistant breast cancer subtype., Methods: To determine the factors associated with clinical CNS relapse, we examined response to neoadjuvant chemotherapy (chemosensitivity), time to relapse and sites of relapse in a cohort of 140 patients without evidence of metastasis at presentation., Results: At 5 years (interquartile range 3-6 years), 44 (31%) patients developed distant metastases, including 13 with CNS metastases. CNS relapse was early (median 24 months after diagnosis) and associated with relapse in bone and liver, suggesting hematogenous dissemination. Those with CNS relapse were younger at diagnosis (40 versus 49 years) and more likely to have lymphovascular invasion in the primary tumor compared with non-CNS metastases. Response to neoadjuvant chemotherapy was not different (69% versus 73% response rate) between the two groups. Extent of residual disease after chemotherapy was strongly associated with relapse outside the CNS but not CNS relapses. The CNS was an isolated or dominant site of metastasis in 8 of 13. Despite treatment, most patients with CNS involvement died of neurologic causes a median of 6 months later., Conclusion: Breast cancers that develop CNS metastases differ from those that develop metastases elsewhere. Both tumor behavior and reduced chemotherapy accessibility to the CNS may contribute to increased CNS involvement in breast cancer patients treated with multimodality therapy.
- Published
- 2004
- Full Text
- View/download PDF
27. Mechanisms of disease: Radiosensitization by epidermal growth factor receptor inhibitors.
- Author
-
Sartor CI
- Subjects
- Combined Modality Therapy, DNA Repair, Down-Regulation drug effects, Humans, Signal Transduction drug effects, Antineoplastic Agents therapeutic use, Epidermal Growth Factor therapeutic use, ErbB Receptors antagonists & inhibitors, Neoplasms drug therapy, Neoplasms radiotherapy, Radiation-Sensitizing Agents therapeutic use
- Abstract
The epidermal growth factor receptor (EGFR) inhibitors are among the most intensely studied new molecular therapeutic agents. Although response rates have been somewhat disappointing when EGFR inhibitors are used as single-agent therapy for advanced disease, these inhibitors may be more effective as chemo- and radiosensitizers. The first phase III randomized trial evaluating EGFR inhibitors as radiosensitizers in patients with locally advanced head and neck cancer was strongly positive, indicating significant potential of this class of agents to improve outcome with radiotherapy. However, optimal implementation of EGFR inhibitors as radiosensitizers depends, in part, on a better understanding of the mechanisms of radiosensitization. Preclinical studies provide important observations with regard to potential mechanisms. The phenotypic cellular changes associated with EGFR inhibition are impressively consistent between different model systems, with almost all studies showing that EGFR inhibitors affect proliferation, angiogenesis, and cell survival. Whether EGFR inhibitors influence response to radiation directly, or whether the improved response is a result of additive effects of the two modalities, remains unclear. However, cell-cycle arrest, endothelial cell sensitivity, and apoptotic potential are all important factors in radiation response of epithelial tumors. Furthermore, less-studied effects of EGFR inhibitors on DNA repair suggest that modulation of DNA damage response to cytotoxic injury might result in radio- or chemosensitization. This review will explore potential mechanisms of radiosensitization by EGFR inhibitors.
- Published
- 2004
- Full Text
- View/download PDF
28. High-resolution axillary ultrasound is a poor prognostic test for determining pathologic lymph node status in patients undergoing neoadjuvant chemotherapy for locally advanced breast cancer.
- Author
-
Klauber-Demore N, Kuzmiak C, Rager EL, Ogunrinde OB, Ollila DW, Calvo BF, Kim HJ, Meyer A, Dees C, Graham M 2nd, Collichio FA, Sartor CI, Metzger R, and Carey LA
- Subjects
- Adult, Aged, Axilla, Breast Neoplasms pathology, Female, Humans, Lymphatic Metastasis, Middle Aged, Neoplasm Staging, Prognosis, Ultrasonography, Breast Neoplasms diagnostic imaging, Breast Neoplasms drug therapy
- Abstract
Background: The purpose of this study was to evaluate the efficacy of high-resolution axillary ultrasound in detecting axillary lymph node metastases after neoadjuvant chemotherapy in patients with locally advanced breast cancer., Methods: Fifty-three patients with stage II or III breast cancer undergoing neoadjuvant chemotherapy who had a physical examination, high-resolution axillary ultrasound, and axillary lymph node dissection from January 1999 to September 2003 were included in this study., Results: The positive predictive value of the postchemotherapy ultrasound for predicting pathologic nodal involvement was 83%, but the negative predictive value was only 52%. Postchemotherapy physical examination was also poor at predicting pathologic nodal involvement with a positive predictive value of 93% and a negative predictive value of only 58%., Conclusions: A negative post-neoadjuvant chemotherapy high-resolution axillary ultrasound or physical examination does not predict pathologic node status, and this test has limited value in this setting.
