Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cancer Res Commun ; 4(2): 530-539, 2024 02 26.
Article in English | MEDLINE | ID: mdl-38345536

ABSTRACT

PURPOSE: Single-agent checkpoint inhibition is effective in a minority of patients with platinum-refractory urothelial carcinoma; therefore, the efficacy of combining low-dose paclitaxel with pembrolizumab was tested. MATERIALS AND METHODS: This was a prospective, single-arm phase II trial with key inclusion criteria of imaging progression within 12 months of platinum therapy and Eastern Cooperative Oncology Group ≤1. Treatment was pembrolizumab 200 mg day 1 and paclitaxel 80 mg/m2 days 1 and 8 of a 21-day cycle for up to eight cycles unless progression or unacceptable adverse events (AE). The primary endpoint was overall response rate (ORR) with overall survival (OS), 6-month progression-free survival (PFS), and safety as key secondary endpoints. Change in circulating immune cell populations, plasma, and urinary miRs were evaluated. RESULTS: Twenty-seven patients were treated between April 2016 and June 2020, with median follow-up of 12.4 months. Baseline median age was 68 years, with 81% men and 78% non-Hispanic White. ORR was 33% by intention to treat and 36% in imaging-evaluable patients with three complete responses. Six-month PFS rate was 48.1% [95% confidence interval (CI): 28.7-65.2] and median OS 12.4 months (95% CI: 8.7 months to not reached). Common ≥ grade 2 possibly-related AEs were anemia, lymphopenia, hyperglycemia, and fatigue; grade 3/4 AEs occurred in 56%, including two immune-mediated AEs (pneumonitis and nephritis). Responding patients had a higher percentage of circulating CD4+IFNγ+ T cells. Levels of some miRs, including plasma miR 181 and miR 223, varied in responders compared with nonresponders. CONCLUSIONS: The addition of low-dose paclitaxel to pembrolizumab is active and safe in platinum-refractory urothelial carcinoma. SIGNIFICANCE: We found that combining pembrolizumab with low-dose paclitaxel may be effective in patients with urothelial carcinoma progressing on platinum chemotherapy, with favorable safety profiles.


Subject(s)
Antibodies, Monoclonal, Humanized , Carcinoma, Transitional Cell , MicroRNAs , Urinary Bladder Neoplasms , Male , Humans , Aged , Female , Paclitaxel/adverse effects , Carcinoma, Transitional Cell/drug therapy , Platinum/pharmacology , Urinary Bladder Neoplasms/drug therapy , Prospective Studies , Antineoplastic Combined Chemotherapy Protocols/adverse effects , MicroRNAs/therapeutic use
2.
Am J Clin Oncol ; 44(6): 239-246, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33867481

ABSTRACT

OBJECTIVE: Preclinical studies with muscadine grape extract (MGE) show antitumor activity and decreased systemic inflammation. This phase I study (NCT02583269) assessed safety and tolerability of a proprietary MGE preparation in patients with advanced solid tumors. METHODS: Patients with metastatic or unresectable cancers who were progressing on standard therapies were assigned to MGE in a standard 3+3 design. Five dose levels were tested (320 to 1600 mg total phenolics/d). Safety and maximum-tolerated dose were assessed after 4 weeks. Patients were evaluated for response at 8 weeks and continued on MGE if clinically stable. Secondary outcomes were response, survival, adherence, fatigue, and quality of life (QOL). RESULTS: In total, 23 patients (lung, n=7; gastrointestinal, n=7; genitourinary, n=6; other, n=3) received MGE capsules by mouth twice daily. The cohort [median age 72 years, 48% Eastern Cooperative Oncology Group (ECOG) 2] was heavily pretreated. After 4 weeks on MGE, possibly attributable adverse events grade 2 or higher were fatigue (n=1), decreased lymphocyte count (n=1), and constipation (n=2), including 1 dose-limiting toxicity for grade 3 constipation. Maximum-tolerated dose was not reached. No partial responses were observed. Median time on therapy was 8 weeks, with 29% of patients treated beyond 16 weeks and a median overall survival of 7.2 months. QOL and fatigue levels were stable from baseline to 8 weeks. Higher MGE dose was correlated with improvement in self-reported physical well-being QOL at 8 weeks (r=0.6; P=0.04). CONCLUSIONS: MGE is safe and well-tolerated in heavily pretreated and older cancer patients.  The potential anticancer properties and the effects of MGE on physical well-being and QOL metrics will be evaluated in future studies.


