Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Adv Radiat Oncol ; 1(1): 59-66, 2016.
Article in English | MEDLINE | ID: mdl-28799570

ABSTRACT

PURPOSE/OBJECTIVES: We sought to assess the utility of docetaxel administered concurrently with salvage radiation therapy (SRT) following postprostatectomy biochemical failure (BF). METHODS AND MATERIALS: Men with postprostatectomy BF were accrued on a single-arm phase 2 clinical trial. SRT doses ranged from 64.8 to 70.2 Gy and were delivered in 1.8-Gy fractions to the prostate bed alone as the clinical target volume with a +1-cm uniform planning target volume expansion. The primary endpoint was progression-free survival at 4 years compared with the Stephenson nomogram estimate. Kaplan-Meier methods were used to assess late toxicity, BF, and distant metastases. An unplanned matched-pair analysis was performed with 19 patients treated with SRT alone. RESULTS: Nineteen men were accrued and treated. Median follow-up was 4.8 years. Median pre-RT prostate-specific antigen level was 0.7 ng/mL (interquartile range, 0.4-1.3 ng/mL). All 8 cycles of docetaxel were completed in 17 (89%) patients. Acute grade 1-4 toxicities were observed in 79%, 50%, 58%, and 11%, respectively. A total of 68% of acute grade 1 toxicities were related to fatigue, urinary, or bowel symptoms. For grade 2 toxicities, 76% were related to neutropenia, fatigue, or urinary symptoms. Acute grade 3 and 4 toxicities were most commonly neutropenia (84% and 100%, respectively). All late toxicities were grade 1 to 2 with 89% related to bowel or urinary function. Predicted 4-year progression-free survival was 39% and observed was 42% (90% confidence interval [CI], 24-60). Matched-pair analysis demonstrated no significant improvement in BF (P = .96, hazard ratio, 0.98; 90% CI, 0.4-2.3) or distant metastases (P = .09; hazard ratio, 0.3; 90% CI, 0.07-1.2), and no difference between late bowel (P = .60) or urinary toxicity (P = .41). CONCLUSIONS: Docetaxel can safely be administered concurrently with SRT without significantly impacting posttreatment toxicity. Neutropenia was the most significant acute toxicity. Given the small sample size, no clear clinical benefit was observed. Larger studies are needed to determine the efficacy of concurrent docetaxel in this setting.

2.
Radiat Oncol ; 7: 82, 2012 Jun 09.
Article in English | MEDLINE | ID: mdl-22681643

ABSTRACT

BACKGROUND: Low-risk prostate cancer (PCa) patients have excellent outcomes, with treatment modality often selected by perceived effects on quality of life. Acute urinary symptoms are common during external beam radiotherapy (EBRT), while chronic symptoms have been linked to urethral dose. Since most low-risk PCa occurs in the peripheral zone (PZ), we hypothesized that EBRT using urethral sparing intensity modulated radiation therapy (US-IMRT) could improve urinary health-related quality of life (HRQOL) while maintaining high rates of PCa control. METHODS: Patients with National Comprehensive Cancer Network (NCCN) defined low-risk PCa with no visible lesion within 5 mm of the prostatic urethra on MRI were randomized to US-IMRT or standard (S-) IMRT. Prescription dose was 75.6 Gy in 41 fractions to the PZ + 3-5 mm for US-IMRT and to the prostate + 3 mm for S-IMRT. For US-IMRT, mean proximal and distal urethral doses were limited to 65 Gy and 74 Gy, respectively. HRQOL was assessed using the Expanded Prostate Cancer Index (EPIC) Quality of Life questionnaire. The primary endpoint was change in urinary HRQOL at 3 months. RESULTS: From June 2004 to November 2006, 16 patients were randomized, after which a futility analysis concluded that continued accrual was unlikely to demonstrate a difference in the primary endpoint. Mean change in EPIC urinary HRQOL at 3 months was -0.5 ± 11.2 in the US-IMRT arm and +3.9 ± 15.3 in the S-IMRT arm (p = 0.52). Median PSA nadir was higher in the US-IMRT arm (1.46 vs. 0.78, p = 0.05). At 4.7 years median follow-up, three US-IMRT and no S-IMRT patients experienced PSA failure (p = 0.06; HR 8.8, 95% CI 0.9-86). Two out of 3 patients with PSA failure had biopsy-proven local failure, both located contralateral to the original site of disease. CONCLUSIONS: Compared with S-IMRT, US-IMRT failed to improve urinary HRQOL and resulted in higher PSA nadir and inferior biochemical control. The high rate of PSA failure and contralateral local failures in US-IMRT patients, despite careful selection of MRI-screened low-risk patients, serve as a cautionary tale for focal PCa treatments.


Subject(s)
Adenocarcinoma/radiotherapy , Prostatic Neoplasms/radiotherapy , Radiotherapy, Intensity-Modulated/methods , Urethra/radiation effects , Humans , Male , Quality of Life
3.
J Natl Cancer Inst ; 101(4): 228-36, 2009 Feb 18.
Article in English | MEDLINE | ID: mdl-19211454

ABSTRACT

BACKGROUND: The identification of surrogate endpoints for prostate cancer-specific survival may shorten the length of clinical trials for prostate cancer. We evaluated distant metastasis and general clinical treatment failure as potential surrogates for prostate cancer-specific survival by use of data from the Radiation Therapy and Oncology Group 92-02 randomized trial. METHODS: Patients (n = 1554 randomly assigned and 1521 evaluable for this analysis) with locally advanced prostate cancer had been treated with 4 months of neoadjuvant and concurrent androgen deprivation therapy with external beam radiation therapy and then randomly assigned to no additional therapy (control arm) or 24 additional months of androgen deprivation therapy (experimental arm). Data from landmark analyses at 3 and 5 years for general clinical treatment failure (defined as documented local disease progression, regional or distant metastasis, initiation of androgen deprivation therapy, or a prostate-specific antigen level of 25 ng/mL or higher after radiation therapy) and/or distant metastasis were tested as surrogate endpoints for prostate cancer-specific survival at 10 years by use of Prentice's four criteria. All statistical tests were two-sided. RESULTS: At 3 years, 1364 patients were alive and contributed data for analysis. Both distant metastasis and general clinical treatment failure at 3 years were consistent with all four of Prentice's criteria for being surrogate endpoints for prostate cancer-specific survival at 10 years. At 5 years, 1178 patients were alive and contributed data for analysis. Although prostate cancer-specific survival was not statistically significantly different between treatment arms at 5 years (P = .08), both endpoints were consistent with Prentice's remaining criteria. CONCLUSIONS: Distant metastasis and general clinical treatment failure at 3 years may be candidate surrogate endpoints for prostate cancer-specific survival at 10 years. These endpoints, however, must be validated in other datasets.


Subject(s)
Biomarkers, Tumor/blood , Endpoint Determination , Neoplasms, Hormone-Dependent/mortality , Neoplasms, Hormone-Dependent/pathology , Prostate-Specific Antigen/blood , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Aged , Androgen Antagonists/administration & dosage , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemotherapy, Adjuvant , Disease-Free Survival , Dose Fractionation, Radiation , Endpoint Determination/methods , Flutamide/administration & dosage , Follow-Up Studies , Goserelin/administration & dosage , Humans , Karnofsky Performance Status , Male , Middle Aged , Neoadjuvant Therapy/methods , Neoplasms, Hormone-Dependent/blood , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/radiotherapy , Neoplasms, Hormone-Dependent/surgery , Prostatic Neoplasms/blood , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/surgery , Radiotherapy, Adjuvant , Research Design , Treatment Failure , Treatment Outcome
4.
Clin Cancer Res ; 15(2): 589-96, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19147764

ABSTRACT

PURPOSE: The PI3K/Akt/mTOR prosurvival pathway is frequently up-regulated in soft tissue sarcoma. Mammalian target of rapamycin (mTOR) inhibitors, such as rapamycin, have recently shown clinical benefit in soft tissue sarcoma, and mTOR inhibition has also been associated with radiosensitization of carcinoma and endothelial cells. This study tested the hypothesis that rapamycin radiosensitizes soft tissue sarcoma and endothelial cells in vitro and in vivo through the inhibition of mTOR. EXPERIMENTAL DESIGN: Colony formation assays were done to determine the radiosensitizing properties of rapamycin on three human soft tissue sarcoma cell lines (SK-LMS-1, SW-872, and HT-1080) and human dermal microvascular endothelial cells (HDMEC). The functional effects of rapamycin and radiation on the endothelial compartment were evaluated with microvascular sprouting assays. The in vivo radiosensitizing activity of rapamycin was assessed with s.c. SK-LMS-1 nude mice xenografts treated with concurrent daily rapamycin, radiation, or both for three weeks. RESULTS: In vitro radiosensitization was shown in all three soft tissue sarcoma cell lines with minimally cytotoxic doses of rapamycin. SK-LMS-1 xenografts displayed significant tumor growth delay with rapamycin and radiation compared with either treatment alone. Radiation resulted in transient increased mTOR function, whereas rapamycin abolished this signaling in irradiated and unirradiated samples. In HDMEC, rapamycin and radiation reduced microvessel sprouting, but did not alter colony formation. CONCLUSIONS: Minimally cytotoxic concentrations of rapamycin inhibited the mTOR cascade in culture and in vivo while radiosensitizing soft tissue sarcoma, and produced synergistic effects with radiation on HDMEC microvessel formation. By targeting both tumor and endothelial compartments, rapamycin produced potent radiosensitization of soft tissue sarcoma xenografts. Clinical trials combining rapamycin and radiotherapy in soft tissue sarcoma are warranted.


Subject(s)
Protein Kinases/metabolism , Sarcoma/blood supply , Sarcoma/metabolism , Animals , Antineoplastic Agents/pharmacology , Carcinoma , Cell Line, Tumor , Dose-Response Relationship, Radiation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium/pathology , Humans , Mice , Mice, Nude , Microcirculation , Models, Biological , Neoplasm Transplantation , Radiation-Sensitizing Agents/pharmacology , Sarcoma/drug therapy , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
5.
Urology ; 73(1): 158-62, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18722651

ABSTRACT

OBJECTIVES: To examine the effect of concurrent statin use during definitive radiotherapy (RT) on the biochemical outcomes for localized prostate cancer. METHODS: A total of 968 patients treated with RT had information about medication use available. Of these, 23% had been taking using statins during RT. Progression-free survival (PFS) was determined by a biochemical failure definition of prostate-specific antigen nadir plus 2 ng/mL, clinical failure, start of androgen deprivation therapy, or death. RESULTS: The mean patient age was 68 years. The median radiation dose was 76 Gy. Of the patients, 29% underwent androgen deprivation therapy. The 5-year overall survival rate was 83%. The median PFS time was 7.8 years versus 6.4 years, and the 5-year PFS rate was 70% versus 59% in favor of the statin users (P = 0.03). The analysis by risk group demonstrated no significant statin effect in any of the three risk strata. Stratification by hydrophilic versus hydrophobic statin agents revealed similar results. Multivariate analysis revealed that T stage (P <0.0001), pretreatment prostate-specific antigen level (P <0.0001), and Gleason score (P = 0.0026) were significant predictors of PFS; however, statin use (P = 0.48), androgen deprivation therapy (P = 0.95), pelvic RT (P = 0.96), radiation dose (P = 0.13), age (P = 0.19), and year of treatment (P = 0.07) were not. CONCLUSIONS: Statin use did not affect PFS after adjusting for differences in treatment year and multiple prognostic factors. However, T stage, baseline prostate-specific antigen level, and Gleason score were critical determinants of prostate-specific antigen failure. These results did not differ when hydrophilic pravastatin was excluded.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/drug therapy , Aged , Combined Modality Therapy , Humans , Male , Prostatic Neoplasms/radiotherapy , Retrospective Studies , Treatment Outcome
6.
Int J Radiat Oncol Biol Phys ; 72(1): 153-60, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18722266

ABSTRACT

PURPOSE: External beam radiotherapy (RT) is often used in an attempt to cure localized prostate cancer (PCa), but it is only palliative against disseminated disease. Raf kinase inhibitory protein (RKIP) is a metastasis suppressor whose expression is reduced in approximately 50% of localized PCa tissues and is absent in metastases. Chemotherapeutic agents have been shown to induce tumor apoptosis through induction of RKIP expression. Our goal was to test whether RT similarly induces apoptosis through induction of RKIP expression. METHODS AND MATERIALS: The C4-2B PCa cell line was engineered to overexpress or underexpress RKIP. The engineered cells were tested for apoptosis in cell culture and tumor regression in mice after RT. RESULTS: RT induced both RKIP expression and apoptosis of PCa cells. Overexpression of RKIP sensitized PCa cells to radiation-induced apoptosis. In contrast, short-hairpin targeting of RKIP, so that RT could not induce RKIP expression, protected cells from radiation-induced apoptosis. In a murine model, knockdown of RKIP in PCa cells diminished radiation-induced apoptosis. Molecular concept mapping of genes altered on manipulation of RKIP expression revealed an inverse correlation with the concept of genes altered by RT. CONCLUSION: The data presented in this report indicate that the loss of RKIP, as seen in primary PCa tumors and metastases, confers protection against radiation-induced apoptosis. Therefore, it is conceivable that the loss of RKIP confers a growth advantage on PCa cells at distant sites, because the loss of RKIP would decrease apoptosis, favoring proliferation.


Subject(s)
Apoptosis/radiation effects , Neoplasm Proteins/deficiency , Phosphatidylethanolamine Binding Protein/deficiency , Prostatic Neoplasms/radiotherapy , Radiation Tolerance/physiology , Animals , Apoptosis/physiology , Cell Line, Tumor , Enzyme Induction/radiation effects , Male , Mice , Neoplasm Metastasis , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Phosphatidylethanolamine Binding Protein/biosynthesis , Phosphatidylethanolamine Binding Protein/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Prostatic Neoplasms/enzymology , Severe Combined Immunodeficiency
7.
Transl Oncol ; 1(1): 50-6, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18607508

ABSTRACT

BACKGROUND/PURPOSE: Local and systemic control of soft tissue sarcoma (STS) remains a clinical challenge, particularly for retroperitoneal, deep truncal, or advanced extremity disease. 2',2'-Difluoro-2'-deoxycytidine (gemcitabine) is a potent radiosensitizer in many tumor types, but it has not been studied in human STS. The purpose of this study was to determine the radiosensitizing potential of gemcitabine in preclinical models of human STS. MATERIALS AND METHODS: The in vitro radiosensitizing activity of gemcitabine was assessed with clonogenic survival assay on three human STS cell lines: SK-LMS-1 (leiomyosarcoma), SW-872 (liposarcoma), and HT-1080 (fibrosarcoma). Cell cycle distribution was determined using dual-channel flow cytometry. The in vivo radiosensitizing activity of gemcitabine was assessed with subcutaneous SK-LMS-1 nude mice xenografts. Tumor-bearing mice were treated with concurrent weekly gemcitabine and fractionated daily radiotherapy (RT) (2 Gy daily) for 3 weeks (a total dose of 30 Gy). RESULTS: The 50% inhibitory concentration (IC(50)) of gemcitabine for the human STS cell lines ranged from 10 to 1000 nM. Significant in vitro radiosensitization was demonstrated in all three human STS cell lines using gemcitabine concentrations at and below the IC(50). Maximal radiosensitization was associated with accumulation of cells in early S-phase. SK-LMS-1 xenografts displayed significant tumor growth delay with combined gemcitabine and RT compared to either treatment alone. Treatment related toxicity was greatest in the gemcitabine plus RT arm, but remained at an acceptable level. CONCLUSIONS: Gemcitabine is a potent radiosensitizer in preclinical models of human STS. Clinical trials combining gemcitabine and RT in human STS are warranted.

8.
Urology ; 71(2): 313-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18308110

ABSTRACT

OBJECTIVES: The Radiation Therapy Oncology Group 9413 trial has shown an improvement in progression-free survival (PFS) with external beam radiotherapy that included the pelvic nodes. In clinical practice, two pelvic field size designs are in use. We examined the effect of differences in the level of pelvic nodal coverage using three-dimensional conformal radiotherapy on biochemical failure-free survival (BFFS) and PFS in early-stage prostate cancer. METHODS: The patients were identified retrospectively who had undergone whole pelvis (WP) or minipelvis (MP) three-dimensional conformal radiotherapy. Biochemical failure was defined as the nadir prostate-specific antigen (PSA) level plus 2 ng/mL. RESULTS: Of the 669 patients identified, 384 had undergone MP (57%) and 285 WP (42%) treatment, with a median PSA follow-up of 56 months. Of the 669 patients, 11%, 35%, and 54% were at low, intermediate, and high risk, respectively. The median dose was 75 Gy for the MP and 71 Gy for the WP groups. Of the MP and WP groups, 52% and 36% underwent hormonal therapy. The median BFFS and 5-year BFFS rate was 128 months and 73% for the MP group and 96 months and 58% for the WP group. The median PFS and 5-year PFS rate was 128 months and 72% for the MP group and 83 months and 56% for the WP group. Multivariate analysis revealed no difference between MP and WP treatment. However T stage, pretreatment PSA level, and Gleason score were significant predictors of BFFS and PFS. CONCLUSIONS: We observed no difference in outcomes between patients undergoing WP versus MP using three-dimensional conformal radiotherapy. Therefore, the exclusion of the common iliac lymph nodes in the treatment of patients with high-risk prostate cancer might be acceptable.


Subject(s)
Prostatic Neoplasms/radiotherapy , Radiotherapy, Conformal/methods , Aged , Humans , Lymph Nodes , Male , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Retrospective Studies , Treatment Outcome
9.
Int J Radiat Oncol Biol Phys ; 69(1): 103-10, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17560050

ABSTRACT

PURPOSE: To determine the long-term outcomes after multimodality treatment of retroperitoneal, pelvic, and deep truncal sarcomas and to identify the factors associated with local control (LC), distant metastasis (DM), and overall survival (OS). METHODS AND MATERIALS: A total of 85 patients with retroperitoneal, pelvic, and deep truncal sarcomas were treated with radiotherapy (RT) between 1987 and 2005. A retrospective analysis of LC, DM, and OS was conducted using log-rank and Cox regression statistical methods. RESULTS: The 2- and 5-year LC, DM, and OS rates were 66% and 51%, 38% and 58%, and 70% and 34%, respectively. Negative surgical margins and a higher radiation dose were associated with greater LC rates on both univariate and multivariate analyses, and female gender was significantly associated with greater LC on multivariate analysis only. None of the analyzed risk factors was significantly associated with DM, although patients with high-grade tumors showed a trend toward an increased risk of DM. Gross residual disease after resection and high tumor grade were associated with worse OS rates on univariate and multivariate analyses, and male gender was significantly associated with worse OS on multivariate analysis only. A time-dependent analysis of LC in relation to DM demonstrated that patients with local failure had a hazard ratio of 19.7 for DM compared with patients without local failure (p < 0.0001). Of the 85 patients, 5 and 8, respectively, had clinically significant acute and late toxicity. CONCLUSION: The results of this study emphasize the importance of LC in patients with retroperitoneal sarcoma. Radiation dose escalation or radiosensitization strategies to enhance LC are warranted.


Subject(s)
Neoplasm Recurrence, Local/radiotherapy , Pelvic Neoplasms/radiotherapy , Retroperitoneal Neoplasms/radiotherapy , Sarcoma/radiotherapy , Adult , Aged , Analysis of Variance , Combined Modality Therapy , Female , Humans , Male , Middle Aged , Pelvic Neoplasms/pathology , Pelvic Neoplasms/surgery , Radiotherapy/adverse effects , Retroperitoneal Neoplasms/pathology , Retroperitoneal Neoplasms/surgery , Retrospective Studies , Risk Factors , Sarcoma/pathology , Sarcoma/secondary , Sarcoma/surgery , Sex Factors , Survival Analysis
10.
Urology ; 69(5): 936-40, 2007 May.
Article in English | MEDLINE | ID: mdl-17482938

ABSTRACT

OBJECTIVES: To examine the prognostic value of percent positive cores (PPC) in prostate cancer patients treated with external beam radiotherapy (RT). METHODS: An institutional review board-approved, retrospective analysis was conducted on 814 patients treated with RT with or without hormonal therapy between 1984 and 2002. Percent positive cores (number of positive cores divided by total number of cores) was calculable for 591 patients with a median follow-up of 65 months. Univariate and multivariable analyses were performed using Kaplan-Meier and Cox proportional hazard methods relating PPC to other risk factors, biochemical/clinical disease-free survival (PSA-DFS), prostate cancer-specific survival (DSS), and overall survival (OS). RESULTS: Percent positive cores was associated with stage, Gleason score (GS), pretreatment serum prostate-specific antigen (PSA) level, and use of adjunctive androgen suppression therapy. The 5-year PSA-DFS, DSS, and OS rates were 80%, 99%, and 91%, respectively, for patients with PPC less than 50%, compared with 56%, 94%, and 87% for patients with PPC 50% or greater (P <0.0001, <0.004, and <0.04, respectively). Multivariable analysis revealed that PPC, stage, GS, PSA, and androgen suppression therapy were all significantly associated with PSA-DFS, whereas only GS was associated with DSS and OS. For high, intermediate, and low-risk patients, 5-year PSA-DFS was 62% versus 39%, 80% versus 59%, and 90% versus 82% for PPC less than 50% versus PPC 50% or greater, respectively. CONCLUSIONS: Percent positive cores predicts outcome of prostate cancer patients treated with RT, independently of other known prognostic factors. Percent positive cores may have particular use for further risk stratification within established clinical risk categories.


Subject(s)
Biopsy, Needle/methods , Brachytherapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Adult , Aged , Analysis of Variance , Dose Fractionation, Radiation , Humans , Male , Middle Aged , Probability , Prognosis , Prostate-Specific Antigen/blood , Prostatic Neoplasms/mortality , Radiotherapy Dosage , Retrospective Studies , Risk Assessment , Survival Analysis , Treatment Outcome
11.
Int J Radiat Oncol Biol Phys ; 68(3): 801-8, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17379445

ABSTRACT

PURPOSE: Full-dose gemcitabine and concurrent radiotherapy is a promising treatment approach in unresectable pancreatic cancer. This study was conducted to assess the pattern of failure and toxicity associated with the use of conformal treatment volumes, omitting prophylactic lymph node irradiation. METHODS AND MATERIALS: Seventy-four patients with locally advanced pancreatic cancer were treated between 1997 and 2005 with full-dose (1000 mg/m(2), Days 1, 8, and 15) gemcitabine and concurrent radiotherapy (36 Gy [median] in 15 daily fractions). The planning target volume (PTV) was limited to the gross tumor volume (GTV) plus 1-cm margin. Patient computed tomography (CT) scans were systematically reviewed to determine the pattern of failure. Kaplan-Meier and Cox-regression models were used to analyze freedom from local progression (FFLP), distant failure, overall survival (OS), and toxicity. RESULTS: With a median follow-up of 10.6 months (20.6 months in living patients), the 1-year and 2-year FFLP rates were 64% and 38%, respectively. Four patients (5%) failed in the peripancreatic lymph nodes (3 in-field and 1 marginal failure). Median OS was 11.2 months. Analyzed as a time-dependent covariate, local failure was a significant predictor of OS (p = 0.0074). Sixteen patients (22%) had significant gastrointestinal (GI) toxicity (> or = Grade 3). PTV correlated with significant GI toxicity (p = 0.007). CONCLUSIONS: Freedom from local progression in unresectable pancreatic cancer is suboptimal. In conjunction with full-dose gemcitabine, the use of conformal fields encompassing only the GTV helps reduce toxicity and does not result in marginal failures. Our findings provide rationale for intensification of local therapy in conjunction with more effective systemic therapy.


Subject(s)
Deoxycytidine/analogs & derivatives , Gastrointestinal Diseases/epidemiology , Neoplasm Recurrence, Local/mortality , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/therapy , Radiation Injuries/epidemiology , Radiotherapy, Adjuvant/mortality , Adult , Aged , Antimetabolites, Antineoplastic/administration & dosage , Comorbidity , Deoxycytidine/administration & dosage , Female , Humans , Incidence , Male , Michigan/epidemiology , Middle Aged , Neoplasm Recurrence, Local/prevention & control , Pancreatic Neoplasms/surgery , Prognosis , Radiation-Sensitizing Agents/administration & dosage , Risk Assessment/methods , Risk Factors , Survival Analysis , Survival Rate , Treatment Outcome , Gemcitabine
12.
Cancer Res ; 66(24): 11632-43, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17178857

ABSTRACT

The 8p11-p12 genomic region is amplified in 15% of breast cancers and harbors several candidate oncogenes. However, functional evidence for a transforming role for these genes is lacking. We identified 21 genes from this region as potential oncogenes based on statistical association between copy number and expression. We further showed that three of these genes (LSM1, BAG4, and C8orf4) induce transformed phenotypes when overexpressed in MCF-10A cells, and overexpression of these genes in combination influences the growth factor independence phenotype and the ability of the cells to grow under anchorage-independent conditions. Thus, LSM1, BAG4, and C8orf4 are breast cancer oncogenes that can work in combination to influence the transformed phenotype in human mammary epithelial cells.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/genetics , Chromosomes, Human, Pair 8 , Neoplasm Proteins/genetics , Oncogenes , Proto-Oncogene Proteins/genetics , RNA-Binding Proteins/genetics , Cell Division , Cell Line, Tumor , Cell Transformation, Neoplastic , Chromosome Mapping , Culture Media , Female , Humans , Lentivirus/genetics , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
13.
Urology ; 68(6): 1257-62, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17141830

ABSTRACT

OBJECTIVES: To determine whether nadir prostate-specific antigen (PSA) levels within 12 months (nadir PSA12) after completion of radiotherapy (RT) can be used as an early marker of recurrence risk. METHODS: A total of 4839 patients were treated with RT and without hormonal therapy from 1986 to 1995 for Stage T1-T2 prostate cancer at nine institutions. Of these 4839 patients, 4833, with a median follow-up of 6.3 years, met the criteria for analysis. The study endpoints included freedom from PSA failure, initiation of androgen deprivation, or documented local or distant failure (PSA-DFS); freedom from clinically apparent distant metastasis (DMFS); and overall survival (OS). RESULTS: Patients with a nadir PSA12 of 2.0 ng/mL or less had an 8-year PSA-DFS, DMFS, and OS rate of 55%, 95%, and 73%, respectively, compared with 40%, 88%, and 69%, respectively, for patients with a nadir PSA12 of more than 2.0 ng/mL. Multivariate analysis confirmed that a nadir PSA12 of greater than 2 ng/mL was an independent predictor of PSA-DFS, DMFS, and OS. Classification and regression tree analysis identified the nadir PSA12 levels after RT associated with PSA-DFS, DMFS, and OS. Nadir PSA12, combined with the pretreatment PSA level, identified patients at particularly high risk of distant metastasis. CONCLUSIONS: The results of this large, multi-institutional study have demonstrated that nadir PSA12 is predictive of clinical outcomes for patients with localized prostate cancer after RT. A high pretreatment PSA level and high nadir PSA12 will identify patients at particularly high risk who might benefit from early adjuvant therapy.


Subject(s)
Adenocarcinoma/blood , Adenocarcinoma/radiotherapy , Biomarkers, Tumor/blood , Neoplasm Recurrence, Local/blood , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/radiotherapy , Adenocarcinoma/pathology , Biopsy , Follow-Up Studies , Humans , Male , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , Prostatic Neoplasms/pathology , Retrospective Studies , Risk Factors , Time Factors , Treatment Failure
14.
Cancer Metastasis Rev ; 25(4): 669-75, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17160556

ABSTRACT

Cancer metastasis to the bone develops commonly in patients with a variety of malignancies, and is a major cause of morbidity and diminished quality of life in a significant proportion of cancer patients. The effective treatment of bone metastasis requires cooperation between medical, surgical and radiation oncologists. Radiotherapy, either in the form of targeted external beam radiation therapy, or systemic administration of radionuclides, plays a central role in treatment of symptomatic bone metastases. The appropriate external beam treatment techniques, dose and fractionation regimens for the treatment of symptomatic, localized bone metastasis have been established in prospective clinical trials. Large-field, hemi-body irradiation has been utilized for treatment of symptoms related to more widely disseminated bone metastases, but has been associated with substantial toxicity. Strontium-89 and Samarium-153 are widely available systemically administered radionuclides that are useful for the treatment of widely disseminated disease, and have largely supplanted the use of hemi-body irradiation. Combined with appropriate medical and surgical interventions, as well as the appropriate use of analgesics, radiotherapy is a well-tolerated and highly effective treatment for the palliation of symptomatic bone metastases.


Subject(s)
Bone Neoplasms/radiotherapy , Bone Neoplasms/secondary , Radiation Oncology , Bone Neoplasms/diagnostic imaging , Humans , Radiography
15.
J Urol ; 176(4 Pt 1): 1409-14; discussion 1414, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16952645

ABSTRACT

PURPOSE: Radiotherapy for biochemical prostate cancer recurrence after prostatectomy achieves durable salvage rates of only 40% to 50%. Improved methods of identifying patients unlikely to benefit from salvage radiotherapy are needed. Altered expression of the adhesion molecule E-cadherin may be associated with the invasive and metastatic phenotype. We examined the relationship between E-cadherin expression and outcomes after salvage radiotherapy. MATERIALS AND METHODS: E-cadherin expression was examined by immunohistochemistical analysis of a tissue microarray of prostatectomy tissues from patients who underwent salvage radiotherapy. The relation between E-cadherin staining, other risk factors and biochemical failure after salvage radiotherapy was analyzed using Kaplan-Meier and Cox regression methods. RESULTS: Of 37 analyzable cases 25 showed aberrant E-cadherin expression, while the remainder had normal expression. At a median clinical followup of 40 months univariate analysis demonstrated that E-cadherin staining was not associated with Gleason score, extracapsular extension, surgical margin status, pre-prostatectomy or pre-radiotherapy prostate specific antigen, complete biochemical response after radiotherapy or adjunctive hormonal therapy but it was associated with seminal vesicle invasion. Two-year failure-free survival was 55% in patients with aberrant E-cadherin expression compared with 92% in patients with normal E-cadherin expression (p = 0.02). Multivariate analysis confirmed that aberrant E-cadherin expression was associated with salvage radiotherapy failure (p = 0.03). CONCLUSIONS: Aberrant E-cadherin staining is associated with increased biochemical failure rates after salvage radiotherapy. Patients with biochemical failure after prostatectomy and aberrant E-cadherin expression are likely to have subclinical disseminated disease. Early systemic therapy may be warranted in these patients.


Subject(s)
Cadherins/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/radiotherapy , Salvage Therapy , Follow-Up Studies , Humans , Male , Middle Aged , Predictive Value of Tests , Prostate-Specific Antigen/blood , Prostatectomy , Prostatic Neoplasms/surgery , Treatment Outcome
17.
Cancer ; 107(3): 554-62, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16795069

ABSTRACT

BACKGROUND: Craniofacial osteosarcoma differs from long bone osteosarcoma in that patients are older, tumors are often low grade, and prognosis is more favorable. Although most are sporadic, some tumors occur in association with prior radiation therapy. The purpose of the current study was to compare clinicopathologic and prognostic features of primary and radiation-associated osteosarcoma. METHODS: The study group consisted of 15 primary and 6 radiation-associated osteosarcomas. Clinical and follow-up data were obtained in every case. Tissue microarrays were immunohistochemically stained for p53, pRB, Ki-67 (MIB-1), and ezrin. DNA was sequenced for TP53 mutations. RESULTS: All radiation-associated osteosarcomas were high grade and half were fibroblastic. In contrast, 47% of primary craniofacial osteosarcomas were high grade and only 1 was fibroblastic. All radiation-associated osteosarcomas recurred, half the patients died of disease, 2 were alive with unresectable tumors, whereas only 1 was alive without disease. In contrast, 80% of patients with primary tumors were alive without disease, 33% had local recurrences, and 13% died of disease. Radiation-associated tumors overexpressed p53 more often (33% vs. 13%), more often had TP53 mutations (33% vs. 8%), had higher proliferative activity (67% vs. 0% showing >50% MIB-1 staining), and expressed ezrin more frequently (83% vs. 40%) than primary tumors. Compared with a control group of 24 high- and 7 low-grade primary extremity osteosarcomas, radiation-associated tumors marked as the high-grade tumors. CONCLUSIONS: Craniofacial radiation-associated osteosarcomas are high-grade tumors that behave more aggressively than most primary craniofacial osteosarcomas. In addition, they demonstrate higher expression rates of adverse prognostic indicators, further highlighting the distinction.


Subject(s)
Biomarkers, Tumor/blood , Neoplasms, Radiation-Induced/pathology , Osteosarcoma/pathology , Skull Neoplasms/pathology , Tumor Suppressor Protein p53/blood , Adolescent , Adult , Aged , Biomarkers, Tumor/genetics , Child , Female , Follow-Up Studies , Humans , Immunohistochemistry , Male , Middle Aged , Mutation , Neoplasm Recurrence, Local , Neoplasms, Radiation-Induced/genetics , Osteosarcoma/genetics , Prognosis , Skull Neoplasms/genetics , Survival Rate , Tumor Suppressor Protein p53/genetics
18.
Otol Neurotol ; 27(3): 337-41, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16639271

ABSTRACT

OBJECTIVE: To highlight a case of cochlear implantation in the setting of ipsilateral petrous apex chondrosarcoma. BACKGROUND: A patient with bilateral progressive hearing loss was incidentally found to have a destructive right petrous apex lesion on computed tomography before cochlear implantation. The patient had no associated symptoms and a magnetic resonance imaging scan was obtained, narrowing the differential diagnosis. A middle cranial fossa approach was performed for synchronous biopsy of the lesion and cochlear implantation. RESULTS: Frozen sections revealed a low-grade chondroid lesion, and a Med-El Combi 40+ cochlear implant with a split electrode array was inserted via the middle fossa. Final pathologic examination revealed a Grade I chondrosarcoma. The patient suffered no complications postoperatively and was followed-up over 5 years with serial computed tomographic scans and clinical examinations. No additional treatment was administered. Eighteen months postoperatively, the patient experienced episodic vertigo. There were no new findings on computed tomography, and the vertigo improved with a low-salt diet. Otherwise, the patient had excellent hearing results, and the lesion has not progressed under observation. CONCLUSION: The implications of observing low-grade chondrosarcomas in well-selected patients and the unique aspect of cochlear implantation on the affected side are discussed.


Subject(s)
Chondrosarcoma/diagnosis , Cochlear Implantation , Hearing Loss, Bilateral/surgery , Petrous Bone , Skull Neoplasms/diagnosis , Acoustic Stimulation , Aged , Biopsy , Chondrosarcoma/surgery , Female , Humans , Magnetic Resonance Imaging , Petrous Bone/pathology , Petrous Bone/surgery , Skull Neoplasms/surgery , Tomography, X-Ray Computed , Treatment Outcome
19.
Urology ; 67(2): 423.e5-423.e8, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16461109

ABSTRACT

Sarcomatoid carcinomas of the prostate are rare malignancies composed of carcinomatous and sarcomatous elements. Their etiology is uncertain and may represent a single malignant process or a mixture of two distinct malignancies. We report a clinical case of a patient who presented with locally advanced disease and was treated with hormonal and cytotoxic chemotherapy, but ultimately developed distant metastasis and died of the disease. A loss-of-heterozygosity analysis of the primary and metastatic tissues provided compelling evidence that the carcinomatous and sarcomatous elements are clonally related, supporting the hypothesis that a single malignant process underlies the etiology of sarcomatoid carcinoma of the prostate.


Subject(s)
Carcinoma/pathology , Prostatic Neoplasms/pathology , Carcinoma/genetics , Humans , Male , Middle Aged , Neoplasm Metastasis , Prostatic Neoplasms/genetics , Sarcoma/genetics , Sarcoma/pathology
20.
Int J Radiat Oncol Biol Phys ; 64(4): 1140-50, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16198506

ABSTRACT

PURPOSE: To determine the significance of prostate-specific antigen (PSA) nadir (nPSA) and the time to nPSA (T(nPSA)) in predicting biochemical or clinical disease-free survival (PSA-DFS) and distant metastasis-free survival (DMFS) in patients treated with definitive external beam radiotherapy (RT) for clinical Stage T1b-T2 prostate cancer. METHODS AND MATERIALS: Nine participating institutions submitted data on 4839 patients treated between 1986 and 1995 for Stage T1b-T2cN0-NxM0 prostate cancer. All patients were treated definitively with RT alone to doses > or =60 Gy, without neoadjuvant or planned adjuvant androgen suppression. A total of 4833 patients with a median follow-up of 6.3 years met the criteria for analysis. Two endpoints were considered: (1) PSA-DFS, defined as freedom from PSA failure (American Society for Therapeutic Radiology and Oncology definition), initiation of androgen suppression after completion of RT, or documented local or distant failure; and (2) DMFS, defined as freedom from clinically apparent distant failure. In patients with failure, nPSA was defined as the lowest PSA measurement before any failure. In patients without failure, nPSA was the lowest PSA measurement during the entire follow-up period. T(nPSA) was calculated from the completion of RT to the nPSA date. RESULTS: A greater nPSA level and shorter T(nPSA) were associated with decreased PSA-DFS and DMFS in all patients and in all risk categories (low [Stage T1b, T1c, or T2a, Gleason score < or =6, and PSA level < or =10 ng/mL], intermediate [Stage T1b, T1c, or T2a, Gleason score < or =6, and PSA level >10 but < or =20 ng/mL, or Stage T2b or T2c, Gleason score < or =6, and PSA level < or =20 ng/mL, or Gleason score 7 and PSA level < or =20 ng/mL], and high [Gleason score 8-10 or PSA level >20 ng/mL]), regardless of RT dose. The 8-year PSA-DFS and DMFS rate for patients with nPSA <0.5 ng/mL was 75% and 97%; nPSA > or =0.5 but <1.0 ng/mL, 52% and 96%; nPSA > or =1.0 but <2.0 ng/mL, 40% and 91%; and nPSA > or =2.0 ng/mL, 17% and 73%, respectively. The 8-year PSA-DFS and DMFS rate for patients with T(nPSA) <6 months was 27% and 66%; T(nPSA) > or =6 but <12 months, 31% and 85%; T(nPSA) > or =12 but <24 months, 42% and 94%; and T(nPSA) > or =24 months, 75% and 99%, respectively. A shorter T(nPSA) was associated with decreased PSA-DFS and DMFS, regardless of the nPSA. Both nPSA and T(nPSA) were significant predictors of PSA-DFS and DMFS in multivariate models incorporating clinical stage, Gleason score, initial PSA level, and RT dose. The significance of nPSA and T(nPSA) was supported by landmark analysis, as well as by analysis of nPSA and T(nPSA) as time-dependent covariates. A dose > or =70 Gy was associated with a lower nPSA level and longer T(nPSA) in all risk categories, and a greater dose was significantly associated with greater PSA-DFS and DMFS in multivariate analysis. Regression analysis confirmed that higher clinical stage, Gleason score, and initial PSA were associated with a greater nPSA level. CONCLUSION: The results of this large, multi-institutional analysis of 4833 patients have provided important evidence that nPSA and T(nPSA) after definitive external beam RT are not only predictive of a predominantly PSA endpoint (PSA-DFS), but are also predictive of distant metastasis in all clinical risk categories. Greater RT doses were associated with lower nPSA, longer T(nPSA), and improved PSA-DFS and DMFS.


Subject(s)
Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/radiotherapy , Disease-Free Survival , Humans , Male , Multivariate Analysis , Neoplasm Staging , Prostatic Neoplasms/pathology , Reference Values , Regression Analysis , Treatment Failure
SELECTION OF CITATIONS
SEARCH DETAIL
...