- Published
- 2004
- Full Text
- View/download PDF
29. Involvement of Ras activation in human breast cancer cell signaling, invasion, and anoikis.
- Author
-
Eckert LB, Repasky GA, Ulkü AS, McFall A, Zhou H, Sartor CI, and Der CJ
- Subjects
- Anoikis genetics, Breast Neoplasms enzymology, Breast Neoplasms genetics, Cell Line, Tumor, Cyclooxygenase 2, Estrogen Receptor alpha, Gene Expression Regulation, Neoplastic, Humans, Isoenzymes biosynthesis, Isoenzymes genetics, Isoenzymes metabolism, MAP Kinase Signaling System physiology, Membrane Proteins, Mitogen-Activated Protein Kinases metabolism, Neoplasm Invasiveness, Phosphatidylinositol 3-Kinases metabolism, Prostaglandin-Endoperoxide Synthases biosynthesis, Prostaglandin-Endoperoxide Synthases genetics, Prostaglandin-Endoperoxide Synthases metabolism, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-akt, Receptor, ErbB-2 biosynthesis, Receptor, ErbB-2 genetics, Receptor, ErbB-2 physiology, Receptors, Estrogen biosynthesis, Signal Transduction physiology, ras Proteins genetics, Anoikis physiology, Breast Neoplasms pathology, ras Proteins physiology
- Abstract
Although mutated forms of ras are not associated with the majority of breast cancers (<5%), there is considerable experimental evidence that hyperactive Ras can promote breast cancer growth and development. Therefore, we determined whether Ras and Ras-responsive signaling pathways were activated persistently in nine widely studied human breast cancer cell lines. Although only two of the lines harbor mutationally activated ras, we found that five of nine breast cancer cell lines showed elevated active Ras-GTP levels that may be due, in part, to HER2 activation. Unexpectedly, activation of two key Ras effector pathways, the extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase and phosphatidylinositol 3'-kinase/AKT signaling pathways, was not always associated with Ras activation. Ras activation also did not correlate with invasion or the expression of proteins associated with tumor cell invasion (estrogen receptor alpha and cyclooxygenase 2). We then examined the role of Ras signaling in mediating resistance to matrix deprivation-induced apoptosis (anoikis). Surprisingly, we found that ERK and phosphatidylinositol 3'-kinase/AKT activation did not have significant roles in conferring anoikis resistance. Taken together, these observations show that Ras signaling exhibits significant cell context variations and that other effector pathways may be important for Ras-mediated oncogenesis, as well as for anoikis resistance, in breast cancer. Additionally, because ERK and AKT activation are not strictly associated with Ras activation, pharmacological inhibitors of these two signaling pathways may not be the best approach for inhibition of aberrant Ras function in breast cancer treatment.
- Published
- 2004
- Full Text
- View/download PDF
30. Effects of the EGFR/HER2 kinase inhibitor GW572016 on EGFR- and HER2-overexpressing breast cancer cell line proliferation, radiosensitization, and resistance.
- Author
-
Zhou H, Kim YS, Peletier A, McCall W, Earp HS, and Sartor CI
- Subjects
- Breast Neoplasms metabolism, Breast Neoplasms radiotherapy, Cell Division drug effects, Cell Division radiation effects, Cell Line, Tumor drug effects, Cell Line, Tumor radiation effects, Drug Resistance, Neoplasm, ErbB Receptors metabolism, Female, Humans, Lapatinib, Neoplasm Proteins metabolism, Phosphorylation, Radiation Tolerance, Receptor, ErbB-2 metabolism, Signal Transduction drug effects, Tumor Stem Cell Assay, Breast Neoplasms drug therapy, ErbB Receptors antagonists & inhibitors, Neoplasm Proteins antagonists & inhibitors, Quinazolines pharmacology, Receptor, ErbB-2 antagonists & inhibitors
- Abstract
Purpose: Two members of the epidermal growth factor receptor family, EGFR and HER2, have been implicated in radioresistance in breast cancer and other malignancies. To gauge the potential clinical utility of targeting both EGFR and HER2 to control growth and radiosensitize human breast cancers, we examined the effect of a dual EGFR/HER2 inhibitor, GW572016, on the proliferation and radiation response of either EGFR- or HER2-overexpressing human breast cancer cell lines., Methods and Materials: Primary human breast cancer cell lines that endogenously overexpress EGFR or HER2 and luminal mammary epithelial H16N2 cells stably transfected with HER2 were evaluated for the effect of GW572016 on inhibition of ligand-induced or constitutive receptor phosphorylation, proliferation, radiosensitization, and inhibition of downstream signaling., Results: GW572016 inhibited constitutive and/or ligand-induced EGFR or HER2 tyrosine phosphorylation of all five cell lines, which correlated with the antiproliferative response in all but one cell line. GW572016 radiosensitized EGFR-overexpressing cell lines, but HER2-overexpressing cells were unable to form colonies after brief exposure to GW572016 even in the absence of radiation, and thus could not be evaluated for radiosensitization. One cell line was resistant to the antiproliferative and radiosensitizing effects of GW572016, despite receptor inhibition. Exploration of potential mechanisms of resistance in SUM185 cells revealed failure of GW572016 to inhibit downstream ERK and Akt activation, despite inhibition of HER2 phosphorylation. In contrast, sensitive HER2-overexpressing cell lines demonstrated inhibition of both ERK and Akt phosphorylation., Conclusion: GW572016 potently inhibits receptor phosphorylation in either EGFR- or HER2-overexpressing cell lines and has both antiproliferative and radiosensitizing effects. Resistance to GW572016 was not due to a lack of receptor inhibition, but rather with a lack of inhibition of ERK and Akt, suggesting that measurement of inhibition of crucial signaling pathways may better predict response than inhibition of receptor phosphorylation. The SUM185 cell line provides a valuable model for studying mechanisms of resistance of EGFR/HER2 inhibitor therapy.
