scholarly journals Patients with Advanced Head and Neck Cancers Have Similar Progression-Free Survival on Phase I Trials and Their Last Food and Drug Administration–Approved Treatment

2010 ◽  
Vol 16 (15) ◽  
pp. 4031-4037 ◽  
Author(s):  
Ignacio Garrido-Laguna ◽  
Filip Janku ◽  
Gerald S. Falchook ◽  
Siqing Fu ◽  
David S. Hong ◽  
...  
2018 ◽  
Vol 36 (11) ◽  
pp. 1064-1072 ◽  
Author(s):  
Vanita Noronha ◽  
Amit Joshi ◽  
Vijay Maruti Patil ◽  
Jaiprakash Agarwal ◽  
Sarbani Ghosh-Laskar ◽  
...  

Purpose Chemoradiation with cisplatin 100 mg/m2 given once every 3 weeks is the standard of care in locally advanced head and neck squamous cell cancer (LAHNSCC). Increasingly, low-dose once-a-week cisplatin is substituted because of perceived lower toxicity and convenience. However, there is no level 1 evidence of comparable efficacy to cisplatin once every 3 weeks. Patients and Methods In this phase III randomized trial, we assessed the noninferiority of cisplatin 30 mg/m2 given once a week compared with cisplatin 100 mg/m2 given once every 3 weeks, both administered concurrently with curative intent radiotherapy in patients with LAHNSCC. The primary end point was locoregional control (LRC); secondary end points included toxicity, compliance, response, progression-free survival, and overall survival. Results Between 2013 and 2017, we randomly assigned 300 patients, 150 to each arm. Two hundred seventy-nine patients (93%) received chemoradiotherapy in the adjuvant setting. At a median follow-up of 22 months, the estimated cumulative 2-year LRC rate was 58.5% in the once-a-week arm and 73.1% in the once-every-3-weeks arm, leading to an absolute difference of 14.6% (95% CI, 5.7% to 23.5%); P = .014; hazard ratio (HR), 1.76 (95% CI, 1.11 to 2.79). Acute toxicities of grade 3 or higher occurred in 71.6% of patients in the once-a-week arm and in 84.6% of patients in the once-every-3-weeks arm ( P = .006). Estimated median progression-free survival in the once-a-week arm was 17.7 months (95% CI, 0.42 to 35.05 months) and in the once-every-3-weeks arm, 28.6 months (95% CI, 15.90 to 41.30 months); HR, 1.24 (95% CI, 0.89 to 1.73); P = .21. Estimated median overall survival in the once-a-week arm was 39.5 months and was not reached in the once-every-3-weeks arm (HR, 1.14 [95% CI, 0.79 to 1.65]; P = .48). Conclusion Once-every-3-weeks cisplatin at 100 mg/m2 resulted in superior LRC, albeit with more toxicity, than did once-a-week cisplatin at 30 mg/m2, and should remain the preferred chemoradiotherapy regimen for LAHNSCC in the adjuvant setting.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. e13020-e13020
Author(s):  
Ishwaria Mohan Subbiah ◽  
Apostolia Maria Tsimberidou ◽  
Aung Naing ◽  
Vivek Subbiah ◽  
Ahmed Omar Kaseb ◽  
...  

e13020 Background: Patients with advanced cholangiocarcinoma (CC) and gallbladder carcinoma (GC) have few effective therapeutic options. We compared prognostic factors and clinical outcomes of CC/GC pts treated on phase I trials with that of their first-, second-line and last Food and Drug Administration (FDA)-approved therapy given in setting of metastatic disease. Methods: We retrospectively reviewed electronic medical records of patients with GC and CC evaluated in the phase I program clinic from November 2004 to March 2011. Results: Of the 72 patients with CC or GC, 32 (44%) were not enrolled on a trial mainly due to clinical deterioration (n=25). Of 40 treated patients (GC=6; CC=34; median age 60 years; median prior systemic therapies = 3), 8 (20%) had stable disease (SD) > 6 months; 3 (8%) achieved a partial response (PR); SD > 6 months/PR was observed mainly on protocols with hepatic arterial infusion drug administration and/or angiogenic inhibitors, anti-her2/neu agents or a novel MAPK/ERK kinase (MEK) inhibitor. Median progression-free survival (PFS) on phase I trials was 2.0 months (95% CI 1.7, 2.8) versus 3.0 months (95% CI 2.4, 5.0; p=0.95), 3.0 months (95% CI 2.3, 4.6; p=0.98), and 3.0 months (95% CI 2.4, 3.9; p=0.79) for their first-, second-, and last systemic therapy with FDA-approved agents given in the metastatic setting, respectively. In univariate analysis, factors associated with a shorter Phase I PFS were > 3 metastatic sites, elevated ALT (>56 IU/L), serum creatinine (>1.6mg/dL), and CA19-9 (>35U/mL). Conclusions: In heavily pretreated patients, PFS in the clinical trials setting remained poor but did not differ significantly from that of their first-line, second-line, and last prior therapy with FDA-approved agents. Response rate (SD >6 months/PR) of 28% was seen in trials with locoregional treatment or inhibitors of angiogenesis, her2/neu or MEK.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 5557-5557
Author(s):  
Magda Mostafa ◽  
Hesham Atif ◽  
Mahmoud Fawzy ◽  
Amr Yehia Sakr ◽  
Ahmed Alashwah

5557 Background: In locally advanced head and neck squamous cell carcinoma (HNSCC) weekly cisplatin concurrent with radiation therapy is the standared treatment. However some patients cannot tolerate cisplatin. So we conduct a prospective randomized trial comparing cisplatin versus gemcitabine. Methods: This trial was done in Kasr El-Ainy Center of Clinical Oncology and Radiation therapy (NEMROCK), during the period from March 2010 till June 2011. Sixty patients with locally advanced HNSCC were randomized to receive Cisplatin (30 mg/m2) weekly for 6 consecutive weeks (30 patients) or Gemcitabine (50 mg/m2) weekly for 6 consecutive weeks (30 patients) both concomitant with radiation therapy reaching a dose of 70 Gy over 7 weeks. Primary end points include response rate, progression free survival and toxicity. Toxicities were graded according to NCI-CTCAE v3.0. Results: Thewhole study group included 48 (80%) males and 12 (20%) females. Mean age was 47.9 (± 6.5) years (range 26-61). Both arms were comparable regarding their age, gender, performance status and stage. There were 9 (30%) CR, 7 (23.3%) PR, 2 (6.7%) SD and 12 (40%) PD in cisplatin arm versus 12 (40%) CR, 4 (13.3%) PR, 1 (3.3%) SD and 11 (36.7%) PD in gemcitabine arm. Median progression free survival (PFS) in cisplatin arm was 9 months versus 11months in gemcitabine arm with a hazard ratio of 0.08 (95% CI 0.005 – 1.47). We did not reach median overall survival. Radiotherapy induced skin toxicity (slight or patchy atrophy), nausea, vomiting, mucositis, salivary gland affection and weight loss were equally distributed in both arms. Dysphagia and fatigue were markedly higher in gemcitabine arm. While infection and neutropenia were slightly higher in cisplatin arm. Conclusions: Weekly gemcitabine 50mg/m2 concomitant with radiotherapy was found to be of equal efficacy and toxicity comparable with weekly cisplatin in the treatment of locally advanced HNSCC.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5982-5982 ◽  
Author(s):  
Angela Solaman ◽  
Andrew Walker ◽  
Faith E Davies

Abstract Background: In 2012, the European Medicines Agency (EMA) recommended the use of progression-free survival (PFS) 2 as a secondary endpoint in trials evaluating maintenance treatment in hematology oncology trials. PFS2 is defined by the EMA as time from randomization to objective tumor progression on next-line treatment or death from any cause. Potential benefits of PFS2 may be found in cases where experimental treatments appear to improve PFS but not overall survival (OS), particularly in situations with improved survival but where measurement of OS is confounded, for example when drugs are trialed in early lines of therapy and subsequently coupled with improved salvage treatments. However, in practice the utility of this endpoint may seem unclear in cases where tumor cells undergo molecular or microenvironment changes without obvious clinical progression. Currently, the US Food and Drug Administration (FDA) does not recognize this end point and in our experience, healthcare providers are unsure of its definition or value. Objective: To understand the current definitions and limitations of PFS2 from a review of the literature and registered clinical trials in the context of PFS. Methods: We undertook a pragmatic literature review relating to PFS and PFS2. Sources investigated included peer-reviewed literature, clinicaltrials.gov, horizon scanning and health technology appraisal (HTA) activities, and 'grey literature' sources. After review of the search hits, information was grouped according to the therapy area and type of information and the main themes and questions identified. Results: We identified a range of specific challenges and issues regarding each endpoint. For PFS, we noted ongoing debates across disease areas. These principally related to the interpretation of clinical trial evidence: expert clinical opinion routinely highlighted concerns about statistical issues and bias arising from censoring, missing data, subjectivity and assessment time. Two other common issues were the need for adequate sample size to detect differences in OS and a lack of evidence relating prolonged PFS to better health-related quality of life (HRQoL). For PFS2, more basic challenges and needs were identified than for PFS. Only the EMA provides a clear definition; the US Food and Drug Administration (FDA) and other regulatory agencies do not appear to have a definition or policy regarding PFS2 at this time. On the other hand, PFS2 is increasingly incorporated into oncology trials as a secondary endpoint in both hematologic and solid tumors. As of 1 August 2016, there were 41 oncology trials registered on clinicaltrials.gov where PFS2 is included. Of these, 20 trials have been registered or updated since January 2015. The implications of routinely generating and using PFS2 data for regulatory and market access purposes therefore are currently unclear. Most of the current information is related to PFS2 data in trials of lenalidomide in multiple myeloma. Based on commentaries and discussions in the literature, there is an ongoing need to understand the clinical and economic rationales for using PFS2 data to support benefit-risk assessments (e.g. relating effect sizes to clinical outcomes) and to determine how best to communicate these potential benefits meaningfully. There is also a need to understand how PFS2 may relate to patient-centered benefits, such as HRQoL, which may help to identify patient preferences in the context of treatment choice. This information has been used to develop a physician questionnaire that would help identify insights and opinions about PFS2 in the following areas: (1) use of surrogate endpoints, (2) treatment choice in relapsing cancers, (3) implications of PFS2 for clinical practice and (4) future needs for PFS2. Results from this primary research will be presented subsequently. Conclusions: PFS2 is increasingly included as a secondary endpoint in oncology trials assessing the benefits of maintenance or sequential treatments. However, there is limited guidance from regulatory bodies on using and interpreting PFS2 data even though there are post-approval implications regarding potential positioning of drugs within treatment pathways. To date, the EMA is the only regulatory agency that has considered the relevance of PFS2 as a clinical endpoint. The clinical importance of PFS2 needs greater understanding to help physicians apply the data in patient management. Disclosures Solaman: Celgene: Consultancy. Walker:Celgene: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Amgen: Consultancy, Honoraria. Davies:Janssen: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Celgene: Consultancy, Honoraria.


2019 ◽  
Vol 3 (4) ◽  
Author(s):  
Daphne Day ◽  
Christina Guo ◽  
Yada Kanjanapan ◽  
Ben Tran ◽  
Anna Spreafico ◽  
...  

