Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Bruce A. Bach is active.

Publication


Featured researches published by Bruce A. Bach.


Lancet Oncology | 2013

Cisplatin and fluorouracil with or without panitumumab in patients with recurrent or metastatic squamous-cell carcinoma of the head and neck (SPECTRUM) : an open-label phase 3 randomised trial

Jan B. Vermorken; Jan Stöhlmacher-Williams; Irina Davidenko; Lisa Licitra; Eric Winquist; Cristian Villanueva; Paolo Foa; Sylvie Rottey; K. Składowski; Makoto Tahara; V. R. Pai; Sandrine Faivre; Cesar R. Blajman; Arlene A. Forastiere; Brian N Stein; Kelly S. Oliner; Zhiying Pan; Bruce A. Bach

BACKGROUND Previous trials have shown that anti-EGFR monoclonal antibodies can improve clinical outcomes of patients with recurrent or metastatic squamous-cell carcinoma of the head and neck (SCCHN). We assessed the efficacy and safety of panitumumab combined with cisplatin and fluorouracil as first-line treatment for these patients. METHODS This open-label phase 3 randomised trial was done at 126 sites in 26 countries. Eligible patients were aged at least 18 years; had histologically or cytologically confirmed SCCHN; had distant metastatic or locoregionally recurrent disease, or both, that was deemed to be incurable by surgery or radiotherapy; had an Eastern Cooperative Oncology Group performance status of 1 or less; and had adequate haematological, renal, hepatic, and cardiac function. Patients were randomly assigned according to a computer-generated randomisation sequence (1:1; stratified by previous treatment, primary tumour site, and performance status) to one of two groups. Patients in both groups received up to six 3-week cycles of intravenous cisplatin (100 mg/m(2) on day 1 of each cycle) and fluorouracil (1000 mg/m(2) on days 1-4 of each cycle); those in the experimental group also received intravenous panitumumab (9 mg/kg on day 1 of each cycle). Patients in the experimental group could choose to continue maintenance panitumumab every 3 weeks. The primary endpoint was overall survival and was analysed by intention to treat. In a prospectively defined retrospective analysis, we assessed tumour human papillomavirus (HPV) status as a potential predictive biomarker of outcomes with a validated p16-INK4A (henceforth, p16) immunohistochemical assay. Patients and investigators were aware of group assignment; study statisticians were masked until primary analysis; and the central laboratory assessing p16 status was masked to identification of patients and treatment. This trial is registered with ClinicalTrials.gov, number NCT00460265. FINDINGS Between May 15, 2007, and March 10, 2009, we randomly assigned 657 patients: 327 to the panitumumab group and 330 to the control group. Median overall survival was 11·1 months (95% CI 9·8-12·2) in the panitumumab group and 9·0 months (8·1-11·2) in the control group (hazard ratio [HR] 0·873, 95% CI 0·729-1·046; p=0·1403). Median progression-free survival was 5·8 months (95% CI 5·6-6·6) in the panitumumab group and 4·6 months (4·1-5·4) in the control group (HR 0·780, 95% CI 0·659-0·922; p=0·0036). Several grade 3 or 4 adverse events were more frequent in the panitumumab group than in the control group: skin or eye toxicity (62 [19%] of 325 included in safety analyses vs six [2%] of 325), diarrhoea (15 [5%] vs four [1%]), hypomagnesaemia (40 [12%] vs 12 [4%]), hypokalaemia (33 [10%] vs 23 [7%]), and dehydration (16 [5%] vs seven [2%]). Treatment-related deaths occurred in 14 patients (4%) in the panitumumab group and eight (2%) in the control group. Five (2%) of the fatal adverse events in the panitumumab group were attributed to the experimental agent. We had appropriate samples to assess p16 status for 443 (67%) patients, of whom 99 (22%) were p16 positive. Median overall survival in patients with p16-negative tumours was longer in the panitumumab group than in the control group (11·7 months [95% CI 9·7-13·7] vs 8·6 months [6·9-11·1]; HR 0·73 [95% CI 0·58-0·93]; p=0·0115), but this difference was not shown for p16-positive patients (11·0 months [7·3-12·9] vs 12·6 months [7·7-17·4]; 1·00 [0·62-1·61]; p=0·998). In the control group, p16-positive patients had numerically, but not statistically, longer overall survival than did p16-negative patients (HR 0·70 [95% CI 0·47-1·04]). INTERPRETATION Although the addition of panitumumab to chemotherapy did not improve overall survival in an unselected population of patients with recurrent or metastatic SCCHN, it improved progression-free survival and had an acceptable toxicity profile. p16 status could be a prognostic and predictive marker in patients treated with panitumumab and chemotherapy. Prospective assessment will be necessary to validate our biomarker findings. FUNDING Amgen Inc.