- Published
- 2004
- Full Text
- View/download PDF
31. Epidermal growth factor receptor autocrine signaling in RIE-1 cells transformed by the Ras oncogene enhances radiation resistance.
- Author
-
Grana TM, Sartor CI, and Cox AD
- Subjects
- Animals, Cell Line, Transformed, Culture Media, Conditioned, Epithelial Cells metabolism, Epithelial Cells physiology, Epithelial Cells radiation effects, ErbB Receptors antagonists & inhibitors, ErbB Receptors metabolism, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, Phosphorylation, Radiation Tolerance genetics, Rats, Signal Transduction drug effects, Signal Transduction physiology, Transforming Growth Factor alpha pharmacology, ErbB Receptors physiology, Genes, ras physiology, Radiation Tolerance physiology
- Abstract
Oncogenic forms of the small GTPase Ras increase the resistance of cells to killing by ionizing radiation (IR). Although not all of the signaling pathways for radioresistance are well defined, it is now clear that Ras-dependent signaling pathways involved in radioresistance include those mediated by phosphatidylinositol 3'-kinase (PI3-K) and Raf. Nevertheless, PI3-K and Raf together are not sufficient to reconstitute all of the resistance conferred by Ras, indicating that other effectors must also contribute. We show here that Ras-driven autocrine signaling through the epidermal growth factor receptor (EGFR) also contributes to radioresistance in Ras-transformed cells. Conditioned media (CM) collected from RIE-1 rat intestinal epithelial cells expressing oncogenic Ras increased the survival of irradiated cells. Ras-CM contains elevated levels of the EGFR ligand transforming growth factor alpha (TGF-alpha). Both Ras-CM and TGF-alpha stimulated EGFR phosphorylation, and exogenous TGF-alpha mimicked the effects of Ras-CM to increase radioresistance. Blocking EGFR signaling with the EGFR/HER-2 kinase inhibitor (KI) GW572016 decreased the postradiation survival of irradiated Ras-transformed cells and normal cells but had no effect on the survival of unirradiated cells. Ras-CM and TGF-alpha also increase PI3-K activity downstream of the EGFR and increase postradiation survival, both of which are abrogated by GW572016. Thus, Ras utilizes autocrine signaling through EGFR to increase radioresistance, and the EGFR KI GW572016 acts as a radiosensitizer. The observation that Ras-transformed cells can be sensitized to killing by ionizing radiation with GW572016 demonstrates that EGFR KIs could potentially be used to radiosensitize tumors in which radioresistance is dependent on Ras-driven autocrine signaling through EGFR.
- Published
- 2003
32. Randomized trial of dose-dense versus conventionally scheduled and sequential versus concurrent combination chemotherapy as postoperative adjuvant treatment of node-positive primary breast cancer: first report of Intergroup Trial C9741/Cancer and Leukemia Group B Trial 9741.
- Author
-
Citron ML, Berry DA, Cirrincione C, Hudis C, Winer EP, Gradishar WJ, Davidson NE, Martino S, Livingston R, Ingle JN, Perez EA, Carpenter J, Hurd D, Holland JF, Smith BL, Sartor CI, Leung EH, Abrams J, Schilsky RL, Muss HB, and Norton L
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Breast Neoplasms surgery, Chemotherapy, Adjuvant, Cyclophosphamide administration & dosage, Doxorubicin administration & dosage, Drug Administration Schedule, Female, Filgrastim, Granulocyte Colony-Stimulating Factor therapeutic use, Humans, Middle Aged, Multivariate Analysis, Paclitaxel administration & dosage, Proportional Hazards Models, Recombinant Proteins, Survival Analysis, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Breast Neoplasms drug therapy
- Abstract
Purpose: Using a 2 x 2 factorial design, we studied the adjuvant chemotherapy of women with axillary node-positive breast cancer to compare sequential doxorubicin (A), paclitaxel (T), and cyclophosphamide (C) with concurrent doxorubicin and cyclophosphamide (AC) followed by paclitaxel (T) for disease-free (DFS) and overall survival (OS); to determine whether the dose density of the agents improves DFS and OS; and to compare toxicities., Patients and Methods: A total of 2,005 female patients were randomly assigned to receive one of the following regimens: (I) sequential A x 4 (doses) --> T x 4 --> C x 4 with doses every 3 weeks, (II) sequential A x 4 --> T x 4 --> C x 4 every 2 weeks with filgrastim, (III) concurrent AC x 4 --> T x 4 every 3 weeks, or (IV) concurrent AC x 4 --> T x 4 every 2 weeks with filgrastim., Results: A protocol-specified analysis was performed at a median follow-up of 36 months: 315 patients had experienced relapse or died, compared with 515 expected treatment failures. Dose-dense treatment improved the primary end point, DFS (risk ratio [RR] = 0.74; P =.010), and OS (RR = 0.69; P =.013). Four-year DFS was 82% for the dose-dense regimens and 75% for the others. There was no difference in either DFS or OS between the concurrent and sequential schedules. There was no interaction between density and sequence. Severe neutropenia was less frequent in patients who received the dose-dense regimens., Conclusion: Dose density improves clinical outcomes significantly, despite the lower than expected number of events at this time. Sequential chemotherapy is as effective as concurrent chemotherapy.
- Published
- 2003
- Full Text
- View/download PDF
33. The EGF receptor family--multiple roles in proliferation, differentiation, and neoplasia with an emphasis on HER4.
- Author
-
Earp HS 3rd, Calvo BF, and Sartor CI
- Subjects
- Animals, Breast Neoplasms etiology, Breast Neoplasms genetics, Breast Neoplasms physiopathology, Cell Differentiation physiology, Cell Division physiology, ErbB Receptors genetics, Female, Humans, Ligands, Neuregulin-1 physiology, Receptor, ErbB-4, Signal Transduction, Tumor Cells, Cultured, ErbB Receptors physiology, Neoplasms etiology
- Abstract
The EGF Receptor (EGFR), the first transmembrane receptor tyrosine kinase cloned and sequenced, and its closely related family members HER2, HER3, and HER4, play myriad roles in mammalian growth and development. Receptor activation involves ligand binding to separate receptors followed by formation of active dimers. These receptors can signal as homodimers or they can subtly alter signaling output by heterodimerizing with other family members. Adding complexity, these receptors with varying specificity bind at least 10 ligands from two ligand families, the EGF and neuregulin/heregulin families. This signaling system's impact on human neoplasia is underscored by the following: i.) EGFR is overexpressed or activated by autocrine or paracrine growth factor loops in at least 50% of epithelial malignancies; ii.) HER2 is amplified and dramatically overexpressed in approximately 20%-25% or breast cancers; iii) HER3 and HER4 are variably expressed in breast and other cancers. Overexpression and/or activation of EGFR, HER2 and HER3 has been correlated with poor tumor prognosis; antibody and small molecule inhibitors of their activity are being tested as therapy in cancer patients. However, the signaling complexity engendered by four interacting receptors and ten ligands makes it difficult to definitively measure receptor signaling output in human tumors and even makes mechanistic studies of the family's role in normal physiology and neoplastic transformation a challenge. In spite of the literature's emphasis on growth control, activation by some EGF receptor family member ligands can produce tumor cell differentiation, characterized by growth cessation and differentiation gene product synthesis. The present work delineates a role for HER4 in breast cancer cell differentiation and demonstrates that HER4 is both necessary and sufficient to produce an anti-proliferative signal. These
- Published
- 2003
34. Epidermal growth factor family receptors and inhibitors: radiation response modulators.
- Author
-
Sartor CI
- Subjects
- Down-Regulation drug effects, Down-Regulation physiology, Humans, Receptor Protein-Tyrosine Kinases drug effects, Receptor Protein-Tyrosine Kinases physiology, Transcriptional Activation drug effects, Transcriptional Activation physiology, Epidermal Growth Factor therapeutic use, ErbB Receptors drug effects, ErbB Receptors physiology, Head and Neck Neoplasms drug therapy, Head and Neck Neoplasms physiopathology
- Abstract
Growing evidence suggests that epidermal growth factor family receptors (HERs) play a significant role in radiation response. EGFR expression levels and activation by ligand correlate with radioresistance, and exogenous HER2 expression alters radiation response. Preclinical studies of anti-EGFR anti-HER2 antibodies, and kinase inhibitors that inhibit EGFR, both EGFR and HER2, or all 4 family members show potential for clinical radiosensitization. Early-phase clinical trials of the anti-EGFR antibody, C225, prove the combination of C225 and radiotherapy to be well tolerated and promising. A phase 3 randomized trial in head and neck cancer is underway, and clinical investigation of other HER inhibitors is in progress. The mechanisms(s) of radiation response modulation by HERs appear complex and diverse. Signal transduction initiated by receptor activation promotes survival and proliferation after ionizing radiation, and HER inhibitors affect cellular responses to ionizing radiation (IR) in diverse ways, including inducing apoptosis, cell cycle arrest, and impeding DNA repair. HER signaling and inhibition also affect tumor-stroma interactions, particularly angiogenesis and endothelial survival after IR. Further investigation of the radiation response modulation by EGFR family members and their inhibitors will lead to optimization of this promising therapeutic approach., (Copyright 2003, Elsevier Science (USA). All rights reserved.)
