scholarly journals Blinded Independent Central Review of Progression-Free Survival in Phase III Clinical Trials: Important Design Element or Unnecessary Expense?

2008 ◽  
Vol 26 (22) ◽  
pp. 3791-3796 ◽  
Author(s):  
Lori E. Dodd ◽  
Edward L. Korn ◽  
Boris Freidlin ◽  
C. Carl Jaffe ◽  
Lawrence V. Rubinstein ◽  
...  

Progression-free survival is an important end point in advanced disease settings. Blinded independent central review (BICR) of progression in randomized clinical trials has been advocated to control bias that might result from errors in progression assessments. However, although BICR lessens some potential biases, it does not remove all biases from evaluations of treatment effectiveness. In fact, as typically conducted, BICRs may introduce bias because of informative censoring, which results from having to censor unconfirmed locally determined progressions. In this article, we discuss the rationale for BICR and different ways of implementing independent review. We discuss the limitations of these approaches and review published trials that report implementing BICR. We demonstrate the existence of informative censoring using data from a randomized phase II trial. We conclude that double-blinded trials with consistent application of measurement criteria are the best means of ensuring unbiased trial results. When such designs are not practical, BICR is not recommended as a general strategy for reducing bias. However, BICR may be useful as an auditing tool to assess the reliability of marginally positive results.

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 32-33
Author(s):  
Zahoor Ahmed ◽  
Karun Neupane ◽  
Rabia Ashraf ◽  
Amna Khan ◽  
Moazzam Shahzad ◽  
...  

Introduction: Daratumumab (Dara) is a human anti-CD38 monoclonal antibody approved for multiple myeloma (MM) treatment. Dara has a promising efficacy and a favorable safety profile in newly diagnosed MM (NDMM) patients. This study is focused on the efficacy and safety of Dara when added to the standard care regimen in transplant ineligible NDMM in phase III clinical trials. Methods: We performed a comprehensive database search on four major databases (PubMed, Embase, Cochrane, and Clinicaltrials.gov). Our search strategy included MeSH (Medical Subject Headings) terms and key words for multiple myeloma and Dara including trade names and generic names from date of inception to May 2020. Initial search revealed 587 articles. After excluding review articles, duplicates, and non-relevant articles, two phase III clinical trials were included which reported overall response rate (ORR), and progression free survival (PFS) of transplant ineligible NDMM patients with Dara addition to standard care regimen. Odds ratios (OR) of ORR were computed and hazard ratios (HR) of PFS (along with 95% confidence intervals; CI) were extracted to compute a pooled HR using a fixed effect model in RevMan v.5.4. Results: A total of 1453 transplant ineligible NDMM patients were enrolled and evaluated in two phase III randomized clinical trials. Seven hundred and eighteen patients were in Dara group and 735 patients were in control group. Bahlis et al. (2019) studied Dara + lenolidamide (R) and dexamethasone (d) vs Rd in NDMM pts (n=737) in MAIA phase III trial. Similarly, Mateos et al. (2018) reported the role of Dara + bortezomib (V) + melphalan (M), and prednisone (P) vs VMP in NDMM pts (n=706) in a phase III trial (Alcyone). A pooled analysis of these phase III trials showed ORR (OR: 3.26, 95% CI 2.36-4.49; p < 0.00001, I2 = 0%), and progression free survival (PFS) (HR: 0.53, 95% CI 0.43-0.65; p < 0.00001, I2 = 0%). Achievement of minimal residual disease (MRD) negative status was significant in Dara based regimen as compared to control group (OR: 4.49, 95% CI 3.31-6.37; p < 0.00001, I2 = 0%). Dara addition to standard care regimen (Rd and VMP) decreased the risk of progression/death to 42% (HR: 0.58, 95% CI 0.48-0.70; p < 0.00001, I2 = 0%). The addition of Dara increased the risk of neutropenia (OR: 1.41, 95% CI 1.07-1.85; p < 0.02, I2 = 44%), and pneumonia (OR: 2.25, 95% CI 1.54-3.29; p < 0.0001, I2 = 37%) vs control group. However, decreased risk of anemia (OR: 0.64, 95% CI 0.49-0.85: p < 0.002, I2=30%) was observed in Dara group vs control group (Figure 1). Conclusion: Addition of Dara to the standard care regimen for transplant ineligible NDMM achieved the surrogate end points with improved efficacy and MRD negative status with manageable toxicity. However, data from more randomized controlled trials is needed. Table Disclosures Anwer: Incyte, Seattle Genetics, Acetylon Pharmaceuticals, AbbVie Pharma, Astellas Pharma, Celegene, Millennium Pharmaceuticals.: Honoraria, Research Funding, Speakers Bureau.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 6622-6622
Author(s):  
Andrea Bonetti ◽  
Jcopo Giuliani

6622 Background: In Western Countries, colorectal cancer (CRC) is the second most common cause of death from cancer. In light of the relevant expenses of drugs it might be interesting to make a balance between the cost of the drugs and clinical parameters of interest such as progression free survival (PFS). Methods: Phase III randomized clinical trials (RCTs) that compared at least two front-line chemotherapy regimens for mCRC patients were evaluated. Differences in PFS between the different arms were calculated and compared with the pharmacological costs (at the Pharmacy of our Hospital) needed to get one month of PFS. Subsequently we applied the ESMO-MCBS (a 1 to 5 scale) to the above phase III RCTs. Results: Overall 28 phase III RCTs, including 19 958 patients, were analyzed. The FOLFOX resulted the least expensive (56 € per month of PFS gained) while the addition of irinotecan to FOLFOX (FOLFOXIRI) increased only marginally the costs (90 € per month of PFS gained). Treatments including the monoclonal antibodies showed a cost per month of PFS gained of 2823 € (FOLFIRI with cetuximab in KRAS wild-type patients and liver-only metastases), of € 15 822 (FOLFOX with panitumumab in KRAS wild type) and of 13 383 € (FOLFOX with bevacizumab). According to the ESMO-MCBS the treatments including an EGFR-inhibitor (cetuximab or panitunumab) were associated with a score of 4 while the inclusion of bevacizumab reached a score of 3. The remaining phase III RCTs obtained a low (grade 1-2) score.Dividing the costs per month of PFS gained with the grade of ESMO-MCBS, for each RCTs, we obtained the costs of each point of ESMO-MCBS per month of PFS gained. FOLFOX was confirmed as being the least expensive (18.7 €) while among treatments including a targeted biological agent panitunumab in combination with FOLFOX in K-RAS wild type patients was less expensive (3955 €) than the combinations FOLFOX-bevacizumab (13 383 €) and FOLFIRI-cetuximab in K-RAS wild type patients (21 854.6 €). Conclusions: Our data demonstrate a huge difference in cost per month of PFS gained and per each point of the ESMO-MCBS in modern front line treatments in mCRC.


2017 ◽  
pp. 1-12 ◽  
Author(s):  
Dawn L. Hershman ◽  
Cathee Till ◽  
Jason D. Wright ◽  
Scott Ramsey ◽  
William E. Barlow ◽  
...  

Purpose Diabetes is common, increases with age, and may affect outcomes among people with cancer. Understanding the association between diabetes and cancer outcome is challenging, because patients with diabetes have increased all-cause mortality compared with patients without diabetes. Methods We systematically examined the phase III trial database of SWOG to identify patients enrolled in trials during the period from 1999 to 2011. We linked the SWOG clinical records to Medicare claims data according to Social Security number, sex, and date of birth. Medicare claims were used to identify diabetes with at least 6 months of continuous Medicare coverage immediately before registration. Multivariable Cox regression was used to compare survival outcomes between patients with and without diabetes for each of 10 tumor cohorts. The primary outcome was overall survival. We also examined progression-free survival and cancer-free survival. Results In total, 6,422 patients from 15 trials were ≥ 65.5 years of age, of whom 3,173 patients (49%) met the criteria for linkage to Medicare claims. Thirty percent (n = 952) had claims for diabetes before registration. Patients with diabetes were more likely to be black ( P < .001), but no other differences in demographic characteristics were observed. In multivariable Cox regression, no association was found between baseline diabetes and overall or progression-free survival; in one case, patients with diabetes had marginally worse cancer-free survival (advanced non–small-cell lung cancer; P = .05). A global test found that baseline diabetes was associated with worse overall survival ( P = .03) across the entire panel of analyses. Conclusion Diabetes is common among elderly patients enrolled in clinical trials. Unlike prior observational studies, among patients treated with uniform treatment regimens, and controlling for known prognostic factors, we did not observe an association between diabetes and progression-free or cancer-free survival.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 6115-6115 ◽  
Author(s):  
A. Kumar ◽  
H. P. Soares ◽  
L. Balducci ◽  
B. Djulbegovic

6115 Background: Elderly patients share the majority of the disease burden in cancer. Although 61% of new cancer cases occur among elderly, yet elderly comprise only 25% of the patients enrolled in clinical trials. A systematic review to assess the accurate participation of elderly in randomized clinical trials (RCT) has not been done. Methods: We reviewed all consecutively completed phase III RCTs conducted by 5 National Cancer Institute sponsored cooperative groups. Published papers and study protocols were used to ensure the accuracy of the data extraction. We used a cut-off of age ≥65 to define elderly patients. For trials that did not have an exclusive ≥65 age criteria for enrollment, data were extracted on number of participants ≥65. Outcome between the innovative and the standard treatment was compared. Data were extracted as per the methods recommended by the Cochrane Collaboration. Results: Out of 413 studies, only two trials exclusively enrolled elderly patients (0.5%). 57% (n=235) of the trials did not have any stratification by age. Only 10% of the studies had a stratification age at ≥65 (n=42). Contrary to the general notion that elderly may not respond well to newer chemotherapy combination treatments, overall survival in these two trials exclusively enrolling elderly favored the newer treatments [Hazard ratio (HR) 0.62(95%CI 0.44, 0.87)]. In addition, trials enrolling >40% of elderly had favorable outcomes involving innovative treatments for survival and event-free survival [HR survival 0.85(95%CI 0.77, 0.94), HR event-free survival was 0.76(95%CI 0.64, 0.92)]. Treatment related mortality was similar in the innovative and the standard arms [HR 0.87(95%CI 0.39, 1.96)]. Conclusions: Our data indicate that enrollment of elderly in experimental RCTs is not associated with increased harms to this patient population. Increased participation of elderly may help in finding new treatments that are clinically applicable specifically to this cohort of patients. No significant financial relationships to disclose.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2715-2715
Author(s):  
Brian G. Van Ness ◽  
Christine Ramos ◽  
Vipin Kumar ◽  
Michael Steinbach ◽  
Brian GM Durie ◽  
...  

Abstract The Bank On A Cure (BOAC) has established DNA banks from multiple cooperative and institutional clinical trials, and platforms for examining the association of genetic variations (SNPs) with disease risk and outcomes in myeloma. We have previously described the development and content of a novel custom SNP panel that contains 3,404 SNPs in 983 genes, representing cellular functions and pathways that may influence disease response, toxicities, complications, and survival. Although survival certainly varies according to tumor heterogeneity (ie. chromosomal abnormalities, gene expression variations) germline variations that influence the microenvironment, drug distribution, drug transport and metabolism, may also have an association with event free survival outcomes. To explore SNP associations with progression free survival (PFS) we compared the BOAC SNP profiles of short term PFS (less than 1 year, n=70) versus long term PFS (greater than 3 years, n=73) in two phase III clinical trials (ECOG E9487 and SWOG S9321). A variety of analytical approaches was undertaken including univariate rank ordering, recursive partitioning, and support vector machine learning tools (SVM). Each of these approaches has advantages and limitations in dealing with type I false positive errors as well as sensitivity and specificity. We included subset validation approaches and randomization of classes to address how robust and predictive different approaches were. From our analysis we conclude germline genomic variations do have an impact on progression free survival, with a subset of SNPs from the panel reaching 76% predictive association and hazard ratios of PFS of 9.6 (CI 4.5, 20.5), p&lt;0.001, using SVM analysis. Based on univariate approaches, we find the most significant variations associated with PFS differences were genes that could be functionally categorized as pharmacologic. The presentation will focus on the analytical approaches, and refinements necessary to assure predictive value compared to random associations. Notwithstanding the clear importance of tumor cell variations in genetic deregulation, we conclude that various functions within the bone marrow and drug response likely interplay as a complex influence on disease progression, response, and survival. This suggests combining gene expression profiles of the tumors with germline SNP profiles may provide more accurate prognosis. These combined analytical approaches are currently being developed with BOAC data bases, and examples will be discussed.


2014 ◽  
Vol 32 (27) ◽  
pp. 3068-3074 ◽  
Author(s):  
Federico Campigotto ◽  
Edie Weller

Informative censoring in a progression-free survival (PFS) analysis arises when patients are censored for initiation of an effective anticancer treatment before the protocol-defined progression, and these patients are at a different risk for treatment failure than those who continue on therapy. This may cause bias in the estimated PFS when using the Kaplan-Meier method for analysis. Although there are several articles that discuss this issue from a theoretical perspective or in randomized phase III studies, there are little data to demonstrate the magnitude of the bias on the estimated quantities from a phase II trial. This article describes the issues by using two oncology phase II trials as examples, evaluates the impact of the bias using simulations, and provides recommendations. The two trials were selected because they demonstrate two different reasons for censoring. Simulations show that the magnitude of the bias depends primarily on the proportion of patients who are informatively censored and secondarily on the hazard ratio between the group of patients who remain on study and the group of patients who are informatively censored. Recommendations include using an alternative end point, which includes inadequate response and initial signs of clinical progression as treatment failure, and a competing risk analysis for studies in which competing events preclude or modify the probability of observing the primary event of interest. If informative censoring cannot be avoided, then all patients should be observed until progression, and sensitivity analyses should be used as appropriate.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5469-5469
Author(s):  
Stefano Molica ◽  
Diana Giannarelli ◽  
Luciano Levato ◽  
Emili Montserrat

Background: Recent studies have demonstrated a prolonged progression-free survival (PFS) in CLL patients receiving ibrutinib-based regimes as upfront therapy. These studies, however, are limited in number and heterogeneous regarding patients' selection criteria, treatment combinations and schedules. Because of this, we conducted a meta-analysis of randomized clinical trials using ibrutinib as upfront treatment to evaluate the strength of the evidence and patients' subgroups who benefit the most of such treatment. Materials & Methods: A systematic literature search was performed using PubMed to identify full length reports dealing with the randomized clinical trials (RCTs) of ibrutinib, used alone or in combination as upfront therapy of CLL prior to August 2nd, 2019. A complementary manual search of the ASH, EHA and ASCO conference proceedings was also performed. The search strategy used both Medical Subject Headings (MESH) terms and free text words to increase the sensitivity of the search. The electronic search yielded 4 full-text articles assessed for eligibility. Results: In total 4 RCTs (i.e., RESONATE2, ALLIANCE, ILLUMINATE, ECOG-ACRIN) including 1574 untreated CLL patients compared head-to-head an ibrutinib-based regimen to CT or CIT. Inclusion criteria were either age 65 years or older or coexisting conditions for 1045 patients and age younger than 70 years in absence of coexisting conditions for 529 patients. Among patients treated with an ibrutinib-based regimen, 318 received ibrutinib as single agent and 649 ibrutinib in association with anti-CD20 monoclonal antibodies (i.e., 536 ibrutinib + rituximab [R], 113 ibrutinib + obinotuzumab [Obino]). The CT or CIT control arms included 607 patients and consisted of chlorambucil (CLB)(n=133), CLB+Obino (n=116), bendamustine and R (BR) (n=183) and fludarabine, cyclophosphamide and R (FCR)(n=175). All four studies included in the quantitative meta-analysis had enough data to assess PFS. Results indicate that treatment with ibrutinib-based regimens improved PFS compared with CT or CIT. The pooled HR for PFS in ibrutinib-treated patients was 0.331 (95% confidence interval [CI]: 0.272-0.403; P=0.000). The I2 statistic for heterogeneity (i.e. 0%; P=0.50) and Q values (i.e. 3.35) indicated a high level of homogeneity of results across studies. Of note, data were robust with no evidence of obvious publication bias (i.e., Funnel plot analysis indicate several missing studies that would bring p-value up to >0.05=156). Overall survival (OS) was evaluated in 3 studies accounting for 1017 patients and revealed an HR of 0.289 (95% CI, 0.071-0.1175), with definite heterogeneity across studies (I2=82.7%; Q=11.6; P=0.003). Next, we evaluated the magnitude of improvement in PFS obtained with ibrutinib-based regimens in patients with high-risk genetic features such as 11q deletion and unmutated IGHV status. Data for PFS by 11q deletion status, restricted to 3 studies (n=206), revealed a significantly lower risk of progression for 11q deleted patients treated with ibrutinib-based regimens (HR,0.159; 95% CI: 0.077-0.327; I2 = 42%; P=0.18; Q = 3.46) (Fig 1A). Of note, a lower risk of progression was observed in both patients with unmutated (n=522) (HR, 0.178; 95% CI, 0.121-0.261; p=.000; I2=11%; Q=2.21;P=0.32)(Fig 1B) and mutated IGHV (n= 287)(HR, 0.270;95% CI, 0.149-0.489; I2=0%; P=0.38;Q=1.91)(Fig 1B) who received ibrutinib in upfront. Despite slight differences of the HR values between patients with mutated and unmutated IGHV, interaction analysis suggests that the magnitude of PFS improvement is comparable in both groups (HR, 1.50; 95% CI, 0.74-3.04; P=0.87). Conclusions This meta-analysis validates the notion that in comparison with CT or CIT ibrutinib-based regimens, given as upfront therapy, abrogate the negative impact of 11q deletion and unmutated IGHV on PFS. Even more, 11q deletion could be a favorable predictive biomarker for ibrutinib therapy. Because of design and selection criteria of studies included in this meta-analysis, some aspects such as the impact on response and its duration in patients with 17p deletion/TP53 mutations need further evaluation. Nonetheless, our results should inform updated algorithms of upfront CLL treatment. Figure 1 Disclosures Molica: Gilead, AbbVie, Jansen, Roche: Other: Advisory Board. Levato:Novartis: Honoraria; BMS: Honoraria; Incyte: Honoraria; Pfizer: Honoraria.


Sign in / Sign up

Export Citation Format

Share Document