Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Endocr Relat Cancer ; 30(11)2023 11 01.
Article in English | MEDLINE | ID: mdl-37702588

ABSTRACT

We assessed the efficacy and safety of combining bevacizumab with temsirolimus in patients with advanced extra-pancreatic neuroendocrine tumors. This NCI-sponsored multicenter, open-label, phase II study (NCT01010126) enrolled patients with advanced, recurrent, or metastatic extra-pancreatic neuroendocrine tumors. All patients were treated with temsirolimus and bevacizumab until disease progression or unacceptable toxicity. Temsirolimus 25 mg was administered i.v. on days 1, 8, 15, and 22 and bevacizumab 10 mg/kg i.v. on days 1 and 15 of a 4-week cycle. Discontinuation of temsirolimus or bevacizumab did not require discontinuation of the other agent. The primary endpoints were objective response rate and 6-month progression-free survival rate. Fifty-nine patients were enrolled in this study, and 54 were evaluated for efficacy and adverse events. While median progression-free survival was 7.1 months, the median duration of treatment with temsirolimus was 3.9 months and that with bevacizumab was 3.5 months. The objective response rate of combination therapy was 2%, and 6-month progression-free survival was 48%. The most frequently reported grade 3-4 adverse events included fatigue (13%), hypertension (13%), and bleeding (13%). Close to 54% of the patients discontinued treatment due to adverse events, refusal of further treatment, or treatment delays. Three deaths occurred in the study, of which two were due to treatment-related bowel perforations. Given the minimal efficacy and increased toxicity seen with the combination of bevacizumab and temsirolimus, we do not recommend the use of this regimen in patients with advanced extra-pancreatic neuroendocrine tumors.


Subject(s)
Neoplasms, Second Primary , Neuroendocrine Tumors , Pancreatic Neoplasms , Humans , Bevacizumab/adverse effects , Neuroendocrine Tumors/drug therapy , Combined Modality Therapy , Pancreatic Neoplasms/drug therapy
2.
J Clin Oncol ; 41(7): 1470-1491, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36603169

ABSTRACT

PURPOSE: To develop recommendations involving targeted therapies for patients with advanced gastroesophageal cancer. METHODS: The American Society of Clinical Oncology convened an Expert Panel to conduct a systematic review of relevant studies and develop recommendations for clinical practice. RESULTS: Eighteen randomized controlled trials met the inclusion criteria for the systematic review. RECOMMENDATIONS: For human epidermal growth factor receptor 2 (HER2)-negative patients with gastric adenocarcinoma (AC) and programmed death-ligand 1 (PD-L1) combined positive score (CPS) ≥ 5, first-line therapy with nivolumab and chemotherapy (CT) is recommended. For HER2-negative patients with esophageal or gastroesophageal junction (GEJ) AC and PD-L1 CPS ≥ 5, first-line therapy with nivolumab and CT is recommended. First-line therapy with pembrolizumab and CT is recommended for HER2-negative patients with esophageal or GEJ AC and PD-L1 CPS ≥ 10. For patients with esophageal squamous cell carcinoma and PD-L1 tumor proportion score ≥ 1%, nivolumab plus CT, or nivolumab plus ipilimumab is recommended; for patients with esophageal squamous cell carcinoma and PD-L1 CPS ≥ 10, pembrolizumab plus CT is recommended. For patients with HER2-positive gastric or GEJ previously untreated, unresectable or metastatic AC, trastuzumab plus pembrolizumab is recommended, in combination with CT. For patients with advanced gastroesophageal or GEJ AC whose disease has progressed after first-line therapy, ramucirumab plus paclitaxel is recommended. For HER2-positive patients with gastric or GEJ AC who have progressed after first-line therapy, trastuzumab deruxtecan is recommended. In all cases, participation in a clinical trial is recommended as it is the panel's expectation that targeted treatment options for gastroesophageal cancer will continue to evolve.Additional information is available at www.asco.org/gastrointestinal-cancer-guidelines.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Stomach Neoplasms , Humans , Esophageal Neoplasms/pathology , Nivolumab/therapeutic use , B7-H1 Antigen/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Stomach Neoplasms/pathology , Esophagogastric Junction/pathology , Immunotherapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
3.
Cancer Chemother Pharmacol ; 90(3): 217-228, 2022 09.
Article in English | MEDLINE | ID: mdl-35907014

ABSTRACT

PURPOSE: Aurora Kinase A (AKA) inhibition with gemcitabine represents a potentially synergistic cancer treatment strategy via mitotic catastrophe. The feasibility, safety, and preliminary efficacy of alisertib (MLN8237), an oral AKA inhibitor, with gemcitabine was evaluated in this open-label phase I trial with dose escalation and expansion. METHODS: Key inclusion criteria included advanced solid tumor with any number of prior chemotherapy regimens in the dose escalation phase, and advanced pancreatic adenocarcinoma with up to two prior chemotherapy regimens. Four dose levels (DLs 1-4) of alisertib (20, 30, 40, or 50 mg) were evaluated in 3 + 3 design with gemcitabine 1000 mg/m2 on days 1, 8, and 15 in 28-day cycles. RESULTS: In total, 21 subjects were treated in dose escalation and 5 subjects were treated in dose expansion at DL4. Dose-limiting toxicities were observed in 1 of 6 subjects each in DL3 and DL4. All subjects experienced treatment-related adverse events. Grade ≥ 3 treatment-related adverse events were observed in 73% of subjects, with neutropenia observed in 54%. Out of 22 subjects evaluable for response, 2 subjects (9%) had partial response and 14 subjects (64%) had stable disease. Median PFS was 4.1 months (95% CI 2.1-4.5). No significant changes in pharmacokinetic parameters for gemcitabine or its metabolite dFdU were observed with alisertib co-administration. CONCLUSIONS: This trial established the recommended phase 2 dose of alisertib 50 mg to be combined with gemcitabine. Gemcitabine and alisertib are a feasible strategy with potential for disease control in multiple heavily pre-treated tumors, though gastrointestinal and hematologic toxicity was apparent.


Subject(s)
Adenocarcinoma , Neoplasms , Pancreatic Neoplasms , Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Azepines , Deoxycytidine/analogs & derivatives , Humans , Maximum Tolerated Dose , Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Pyrimidines , Gemcitabine , Pancreatic Neoplasms
4.
Clin Lung Cancer ; 23(1): 60-71, 2022 01.
Article in English | MEDLINE | ID: mdl-34753703

ABSTRACT

BACKGROUND: The phase III S0819 trial investigated addition of cetuximab to first-line chemotherapy (CT) in NSCLC. Subgroup analyses suggested an OS benefit among patients with EGFR copy number gain in squamous cell carcinomas (SCC), (HR = 0.58 [0.39-0.86], P = .0071). A more detailed model based on EGFR FISH, EGFR IHC and KRAS mutation status was evaluated to yield a more precise predictive paradigm of cetuximab-based therapy in advanced NSCLC. METHODS: FISH was performed using the Colorado Scoring Criteria; H-Score was used to quantify EGFR IHC expression (cut-off ≥ 200). A Cox model was used to assess treatment effects for OS and PFS within biomarker and clinical subgroups. KRAS mutation was analyzed using Therascreen. The false discovery rate controlled for multiple comparisons. S0819 ClinicalTrials.gov Identifier: NCT00946712. RESULTS: Of 1,313 eligible patients, assay results were obtained for FISH on 976 patients (41% positive), for IHC on 945 patients (31% positive), and KRAS mutation status on 627 patients (26% positive). In SCC patients, OS was significantly improved with addition of cetuximab when both EGFR FISH and EGFR IHC were positive (N = 58), (OS HR: 0.32 [95% CI 0.18-0.59]; P = .0002, q = 0.08), median 12.6 versus 4.6 months. The results were independent of KRAS mutation status. In Non-SCC, no predictive value of EGFR IHC, EGFR FISH status and/or KRAS status was seen. CONCLUSIONS: In NSCLC SCC, a combination index of EGFR FISH plus EGFR IHC results was associated with improved OS when cetuximab was added to CT, representing a potential predictive molecular paradigm for patients suitable for EGFR-antibody therapy.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Cetuximab/therapeutic use , Gene Expression , Lung Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , ErbB Receptors/genetics , Female , Forecasting , Humans , Male , Middle Aged , Treatment Outcome , Young Adult
5.
Cancer Chemother Pharmacol ; 88(2): 335-341, 2021 08.
Article in English | MEDLINE | ID: mdl-33993383

ABSTRACT

PURPOSE: Aurora kinases are overexpressed or amplified in numerous malignancies. This study was designed to determine the safety and tolerability of the Aurora A kinase inhibitor alisertib (MLN8237) when combined with weekly irinotecan. METHODS: In this single-center phase 1 study, adult patients with refractory advanced solid tumors received 100 mg/m2 irinotecan intravenously on day 1 and 8 of a 21-day cycle. Alisertib at planned escalating dose levels of 20-60 mg was administered orally twice per day on days 1-3 and 8-10. Patients homozygous for UGT1A1*28 were excluded. The primary objective was the safety of alisertib when combined with irinotecan to determine the maximum tolerated dose (MTD). Secondary objectives included overall response rate by RECIST and pharmacokinetics in a planned expansion cohort of patients with colorectal cancer treated at the MTD. RESULTS: A total of 17 patients enrolled at three dose levels. Dose-limiting toxicities included diarrhea, dehydration, and neutropenia. The MTD of alisertib combined with weekly irinotecan was 20 mg twice per day on days 1-3 and 8-10. One fatal cardiac arrest at the highest dose level tested was deemed possibly related to drug treatment. One partial response in 11 efficacy evaluable patients (9%) occurred in a patient with small cell lung cancer. The study was terminated prior to the planned expansion in patients with colorectal cancer. CONCLUSION: In contrast to prior results in a pediatric population, adult patients did not tolerate alisertib combined with irinotecan at clinically meaningful doses due to hematologic and gastrointestinal toxicities. The study was registered with ClinicalTrials.gov under study number NCT01923337 on Aug 15, 2013.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azepines/therapeutic use , Irinotecan/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Adult , Aged , Aged, 80 and over , Cohort Studies , Drug Administration Schedule , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Young Adult
6.
J Adolesc Young Adult Oncol ; 10(5): 521-533, 2021 10.
Article in English | MEDLINE | ID: mdl-33512275

ABSTRACT

Purpose: We sought to enumerate secondary medical conditions from hospitalization records in adolescent and young adult (AYA) differentiated thyroid cancer (DTC) survivors and identify characteristics of patients with increased likelihood of subsequent medical diagnoses. Methods: Using data from the California Cancer Registry and statewide hospitalization data, we examined incident oncologic, endocrine, pulmonary, hematologic, and cardiovascular diagnoses in 12,312 AYA (aged 15-39) patients diagnosed with DTC in 1996-2012 and surviving >2 years after diagnosis with follow-up through 2014. We calculated the cumulative incidence of each condition accounting for the competing risk of death and used multivariable Cox proportional hazards regression to evaluate sociodemographic and clinical characteristics associated with each incident condition. Results: The 10-year cumulative incidences of multiple medical conditions were particularly high in blacks and Hispanics. Asian/Pacific Islander survivors were most likely to develop subsequent cancers. Men had higher rates of cardiovascular and diabetes diagnoses than women, but lower rates of asthma and cytopenias. Low socioeconomic status and/or public or no insurance were associated with a higher risk of several diagnoses. More extensive disease stage and thyroid surgery increased the risk of calcium and phosphorus metabolism disorders. Neck reoperation associated with the risk of cytopenias, as well as subsequent endocrine, cardiovascular, and respiratory diagnoses. Conclusion: The incidence of medical conditions after thyroid cancer diagnosis and treatment differ among racial/ethnic groups and sexes. Those residing in lower socioeconomic neighborhoods, those with public or no insurance, and those who require further neck surgery have substantially higher burdens of subsequent medical diagnoses.


Subject(s)
Thyroid Neoplasms , Adolescent , Female , Hispanic or Latino , Humans , Incidence , Male , Social Class , Survivors , Thyroid Neoplasms/epidemiology , Young Adult
7.
J Natl Compr Canc Netw ; 18(11): 1446-1452, 2020 11.
Article in English | MEDLINE | ID: mdl-33152701

ABSTRACT

Organ donors are systematically screened for infection, whereas screening for malignancy is less rigorous. The true incidence of donor-transmitted malignancies is unknown due to a lack of universal tumor testing in the posttransplant setting. Donor-transmitted malignancy may occur even when not suspected based on donor or recipient factors, including age and time to cancer diagnosis. We describe the detection of a gastrointestinal adenocarcinoma transmitted from a young donor to 4 transplant recipients. Multidimensional histopathologic and genomic profiling showed a CDH1 mutation and MET amplification, consistent with gastric origin. At the time of writing, one patient in this series remains alive and without evidence of cancer after prompt organ explant after cancer was reported in other recipients. Because identification of a donor-derived malignancy changes management, our recommendation is to routinely perform short tandem repeat testing (or a comparable assay) immediately upon diagnosis of cancer in any organ transplant recipient. Routine testing for a donor-origin cancer and centralized reporting of outcomes are necessary to establish a robust evidence base for the future development of clinical practice guidelines.


Subject(s)
Neoplasms , Organ Transplantation , Transplant Recipients , Humans , Incidence , Neoplasms/diagnosis , Neoplasms/genetics , Organ Transplantation/adverse effects , Tissue Donors
8.
Mol Cancer Ther ; 19(4): 1070-1079, 2020 04.
Article in English | MEDLINE | ID: mdl-32029633

ABSTRACT

FOLFOX is one of the most effective treatments for advanced colorectal cancer. However, cumulative oxaliplatin neurotoxicity often results in halting the therapy. Oxaliplatin functions predominantly via the formation of toxic covalent drug-DNA adducts. We hypothesize that oxaliplatin-DNA adduct levels formed in vivo in peripheral blood mononuclear cells (PBMC) are proportional to tumor shrinkage caused by FOLFOX therapy. We further hypothesize that adducts induced by subtherapeutic "diagnostic microdoses" are proportional to those induced by therapeutic doses and are also predictive of response to FOLFOX therapy. These hypotheses were tested in colorectal cancer cell lines and a pilot clinical study. Four colorectal cancer cell lines were cultured with therapeutically relevant (100 µmol/L) or diagnostic microdose (1 µmol/L) concentrations of [14C]oxaliplatin. The C-14 label enabled quantification of oxaliplatin-DNA adduct level with accelerator mass spectrometry (AMS). Oxaliplatin-DNA adduct formation was correlated with oxaliplatin cytotoxicity for each cell line as measured by the MTT viability assay. Six colorectal cancer patients received by intravenous route a diagnostic microdose containing [14C]oxaliplatin prior to treatment, as well as a second [14C]oxaliplatin dose during FOLFOX chemotherapy, termed a "therapeutic dose." Oxaliplatin-DNA adduct levels from PBMC correlated significantly to mean tumor volume change of evaluable target lesions (5 of the 6 patients had measurable disease). Oxaliplatin-DNA adduct levels were linearly proportional between microdose and therapeutically relevant concentrations in cell culture experiments and patient samples, as was plasma pharmacokinetics, indicating potential utility of diagnostic microdosing.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/blood , Carbon Radioisotopes/analysis , Colorectal Neoplasms/pathology , DNA Adducts/blood , Liver Neoplasms/secondary , Oxaliplatin/blood , Apoptosis , Cell Proliferation , Colorectal Neoplasms/blood , Colorectal Neoplasms/drug therapy , Dose-Response Relationship, Drug , Fluorouracil/administration & dosage , Humans , Leucovorin/administration & dosage , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Liver Neoplasms/blood , Liver Neoplasms/drug therapy , Oxaliplatin/administration & dosage , Patient Selection , Pilot Projects , Prognosis , Tumor Cells, Cultured
9.
J Clin Oncol ; 37(18): 1578-1589, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31021656

ABSTRACT

PURPOSE: An ASCO provisional clinical opinion offers timely clinical direction to ASCO's membership after publication or presentation of potentially practice-changing data from major studies. This provisional clinical opinion addresses the role of treatment deintensification in the management of p16+ oropharyngeal cancer (OPC). CLINICAL CONTEXT: For patients with p16+ OPC, current treatment approaches are well established. In the good-prognosis subset of nonsmoking p16+ patients with early-stage disease, these treatments have been highly successful, albeit with significant associated acute and late toxicity. Deintensification of surgical, radiation, and medical treatment in an effort to reduce toxicity while preserving high survival rates is an appropriate therapeutic objective currently being explored in patients who are experiencing the best treatment results. However, careful delineation of this good-risk subset is essential. While the current eighth edition of the American Joint Committee on Cancer staging system is prognostically robust, it should not be interpreted as reason to alter therapeutic decisions or justify treatment deintensification. The development of transoral surgical techniques and the adoption of intensity-modulated radiation therapy planning have been transformative in disease management and suggest potentially beneficial approaches. Recent advances in systemic treatments have been notable. The optimal integration and modification of these modalities to ameliorate toxicity has not been defined and remains an important focus of current investigation. PROVISIONAL CLINICAL OPINION: The hypothesis that de-escalation of treatment intensity for patients with p16+ OPC can reduce long-term toxicity without compromising survival is compelling and necessitates careful study and the analysis of well-designed clinical trials before changing current treatment standards. Treatment deintensification for these patients should only be undertaken in a clinical trial. Additional information is available at www.asco.org/head-neck-cancer-guidelines .


Subject(s)
Oropharyngeal Neoplasms/therapy , Humans
10.
Kidney Cancer ; 3(1): 51-61, 2019 Feb 05.
Article in English | MEDLINE | ID: mdl-30854497

ABSTRACT

BACKGROUND: In renal cell carcinoma (RCC), angiopoietin (Ang) 2 is elevated at the time of progression on anti-vascular endothelial growth factor (VEGF) therapy and may contribute to resistance. OBJECTIVE: We tested trebananib, an Ang 1 and 2 neutralizing peptibody in patients with RCC progressing on anti-VEGF treatment. METHODS: Patients with measurable RCC progressing despite an anti-VEGF agent within 12 weeks, any number of prior treatments, and good PS were randomized to trebananib 15 mg/kg IV weekly without (Arm A) or with (Arm B) continuation of the prior anti-VEGF agent. The primary endpoint for each arm was tumor response (RECIST 1.1). Secondary endpoints included progression free survival and adverse events. RESULTS: Of 41 enrolled patients, 35 were eligible and started treatment (17 Arm A, 18 Arm B) with median age 60 (46-76) and 3 prior treatments (1-8). Four died prior to documented progression and 27 progressed as their first event. Both arms were stopped after interim analysis, 2 responses (11%; 95% C.I. 1-35%) were observed in Arm B. Median PFS of 2.7 (95% C.I. 2.3-4.7) months in Arm A and 5.2 (95% C.I. 2.7-10.8) months in Arm B did not support continued study. Common adverse events including fatigue, nausea, and increased creatinine were generally grade 1-2 and numerically higher in Arm B. The most common grade 3 or higher adverse events were hypertension and dyspnea. CONCLUSIONS: While tolerable, trebananib either without or with continued anti-VEGF therapy did not show promising activity in RCC patients who recently progressed on anti-VEGF therapy alone.

11.
Cancer Chemother Pharmacol ; 82(4): 723-732, 2018 10.
Article in English | MEDLINE | ID: mdl-30128950

ABSTRACT

PURPOSE: Tyrosine kinase inhibitors (TKI) that target MET signaling have shown promise in various types of cancer, including lung cancer. Combination strategies have been proposed and developed to increase their therapeutic index. Based on preclinical synergy between inhibition of MET and topoisomerase I, a phase I study was designed to explore the combination of topotecan with the MET TKI tivantinib. METHODS: Eligible patients with advanced solid malignancies for which there was no known effective treatment received topotecan at doses of 1.0-1.5 mg/m2/day for five consecutive days in 21-day cycles with continuous, oral tivantinib given at escalating doses of 120-360 mg orally twice daily. Pharmacokinetic analyses of tivantinib were included. Circulating tumor cells (CTC) were collected serially to identify peripheral changes in MET phosphorylation. RESULTS: The trial included 18 patients, 17 of whom received treatment. At the planned doses, the combination of topotecan and tivantinib was not tolerable due to thrombocytopenia and neutropenia. The addition of G-CSF to attenuate neutropenia did not improve tolerability. Greater tivantinib exposure, assessed through pharmacokinetic analysis, was associated with greater toxicity. No responses were seen. MET phosphorylation was feasible in CTC, but no changes were seen with therapy. CONCLUSIONS: The combination of topotecan and oral tivantinib was not tolerable in this patient population.


Subject(s)
Neoplasms/drug therapy , Neutropenia , Pyrrolidinones , Quinolines , Thrombocytopenia , Topotecan , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Drug Monitoring/methods , Female , Humans , Male , Middle Aged , Neoplasm Staging , Neoplasms/classification , Neoplasms/metabolism , Neoplasms/pathology , Neutropenia/chemically induced , Neutropenia/diagnosis , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , Pyrrolidinones/administration & dosage , Pyrrolidinones/adverse effects , Pyrrolidinones/pharmacokinetics , Quinolines/administration & dosage , Quinolines/adverse effects , Quinolines/pharmacokinetics , Thrombocytopenia/chemically induced , Thrombocytopenia/diagnosis , Topotecan/administration & dosage , Topotecan/adverse effects , Topotecan/pharmacokinetics , Treatment Failure , Treatment Outcome
12.
Thyroid ; 28(9): 1143-1152, 2018 09.
Article in English | MEDLINE | ID: mdl-29929451

ABSTRACT

BACKGROUND: In an era of rising differentiated thyroid cancer incidence, the rate and impact of neck reoperation may inform the intensity of earlier interventions and surveillance. This study sought to define predictors of neck reoperation and to assess its impact on survival. METHODS: Using the California Cancer Registry linked to the California Office of Statewide Health Planning and Development records, a retrospective cohort study was performed of 24,230 patients with total or near-total thyroidectomy for papillary or follicular thyroid cancer between 1991 and 2008 and follow-up through 2013. The primary outcome was neck reoperation 91 days to 5 years after the initial thyroid surgery. Using logistic and Cox proportional hazards regression, the impact of sociodemographics, tumor staging, and hospital thyroid cancer surgery volume on neck reoperation and survival was determined. RESULTS: Neck reoperation was identified in 1231 (5.1%) patients in increasing odds from 1991 to 2008. In multivariable models, male sex, papillary thyroid cancer, and advancing tumor stage were associated with neck reoperation. Among men, neck reoperation was associated with Asian/Pacific Islander (odds ratio [OR] = 1.44 [confidence interval (CI) 1.07-1.94]) race/ethnicity. Among women, neck reoperation was associated with younger age (15-34 years; OR = 1.50 [CI 1.17-1.92] versus ≥55 years), and Asian/Pacific Islander (OR = 1.24 [CI 1.02-1.51]) or Hispanic (OR = 1.20 [CI 1.00-1.44]) race/ethnicity. After controlling for baseline characteristics, neck reoperation predicted worse thyroid cancer-specific survival (hazard ratio = 4.26 [CI 3.50-5.19]). The effect differed between men and women, and was most pronounced among women who received radioiodine in initial treatment (hazard ratio = 8.32 [CI 6.14-11.27]). CONCLUSIONS: Neck reoperation is becoming increasingly frequent and is strongly predictive of mortality. Advancing tumor stage, Asian/Pacific Islander race/ethnicity, male sex, as well as younger age and Hispanic ethnicity among women predict a higher risk for neck reoperation and subsequent mortality, reflecting a higher risk of persistent or more biologically aggressive disease.


Subject(s)
Adenocarcinoma, Follicular/surgery , Thyroid Cancer, Papillary/surgery , Thyroid Neoplasms/surgery , Thyroidectomy , Adenocarcinoma, Follicular/mortality , Adenocarcinoma, Follicular/pathology , Adolescent , Adult , Age Factors , Female , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , Reoperation , Retrospective Studies , Risk Factors , Sex Factors , Thyroid Cancer, Papillary/mortality , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/mortality , Thyroid Neoplasms/pathology , Young Adult
13.
Cancer Chemother Pharmacol ; 81(5): 957-963, 2018 05.
Article in English | MEDLINE | ID: mdl-29520435

ABSTRACT

PURPOSE: The insulin-like growth factor (IGF) pathway is activated in hepatocarcinogenesis. Cixutumumab is a monoclonal antibody against human insulin-like growth factor-1 receptor (IGF-1R). Given the cross-talk between the IGF and VEGF pathways, we performed a phase I study of the combination of cixutumumab and sorafenib in hepatocellular cancer (HCC). METHODS: Eligible patients with no prior systemic therapy for advanced HCC and Child-Pugh A to B7 were treated with sorafenib 400 mg BID and escalating doses of cixutumumab (2, 4, or 6 mg/kg IV weekly) in a 3 + 3 design. Dose limiting toxicity (DLT) was defined as treatment-related grade 3 or 4 non-hematologic toxicity (except for a subset of manageable toxicities) or any grade 4 hematologic toxicities. RESULTS: In 21 patients enrolled, there were 3 DLTs; grade 3 hyperglycemia, grade 3 hypophosphatemia, and grade 5 peritonitis. The maximum tolerated dose of cixutumumab was 4 mg/kg IV weekly with standard dose sorafenib. Eighteen of 21 (86%) patients had grade 3 or above toxicities attributed to treatment. One patient also experienced grade 4 colonic perforation and grade 5 peritonitis. The median number of cycles completed was 4 (0-26). Of 16 patients evaluable for response, 81% achieved stable disease. The median progression free survival was 6.0 months (95% CI 3.6-undefined) and the median overall survival was 10.5 months (95% CI 7.1-undefined). CONCLUSIONS: While the combination of cixutumumab and sorafenib had a toxicity profile similar to that of sorafenib monotherapy, it manifested limited clinical efficacy in unselected patients with HCC.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Sorafenib/administration & dosage , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Dose-Response Relationship, Drug , Feasibility Studies , Female , Humans , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Maximum Tolerated Dose , Middle Aged , Progression-Free Survival , Sorafenib/adverse effects
14.
Methods Mol Biol ; 1765: 281-297, 2018.
Article in English | MEDLINE | ID: mdl-29589315

ABSTRACT

Concurrent with an expansion in the number of agents available for the treatment of advanced CRC, there has been an increase in our understanding of selection biomarkers to optimize the management of patients with this disease. For CRC patients being considered for anti-EGFR therapy, expanded RAS testing is the standard of care to determine the subset of patients who can benefit from cetuximab or panitumumab in conjunction with chemotherapy. A small fraction of patients have HER2 amplification where emerging data suggest treatment with drugs targeting this alteration. Although advanced CRC patients who harbor the BRAF V600E mutation have a poorer prognosis, they are eligible for combinatorial therapy targeting EGFR/BRAF or BRAF/MEK within the MAP kinase signaling pathway. Once primarily thought to be a negative prognostic marker, BRAF V600E mutation is now considered as a positive predictive factor with an opportunity for clinical intervention. A growing body of evidence also supports MSI testing as clinical benefits with immune checkpoint blockade by cancer immunotherapy have been demonstrated in MSI-high patients whose tumors exhibit high mutational burden. It has been established that UGT1A1*28 polymorphism is associated with irinotecan toxicity, but this test is rarely performed as the management strategy has not been identified. No established predictive biomarker for anti-VEGF therapy has yet to be discovered.It is becoming increasingly apparent that our growing understanding of biomarkers is revolutionizing and improving our strategies in the treatment of advanced CRC. Traditional nonselective cytotoxic chemotherapy is gradually being augmented and even in some cases supplanted by selective targeted agents based on our increasing understanding of tumor signaling and mechanism at the molecular level. The prospect of personalized medicine in directing treatment approaches that are optimally beneficial for patients brings tremendous excitement to the growing field of cancer therapeutics. As discussed in this chapter, the concurrent development of molecular biomarkers with new treatment strategies holds great promise of precision medicine in improving outcomes for patients with advanced CRC.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Colorectal Neoplasms/therapy , Immunotherapy/methods , Precision Medicine/methods , Antineoplastic Agents/pharmacology , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Humans , Microsatellite Instability , Molecular Targeted Therapy/methods , Mutation , Prognosis , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Treatment Outcome , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics
16.
Lancet Oncol ; 19(1): 101-114, 2018 01.
Article in English | MEDLINE | ID: mdl-29169877

ABSTRACT

BACKGROUND: EGFR antibodies have shown promise in patients with advanced non-small-cell lung cancer (NSCLC), particularly with squamous cell histology. We hypothesised that EGFR copy number by fluorescence in-situ hybridisation (FISH) can identify patients most likely to benefit from these drugs combined with chemotherapy and we aimed to explore the activity of cetuximab with chemotherapy in patients with advanced NSCLC who are EGFR FISH-positive. METHODS: We did this open-label, phase 3 study (SWOG S0819) at 277 sites in the USA and Mexico. We randomly assigned (1:1) eligible patients with treatment-naive stage IV NSCLC to receive paclitaxel (200 mg/m2; every 21 days) plus carboplatin (area under the curve of 6 by modified Calvert formula; every 21 days) or carboplatin plus paclitaxel and bevacizumab (15 mg/kg; every 21 days), either with cetuximab (250 mg/m2 weekly after loading dose; cetuximab group) or without (control group), stratified by bevacizumab treatment, smoking status, and M-substage using a dynamic-balancing algorithm. Co-primary endpoints were progression-free survival in patients with EGFR FISH-positive cancer and overall survival in the entire study population. We analysed clinical outcomes with the intention-to-treat principle and analysis of safety outcomes included patients who received at least one dose of study drug. This study is registered with ClinicalTrials.gov (number NCT00946712). FINDINGS: Between Aug 13, 2009, and May 30, 2014, we randomly assigned 1313 patients to the control group (n=657; 277 with bevacizumab and 380 without bevacizumab in the intention-to-treat population) or the cetuximab group (n=656; 283 with bevacizumab and 373 without bevacizumab in the intention-to-treat population). EGFR FISH was assessable in 976 patients and 400 patients (41%) were EGFR FISH-positive. The median follow-up for patients last known to be alive was 35·2 months (IQR 22·9-39·9). After 194 progression-free survival events in the cetuximab group and 198 in the control group in the EGFR FISH-positive subpopulation, progression-free survival did not differ between treatment groups (hazard ratio [HR] 0·92, 95% CI 0·75-1·12; p=0·40; median 5·4 months [95% CI 4·5-5·7] vs 4·8 months [3·9-5·5]). After 570 deaths in the cetuximab group and 593 in the control group, overall survival did not differ between the treatment groups in the entire study population (HR 0·93, 95% CI 0·83-1·04; p=0·22; median 10·9 months [95% CI 9·5-12·0] vs 9·2 months [8·7-10·3]). In the prespecified analysis of EGFR FISH-positive subpopulation with squamous cell histology, overall survival was significantly longer in the cetuximab group than in the control group (HR 0·58, 95% CI 0·36-0·86; p=0·0071), although progression-free survival did not differ between treatment groups in this subgroup (0·68, 0·46-1·01; p=0·055). Overall survival and progression-free survival did not differ among patients who were EGFR FISH non-positive with squamous cell histology (HR 1·04, 95% CI 0·78-1·40; p=0·77; and 1·02, 0·77-1·36; p=0·88 respectively) or patients with non-squamous histology regardless of EGFR FISH status (for EGFR FISH-positive 0·88, 0·68-1·14; p=0·34; and 0·99, 0·78-1·27; p=0·96; respectively; and for EGFR FISH non-positive 1·00, 0·85-1·17; p=0·97; and 1·03, 0·88-1·20; p=0·69; respectively). The most common grade 3-4 adverse events were decreased neutrophil count (210 [37%] in the cetuximab group vs 158 [25%] in the control group), decreased leucocyte count (103 [16%] vs 74 [20%]), fatigue (81 [13%] vs 74 [20%]), and acne or rash (52 [8%] vs one [<1%]). 59 (9%) patients in the cetuximab group and 31 (5%) patients in the control group had severe adverse events. Deaths related to treatment occurred in 32 (6%) patients in the cetuximab group and 13 (2%) patients in the control group. INTERPRETATION: Although this study did not meet its primary endpoints, prespecified subgroup analyses of patients with EGFR FISH-positive squamous-cell carcinoma cancers are encouraging and support continued evaluation of anti-EGFR antibodies in this subpopulation. FUNDING: National Cancer Institute and Eli Lilly and Company.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Cetuximab/administration & dosage , Lung Neoplasms/drug therapy , Paclitaxel/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carboplatin/adverse effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cetuximab/adverse effects , Disease Progression , Disease-Free Survival , ErbB Receptors/genetics , Female , Humans , In Situ Hybridization, Fluorescence , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Mexico , Middle Aged , Mutation , Paclitaxel/adverse effects , Risk Factors , Time Factors , Treatment Outcome , United States
17.
Cancer Epidemiol Biomarkers Prev ; 26(10): 1558-1563, 2017 10.
Article in English | MEDLINE | ID: mdl-28939585

ABSTRACT

Introduction: Both colon cancer and dementia are prevalent among the elderly and have a high risk of cooccurrence. Previous studies found that patients with dementia were treated less aggressively. In this study, we hypothesized that presence of preexisting dementia was associated with worse survival for stage III colon cancer patients, and that postoperative chemotherapy was on the causal pathway.Methods: We defined preexisting dementia in Surveillance Epidemiology and End Results Medicare data through either a formal diagnosis or a prescription for dementia drugs or both before the diagnosis of cancer. We applied multivariable Cox regression to estimate the effect of preexisting dementia on survival, adjusting for demographic factors, tumor characteristics, and receipt of chemotherapy. We assessed mediating effects in the context of the counterfactual framework using the accelerated failure time model.Results: There were 4,573 patients diagnosed with stage III colon cancer between 2007 and 2009 identified. A preexisting diagnosis of dementia significantly increased the risk of death by 45% (HR = 1.45, 95% CI: 1.29-1.63). Patients with either a formal diagnosis of dementia or a related prescription had significantly lower cause-specific survival than their cognitively healthy counterparts. Receipt of chemotherapy was a significant mediator on the causal pathway. The effect of presence of dementia was mediated by receipt of chemotherapy by 13% for preexisting dementia.Conclusions: Preexisting dementia is significantly associated with worse survival for stage III colon cancer patients, and its deleterious effect is partially explained by decreased likelihood of postoperative chemotherapy receipt.Impact: This is the first study that provides estimate of the mediating effect of diminished chemotherapy in patients with stage III colon cancer and dementia, simultaneously demonstrating the cancer-specific survival benefit of chemotherapy in the presence of dementia. Cancer Epidemiol Biomarkers Prev; 26(10); 1558-63. ©2017 AACR.


Subject(s)
Chemotherapy, Adjuvant/adverse effects , Dementia/etiology , Aged , Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , Female , Humans , Male , Neoplasm Staging , Survival Analysis
18.
Cancer Treat Res Commun ; 10: 21-26, 2017.
Article in English | MEDLINE | ID: mdl-28736761

ABSTRACT

BACKGROUND: Prior work identified the fibroblast growth factor (FGF) pathway as a mediator of resistance to anti-vascular endothelial growth factor (VEGF) therapy. We tested dovitinib, an inhibitor of both FGF and VEGF receptors, in patients progressing on anti-VEGF treatment. METHODS: Patients with measurable advanced colorectal or non-small cell lung cancer with progression despite anti-VEGF treatment within 56 days, good performance status and adequate organ function were eligible. A research tumor biopsy was followed by treatment with dovitinib 500 mg on a 5-day on/2-day off schedule for 28-day cycles. The primary endpoint of tumor response was evaluated every 2 cycles. Secondary endpoints included toxicity and 8-week disease control rate. Intratumor mRNA expression of angiogenic mediators was analyzed using a next generation sequencing based expression array. RESULTS: Ten patients treated previously with bevacizumab or ziv-aflibercept enrolled. The study closed with termination of dovitinib development. No responses were observed in 7 evaluable patients. The best response was stable disease in 1 patient. Common toxicities included gastrointestinal, metabolic, and biochemical derangements. All patients experienced at least one grade ≥ 3 treatment-related adverse event, most commonly fatigue, elevated GGT, and lymphopenia. Expression of multiple angiogenic mediators was common in tumors progressing on anti-VEGF therapy including high levels of FGFR1 and VEGFA. CONCLUSIONS: We found no evidence for the activity of dovitinib in patients who had recently progressed on anti-VEGF therapy and toxicities were significant. In tumors progressing despite anti-VEGF therapy, a multitude of pro-angiogenic mediators are expressed, including members of the FGF pathway.

19.
Gland Surg ; 5(4): 398-404, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27563561

ABSTRACT

BACKGROUND: Race, gender and socioeconomic disparities have been suggested to adversely influence stage at presentation, treatment options and outcomes in patients with cancer. Underserved minorities and those with a low socioeconomic status (SES) present with more advanced disease and have worse outcomes for differentiated thyroid cancer, but this relationship has never been evaluated for medullary thyroid cancer (MTC). METHODS: We used the California Cancer Registry (CCR) to evaluate disparities in the presentation, treatment and outcomes of patients diagnosed with MTC. RESULTS: We identified 634 patients with MTC diagnosed between 1988 and 2011. Almost everyone (85%) underwent thyroidectomy with 50% having a central lymph node dissection (CLND). There were no statistically significant differences by age, race or SES in mean tumor size or the proportion of patients diagnosed with localized disease, but men were diagnosed with larger tumors than women and were less likely to be diagnosed at a localized stage. Younger patients and women were more likely to be treated with a thyroidectomy. There were no statistically significant differences in surgical treatment by race or SES. Patients in the highest SES category had a better overall survival, but not disease specific survival, than those in the lowest SES (HR =0.3, CI =0.1-0.7). Patients treated with thyroidectomy had a better overall and cause specific survival, but the effect of CLND was not statistically significant after adjustment for other factors. CONCLUSIONS: In MTC, we did not find that race, gender or SES influenced the presentation, treatment or outcomes of patients with MTC. Men with MTC present with larger tumors and are less likely to have localized disease. Half of the MTC patients in California do not undergo a CLND at the time of thyroidectomy, which may suggest a lack appropriate care across a range of healthcare systems.

20.
Anticancer Res ; 36(6): 3013-7, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27272819

ABSTRACT

AIM: To evaluate Radiation Therapy Oncology Group planning target volume margins of 7-10 mm for radiation therapy in anorectal cancer using prone belly-board positioning without image guidance. PATIENTS AND METHODS: 375 kV cone beam computed tomography image-guided radiotherapy (IGRT) images from 20 patients treated for anorectal cancer were retrospectively analyzed for setup shifts. We calculated the total translational shift for each patient and the frequency with which setup shifts exceeded 7 mm and 10 mm. RESULTS: A total of 42.7% of treatments required shifts >7 mm and 20.8% >10 mm. The mean translational shift was 7.1 mm. 70% of patients experienced shifts ≥7 mm in 20% or more of their treatments and 25% of ≥10 mm in 20% or more of their treatments; 15% experienced shifts ≥10 mm in over half of their treatments. van Herk calculations suggest margins of 12.8 mm are necessary for accuracy without IGRT. CONCLUSION: IGRT using a prone belly board and 7-10 mm margins requires daily image-guidance to prevent planning target volume misses and ensure optimal dose delivery.


Subject(s)
Cone-Beam Computed Tomography/methods , Patient Positioning , Radiotherapy, Image-Guided/methods , Rectal Neoplasms/radiotherapy , Adult , Aged , Aged, 80 and over , Body Mass Index , Female , Humans , Male , Middle Aged , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...