European Journal of Cancer | 2012

First-line treatment of metastatic or locally advanced unresectable soft tissue sarcomas with conatumumab in combination with doxorubicin or doxorubicin alone: A Phase I/II open-label and double-blind study

George D. Demetri; Axel Le Cesne; Sant P. Chawla; Thomas Brodowicz; Robert G. Maki; Bruce A. Bach; D. Smethurst; Sarah Bray; Yong-jiang Hei; Jean-Yves Blay

BACKGROUND Conatumumab is a fully human monoclonal agonist antibody that binds to death receptor 5 and induces apoptosis in sensitive cells. This study evaluated the safety and efficacy of doxorubicin ± conatumumab as first-line systemic therapy for metastatic or locally advanced/unresectable soft-tissue sarcoma. METHODS In Phase I, six patients received doxorubicin (75 mg/m2) with conatumumab (15 mg/kg) every 3 weeks. In Phase II, patients were randomised (2:1) to receive doxorubicin with either double-blind conatumumab 15 mg/kg (conatumumab-doxorubicin; n=86) or placebo (placebo-doxorubicin; n=42). Patients who progressed on placebo-doxorubicin could receive open-label conatumumab monotherapy post-chemotherapy (n=21). FINDINGS The expected histopathologic subtypes (e.g. leiomyosarcoma, liposarcoma, others) were represented in this trial. No unexpected adverse events were noted in either Phase I or II. Median progression-free survival in Phase II was 5.6 and 6.4 months in the conatumumab-doxorubicin and placebo-doxorubicin arms, respectively (stratified HR: 1.00; p=0.973), with more early progressions noted in the first 3.5 months in the conatumumab-doxorubicin arm. Median overall survival was not reached after 8.6 months median follow-up in either arm. Common adverse events were nausea (conatumumab-doxorubicin: 66%; placebo-doxorubicin: 80%), alopecia (55%; 63%), fatigue (60%; 38%) and neutropenia (32%; 50%). Post-chemotherapy results were not notably improved by conatumumab dosing. INTERPRETATION Addition of conatumumab to doxorubicin appeared to be safe but did not improve disease control in a heterogeneous unselected group of patients with soft tissue sarcomas. The results of this trial are very useful for estimating the outcomes of first-line therapy of sarcoma patients treated with standard doxorubicin.


Gynecologic Oncology | 2016

Final results of a phase 3 study of trebananib plus weekly paclitaxel in recurrent ovarian cancer (TRINOVA-1): Long-term survival, impact of ascites, and progression-free survival-2☆☆☆☆☆☆

Bradley J. Monk; Andres Poveda; Ignace Vergote; Francesco Raspagliesi; Keiichi Fujiwara; Duk Soo Bae; Isabelle Ray-Coquard; Diane Provencher; Beth Y. Karlan; Catherine Lhommé; Gary Richardson; Dolores Gallardo Rincon; Robert L. Coleman; Christian Marth; Arija Brize; Michel Fabbro; Andrés Redondo; Aristotelis Bamias; Haijun Ma; Florian D. Vogl; Bruce A. Bach; Amit M. Oza

PURPOSE Trebananib, a peptibody that blocks binding of angiopoietin-1 and -2 to Tie2, significantly prolonged progression-free survival (PFS) in patients with recurrent epithelial ovarian cancer in the phase 3 TRINOVA-1 study. We report overall survival (OS) in the intent-to-treat population and clinically relevant subgroups and time to second disease progression (PFS-2). PATIENTS AND METHODS Women with recurrent disease (platinum-free interval<12months) were randomized to receive intravenous paclitaxel 80mg/m(2) (3weeks on/1week off) plus intravenous trebananib 15mg/kg or placebo, weekly. OS in the intent-to-treat population was a key secondary endpoint. Exploratory analysis of PFS-2 was conducted according to guidance by the European Medicines Agency. RESULTS Median OS was not significantly improved with trebananib compared with placebo (19.3 versus 18.3months; HR, 0.95; 95% CI, 0.81-1.11; P=0.52) in the intent-to-treat population (n=919). In subgroup analysis, trebananib improved median OS compared with placebo (14.5 versus 12.3months; HR, 0.72; 95% CI, 0.55-0.93; P=0.011) in patients with ascites at baseline (n=295). In the intent-to-treat population, trebananib significantly improved median PFS-2 compared with placebo (12.5 versus 10.9months; HR, 0.85; 95% CI, 0.74-0.98; P=0.024). The incidence and type of adverse events in this updated analysis was consistent with that described in the primary analysis; no new safety signals were detected. CONCLUSIONS OS was not significantly longer in the intent-to-treat population, although there was an improvement in OS in patients with ascites receiving trebananib. PFS-2 confirmed that the PFS benefit associated with trebananib was maintained through the second disease progression independent of the choice of subsequent therapy.