- Published
- 2003
- Full Text
- View/download PDF
35. Molecular targets as therapeutic strategies in the management of breast cancer.
- Author
-
Sartor CI
- Subjects
- Breast Neoplasms blood supply, Breast Neoplasms metabolism, Epidermal Growth Factor metabolism, Female, Humans, NF-kappa B antagonists & inhibitors, Neovascularization, Pathologic prevention & control, Angiogenesis Inhibitors therapeutic use, Apoptosis drug effects, Breast Neoplasms therapy, Epidermal Growth Factor antagonists & inhibitors
- Abstract
Therapy directed against specific biologic targets has long been used in the treatment of breast cancer; the estrogen receptor is a validated prognostic and therapeutic target, and antiestrogen therapy has been used effectively for decades. Recently, scientific progress and increased comprehension of mechanisms of breast cancer pathogenesis have led to the proliferation of both potential molecular targets and new therapeutic agents. The success of traztuzumab (Herceptin, Genentech, South San Francisco, CA), an anti-HER2 antibody, has spurred the development of other biologically directed therapeutics. In this overview, I discuss three targets relevant to breast cancer (the epidermal growth factor receptor family, angiogenesis, and NF-kappa B), and therapeutic approaches directed against these targets are discussed., (Copyright 2002, Elsevier Science (USA).)
- Published
- 2002
- Full Text
- View/download PDF
36. Is less more? Lessons in radiation schedules in breast cancer.
- Author
-
Sartor CI and Tepper JE
- Subjects
- Dose Fractionation, Radiation, Female, Humans, Breast Neoplasms radiotherapy
- Published
- 2002
- Full Text
- View/download PDF
37. Ras mediates radioresistance through both phosphatidylinositol 3-kinase-dependent and Raf-dependent but mitogen-activated protein kinase/extracellular signal-regulated kinase kinase-independent signaling pathways.
- Author
-
Grana TM, Rusyn EV, Zhou H, Sartor CI, and Cox AD
- Subjects
- Animals, Epithelial Cells physiology, Epithelial Cells radiation effects, Intestines cytology, Intestines physiology, Intestines radiation effects, JNK Mitogen-Activated Protein Kinases, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3, Mitogen-Activated Protein Kinases metabolism, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation radiation effects, Protein Isoforms, Rats, p38 Mitogen-Activated Protein Kinases, MAP Kinase Signaling System physiology, Phosphatidylinositol 3-Kinases physiology, Proto-Oncogene Proteins c-raf physiology, Radiation Tolerance physiology, ras Proteins physiology
- Abstract
Cells transformed by the oncogenic small GTPase, Ras, display a radioresistant phenotype in response to ionizing radiation (IR). To determine the mechanisms by which Ras mediates radioresistance in epithelial cells, we assessed the importance of three major survival pathways that can be activated by Ras [phosphatidylinositol 3-kinase (PI3-K)>Akt, nuclear factor kappaB (NF-kappaB), and Raf>mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEK)>extracellular signal-regulated kinase] as necessary or sufficient for Ras-mediated radioresistance in matched pairs of RIE-1 rat intestinal epithelial cells expressing oncogenic Ras or empty vector (RIE-Ras and RIE-vector). Inhibiting PI3-K with LY294002 sensitized RIE-1 cells to IR in a dose-dependent manner, indicating that PI3-K is necessary for radioresistance, whereas inhibition of NF-kappaB with the super-repressor IkappaBalpha had little effect on survival. Expression of either the constitutively active catalytic subunit of PI3-K, p110alpha-CAAX, or the Ras effector domain mutant 12V/40C, which retains binding to PI3-K but is impaired in binding to other Ras effectors, was sufficient to confer partial radioresistance. Expression of either a constitutively active form of the serine/threonine kinase Raf-1 or the Ras effector domain mutant 12V/35S, which retains binding to Raf but is impaired in binding to other Ras effectors, was also sufficient to confer partial radioresistance. Surprisingly, however, even complete inhibition of MEK activity by using U0126 resulted in no change in post-IR survival whatsoever. Thus, whereas Raf contributes to Ras-mediated radioresistance, this is accomplished through a MEK-independent pathway. Taken together, these results indicate that multiple pathways, including both PI3-K-dependent and Raf-dependent but MEK-independent signaling, are required for Ras-mediated radioresistance in epithelial cells. Finally, we demonstrate that Ras-mediated radioresistance can be uncoupled from Ras-mediated transformation, in that PI3-K is required for radioresistance but not transformation, whereas MEK and NF-kappaB are required for transformation but not radioresistance in RIE-1 epithelial cells.