Abstract Background Immuno-oncology (IO) is rapidly evolving in early drug development. We aimed to develop and prospectively validate a prognostic index for patients treated in IO phase I trials to assist with patient selection. Methods The development cohort included 192 advanced solid tumor patients treated in 13 IO phase I trials, targeting immune checkpoint and/or co-stimulatory molecules. A prognostic scoring system was developed from multivariate survival analysis of 10 clinical factors, and subsequently validated in two independent validation cohorts (n = 152 and n = 80). Results In the development cohort, median age was 57.5 years (range = 20.4–84.8 years). Median progression-free survival and overall survival (OS) were 13.4 and 73.6 weeks, respectively, 90-day mortality was 16%, and overall response rate was 20%. In multivariate analysis, Eastern Cooperative Oncology Group performance status greater than or equal to 1 (hazard ratio [HR] = 3.2, 95% confidence interval [CI] = 1.8 to 5.7; P < .001), number of metastatic sites greater than 2 (HR = 2.0, 95% CI = 1.3 to 3.1; P = .003), and albumin less than the lower limit of normal (HR = 1.8, 95% CI = 1.2 to 2.7; P = .007) were independent prognostic factors; comprising the Princess Margaret Immuno-oncology Prognostic Index (PM-IPI). Patients with a score of 2–3 compared with patients with a score of 0–1 had shorter OS (HR = 3.4, 95% CI = 1.9 to 6.1; P < .001), progression-free survival (HR = 2.3, 95% CI = 1.7 to 3.2; P < .001), higher 90-day mortality (odds ratio = 8.1, 95% CI = 3.0 to 35.4; P < .001), and lower overall response rate (odds ratio = 0.4, 95% CI = 0.2 to 0.8; P = .019). The PM-IPI retained prognostic ability in both validation cohorts and performed better than previously published phase I prognostic scores for predicting OS in all three cohorts. Conclusions The PM-IPI is a validated prognostic score for patients treated in phase I IO trials and may aid in improving patient selection.


2017 ◽  
Vol 35 (15) ◽  
pp. 1686-1694 ◽  
Author(s):  
Aidan Tan ◽  
Raphael Porcher ◽  
Perrine Crequit ◽  
Philippe Ravaud ◽  
Agnes Dechartres

Purpose We aimed to compare treatment effect sizes between overall survival (OS) and progression-free survival (PFS) in trials of US Food and Drug Administration–approved oncology immunotherapy drugs with results posted at ClinicalTrials.gov . Methods We searched ClinicalTrials.gov for phase II to IV cancer trials of Food and Drug Administration–approved immunotherapy drugs and selected those reporting results for both OS and PFS. For each trial, we extracted the hazard ratios (HRs) with 95% CIs for both outcomes and evaluated the differences by a ratio of HRs (rHRs): the HR for PFS to that for OS. We performed a random effects meta-analysis across trials to obtain a summary rHR. We also evaluated surrogacy of PFS for OS by the coefficient of determination and the surrogacy threshold effect, the minimal value of HR for PFS to predict a non-null effect on OS. Results We identified 51 trials assessing 14 drugs across 15 conditions. Treatment effect sizes were 17% greater, on average, with PFS than with OS (rHR, 0.83; 95% CI, 0.79 to 0.88; I2 = 34.4%; P = .01; τ2 = 0.0129). Nearly one half of the trials (n = 23, 45%) showed statistically significant benefits for PFS but not for OS. Differences were great for trials of obinutuzumab (rHR, 0.21; 95% CI, 0.08 to 0.54), bevacizumab (rHR, 0.75; 95% CI, 0.67 to 0.84), and rituximab (rHR, 0.79; 95% CI, 0.64 to 0.98). The coefficient of determination was 38% and the surrogacy threshold effect was 0.50. Conclusion Treatment effect sizes in trials of immunotherapy drugs were greater for PFS than for OS, with important differences for some drugs, which is consistent with surrogacy metrics. Caution must be taken when interpreting PFS in the absence of OS data.


Sign in / Sign up

Export Citation Format

Share Document