European Journal of Cancer | 2011

Safety and Efficacy of Panitumumab (pmab) in HPV Positive (+) and HPV Negative (−) Recurrent/metastatic (R/M) Squamous Cell Carcinoma of the Head and Neck (SCCHN): Analysis of the Phase 3 SPECTRUM Trial

Jan B. Vermorken; J. Stöhlmacher; K. Oliner; C. Villanueva; P. Foa; Eric Winquist; Lisa Licitra; K. Składowski; Z. Pan; Bruce A. Bach

and GCIG CA125 criteria. 3 core PTB obtained before and after 4 weeks of GSK795, were analysed by immunohistochemistry (IHC) and Reverse Phase Protein Array (RPPA). Sequenom mutation profiling (SMP) of archival tissue and PTB was also done. Results: The most common drug related AE was G1/2 vomiting (33%); there was only one G3 drug related AE: hepatotoxicity. A PK-PET PD relationship was observed between GSK795 plasma Cmax and Ki for best responding lesions. Although no RECISTv1.1 responses were noted, CA125 GCIG criteria PR rate was 20% (n = 2/10). A direct correlation was observed between best CA125 response, best CT response and disease stabilization (p 52 weeks. IHC and RPPA showed PD evidence of PI3K/AKT pathway inhibition (pAKT/PRAS40) irrespective of response status. SMP demonstrated mutations associated with response (PIK3CA) and resistance (Kras, MET) to GSK795. RPPA identified putative predictive (S6), resistance (CCNE1) and response (Bid) biomarkers suitable for validation for AKT inhibitor therapy.


Journal of Bone and Joint Surgery, American Volume | 2015

Incidence Trends in the Diagnosis of Giant Cell Tumor of Bone in Sweden Since 1958.

Julia Rockberg; Bruce A. Bach; Justyna Amelio; Rohini K. Hernandez; P. Sobocki; Jacob Engellau; Henrik C. F. Bauer; Alexander Liede

BACKGROUND The Swedish Cancer Registry (founded in 1958) constitutes a unique resource for epidemiological studies of giant cell tumor of bone with potential for use for population-based studies of incidence over time. The aim of this study was to provide what we believe is the first modern population-based assessment of the incidence trends of giant cell tumor, a unique osteoclastogenic lytic stromal tumor with both benign and malignant histological forms, and to compare the findings with data from the same registry on osteosarcoma, a tumor that may display similar histological characteristics. METHODS Cases were identified with use of codes for pathological bone tumor (International Classification of Diseases [ICD]-7 196). Specific morphological coding distinguishes benign (PAD 741) from malignant giant cell tumor (PAD 746) and osteosarcoma (PAD 766). RESULTS During the period of 1958 to 2011, 4625 bone tumors were reported, including 505 giant cell tumors (383 benign and 122 malignant) and 1152 osteosarcomas. From 1958 to 1982 the ratio of malignant to benign giant cell tumors was 1.3, whereas from 1983 to 2011 the ratio inverted to 0.09, suggesting a change in the reporting or diagnosis of malignant or benign cases. Cases of giant cell tumor diagnosed from 1983 to 2011 displayed an age and sex distribution (median age at diagnosis, 34.0 years; 54% female) that were consistent with those in large published case series but differed from those in 1958 to 1982 (median age at diagnosis, 31.5 years; 48% female). The most current data (1983 to 2011) showed the giant cell tumor incidence in Sweden to be 1.3 per million per year, while the osteosarcoma incidence was 2.3 per million per year. CONCLUSIONS Early Swedish Cancer Registry data (1958 to 1982) revealed a higher proportion of malignant giant cell tumors than seen in large sequential case series and a distinct age and sex profile compared with more recent data (1983 to 2011). This likely represents changes in the diagnostic workup and introduction of multidisciplinary review of giant-cell-containing tumors around 1982. Recent data may reflect the impact of expert centralized biopsy and multidisciplinary case review and more comprehensive reporting of benign giant cell tumors.