Subject(s)
Neoplasms/drug therapy , Plant Extracts/pharmacokinetics , Plant Extracts/therapeutic use , Vitis/chemistry , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Metastasis , Neoplasms/pathology , Prognosis , Tissue Distribution
3.
Case Rep Urol ; 2020: 8846135, 2020.
Article in English | MEDLINE | ID: mdl-33204569

ABSTRACT

Nivolumab plus ipilimumab represents an effective combination of checkpoint inhibitors that can lead to a durable response with minimal toxicity in patients with metastatic renal cell carcinoma (mRCC). We present a case of a pathologic complete response to neoadjuvant nivolumab plus ipilimumab in a patient with a 13.9 cm left renal mass and significant retroperitoneal and iliac lymphadenopathy, classified as intermediate-risk mRCC. We discuss and review the literature on complete responses after systemic therapy and the ability to predict who has undergone a complete response in the face of residual radiographic evidence of disease.

4.
Oral Oncol ; 82: 83-90, 2018 07.
Article in English | MEDLINE | ID: mdl-29909907

ABSTRACT

OBJECTIVES: Cixutumumab (CIX) and cetuximab (CET) monoclonal antibodies block ligand-binding to insulin-like growth factor-1 receptor (IGF-1R) and epidermal growth factor receptor (EGFR) respectively. The objective of this study was to assess the efficacy of CIX alone or combined with CET in recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC) patients. METHODS: In this open-label phase II trial, 91 R/M HNSCC patients who progressed within 90 days of platinum-based chemotherapy, were randomized to CIX 10 mg/kg alone or with CET 500 mg/m2 every 2 weeks. Patients were stratified by prior CET use. The primary endpoint was median progression-free survival (PFS). Exploratory biomarker assessments included relevant markers on archival tumor and serial cytokine/angiogenic-factor profiles in blood. RESULTS: Forty-seven patients were treated with CIX monotherapy and 44 with combination. The median PFS was 1.9 and 2.0 months and clinical benefit rate (complete or partial responses and stable disease) was 5.9% and 15.3%, respectively. There was no exacerbation of CET toxicity by concurrent CIX exposure. Higher tumor expression of IGF-1 was associated with improved PFS in the CIX + CET arm while increased p-EGFR expression correlated with shorter PFS in patients receiving single agent CIX. Higher serum baseline levels of IGF-1 and IGFBP-3 correlated with improved PFS and overall survival (OS) in the CIX arm. Neither regimen resulted in improved PFS or OS compared to historical data with CET alone. CONCLUSION: The results of this study do not support the use of cixutumumab alone or with cetuximab in unselected patients with R/M HNSCC.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cetuximab/therapeutic use , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/pathology , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Cetuximab/administration & dosage , Cetuximab/adverse effects , ErbB Receptors/metabolism , Female , Head and Neck Neoplasms/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/metabolism , Male , Neoplasm Metastasis , Neoplasm Recurrence, Local , Squamous Cell Carcinoma of Head and Neck/metabolism
5.
Clin Cancer Res ; 23(15): 4138-4145, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28377480

ABSTRACT

Purpose: Genetic and preclinical studies have implicated FGFR signaling in the pathogenesis of adenoid cystic carcinoma (ACC). Dovitinib, a suppressor of FGFR activity, may be active in ACC.Experimental Design: In a two-stage phase II study, 35 patients with progressive ACC were treated with dovitinib 500 mg orally for 5 of 7 days continuously. The primary endpoints were objective response rate and change in tumor growth rate. Progression-free survival, overall survival, metabolic response, biomarker, and quality of life were secondary endpoints.Results: Of 34 evaluable patients, 2 (6%) had a partial response and 22 (65%) had stable disease >4 months. Median PFS was 8.2 months and OS was 20.6 months. The slope of the overall TGR fell from 1.95 to 0.63 on treatment (P < 0.001). Toxicity was moderate; 63% of patients developed grade 3-4 toxicity, 94% required dose modifications, and 21% stopped treatment early. An early metabolic response based on 18FDG-PET scans was seen in 3 of 15 patients but did not correlate with RECIST response. MYB gene translocation was observed and significantly correlated with overexpression of MYB but did not correlate with FGFR1 phosphorylation or clinical response to dovitinib.Conclusions: Dovitinib produced few objective responses in patients with ACC but did suppress the TGR with a PFS that compares favorably with those reported with other targeted agents. Future studies of more potent and selective FGFR inhibitors in biomarker-selected patients will be required to determine whether FGFR signaling is a valid therapeutic target in ACC. Clin Cancer Res; 23(15); 4138-45. ©2017 AACR.


Subject(s)
Benzimidazoles/administration & dosage , Carcinoma, Adenoid Cystic/drug therapy , Cell Proliferation/drug effects , Neoplasm Recurrence, Local/drug therapy , Quinolones/administration & dosage , Adult , Aged , Benzimidazoles/adverse effects , Carcinoma, Adenoid Cystic/pathology , Disease-Free Survival , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Recurrence, Local/pathology , Oncogene Proteins v-myb/genetics , Quinolones/adverse effects , Receptor, Fibroblast Growth Factor, Type 1/genetics
6.
Head Neck ; 38(4): 620-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25487882

ABSTRACT

BACKGROUND: Adenoid cystic carcinoma (ACC) is a rare tumor of secretory glands. In this study, recent advances in molecular characterization and in therapeutics are reviewed. METHODS: A search of articles in PubMed and of abstracts from national meetings was performed regarding ACC. RESULTS: Recent genetic analyses found that recurrent chromosome 6:9 translocations in ACC generate an MYB:NFIB gene fusion resulting in overexpression of the MYB oncoprotein. Several other frequent mutations are recently published that may be relevant for drug development. Several trials of targeted drugs are reviewed. Some agents delay tumor progression, but tumor responses remain rare. CONCLUSION: ACCs have a characteristic chromosomal translocation, but also frequently pick up additional mutations. Clinical research is limited by the rarity and slow growth of ACC. Several ongoing trials are testing agents that inhibit fibroblast growth factor receptor signaling or other signaling pathways. Novel treatments based on the recently sequenced tumor genome are under development.


Subject(s)
Carcinoma, Adenoid Cystic/genetics , Chromosomes, Human, Pair 6 , Chromosomes, Human, Pair 9 , Molecular Targeted Therapy/methods , Salivary Gland Neoplasms/genetics , Translocation, Genetic , Carcinoma, Adenoid Cystic/pathology , Carcinoma, Adenoid Cystic/therapy , Clinical Trials as Topic , Female , Genes, myb/genetics , Genetic Predisposition to Disease , Humans , Male , Mutation , Salivary Gland Neoplasms/pathology , Salivary Gland Neoplasms/therapy
8.
Cancer ; 119(13): 2405-12, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23609948

ABSTRACT

BACKGROUND: This study sought to identify novel effectors and markers of localized but potentially life-threatening prostate cancer (PCa), by evaluating chromosomal copy number alterations (CNAs) in tumors from patients who underwent prostatectomy and correlating these with clinicopathologic features and outcome. METHODS: CNAs in tumor DNA samples from 125 patients in the discovery cohort who underwent prostatectomy were assayed with high-resolution Affymetrix 6.0 single-nucleotide polymorphism microarrays and then analyzed using the Genomic Identification of Significant Targets in Cancer (GISTIC) algorithm. RESULTS: The assays revealed 20 significant regions of CNAs, 4 of them novel, and identified the target genes of 4 of the alterations. By univariate analysis, 7 CNAs were significantly associated with early PCa-specific mortality. These included gains of chromosomal regions that contain the genes MYC, ADAR, or TPD52 and losses of sequences that incorporate SERPINB5, USP10, PTEN, or TP53. On multivariate analysis, only the CNAs of PTEN (phosphatase and tensin homolog) and MYC (v-myc myelocytomatosis viral oncogene homolog) contributed additional prognostic information independent of that provided by pathologic stage, Gleason score, and initial prostate-specific antigen level. Patients whose tumors had alterations of both genes had a markedly elevated risk of PCa-specific mortality (odds ratio = 53; 95% CI = 6.92-405, P = 1 × 10(-4)). Analyses of 333 tumors from 3 additional distinct patient cohorts confirmed the relationship between CNAs of PTEN and MYC and lethal PCa. CONCLUSIONS: This study identified new CNAs and genes that likely contribute to the pathogenesis of localized PCa and suggests that patients whose tumors have acquired CNAs of PTEN, MYC, or both have an increased risk of early PCa-specific mortality.


Subject(s)
DNA Copy Number Variations , Genetic Markers , Polymorphism, Single Nucleotide , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Proto-Oncogenes/genetics , Adult , Aged , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Odds Ratio , Predictive Value of Tests , Prognosis , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery
9.
Support Care Cancer ; 21(3): 889-99, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23052911

ABSTRACT

PURPOSE: Appropriate utilization of treatment is a goal for all patients undergoing cancer treatment. Proper treatment maximizes benefit and limits exposure to unnecessary measures. This report describes findings of the feasibility and acceptability of implementing a short, clinic-based decision aid and presents an in-depth clinical profile of the participants. METHODS: This descriptive study used a prospective, quantitative approach to obtain the feasibility and acceptability of a decision aid (DecisionKEYS for Balancing Choices) for use in clinical settings. It combined results of trials of patients with three different common malignancies. All groups used the same decision aid series. Participants included 80 patients with solid tumors (22 with newly diagnosed breast cancer, 19 with advanced prostate cancer, and 39 with advanced lung cancer) and their 80 supporters as well as their physicians and nurses, for a total of 160 participants and 10 health professionals. RESULTS: The decision aid was highly acceptable to patient and supporter participants in all diagnostic groups. It was feasible for use in clinic settings; the overall value was rated highly. Of six physicians, all found the interactive format with the help of the nurse as feasible and acceptable. Nurses also rated the decision aid favorably. CONCLUSIONS: This intervention provides the opportunity to enhance decision making about cancer treatment and warrants further study including larger and more diverse groups. Strengths of the study included a theoretical grounding, feasibility testing of a practical clinic-based intervention, and summative evaluation of acceptability of the intervention by patient and supporter pairs. Further research also is needed to test the effectiveness of the decision aid in diverse clinical settings and to determine if this intervention can decrease overall costs.


Subject(s)
Decision Making , Decision Support Techniques , Neoplasms/therapy , Aged , Attitude of Health Personnel , Feasibility Studies , Female , Humans , Male , Middle Aged , Neoplasms/pathology , Patient Acceptance of Health Care , Prospective Studies
10.
J Oral Pathol Med ; 42(4): 332-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23106397

ABSTRACT

BACKGROUND: Although oral squamous cell carcinomas (OSCCs) commonly overexpress the epidermal growth factor receptor (EGFR), EGFR tyrosine kinase inhibitors (TKIs) exhibit poor efficacy clinically. Activation of the insulin-like growth factor-1 receptor (IGF1R) induces resistance of OSCC cells to EGFR-TKIs in vitro. This study seeks to evaluate the changes in cell cycle status in OSCC cells in response to gefitinib and IGF1R activation. METHODS: SCC-25 OSCC cells were used for in vitro analyses. RESULTS: Gefitinib caused a 50% reduction in S-phase population, and IGF1R activation caused a 2.8-fold increase; combined treatment yielded a baseline S-phase population. Gefitinib treatment increased the cyclin-dependent kinase inhibitor p27, and this was not abrogated by IGF1R activation. pT157-p27 was noted by immunoblot to be decreased on gefitinib treatment, but this was reversed with IGF1R activation. T157 phosphorylation contributes to cytoplasmic localization of p27 where it can promote cell proliferation and cell motility. Using both subcellular fractionation and immunofluorescence microscopy techniques, IGF1R stimulation was noted to increase the relative cytoplasmic localization of p27; this persisted when combined with gefitinib. CONCLUSIONS: IGF1R activation partially reverses the cell cycle arrest caused by gefitinib in OSCC cells. While IGF1R stimulation does not eliminate the gefitinib-induced increase in total p27, its phosphorylation state and subcellular localization are altered. This may contribute to the ability of the IGF1R to rescue OSCC cells from EGFR-TKI treatment and may have important implications for the use of p27 as a biomarker of cell cycle arrest and response to therapy.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cyclin-Dependent Kinase Inhibitor p27/physiology , ErbB Receptors/physiology , Mouth Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Receptor, IGF Type 1/physiology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Nucleus/ultrastructure , Cyclin D/drug effects , Cyclin-Dependent Kinase Inhibitor p27/drug effects , Cytoplasm/ultrastructure , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , Gefitinib , Humans , Insulin-Like Growth Factor I/pharmacology , Oncogene Protein v-akt/physiology , Peptide Fragments/pharmacology , Phosphorylation , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/pharmacology , Quinazolines/administration & dosage , Receptor, IGF Type 1/drug effects , S Phase/drug effects , Subcellular Fractions/ultrastructure
11.
Int J Otolaryngol ; 2012: 685951, 2012.
Article in English | MEDLINE | ID: mdl-22888357

ABSTRACT

We evaluated a panel of 8 immunohistochemical biomarkers as predictors of clinical response to definitive intensity-modulated radiotherapy in patients with oropharyngeal squamous cell carcinoma (OPSCC). 106 patients with OPSCC were treated to a total dose of 66-70 Gy and retrospectively analyzed for locoregional control (LRC), disease-free survival (DFS), and overall survival (OS). All tumors had p16 immunohistochemical staining, and 101 tumors also had epidermal growth factor receptor (EGFR) staining. 53% of the patients had sufficient archived pathologic specimens for incorporation into a tissue microarray for immunohistochemical analysis for cyclophilin B, cyclin D1, p21, hypoxia-inducible factor-1α (HIF-1α), carbonic anhydrase, and major vault protein. Median followup was 27.2 months. 66% of the tumors were p16 positive, and 34% were p16 negative. On univariate analysis, the following correlations were statistically significant: p16 positive staining with higher LRC (P = 0.005) and longer DFS (P < 0.001); cyclin D1 positive staining with lower LRC (P = 0.033) and shorter DFS (P = 0.002); HIF-1α positive staining with shorter DFS (P = 0.039). On multivariate analysis, p16 was the only significant independent predictor of DFS (P = 0.023). After immunohistochemical examination of a panel of 8 biomarkers, our study could only verify p16 as an independent prognostic factor in OPSCC.

12.
Mol Cancer Ther ; 10(11): 2124-34, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21878657

ABSTRACT

Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKI) have poor efficacy in head and neck squamous carcinoma cells (HNSCC). Because the IGF-1 receptor (IGF1R) generates potent prosurvival signals and has been implicated in therapeutic resistance, its ability to induce resistance to EGFR-TKIs was studied in vitro. Five HNSCC cell lines showed reduced sensitivity to the EGFR-TKI gefitinib when the IGF1R was activated. In SCC-25 and Cal27 cells, gefitinib inhibited basal and EGF-stimulated EGFR, extracellular signal-regulated kinase (Erk), and Akt phosphorylation and reduced cell number. This correlated with initiation of apoptosis based on a 4-fold increase in PARP cleavage and a 2.5-fold increase in Annexin V positivity. The apoptotic response and reduction in cell number were blocked by IGF1R activation, which resulted in phosphorylation of both Erk and Akt. In both the cell lines, IGF1R-induced Erk, but not Akt, activation was eliminated by gefitinib. IGF1R-induced gefitinib resistance was unaffected by MAP/Erk kinase inhibition with U0126 but was partially impaired by inhibition of phosphoinositide-3-kinase with LY294002. The IGF1R-TKI PQ401 inhibited growth of SCC-25 and Cal27 cells alone and also acted synergistically with gefitinib. Thus, the IGF1R can make HNSCC cells resistant to EGFR-TKI treatment via a prosurvival mechanism. Of the 8 HNSCC tumor samples studied, all samples expressed the IGF1R and 5 showed detectable IGF1R phosphorylation, suggesting that this receptor may be relevant in vivo, and thus, combined EGFR/IGF1R inhibition may be necessary in some patients for effective targeted molecular therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/metabolism , ErbB Receptors/antagonists & inhibitors , Head and Neck Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor, IGF Type 1/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/genetics , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Head and Neck Neoplasms/genetics , Humans , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/genetics , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck
13.
Cancer ; 116(11): 2645-54, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20225325

ABSTRACT

BACKGROUND: Patients with oropharyngeal squamous cell carcinoma (OPSCC) treated with intensity-modulated radiotherapy (IMRT) were stratified by p16 status, neck dissection, and chemotherapy to correlate these factors with outcomes. METHODS: A total of 112 patients with OPSCC treated with IMRT from 2002 to 2008 were retrospectively analyzed. All patients received RT to 66-70 Gray. Forty-five of the tumors were p16 positive (p16+), 27 were p16 negative (p16-), and 41 had unknown p16 status. Sixty-two patients had postradiation neck dissections. Nine patients with p16- tumors and 28 patients with p16+ tumors received chemotherapy. The distribution of T, N, and stage grouping among the p16+ and p16- patients was not significantly different, and 87.5% patients had stage III/IV disease. RESULTS: The median follow-up was 26.3 months. For patients with p16+ tumors, p16- tumors, and the overall cohort, the actuarial 3-year locoregional progression-free survival rate was 97.8%,73.5%, and 90.5% respectively (P = .006) and the disease-free survival rate was 88.2%, 61.4%, and 81.7%, respectively (P = .004). Patients with p16+ tumors had an 89.5% and 87.5% pathologic complete response (CR) on neck dissection with and without chemotherapy, respectively. In contrast, patients with p16- tumors had a 66.7% and 25.0% pathologic CR on neck dissection with and without chemotherapy, respectively. CONCLUSIONS: In this series, p16 status was found to be a significant predictive biomarker and patients with p16+ tumors had much better outcomes than patients with p16- tumors. Further investigation is warranted to determine whether less intense therapy is appropriate for selected patients with p16+ OPSCC, whereas more aggressive strategies are needed to improve outcomes in patients with p16- disease.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/radiotherapy , Genes, p16 , Oropharyngeal Neoplasms/genetics , Oropharyngeal Neoplasms/radiotherapy , Radiotherapy, Intensity-Modulated , Combined Modality Therapy , Cyclin-Dependent Kinase Inhibitor p16 , Female , Humans , Male , Middle Aged , Neck Dissection , Neoplasm Proteins/metabolism , Radiotherapy Dosage , Treatment Outcome
14.
Arch Otolaryngol Head Neck Surg ; 135(11): 1126-32, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19917926