- Published
- 2002
38. Postmastectomy radiotherapy in women with breast cancer metastatic to one to three axillary lymph nodes.
- Author
-
Sartor CI
- Subjects
- Breast Neoplasms pathology, Female, Humans, Mastectomy, Neoplasm Staging, Radiotherapy, Adjuvant, Randomized Controlled Trials as Topic, Research Design, Risk Factors, Survival Analysis, Breast Neoplasms radiotherapy, Breast Neoplasms surgery, Lymphatic Metastasis radiotherapy
- Abstract
The influence of postmastectomy radiotherapy on survival has long been debated. Early randomized trials established a clear role for adjuvant postmastectomy chest wall radiotherapy (PMCWRT) in reducing locoregional recurrence (LRR), and PMCWRT became standard therapy for patients at high risk of LRR: those with T3 or T4 tumors and four or more involved lymph nodes. However, without effective systemic therapy, distant metastases limited any effect of improved local control on overall outcome, and radiotherapy showed no benefit in survival. In fact, early meta-analyses showed a negative impact of radiotherapy on survival. As data and techniques matured, a favorable influence of PMCWRT on breast cancer-specific mortality emerged but was offset by a radiotherapy-related increase in vascular mortality. Improvements in radiotherapy delivery to increase efficacy and reduce toxicity, restriction of PMCWRT to patients at intermediate or high risk of LRR after mastectomy, and improved distant control of disease with systemic therapy are expected to bring the greatest likelihood of a survival advantage from locoregional control. Three randomized trials with sufficient follow-up meet these criteria. All demonstrate significant improvement in overall survival with PMCWRT. However, the trials were not designed to specifically address the benefit of PMCWRT in patients at intermediate risk of LRR (those with T1 or T2 tumors and one to three involved lymph nodes). These findings have been discussed in a host of publications and conferences in light of historical negative results. This review focuses on the recent data on PMCWRT in patients with one to three involved nodes.
- Published
- 2001
- Full Text
- View/download PDF
39. Her4 mediates ligand-dependent antiproliferative and differentiation responses in human breast cancer cells.
- Author
-
Sartor CI, Zhou H, Kozlowska E, Guttridge K, Kawata E, Caskey L, Harrelson J, Hynes N, Ethier S, Calvo B, and Earp HS 3rd
- Subjects
- Breast Neoplasms metabolism, Cell Differentiation physiology, Cell Division physiology, Cell Size, Female, Flow Cytometry, Heparin-binding EGF-like Growth Factor, Humans, Immunoblotting, Intercellular Signaling Peptides and Proteins, Ligands, Phosphorylation, Phosphotyrosine metabolism, RNA, Messenger metabolism, Receptor, ErbB-2 metabolism, Receptor, ErbB-4, Signal Transduction physiology, Tumor Cells, Cultured, Breast Neoplasms pathology, Cell Differentiation drug effects, Cell Division drug effects, Epidermal Growth Factor pharmacology, ErbB Receptors metabolism, Neuregulin-1 pharmacology
- Abstract
The function of the epidermal growth factor receptor (EGFR) family member HER4 remains unclear because its activating ligand, heregulin, results in either proliferation or differentiation. This variable response may stem from the range of signals generated by HER4 homodimers versus heterodimeric complexes with other EGFR family members. The ratio of homo- and heterodimeric complexes may be influenced both by a cell's EGFR family member expression profile and by the ligand or even ligand isoform used. To define the role of HER4 in mediating antiproliferative and differentiation responses, human breast cancer cell lines were screened for responses to heregulin. Only cells that expressed HER4 exhibited heregulin-dependent antiproliferative responses. In-depth studies of one line, SUM44, demonstrated that the antiproliferative and differentiation responses correlated with HER4 activation and were abolished by stable expression of a kinase-inactive HER4. HB-EGF, a HER4-specific ligand in this EGFR-negative cell line, also induced an antiproliferative response. Moreover, introduction and stable expression of HER4 in HER4-negative SUM102 cells resulted in the acquisition of a heregulin-dependent antiproliferative response, associated with increases in markers of differentiation. The role of HER2 in these heregulin-dependent responses was examined through elimination of cell surface HER2 signaling by stable expression of a single-chain anti-HER2 antibody that sequestered HER2 in the endoplasmic reticulum. In the cell lines with either endogenously (SUM44) or exogenously (SUM102) expressed HER4, elimination of HER2 did not alter HER4-dependent decreases in cell growth. These results suggest that HER4 is both necessary and sufficient to trigger an antiproliferative response in human breast cancer cells.
- Published
- 2001
- Full Text
- View/download PDF
40. Clinical significance of micrometastatic disease in the era of sentinel node.
- Author
-
Ollila DW, Carey LA, and Sartor CI
- Published
- 2001
- Full Text
- View/download PDF
41. Biological modifiers as potential radiosensitizers: targeting the epidermal growth factor receptor family.
- Author
-
Sartor CI
- Subjects
- Animals, Antibodies, Monoclonal, Humanized, Antineoplastic Agents pharmacology, Breast Neoplasms genetics, Breast Neoplasms radiotherapy, Cell Cycle, Cetuximab, ErbB Receptors genetics, Gene Expression, Head and Neck Neoplasms radiotherapy, Humans, Immunologic Factors pharmacology, Radiation-Sensitizing Agents pharmacology, Signal Transduction, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Breast Neoplasms drug therapy, ErbB Receptors antagonists & inhibitors, Genes, erbB-2, Head and Neck Neoplasms drug therapy, Immunologic Factors therapeutic use, Radiation-Sensitizing Agents therapeutic use
- Abstract
The epidermal growth factor receptor family plays an important role in the pathogenesis of human epithelial tumors. Overexpression is associated with poor prognosis and resistance to therapy. Epidermal growth factor receptor family members activate signal transduction pathways that have been implicated in radioresistance, and inhibition of signal transduction pathways involved in epidermal growth factor receptor family member signaling causes radiosensitization. Recent encouraging results indicate that epidermal growth factor receptor family member inhibitors may be specific, effective radiosensitizers in tumors that overexpress one or more of these receptors.
- Published
- 2000
42. Rate of PSA rise predicts metastatic versus local recurrence after definitive radiotherapy.
- Author
-
Sartor CI, Strawderman MH, Lin XH, Kish KE, McLaughlin PW, and Sandler HM
- Subjects
- Adenocarcinoma radiotherapy, Diagnosis, Differential, Humans, Male, Prostatic Neoplasms radiotherapy, Treatment Failure, Adenocarcinoma blood, Adenocarcinoma secondary, Neoplasm Recurrence, Local blood, Prostate-Specific Antigen blood, Prostatic Neoplasms blood, Prostatic Neoplasms pathology
- Abstract
Objective: A rising prostate specific antigen (PSA) following treatment for adenocarcinoma of the prostate indicates eventual clinical failure, but the rate of rise can be quite different from patient to patient, as can the pattern of clinical failure. We sought to determine whether the rate of PSA rise could differentiate future local versus metastatic failure., Methods and Materials: Two thousand six hundred sixty-seven PSA values from 400 patients treated with radiotherapy for localized adenocarcinoma of the prostate were analyzed with respect to PSA patterns and clinical outcome. Patients had received no hormonal therapy or prostate surgery and had > 4 PSA values post-treatment. PSA rate of rise, determined by the slope of the natural log, was classified as gradual [< 0.69 log(ng/ml)/year, or doubling time (DT) > 1 year], moderate [0.69-1.4 log(ng/ml)/year, or DT 6 months-1 year], or rapid [> 1.4 log(ng/ml)/year, or DT < 6 months]., Results: Sixty-one percent of patients had non-rising PSA following treatment; 25% of patients with rising PSA developed clinical failure, and 93% of patients with clinical failure had rising PSA. The rate of rise discerned different clinical failure patterns. Local failure occurred in 23% of patients with moderate rate of rise versus 7% with gradual rise (p = 0.0001). Metastatic disease developed in 46% of those with rapid rise versus 8% with moderate rise (p < 0.0001). By multivariate analysis, in addition to rate of rise, PSA nadir and rate of decline predicted local failure; those with post-treatment nadir of 1-4 ng/ml were five times more likely to experience local failure than nadir < 1 ng/ml (p = 0.0002). Rapid rate of rise was the most significant independent predictor of metastatic failure., Conclusions: The rate of PSA rise following definitive radiotherapy can predict clinical failure patterns, with a rapidly rising PSA indicating metastatic recurrence and moderately rising PSA local recurrence. This information could potentially direct therapy; if the rise predicts metastatic failure hormonal therapy could be considered, while aggressive salvage therapy may benefit subclinical local recurrence identified by a moderate rate of PSA rise.
- Published
- 1997
- Full Text
- View/download PDF
43. Role of epidermal growth factor receptor and STAT-3 activation in autonomous proliferation of SUM-102PT human breast cancer cells.
- Author
-
Sartor CI, Dziubinski ML, Yu CL, Jove R, and Ethier SP
- Subjects
- Adenocarcinoma physiopathology, Breast Neoplasms physiopathology, Cell Division drug effects, Female, Humans, Middle Aged, Phosphotyrosine metabolism, RNA, Neoplasm genetics, Receptors, Estrogen analysis, STAT3 Transcription Factor, Signal Transduction, Tumor Cells, Cultured, Adenocarcinoma pathology, Breast Neoplasms pathology, DNA-Binding Proteins physiology, Epidermal Growth Factor physiology, ErbB Receptors physiology, Receptor Protein-Tyrosine Kinases physiology, Trans-Activators physiology
- Abstract
This report describes the isolation and characterization of a new human breast cancer cell line, SUM-102PT, obtained from a minimally invasive human breast carcinoma. SUM-102PT cells have a near diploid karyotype, and early-passage cells had minor chromosomal abnormalities including a 5, 12 and a 6, 16 reciprocal translocation. The cells were isolated and have been continually cultured in three defined media, one of which contains exogenous epidermal growth factor (EGF). SUM-102PT cells have also been carried in an EGF-free medium supplemented with progesterone. All SUM-102PT cells require EGF receptor (EGFR) activation for continuous growth, because incubation of the cells with EGFR-neutralizing antibodies or with EGFR kinase inhibitors blocks growth of these cells. Southern analysis indicates that the EGFR gene is not amplified in these cells; however, these cells express high levels of EGFR mRNA. Thus, SUM-102PT is representative of a class of human breast cancers characterized by high level EGFR expression in the absence of gene amplification. SUM-102PT cells cultured in EGF-free, progesterone-containing medium express high levels of constitutively active EGFR. Conditioned medium from SUM-102PT cells contains an EGF-like mitogen that binds to a heparin-agarose affinity matrix with high affinity. Northern analysis for various EGF family members indicates that SUM-102PT cells synthesize heparin binding (HB)-EGF mRNA. HB-EGF protein is detectable on the surface of these cells by immunohistochemistry, and SUM-102PT cells are killed by diphtheria toxin, which acts by binding to HB-EGF. Furthermore, HB-EGF antibodies partially neutralize the mitogenic activity of the conditioned medium. Thus, EGFR activation in SUM-102PT cells is mediated, at least in part, by autocrine/juxtacrine stimulation by HB-EGF. SUM-102PT cells also express constitutively active STAT-3 homodimers. Constitutively tyrosine-phosphorylated STAT-3 homodimers were also detected in another breast cancer cell line, MDA468, which has an EGFR amplification and also has constitutive EGFR activity. Thus, SUM-102PT is a new human breast cancer cell line that expresses activated EGFR as a result of an autocrine/juxtacrine interaction with HB-EGF which, in turn, results in activation of STAT-3.