Journal of Bone and Joint Surgery, American Volume | 2014

Regional Variation and Challenges in Estimating the Incidence of Giant Cell Tumor of Bone

Alexander Liede; Bruce A. Bach; Scott Stryker; Rohini K. Hernandez; P. Sobocki; Brian Bennett; Steven S. Wong

BACKGROUND Estimating the incidence of giant cell tumor of bone is challenging because few population-based cancer registries record benign bone tumors. We compared two approaches, the indirect (relative index) estimation approach used in The Burden of Musculoskeletal Diseases in the United States (BMUS) and a direct incidence rate approach (from registries that record giant cell tumor), to estimate giant cell tumor incidence in France, Germany, Italy, Spain, the U.K., Sweden, Australia, Canada, Japan, and the U.S. METHODS Giant cell tumor of bone incidence was calculated with use of the BMUS relative index of giant cell tumor to osteosarcoma in three scenarios (low, base case, and high) from case series. We compared the BMUS approach with the latest data from tumor registries in Australia (1972 to 1996), Japan (2006 to 2008), and Sweden (1993 to 2011) that record giant cell tumors. United Nations population estimates were used to project results to 2013. RESULTS The low scenario in the BMUS approach reflects data from Unni and Inwards; the incidence of giant cell tumor of bone is 0.34 relative to osteosarcoma. As the incidence of osteosarcoma is 31.4% of the total incidence of bone and joint cancers, the incidence of giant cell tumor is 0.11 times that of all bone and joint cancers. The base scenario reflects the series by Mirra et al., with a giant cell tumor incidence of 0.47 relative to osteosarcoma (0.15 to all bone and joint cancers). The high scenario reflects the series by Ward, with an incidence of 0.84 relative to osteosarcoma (0.26 to all bone and joint cancers). Differences among the three series reflect referral to a national center of excellence compared with referral to a local oncology practice. Registry data indicated a giant cell tumor incidence rate per million per year of 1.33 in Australia, 1.03 in Japan, and 1.11 in Sweden in 2013. The estimated incidence rate per million in the ten countries in 2013 ranged from 1.03 (Japan) to 1.17 (Canada) with use of the registry-based approach and from 0.73 (Japan) for the low scenario) to 2.20 (Germany) for the base case with use of the BMUS approach. CONCLUSIONS Giant cell tumor of bone affects approximately one person per million per year in the ten countries studied. Estimates derived with use of age-specific incidences from tumor registries were typically within the range of the low and base case BMUS scenarios. We recommend the registry-derived method for estimating the incidence of giant cell tumor.


Cancer Epidemiology | 2016

Population-based study of giant cell tumor of bone in Sweden (1983–2011)

Justyna Amelio; Julia Rockberg; Rohini K. Hernandez; Patrik Sobocki; Scott Stryker; Bruce A. Bach; Jacob Engellau; Alexander Liede

INTRODUCTION Giant-cell tumor of bone (GCTB) is a locally aggressive histologically benign neoplasm with a less common malignant counterpart. Longitudinal data sources on GCTB are sparse, limited to single institution case series or surgical outcomes studies. The Swedish Cancer Registry is one of the few national population-based databases recording GCTB, representing a unique source to study GCTB epidemiology. We estimated incidence rate (IR) and overall mortality rates based on registry data. MATERIALS AND METHODS We identified patients with a GCTB diagnosis in the Swedish Cancer Registry from 1983 to 2011: benign (ICD-7 196.0-196.9; PAD 741) and malignant (PAD 746). Results were stratified by age at diagnosis, gender, and anatomical lesion location. RESULTS The cohort included 337 GCTB cases (IR of 1.3 per million persons per year). The majority (n=310) had primary benign GCTB (IR of 1.2 per million per year). Median age at diagnosis was 34 years (range 10-88) with 54% (n=183) females. Malignant to benign ratio for women was 0.095 (16/167) and for men 0.077 (11/143). Incidence was highest in the 20-39 years age group (IR of 2.1 per million per year). The most common lesion sites were distal femur and proximal tibia. Mortality at 20 years from diagnosis was 14% (n=48) and was slightly higher for axial (17%; n=6) and pelvic (17%; n=4) lesions. Recurrence occurred in 39% of primary benign cases and 75% of primary malignant cases. CONCLUSIONS In our modern population-based series primary malignant cases were uncommon (8%), peak incidence 20-39 years with slight predominance in women. Recurrence rates remain significant with overall 39% occurring in benign GCTB, and 75% in malignant form. The linkage between databases allowed the first population based estimates of the proportion of patients who received surgery at initial GCTB diagnosis, and those who also received subsequent surgeries.


