Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Redox Biol ; 70: 103022, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38215546

ABSTRACT

PURPOSE: Cisplatin contributes to acute kidney injury (AKI) and chronic kidney disease (CKD) that occurs with greater frequency and severity in older patients. Age-associated cisplatin sensitivity in human fibroblasts involves increased mitochondrial superoxide produced by older donor cells. EXPERIMENTAL DESIGN: Young and old C57BL/6 J murine models of cisplatin-induced AKI and CKD were treated with the SOD mimetic avasopasem manganese to investigate the potential antioxidant and anti-inflammatory effects. Adverse event reporting from a phase 2 and a phase 3 randomized clinical trial (NCT02508389 and NCT03689712) conducted in patients treated with cisplatin and AVA was determined to have established the incidence and severity of AKI. RESULTS: Cisplatin-induced AKI and CKD occurred in all mice, however, was more pronounced in older mice. AVA reduced cisplatin-induced mortality, AKI, and CKD, in older animals. AVA also alleviated cisplatin-induced alterations in mitochondrial electron transport chain (ETC) complex activities and NADPH Oxidase 4 (NOX4) and inhibited the increased levels of the inflammation markers, TNFα, IL1, ICAM-1, and VCAM-1. Analysis of age-stratified subjects treated with cisplatin from clinical trials (NCT02508389, NCT03689712) also supported that the incidence of AKI increased with age and AVA reduced age-associated therapy-induced adverse events (AE), including hypomagnesemia, increased creatinine, and AKI. CONCLUSIONS: Older mice and humans are more susceptible to cisplatin-induced kidney injury, and treatment with AVA mitigates age-associated damage. Mitochondrial ETC and NOX4 activities represent sources of superoxide production contributing to cisplatin-induced kidney injury, and pro-inflammatory cytokine production and endothelial dysfunction may also be increased by superoxide formation.


Subject(s)
Acute Kidney Injury , Organometallic Compounds , Renal Insufficiency, Chronic , Humans , Mice , Animals , Aged , Cisplatin/pharmacology , Antioxidants/pharmacology , Antioxidants/therapeutic use , Superoxides , Mice, Inbred C57BL , Kidney , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Anti-Inflammatory Agents/pharmacology
2.
Expert Opin Investig Drugs ; 32(6): 463-470, 2023.
Article in English | MEDLINE | ID: mdl-37365149

ABSTRACT

INTRODUCTION: Oral mucositis (OM) remains a significant, highly symptomatic, disruptive side effect of radiation and concomitant chemoradiation therapy used for the treatment of squamous cell cancers of the head and neck. Despite its clinical and economic burden, implementation of an effective intervention has been elusive. AREAS COVERED: Increased understanding of the complexity of the biological basis for its pathogenesis has yielded potential druggable targets such as the mitigation of superoxide formation and oxidative stress. Avasopasem manganese is a selective superoxide dismutase mimetic being developed by Galera Therapeutics, which recently submitted a New Drug Application (NDA) to the FDA for a severe OM indication. This review describes the preclinical and clinical studies which led to, and supported the NDA, and assesses the potential utility of avasopasem clinically. EXPERT OPINION: Avasopasem manganese appears to effectively mitigate severe OM associated with concomitant chemoradiation used in the treatment of head and neck cancers, as well as cisplatin-associated renal toxicity in the absence of impairing tumor response.


Subject(s)
Chemoradiotherapy , Organometallic Compounds , Stomatitis , Humans , Stomatitis/drug therapy , Stomatitis/etiology , Superoxide Dismutase , Chemoradiotherapy/adverse effects , Drug Approval , United States Food and Drug Administration , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Organometallic Compounds/pharmacology
3.
Int J Radiat Oncol Biol Phys ; 115(4): 1010, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36822775
4.
Int J Radiat Oncol Biol Phys ; 114(3): 416-421, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35724774

ABSTRACT

PURPOSE: Avasopasem manganese (GC4419), an investigational selective dismutase mimetic radioprotector, reduced duration, incidence, and severity of severe oral mucositis (World Health Organization grade 3-4) in a phase 2b, randomized, double-blind trial of patients receiving concurrent cisplatin (cis) and radiation therapy (RT) for head and neck cancer. We report the secondary endpoints of final 1- and 2-year tumor outcomes and exploratory data on trismus and xerostomia. METHODS AND MATERIALS: Patients with locally advanced oral cavity or oropharynx cancer to be treated with definitive or postop cis and RT were randomized to 1 of 3 arms: 30 mg avasopasem, 90 mg avasopasem, or placebo. Pairwise comparisons of Kaplan-Meier estimates (each active arm separately vs placebo) were made for overall survival, progression-free survival, locoregional control, and distant metastasis-free survival. Xerostomia and trismus data were collected at each follow-up visit and analyzed for trends by post-RT timepoint and treatment group. RESULTS: At a median follow-up for the entire cohort of 25.5 months (25th-75th percentile, 24.6-26.2 months; range, 0.2-31.9 months), Kaplan-Meier estimates of 1- and 2-year overall survival, progression-free survival, locoregional control, and distant metastasis-free survival were not statistically different. No trends were apparent in xerostomia or trismus data. CONCLUSIONS: Avasopasem does not lead to statistically different tumor control outcomes when used concurrently with cis and RT for head and neck cancer. There was no detectable effect on trismus or xerostomia.


Subject(s)
Head and Neck Neoplasms , Stomatitis , Xerostomia , Cisplatin/adverse effects , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Humans , Organometallic Compounds , Stomatitis/etiology , Stomatitis/prevention & control , Trismus/etiology , Trismus/prevention & control , Xerostomia/etiology , Xerostomia/prevention & control
5.
J Clin Med ; 11(6)2022 Mar 16.
Article in English | MEDLINE | ID: mdl-35329988

ABSTRACT

MR-guided adaptive radiotherapy (MRgART) provides opportunities to benefit patients through enhanced use of advanced imaging during treatment for many patients with various cancer treatment sites. This novel technology presents many new challenges which vary based on anatomic treatment location, technique, and potential changes of both tumor and normal tissue during treatment. When introducing new treatment sites, considerations regarding appropriate patient selection, treatment planning, immobilization, and plan-adaption criteria must be thoroughly explored to ensure adequate treatments are performed. This paper presents an institution's experience in developing a MRgART program for a 1.5T MR-linac for the first 234 patients. The paper suggests practical treatment workflows and considerations for treating with MRgART at different anatomical sites, including imaging guidelines, patient immobilization, adaptive workflows, and utilization of bolus.

6.
Lancet Oncol ; 23(2): 259-269, 2022 02.
Article in English | MEDLINE | ID: mdl-35038433

ABSTRACT

BACKGROUND: Trastuzumab is a monoclonal antibody against HER2 (also known as ERBB2). The primary objective of the NRG Oncology/RTOG-1010 trial was to establish whether trastuzumab improves disease-free survival when combined with trimodality treatment (paclitaxel plus carboplatin and radiotherapy, followed by surgery) for patients with untreated HER2-overexpressing oesophageal adenocarcinoma. METHODS: NRG Oncology/RTOG-1010 was an open label, randomised, phase 3 trial for which patients were accrued from 111 NRG-affiliated institutions in the USA. Eligible patients were adults (aged ≥18 years) with newly diagnosed pathologically confirmed oesophageal adenocarcinoma, American Joint Committee on Cancer 7th edition T1N1-2 or T2-3N0-2 stage disease, and a Zubrod performance status of 0-2. Patients were stratified by adenopathy (no vs yes [coeliac absent] vs yes [coeliac present ≤2 cm]) and randomly assigned (1:1) to receive weekly intravenous paclitaxel (50 mg/m2 intravenously over 1 h) and carboplatin (area under the curve 2, intravenously over 30-60 min) for 6 weeks with radiotherapy 50·4 Gy in 28 fractions (chemoradiotherapy) followed by surgery, with or without intravenous trastuzumab (4 mg/kg in week one, 2 mg/kg per week for 5 weeks during chemoradiotherapy, 6 mg/kg once presurgery, and 6 mg/kg every 3 weeks for 13 treatments starting 21-56 days after surgery). The primary endpoint, disease-free survival, was defined as the time from randomisation to death or first of locoregional disease persistence or recurrence, distant metastases, or second primary malignancy. Analyses were done by modified intention to treat. This study is registered with Clinicaltrials.gov, NCT01196390; it is now closed and in follow-up. FINDINGS: 606 patients were entered for HER2 assessment from Dec 30, 2010 to Nov 10, 2015, and 203 eligible patients who were HER2-positive were enrolled and randomly assigned to chemoradiotherapy plus trastuzumab (n=102) or chemoradiotherapy alone (n=101). Median duration of follow-up was 2·8 years (IQR 1·4-5·7). Median disease-free survival was 19·6 months (95% CI 13·5-26·2) with chemoradiotherapy plus trastuzumab compared with 14·2 months (10·5-23·0) for chemoradiotherapy alone (hazard ratio 0·99 [95% CI 0·71-1·39], log-rank p=0·97). Grade 3 treatment-related adverse events occurred in 41 (43%) of 95 patients in the chemoradiotherapy plus trastuzumab group versus 52 (54%) of 96 in the chemoradiotherapy group and grade 4 events occurred in 20 (21%) versus 21 (22%). The most common grade 3 or worse treatment-related adverse events for both groups were haematological (53 [56%] of 95 patients in the chemoradiotherapy plus trastuzumab group vs 55 [57%] of 96 patients in the chemotherapy group) or gastrointestinal disorders (28 [29%] vs 20 [21 %]). 34 (36%) of 95 patients in the chemoradiotherapy plus trastuzumab group and 27 (28%) of 96 patients in the chemoradiotherapy only group had treatment-related serious adverse events. There were eight treatment-related deaths: five (5%) of 95 patients in the chemoradiotherapy plus trastuzumab group (bronchopleural fistula, oesophageal anastomotic leak, lung infection, sudden death, and death not otherwise specified), and three (3%) of 96 in the chemoradiotherapy group (two multiorgan failure and one sepsis). INTERPRETATION: The addition of trastuzumab to neoadjuvant chemoradiotherapy for HER2-overexpressing oesophageal cancer was not effective. Trastuzumab did not lead to increased toxicities, suggesting that future studies combining it with or using other agents targeting HER2 in oesophageal cancer are warranted. FUNDING: National Cancer Institute and Genentech.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Esophageal Neoplasms/drug therapy , Receptor, ErbB-2/analysis , Trastuzumab/therapeutic use , Adenocarcinoma/chemistry , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Carboplatin/administration & dosage , Chemoradiotherapy , Esophageal Neoplasms/chemistry , Esophageal Neoplasms/mortality , Female , Humans , Male , Middle Aged , Paclitaxel/administration & dosage , Trastuzumab/adverse effects
7.
Antioxidants (Basel) ; 10(9)2021 Aug 24.
Article in English | MEDLINE | ID: mdl-34572961

ABSTRACT

Cisplatin is a chemotherapy agent commonly used to treat a wide variety of cancers. Despite the potential for both severe acute and chronic side effects, it remains a preferred therapeutic option for many malignancies due to its potent anti-tumor activity. Common cisplatin-associated side-effects include acute kidney injury (AKI) and chronic kidney disease (CKD). These renal injuries may cause delays and potentially cessation of cisplatin therapy and have long-term effects on renal function reserve. Thus, developing mechanism-based interventional strategies that minimize cisplatin-associated kidney injury without reducing efficacy would be of great benefit. In addition to its action of cross-linking DNA, cisplatin has been shown to affect mitochondrial metabolism, resulting in mitochondrially derived reactive oxygen species (ROS). Increased ROS formation in renal proximal convoluted tubule cells is associated with cisplatin-induced AKI and CKD. We review the mechanisms by which cisplatin may induce AKI and CKD and discuss the potential of mitochondrial superoxide dismutase mimetics to prevent platinum-associated nephrotoxicity.

8.
Radiat Res ; 196(2): 213-224, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34087943

ABSTRACT

Ketogenic diets (KD) are high in fat and low in carbohydrates, forcing cells to utilize mitochondrial fatty acid oxidation for energy production. Since cancer cells demonstrate increased mitochondrial oxidative stress relative to normal cells, we hypothesized that a KD may selectively enhance metabolic oxidative stress in head and neck cancer cells, sensitizing them to radiation and platinum-based chemotherapy without causing increased toxicity in surrounding normal tissues. This hypothesis was tested in preclinical murine xenografts and in a phase 1 clinical trial (NCT01975766). In this study, mice bearing human head and neck cancer xenografts (FaDu) were fed either standard mouse chow or KetoCal® KD (90% fat, 8% carbohydrate, 2% protein) and exposed to ionizing radiation. Tumors were harvested from mice to test for glutathione, a biomarker of oxidative stress. In parallel, patients with locally advanced head and neck cancer were enrolled in a phase 1 clinical trial where they consumed KD and received radiation with concurrent platinum-based chemotherapy. Subjects consumed KetoCal KD via percutaneous endoscopic gastrostomy (PEG) tube and were also allowed to orally consume water, sugar-free drinks, and foods approved by a dietitian. Oxidative stress markers including protein carbonyls and total glutathione were assessed in patient blood samples both pre-KD and while consuming the KD. Mice bearing FaDu xenografts that received radiation and KD demonstrated a slight improvement in tumor growth rate and survival compared to mice that received radiation alone; however a variation in responses was seen dependent on the fatty acid composition of the diet. In the phase 1 clinical trial, a total of twelve patients were enrolled in the study. Four patients completed five weeks of the KD as per protocol (with variance in compliance). Eight patients did not tolerate the diet with concurrent radiation and platinum-chemotherapy (5 were patient decision and 3 were removed from study due to toxicity). The median number of days consuming a KD in patients who did not complete the study was 5.5 (range: 2-8 days). Reasons for discontinuation included "stress of diet compliance" (1 patient), grade 2 nausea (3 patients), and grade 3 fatigue (1 patient). Three patients were removed from the trial due to dose-limiting toxicities including: grade 4 hyperuricemia (2 patients) and grade 3 acute pancreatitis (1 patient). Median weight loss was 2.95% for the KD-tolerant group and 7.92% for patients who did not tolerate the diet. In conclusion, the ketogenic diet shows promise as a treatment combined with radiation in preclinical mouse head and neck cancer xenografts. A phase 1 clinical trial evaluating the safety and tolerability of KD demonstrated difficulty with diet compliance when combined with standard-of-care radiation therapy and cisplatin chemotherapy.


Subject(s)
Diet, Ketogenic/methods , Squamous Cell Carcinoma of Head and Neck/diet therapy , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/radiotherapy , 3-Hydroxyacyl CoA Dehydrogenases/drug effects , 3-Hydroxyacyl CoA Dehydrogenases/radiation effects , Acetyl-CoA C-Acyltransferase/drug effects , Acetyl-CoA C-Acyltransferase/radiation effects , Adult , Aged , Animals , Carbon-Carbon Double Bond Isomerases/drug effects , Carbon-Carbon Double Bond Isomerases/radiation effects , Chemoradiotherapy/adverse effects , Diet, Ketogenic/adverse effects , Enoyl-CoA Hydratase/drug effects , Enoyl-CoA Hydratase/radiation effects , Female , Heterografts , Humans , Male , Mice , Middle Aged , Mitochondria/drug effects , Mitochondria/radiation effects , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Racemases and Epimerases/drug effects , Racemases and Epimerases/radiation effects , Radiation, Ionizing , Squamous Cell Carcinoma of Head and Neck/pathology , Stress, Physiological/drug effects , Stress, Physiological/radiation effects
10.
Laryngoscope ; 131(5): 1019-1025, 2021 05.
Article in English | MEDLINE | ID: mdl-32846018

ABSTRACT

OBJECTIVE: To analyze the clinicodemographic characteristics and treatment outcomes of patients receiving postoperative radiation therapy (PORT) at a different treatment facility rather than the initial surgical facility for head and neck cancer. STUDY DESIGN: Retrospective cohort analysis. METHODS: Utilizing the National Cancer Data Base, 2004 to 2015, patients with a diagnosis of oral cavity/oropharyngeal, hypopharyngeal, and laryngeal squamous cell carcinoma were studied. Multivariate analysis was completed with multivariate regression and Cox proportional hazard model, and survival outcomes were examined using Kaplan-Meier analysis. RESULTS: A total of 15,181 patients who had surgery for a head and neck cancer at an academic/research center were included in the study population. Of the study population, 4,890 (32.2%) patients completed PORT at a different treatment facility. Treatment at a different facility was more common among patients who were ≥65 years old, white, Medicare recipients, those with a greater distance between residence and surgical treatment facility, and with lower income within area of residence (each P < .05). Overall survival was worse in patients completing PORT at a different treatment facility versus at the institution where surgery was completed (61.9% vs. 66.4%; P = .002). CONCLUSIONS: PORT at a different facility was more common in older individuals, Medicare recipients, those with greater distance to travel, and lower-income individuals. Completing PORT outside the hospital where surgery was performed was associated with inferior survival outcomes among head and neck cancer patients. LEVEL OF EVIDENCE: 3 Laryngoscope, 131:1019-1025, 2021.


Subject(s)
Head and Neck Neoplasms/therapy , Otorhinolaryngologic Surgical Procedures/statistics & numerical data , Squamous Cell Carcinoma of Head and Neck/therapy , Academic Medical Centers/statistics & numerical data , Aged , Clinical Decision-Making , Female , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/pathology , Humans , Income/statistics & numerical data , Kaplan-Meier Estimate , Male , Medicare/statistics & numerical data , Middle Aged , Neoplasm Staging , Patient Selection , Radiotherapy, Adjuvant/statistics & numerical data , Retrospective Studies , Squamous Cell Carcinoma of Head and Neck/diagnosis , Squamous Cell Carcinoma of Head and Neck/mortality , Squamous Cell Carcinoma of Head and Neck/pathology , Treatment Outcome , United States/epidemiology
11.
Radiat Res ; 194(2): 124-132, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32845986

ABSTRACT

Patients diagnosed with metastatic sarcoma have limited options for achieving both local and distant tumor control. While SBRT can achieve local control, distant response rates remain low. There is limited evidence demonstrating the safety and efficacy for combining SBRT with concurrent PD-1 checkpoint blockade in metastatic sarcoma. In this prospective case-series, we examined five patients with metastatic sarcoma on pembrolizumab treated concurrently with SBRT from July 1, 2016-October 30, 2018. Acute and chronic toxicity were recorded using Common Terminology Criteria for Adverse Events (CTCAE, version 5.0). SBRT-treated tumor control was assessed using Response Evaluation Criteria in Solid Tumors (RECIST version 1.1). With median follow-up of 14.9 months, three patients with undifferentiated pleomorphic sarcoma, one with intimal, and one with chondroblastic osteosarcoma received SBRT with concurrent pembrolizumab to 10 sites of metastatic disease. No grade 5 toxicities were observed. There was a single incidence of transient grade 4 lymphopenia which resolved without intervention. Grade 3 toxicities included anemia, thrombocytopenia, lymphopenia and colitis. One tumor demonstrated local progression after SBRT, and all others remained stable or with response. In conclusion, combining SBRT with PD-1 inhibition appeared to be safe in this patient population. Expected high rates of treated-tumor local control after SBRT were observed. Two of five patients demonstrated either enhanced local tumor regression, or possible abscopal effect.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Programmed Cell Death 1 Receptor/immunology , Radiosurgery , Sarcoma/therapy , Adult , Aged , Antibodies, Monoclonal, Humanized/immunology , Combined Modality Therapy , Humans , Middle Aged , Neoplasm Metastasis , Sarcoma/immunology , Sarcoma/pathology , Sarcoma/radiotherapy , Treatment Outcome
13.
J Clin Oncol ; 37(34): 3256-3265, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31618127

ABSTRACT

PURPOSE: Oral mucositis (OM) remains a common, debilitating toxicity of radiation therapy (RT) for head and neck cancer. The goal of this phase IIb, multi-institutional, randomized, double-blind trial was to compare the efficacy and safety of GC4419, a superoxide dismutase mimetic, with placebo to reduce the duration, incidence, and severity of severe OM (SOM). PATIENTS AND METHODS: A total of 223 patients (from 44 institutions) with locally advanced oral cavity or oropharynx cancer planned to be treated with definitive or postoperative intensity-modulated RT (IMRT; 60 to 72 Gy [≥ 50 Gy to two or more oral sites]) plus cisplatin (weekly or every 3 weeks) were randomly assigned to receive 30 mg (n = 73) or 90 mg (n = 76) of GC4419 or to receive placebo (n = 74) by 60-minute intravenous administration before each IMRT fraction. WHO grade of OM was assessed biweekly during IMRT and then weekly for up to 8 weeks after IMRT. The primary endpoint was duration of SOM tested for each active dose level versus placebo (intent-to-treat population, two-sided α of .05). The National Cancer Institute Common Terminology Criteria for Adverse Events, version 4.03, was used for adverse event grading. RESULTS: Baseline patient and tumor characteristics as well as treatment delivery were balanced. With 90 mg GC4419 versus placebo, SOM duration was significantly reduced (P = .024; median, 1.5 v 19 days). SOM incidence (43% v 65%; P = .009) and severity (grade 4 incidence, 16% v 30%; P = .045) also were improved. Intermediate improvements were seen with the 30-mg dose. Safety was comparable across arms, with no significant GC4419-specific toxicity nor increase of known toxicities of IMRT plus cisplatin. The 2-year follow-up for tumor outcomes is ongoing. CONCLUSION: GC4419 at a dose of 90 mg produced a significant, clinically meaningful reduction of SOM duration, incidence, and severity with acceptable safety. A phase III trial (ROMAN; ClinicalTrials.gov identifier: NCT03689712) has begun.


Subject(s)
Antineoplastic Agents/administration & dosage , Chemoradiotherapy, Adjuvant/adverse effects , Chemoradiotherapy/adverse effects , Cisplatin/administration & dosage , Mouth Neoplasms/drug therapy , Organometallic Compounds/therapeutic use , Oropharyngeal Neoplasms/drug therapy , Radiation Injuries/prevention & control , Radiation-Protective Agents/therapeutic use , Radiotherapy, Intensity-Modulated/adverse effects , Stomatitis/prevention & control , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Double-Blind Method , Female , Humans , Incidence , Male , Middle Aged , Mouth Neoplasms/epidemiology , Mouth Neoplasms/pathology , Ontario , Oropharyngeal Neoplasms/epidemiology , Oropharyngeal Neoplasms/pathology , Radiation Injuries/diagnosis , Radiation Injuries/epidemiology , Radiation-Protective Agents/adverse effects , Risk Factors , Severity of Illness Index , Stomatitis/diagnosis , Stomatitis/epidemiology , Time Factors , Treatment Outcome , United States
15.
Cancer Med ; 8(6): 2730-2739, 2019 06.
Article in English | MEDLINE | ID: mdl-30968604

ABSTRACT

BACKGROUND: High-dose cisplatin (Cis) is a preferred systemic agent for concurrent chemoradiation (CRT) in locally advanced head and neck squamous cell cancer (LAHNSCC) patients. As some patients are unable to tolerate Cis, this study compares the toxicity and efficacy of weekly cisplatin-paclitaxel (CP) regimen with Cis. METHODS: Patients with LAHNSCC receiving definitive chemoradiation either with Cis (Cisplatin-100 mg/m2 q3w x 3) or CP (Cisplatin-20 mg/m2 ; Paclitaxel-30 mg/m2 qw x7) were included. RESULTS: Cis and CP groups were comprised of 114 and 111 subjects, respectively. Complete response for Cis versus CP groups was 88% versus 88%, respectively. Median follow-up for the study was 58.5 months. After adjusting for potential treatment selection bias, no significant differences were evident between Cis and CP groups for overall survival (hazard ratios [HR] 0.85, 95% CI 0.59-1.21, P = 0.36), progression free survival (HR 0.88, 95% CI 0.62-1.24, P = 0.46), locoregional control (HR 0.77, 95% CI 0.52-1.15, P = 0.21), and distant control (HR 0.87, 95% CI 0.61-1.23, P = 0.42). Patients in the CP group had less acute and chronic toxicities. CONCLUSIONS: Weekly CP regimen can serve as an alternative systemic therapy with radiation in patients with LAHNSCC who are not fit for Cis.


Subject(s)
Antineoplastic Agents/administration & dosage , Cisplatin/administration & dosage , Radiation-Sensitizing Agents/administration & dosage , Squamous Cell Carcinoma of Head and Neck/therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemoradiotherapy/methods , Combined Modality Therapy , Comorbidity , Female , Humans , Male , Middle Aged , Neoplasm Staging , Squamous Cell Carcinoma of Head and Neck/diagnosis , Squamous Cell Carcinoma of Head and Neck/mortality
16.
Tomography ; 5(1): 161-169, 2019 03.
Article in English | MEDLINE | ID: mdl-30854454

ABSTRACT

Radiomics is an image analysis approach for extracting large amounts of quantitative information from medical images using a variety of computational methods. Our goal was to evaluate the utility of radiomic feature analysis from 18F-fluorothymidine positron emission tomography (FLT PET) obtained at baseline in prediction of treatment response in patients with head and neck cancer. Thirty patients with advanced-stage oropharyngeal or laryngeal cancer, treated with definitive chemoradiation therapy, underwent FLT PET imaging before treatment. In total, 377 radiomic features of FLT uptake and feature variants were extracted from volumes of interest; these features variants were defined by either the primary tumor or the total lesion burden, which consisted of the primary tumor and all FLT-avid nodes. Feature variants included normalized measurements of uptake, which were calculated by dividing lesion uptake values by the mean uptake value in the bone marrow. Feature reduction was performed using clustering to remove redundancy, leaving 172 representative features. Effects of these features on progression-free survival were modeled with Cox regression and P-values corrected for multiple comparisons. In total, 9 features were considered significant. Our results suggest that smaller, more homogenous lesions at baseline were associated with better prognosis. In addition, features extracted from total lesion burden had a higher concordance index than primary tumor features for 8 of the 9 significant features. Furthermore, total lesion burden features showed lower interobserver variability.


Subject(s)
Head and Neck Neoplasms/diagnostic imaging , Head and Neck Neoplasms/therapy , Squamous Cell Carcinoma of Head and Neck/diagnostic imaging , Squamous Cell Carcinoma of Head and Neck/therapy , Adult , Aged , Chemoradiotherapy/methods , Dideoxynucleosides , Female , Head and Neck Neoplasms/pathology , Humans , Image Interpretation, Computer-Assisted/methods , Male , Middle Aged , Neoplasm Staging , Observer Variation , Positron-Emission Tomography/methods , Prognosis , Prospective Studies , Radiopharmaceuticals , Squamous Cell Carcinoma of Head and Neck/pathology , Treatment Outcome
17.
Cancer Nurs ; 42(3): 218-228, 2019.
Article in English | MEDLINE | ID: mdl-29649082

ABSTRACT

BACKGROUND: Individuals receiving radiation for head and neck cancer (HNC) often develop painful oral mucositis that impairs function, possibly leading to feeding tubes, hospitalization, and treatment delays. Although pharmacologic medications provide some relief, many report inadequate analgesia and adverse effects. Transcutaneous electrical nerve stimulation (TENS) is a safe, nonpharmacologic intervention; it decreases pain and analgesics and improves function, yet no studies examined TENS for HNC. OBJECTIVE: The aim of this study was to examine the efficacy of TENS for pain and function in HNC patients. METHODS: This study used a randomized, double-blinded crossover design; participants received 3 TENS treatments during weeks 4 to 6 of radiation: active, placebo, and no TENS over the temporomandibular joint and upper cervical region. Pain (McGill Pain Questionnaire, visual analog scale [VAS] resting and function), function (mouth opening, tongue movement, speaking), fatigue (VAS), and treatment effectiveness (VAS) were assessed before and after TENS at 3 visits. RESULTS: Resting pain (McGill Pain Questionnaire and VAS) decreased significantly more after active TENS than placebo or no TENS; changes in function and pain with function did not differ between conditions. Active TENS decreased fatigue significantly more than no TENS and was rated as more effective than placebo TENS. CONCLUSION: Transcutaneous electrical nerve stimulation improves pain in HNC patients receiving radiation but not function or pain with function relative to placebo or no TENS. IMPLICATIONS FOR PRACTICE: Transcutaneous electrical nerve stimulation may be a viable tool for radiation-induced HNC pain to complement pharmacologic approaches. This nonpharmacologic intervention could decrease the debilitating effects of radiation and analgesics, and improve quality of life. Clinical trials should examine the effects and safety of repeated, daily TENS in HNC patients receiving radiation.


Subject(s)
Head and Neck Neoplasms/complications , Head and Neck Neoplasms/therapy , Pain Management/methods , Transcutaneous Electric Nerve Stimulation , Aged , Cross-Over Studies , Double-Blind Method , Female , Humans , Male , Middle Aged , Pain Measurement , Pilot Projects , Treatment Outcome
18.
Semin Radiat Oncol ; 29(1): 72-80, 2019 01.
Article in English | MEDLINE | ID: mdl-30573187

ABSTRACT

Symptomatic normal tissue injury is a common side effect following definitive therapeutic radiation and chemotherapy treatment for a variety of malignancies. These cancer therapy related toxicities may occur acutely during treatment resulting in reduced or missed therapy agent administration or after the completion of therapy resulting in significant chronic morbidities that significantly diminish patient quality of life. Radiation and chemotherapy induce the formation of reactive oxygen species (ROS) both in normal tissues and tumor cells. One type of ROS common to both chemotherapy and radiation therapy is the formation of superoxide (O2•-). Fortunately, due to metabolic differences between cancer and normal cell metabolism, as well as improved targeting techniques, ROS generation following radiation and chemotherapy is generally greater in cancer cells compared to normal tissues. However, the levels of ROS generated in normal tissues are capable of inducing significant toxicity. Thus, several groups are focusing on metabolism-based approaches to mitigate normal tissue effects occurring both during and following cancer therapy. This review will summarize the most current preclinical and clinical data available demonstrating the efficacy of small molecule, superoxide dismutase mimetics in minimizing radiation and chemotherapy-induced normal tissue injury, resulting in enhanced patient outcomes.


Subject(s)
Biomimetic Materials/pharmacology , Neoplasms/radiotherapy , Radiation-Protective Agents/pharmacology , Radiation-Sensitizing Agents/pharmacology , Superoxide Dismutase/pharmacology , Humans , Neoplasms/metabolism , Reactive Oxygen Species/metabolism
20.
Int J Radiat Oncol Biol Phys ; 100(2): 427-435, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29174131

ABSTRACT

PURPOSE: To assess the safety of the superoxide dismutase mimetic GC4419 in combination with radiation and concurrent cisplatin for patients with oral cavity or oropharyngeal cancer (OCC) and to assess the potential of GC4419 to reduce severe oral mucositis (OM). PATIENTS AND METHODS: Patients with locally advanced OCC treated with definitive or postoperative intensity modulated radiation therapy (IMRT) plus cisplatin received GC4419 by 60-minute intravenous infusion, ending <60 minutes before IMRT, Monday through Friday for 3 to 7 weeks, in a dose and duration escalation study. Oral mucositis was assessed twice weekly during and weekly after IMRT. RESULTS: A total of 46 patients received GC4419 in 11 separate dosing and duration cohorts: dose escalation occurred in 5 cohorts receiving 15 to 112 mg/d over 3 weeks (n=20), duration escalation in 3 cohorts receiving 112 mg/d over 4 to 6 weeks (n=12), and then 3 additional cohorts receiving 30 or 90 mg/d over 6 to 7 weeks (n=14). A maximum tolerated dose was not reached. One dose-limiting toxicity (grade 3 gastroenteritis and vomiting with hyponatremia) occurred in each of 2 separate cohorts at 112 mg. Nausea/vomiting and facial paresthesia during infusion seemed to be GC4419 dose-related. Severe OM occurred through 60 Gy in 4 of 14 patients (29%) dosed for 6 to 7 weeks, with median duration of only 2.5 days. CONCLUSIONS: The safety of GC4419 concurrently with chemoradiation for OCC was acceptable. Toxicities included nausea/vomiting and paresthesia. Doses of 30 and 90 mg/d administered for 7 weeks were selected for further study. In an exploratory analysis, severe OM seemed less frequent and briefer than expected.


Subject(s)
Chemoradiotherapy/adverse effects , Mouth Neoplasms/therapy , Oropharyngeal Neoplasms/therapy , Radiotherapy, Intensity-Modulated/adverse effects , Stomatitis/prevention & control , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Stomatitis/etiology , Superoxide Dismutase/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...