- Published
- 1997
44. Interleukin-6 in psoriasis: expression and mitogenicity studies.
- Author
-
Elder JT, Sartor CI, Boman DK, Benrazavi S, Fisher GJ, and Pittelkow MR
- Subjects
- Base Sequence, Cell Division drug effects, Cells, Cultured, Epidermal Growth Factor physiology, Humans, Interleukin-6 pharmacology, Keratinocytes drug effects, Molecular Sequence Data, Nucleic Acid Hybridization, Psoriasis pathology, Interleukin-6 genetics, Psoriasis metabolism, RNA, Messenger analysis
- Abstract
Interleukin-6 (IL-6) is a multifunctional cytokine which has been suggested to function as an autocrine mitogen in psoriatic epidermis. We report here the results of several experiments designed to further examine this hypothesis. Blot hybridization was unable to detect 1.3 kb IL-6 transcripts in RNA extracted from normal or psoriatic epidermal (keratome) biopsies, suggesting that IL-6 expression is very low in normal and psoriatic epidermis. Therefore, qualitative and semiquantitative PCR/Southern blot analyses were performed on keratome-derived RNA, and revealed variable but significantly increased IL-6 mRNA levels in lesional psoriatic relative to normal tissue. To further examine the ability of normal human keratinocytes (NHK) to express IL-6, RNA was extracted from rapidly proliferating secondary NHK cultures. IL-6 transcripts were nearly undetectable by blotting in keratinocytes grown in low-calcium serum-free medium, but low levels could be induced by treatment with 1.8 mM CaCl2. IL-6 transcripts were strongly superinduced after cycloheximide treatment, suggesting that a labile protein regulates IL-6 mRNA levels in these cells. Finally, the mitogenic activity of IL-6 was examined in NHK under varying conditions of cell density and added growth factors. IL-6 did not stimulate high density keratinocyte growth in the presence or absence of other growth factors, but did stimulate clonal growth in epidermal growth factor (EGF)-deficient media at high concentrations (> or = 10 ng/ml). The proliferative effects of IL-6, but not of basic fibroblast growth factor, were abrogated by monoclonal antibodies directed against the EGF receptor.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1992
- Full Text
- View/download PDF
45. Nuclear proteins that bind the human gamma-globin gene promoter: alterations in binding produced by point mutations associated with hereditary persistence of fetal hemoglobin.
- Author
-
Gumucio DL, Rood KL, Gray TA, Riordan MF, Sartor CI, and Collins FS
- Subjects
- Animals, Base Sequence, Binding Sites, Cell Line, Humans, Mice, Molecular Sequence Data, Oligonucleotide Probes, Protein Binding, Restriction Mapping, DNA-Binding Proteins metabolism, Fetal Hemoglobin genetics, Genes, Globins genetics, Mutation, Nuclear Proteins metabolism, Promoter Regions, Genetic
- Abstract
The molecular mechanisms responsible for the human fetal-to-adult hemoglobin switch have not yet been elucidated. Point mutations identified in the promoter regions of gamma-globin genes from individuals with nondeletion hereditary persistence of fetal hemoglobin (HPFH) may mark cis-acting sequences important for this switch, and the trans-acting factors which interact with these sequences may be integral parts in the puzzle of gamma-globin gene regulation. We have used gel retardation and footprinting strategies to define nuclear proteins which bind to the normal gamma-globin promoter and to determine the effect of HPFH mutations on the binding of a subset of these proteins. We have identified five proteins in human erythroleukemia cells (K562 and HEL) which bind to the proximal promoter region of the normal gamma-globin gene. One factor, gamma CAAT, binds the duplicated CCAAT box sequences; the -117 HPFH mutation increases the affinity of interaction between gamma CAAT and its cognate site. Two proteins, gamma CAC1 and gamma CAC2, bind the CACCC sequence. These proteins require divalent cations for binding. The -175 HPFH mutation interferes with the binding of a fourth protein, gamma OBP, which binds an octamer sequence (ATGCAAAT) in the normal gamma-globin promoter. The HPFH phenotype of the -175 mutation indicates that the octamer-binding protein may play a negative regulatory role in this setting. A fifth protein, EF gamma a, binds to sequences which overlap the octamer-binding site. The erythroid-specific distribution of EF gamma a and its close approximation to an apparent repressor-binding site suggest that it may be important in gamma-globin regulation.
- Published
- 1988
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.