Annals of Oncology | 2016

Health-related Quality of Life in Women With Recurrent Ovarian Cancer Receiving Paclitaxel Plus Trebananib or Placebo (TRINOVA-1)

Keiichi Fujiwara; Bradley J. Monk; C Lhommé; Robert L. Coleman; Arija Brize; Isabelle Ray-Coquard; Michel Fabbro; Diane Provencher; A Bamias; Ignace Vergote; A DeCensi; Kathy Zhang; Florian D. Vogl; Bruce A. Bach; Francesco Raspagliesi

BACKGROUND To evaluate the influence of treatment on health-related quality of life (HRQoL) in 919 women with recurrent ovarian cancer enrolled in the TRINOVA-1 study, a randomized, placebo-controlled phase III study that demonstrated that trebananib 15 mg/kg QW plus weekly paclitaxel significantly improved progression-free survival (PFS) compared with placebo plus weekly paclitaxel (7.2 versus 5.4 months; hazard ratio, 0.66; 95% confidence interval 0.57-0.77; P < 0.001). PATIENTS AND METHODS HRQoL was assessed with the Functional Assessment of Cancer Therapy-Ovary [FACT-O; comprising FACT-G and the ovarian cancer-specific subscale (OCS)] and EuroQOL EQ-5D instruments before treatment on day 1 of weeks 1, 5, 9, 13, 17, and every 8 weeks thereafter and at the safety follow-up visit. A pattern-mixture model was used to evaluate the influence of patient dropout on FACT-O and OCS scores over time. RESULTS Of 919 randomized patients, 834 (91%) had a baseline and ≥1 post-baseline HRQoL assessment. At baseline, scores for all instruments were similar for both arms. At 25 weeks, mean ± SD changes from baseline were negligible, with mean ± SD changes typically <1 unit from baseline: -2.4 ± 16.6 in the trebananib arm and -1.6 ± 15.2 in the placebo arm for FACT-O, -0.71 ± 5.5 in the trebananib arm and -0.86 ± 4.9 in the placebo arm for OCS, and -0.02 ± 0.22 in the trebananib arm and 0.02 ± 0.19 in the placebo arm for EQ-5D. Distribution of scores was similar between treatment arms at baseline and over the course of the study. In pattern-mixture models, there was no evidence that patient dropout affected differences in mean FACT-O or OCS scores. Edema had limited effect on either FACT-O or OCS scores in patients with grade ≥2 edema or those with grade 1 or no edema. CONCLUSIONS Our results demonstrate that the improvement in PFS among patients in the trebananib arm in the TRINOVA-1 study was achieved without compromising HRQoL. CLINICALTRIALSGOV IDENTIFIER NCT01204749.


Cancer Research | 2012

Abstract 554: Qualification of a p16INK4A immunohistochemistry (IHC) assay for determination of HPV status in archival Squamous Cell Carcinoma of the Head and Neck (SCCHN) formalin fixed paraffin embedded (FFPE) specimens from a randomized phase 3 trial

Kelly S. Oliner; Mark Ekdahl; Ainura Kyshtoobayeva; Kenneth J. Bloom; Bruce A. Bach