ABSTRACT

OBJECTIVE: To identify factors that predict complete response of cervical nodal disease to radiation therapy (RT) in patients with oropharyngeal squamous cell carcinoma (OP-SCCA). DESIGN: Histologic analysis of prospectively collected specimens and retrospective medical chart review. SETTING: Tertiary referral center. SUBJECTS: Sixty-nine patients with OP-SCCA treated from January 1, 2002, through June 1, 2008. INTERVENTION: Definitive RT, with or without chemotherapy and with or without neck dissection (ND). MAIN OUTCOME MEASURE: Presence of a viable tumor in post-RT ND specimen. RESULTS: Tissue specimens from 69 patients with OP-SCCA treated primarily with RT, with or without chemotherapy, were evaluated. Of these, 47 (68.1%) were strongly and diffusely positive for p16 expression by immunohistochemical analysis, signifying human papillomavirus positivity. Patients with p16-positive and p16-negative tumors (hereinafter, p16+ and p16-, respectively) had similarly sized primary tumors on presentation, but p16+ primary tumors were associated with more advanced neck disease (nodal stages N2c-N3; 31.9% vs 4.5% for p16- tumors) and more contralateral nodes (27.7% vs 4.5% for p16- tumors). Forty-seven patients (59.0%) underwent planned posttreatment ND (a total of 55 NDs). The NDs performed for p16- tumors were significantly more likely to have viable tumor in the specimen (50.0% vs 18.0% for p16+ tumors; P = .02). In addition, p16+ necks with residual viable cancer were characterized by incomplete response on post-RT imaging, tobacco and alcohol use, and extracapsular spread on pretreatment imaging. CONCLUSIONS: In conjunction with other clinical parameters, p16 status can help predict the need for post-RT ND in patients with OP-SCCA. Although close observation may be warranted in selected patients with p16+ tumors, patients with p16- tumors are at much higher risk for residual neck disease, even when initial nodal disease is less advanced.


Subject(s)
Carcinoma, Squamous Cell/radiotherapy , Neoplasm Proteins/metabolism , Oropharyngeal Neoplasms/radiotherapy , Biomarkers, Tumor/metabolism , Biopsy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/secondary , Cyclin-Dependent Kinase Inhibitor p16 , Female , Follow-Up Studies , Humans , Immunohistochemistry , Lymphatic Metastasis , Male , Middle Aged , Neck Dissection , Neoplasm Staging , Oropharyngeal Neoplasms/metabolism , Oropharyngeal Neoplasms/pathology , Prognosis , Retrospective Studies
15.
Anticancer Res ; 29(7): 2869-73, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19596976

ABSTRACT

BACKGROUND: To determine the maximally tolerated dose (MTD) of capecitabine when given concomitantly with carboplatin and intensity-modulated radiation therapy (IMRT) for treatment of localized stage III/IV squamous cell carcinomas of the head and neck (HNSCC). PATIENTS AND METHODS: After six weeks of induction chemotherapy with capecitabine and carboplatin, patients received a second course with concomitant IMRT. The MTD for capecitabine during chemoradiation was determined by a standard phase I trial design. RESULTS: Nine out of the eleven patients qualified for chemoradiation. With weekly carboplatin AUC=1.5 and IMRT, the MTD of capecitabine was 850 mg/m(2)/day when given in divided doses on days 1-14 and 22-35. The dose-limiting toxicity was myleosuppression and other adverse effects were modest. Eight patients experienced complete response after chemoradiation and seven remain relapse-free after 34 months. CONCLUSION: Capecitabine, carboplatin, and IMRT given as described were well tolerated by HNSCC patients and in this pilot study produced a promising rate of disease control.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Capecitabine , Carboplatin/administration & dosage , Combined Modality Therapy , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Humans , Treatment Outcome
18.
Arch Otolaryngol Head Neck Surg ; 132(2): 134-8, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16490869

ABSTRACT

OBJECTIVES: To analyze outcomes and to provide follow-up for our increasing patient cohort with esthesioneuroblastoma. DESIGN: Retrospective cohort analysis. SETTING: Patients were examined from September 1, 1976, to May 30, 2004, in a tertiary care academic hospital. PATIENTS: Fifty consecutive patients diagnosed as having esthesioneuroblastoma were treated with a standardized protocol during a 28-year period. Patients with tumors staged Kadish A or B received preoperative radiotherapy followed by craniofacial resection, while patients with Kadish stage C disease were treated with preoperative sequential chemotherapy and radiotherapy followed by a craniofacial resection. The mean follow-up is 93 months (range, 1-330 months). RESULTS: The disease-free survival was 86.5% and 82.6% at 5 and 15 years, respectively. There were 17 patients (34%) who developed recurrent disease, most of which was locoregional (12 patients [71%]). There was a long interval to relapse (mean, 6 years), with the longest time to regional recurrence being 10 years. Distant relapses occurred sooner, with poorer outcomes. Of these 17 patients, 7 (41%) underwent successful salvage surgery, while 3 remain alive with disease. CONCLUSIONS: Excellent outcomes for esthesioneuroblastoma are achievable. Long-term follow-up is necessary because of the extended interval for recurrent disease; unlike most sinonasal malignancies, surgical salvage is possible.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Esthesioneuroblastoma, Olfactory , Nasal Cavity , Neurosurgical Procedures/methods , Nose Neoplasms , Adolescent , Adult , Aged , Child , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Esthesioneuroblastoma, Olfactory/drug therapy , Esthesioneuroblastoma, Olfactory/radiotherapy , Esthesioneuroblastoma, Olfactory/surgery , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Recurrence, Local , Nose Neoplasms/drug therapy , Nose Neoplasms/radiotherapy , Nose Neoplasms/surgery , Radiotherapy, Adjuvant , Retrospective Studies , Treatment Outcome , Vincristine/administration & dosage
19.
Int J Cancer ; 104(1): 19-27, 2003 Mar 10.
Article in English | MEDLINE | ID: mdl-12532415

ABSTRACT

Overexpressed epidermal growth receptor factor receptors (EGFRs) are thought to contribute to the malignant phenotype of human glioblastomas (GBMs), but the mechanism is not well understood. We found that SKMG-3 cells, a rare GBM cell line that maintains EGFR gene amplification in vitro, produced high levels of EGFR protein. The cells also expressed the related receptors HER2/neu and HER4, but not HER3. Immunoblots and tryptic phosphopeptide maps showed that the SKMG-3 EGFRs were intact and functional and that a subset of these receptors were spontaneously autophosphorylated. EGF treatment stimulated phosphorylation of the EGFRs as well as the downstream effectors Erk, AKT1, stat3 and c-Cbl. Under minimal growth conditions, the unstimulated SKMG-3 cells contained constitutively phosphorylated Erk and AKTI but no detectable stat3 DNA-binding complexes. The EGFR kinase inhibitor PD158780 reduced the constitutive phosphorylation of the receptor and Erk but not that of AKT1. In contrast, inhibition of phosphatidylinositol-3-kinase (PI3K) blocked the constitutive phosphorylation of Erk and AKT-1 but not the EGFR. We conclude that the SKMG-3 cells represent the subset of GBMs with amplified EGFR genes that overexpress intact receptors. The results also suggest that in some GBMs, signals from overexpressed EGFRs contribute to the constitutive phosphorylation of Erk, but these signals may not required for the constitutive activation of PI3K or AKT1.


Subject(s)
ErbB Receptors/physiology , Gene Expression Regulation, Neoplastic , Genes, erbB-1 , Genes, erbB-2 , Glioblastoma/pathology , Neoplasm Proteins/physiology , Receptor, ErbB-2/physiology , Signal Transduction/physiology , Ubiquitin-Protein Ligases , Breast Neoplasms/pathology , Carcinoma, Squamous Cell/pathology , Cells, Cultured , DNA-Binding Proteins/metabolism , Enzyme Activation , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Gene Amplification , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Laryngeal Neoplasms/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neuroglia/cytology , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-cbl , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-4 , STAT3 Transcription Factor , Trans-Activators/metabolism , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...