Introduction: HPV positive SCCHN has a distinct set of molecular alterations and a better prognosis in the locally advanced setting. When HPV is integrated into the host cell genome, HPV protein E7 degrades host retinoblastoma protein, an inhibitor of p16INK4A transcription. Thus, in persistent HPV gene expressing tumors, elevated p16INK4A RNA and protein levels serve as a surrogate of HPV infection applicable to FFPE archival tumor specimens. Our aim was to qualify an existing IHC assay for reliable detection of active HPV expression in SCCHN archival FFPE tumor samples from a randomized phase 3 trial in the recurrent/metastatic (R/M) treatment setting. Methods: The CINtec™ p16INK4A Histology Kit is a semi quantitative, immunocytochemical assay for the evaluation of p16INK4A protein expression in FFPE sections. Banked SCCHN specimens were examined for p16INK4A expression to establish repeatability, reproducibility, and long term analyte stability (blocks and slides). Comparative studies were performed with a p16INK4A reverse transcription quantitative polymerase chain reaction (RT qPCR) assay and an HPV in situ hybridization assay to examine intra assay concordance. Results: 101 banked SCCHN FFPE tumor specimens were utilized for the qualification study. 42 of 101 (42%) SCCHN specimens (all sites) and 17 of 41 (41%) oropharyngeal specimens were HPV positive by p16INK4A. Five slides from each of 6 SCCHN specimens ranging in the percentage of cells expressing p16INK4A (positive, low positive, and negative) were tested on a single day to demonstrate inter assay repeatability. Serial sections from 6 tumors were tested on 3 consecutive working days using multiple automated staining instruments run by different technicians to determine intra assay reproducibility. Inter pathologist scoring, blinded to run, was demonstrably consistent. Positive p16INK4A staining was demonstrated in blocks stored up to 19 years. Analyte stability was demonstrated in cut sections prepared 27 months in advance. Comparative assay studies yielded consistent, though not identical, results. These results led to the design and implementation of a detailed imaging scoring guideline. Conclusions: The CINtecTM p16INK4A Histology Kit was qualified for use as a surrogate for HPV infection in SCCHN specimens. This assay was then utilized to test banked specimens from a randomized, phase 3 study of the anti EGFR antibody panitumumab in patients with HPV positive R/M SCCHN tumors. In this study, patients with HPV positive tumors did not benefit from the addition panitumumab to a platinum + 5 fluorouracil chemotherapy regimen whereas there was a significant improvement in overall survival in the HPV negative group (Vermorken et al., European Multidisciplinary Cancer Congress 2011). Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 554. doi:1538-7445.AM2012-554


Journal of Cancer Research and Clinical Oncology | 2018

Clinical validation of the next-generation sequencing-based Extended RAS Panel assay using metastatic colorectal cancer patient samples from the phase 3 PRIME study

Nitin Udar; Catherine Lofton-Day; Jun Dong; Darcy Vavrek; A. Scott Jung; Kristen Meier; Anita Iyer; Ryan Slaughter; Karen Gutekunst; Bruce A. Bach; Marc Peeters; Jean-Yves Douillard

PurposeTo validate a next-generation sequencing (NGS)-based companion diagnostic using the MiSeqDx® sequencing instrument to simultaneously detect 56 RAS mutations in DNA extracted from formalin-fixed paraffin-embedded metastatic colorectal cancer (mCRC) tumor samples from the PRIME study. The test’s ability to identify patients with mCRC likely to benefit from panitumumab treatment was assessed.MethodsSamples from PRIME, which compared first-line panitumumab + FOLFOX4 with FOLFOX4, were processed according to predefined criteria using a multiplex assay that included input DNA qualification, library preparation, sequencing, and the bioinformatics reporting pipeline. NGS mutational analysis of KRAS and NRAS exons 2, 3, and 4 was performed and compared with Sanger sequencing.ResultsIn 441 samples, positive percent agreement of the Extended RAS Panel with Sanger sequencing was 98.7% and negative percent agreement was 97.6%. For clinical validation (n = 528), progression-free survival (PFS) and overall survival (OS) were compared between patients with RAS mutations (RAS Positive) and those without (RAS Negative). Panitumumab + FOLFOX4 improved PFS in RAS Negative patients (P = 0.02). Quantitative interaction testing indicated the treatment effect (measured by the hazard ratio of panitumumab + FOLFOX4 versus FOLFOX4) differed for RAS Negative versus RAS Positive for PFS (P = 0.0038) and OS (P = 0.0323).ConclusionsNGS allows for broad, rapid, highly specific analyses of genomic regions. These results support use of the Extended RAS Panel as a companion diagnostic for selecting patients for panitumumab, and utilization is consistent with recent clinical guidelines regarding mCRC RAS testing. Overall, approximately 13% more patients were detected with the Extended RAS Panel versus KRAS exon 2 alone.Clinical trial registry identifierNCT00364013 (ClinicalTrials.gov).

Collaboration


Dive into the Bruce A. Bach's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ignace Vergote

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge