Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Ann Oncol ; 33(10): 1052-1060, 2022 10.
Article in English | MEDLINE | ID: mdl-35764271

ABSTRACT

BACKGROUND: In the phase II multicohort CheckMate 142 study, nivolumab plus low-dose (1 mg/kg) ipilimumab provided robust and durable clinical benefit with a manageable safety profile in previously treated patients with microsatellite instability-high/mismatch repair-deficient (MSI-H/dMMR) metastatic colorectal cancer (mCRC) at 13.4- and 25.4-month median follow-up (Overman MJ, Lonardi S, Wong KYM et al. Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J Clin Oncol. 2018;36:773-779. Overman MJ, Lonardi S, Wong KYM, etĀ al. NivolumabĀ plus low-dose ipilimumab in previously treated patients with microsatellite instability-high/mismatch repair deficient metastatic colorectal cancer: long-term follow-up. J Clin Oncol. 2019;37:635). Here, we present results from the 4-year follow-up of these patients. PATIENTS AND METHODS: Patients received nivolumab (3 mg/kg)Ā plus low-dose (1 mg/kg) ipilimumab every 3 weeks (four doses) followed by nivolumab (3 mg/kg) every 2 weeks until disease progression. Primary endpoint was investigator-assessed objective response rate (ORR; as per RECIST version 1.1). RESULTS: A total of 119 patients were treated; 76% had ≥2 prior lines of therapy. Median follow-up was 50.9 months (range 46.9-62.7 months). Median duration of therapy was 24.9 months [95% confidence interval (CI) 15.8-33.2 months]. Investigator-assessed ORR increased from 55% (95% CI 45% to 64%) at 13.4 months to 65% (95% CI 55% to 73%) at 50.9 months with a disease control rate of 81% (95% CI 72% to 87%). The complete response rate increased from 3% at 13.4 months to 13% at 50.9 months. Partial responses were observed in 52% of patients; 21% had stable disease, and 12% had progressive disease. Median time to response was 2.8 months (range 1.1-37.1 months), and median duration of response was not reached (range 1.4+ to 58.0+ months). At data cut-off, 37 (48%) patients had ongoing responses. Median progression-free survival was not reached [95% CI 38.4 months-not estimable (NE)], and median overall survival was not reached (95% CI NE). Grade 3-4 treatment-related adverse events (TRAEs) were observed in 32% of patients; 13% of patients had any-grade TRAEs leading to discontinuation. CONCLUSIONS: The results confirm long-term benefit of nivolumab plus low-dose ipilimumab for previously treated patients with MSI-H/dMMR mCRC. The safety profile was manageable with no new safety signals.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Colonic Neoplasms/drug therapy , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Mismatch Repair/genetics , Follow-Up Studies , Humans , Ipilimumab , Microsatellite Instability , Nivolumab/therapeutic use
2.
Br J Surg ; 108(8): 968-975, 2021 08 19.
Article in English | MEDLINE | ID: mdl-33829254

ABSTRACT

BACKGROUND: Most current models for predicting survival after resection of colorectal liver metastasis include largest diameter and number of colorectal liver metastases as dichotomous variables, resulting in underestimation of the extent of risk variation and substantial loss of statistical power. The aim of this study was to develop and validate a new prognostic model for patients undergoing liver resection including largest diameter and number of colorectal liver metastases as continuous variables. METHODS: A prognostic model was developed using data from patients who underwent liver resection for colorectal liver metastases at MD Anderson Cancer Center and had RAS mutational data. A Cox proportional hazards model analysis was used to develop a model based on largest colorectal liver metastasis diameter and number of metastases as continuous variables. The model results were shown using contour plots, and validated externally in an international multi-institutional cohort. RESULTS: A total of 810 patients met the inclusion criteria. Largest colorectal liver metastasis diameter (hazard ratio (HR) 1.11, 95 per cent confidence interval 1.06 to 1.16; P < 0.001), number of colorectal liver metastases (HR 1.06, 1.03 to 1.09; P < 0.001), and RAS mutation status (HR 1.76, 1.42 to 2.18; P < 0.001) were significantly associated with overall survival, together with age, primary lymph node metastasis, and prehepatectomy chemotherapy. The model performed well in the external validation cohort, with predicted overall survival values almost lying within 10 per cent of observed values. Wild-type RAS was associated with better overall survival than RAS mutation even when liver resection was performed for larger and/or multiple colorectal liver metastases. CONCLUSION: The contour prognostic model, based on diameter and number of lesions considered as continuous variables along with RAS mutation, predicts overall survival after resection of colorectal liver metastasis.


Subject(s)
Colorectal Neoplasms/pathology , Hepatectomy/methods , Liver Neoplasms/surgery , Colorectal Neoplasms/mortality , Colorectal Neoplasms/surgery , Disease-Free Survival , Female , Follow-Up Studies , Humans , Liver Neoplasms/diagnosis , Liver Neoplasms/secondary , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Metastasis , Prognosis , Retrospective Studies , Survival Rate/trends , United States/epidemiology
3.
Ann Oncol ; 30(10): 1580-1590, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31373349

ABSTRACT

Cell-free DNA fragments are shed into the bloodstream by tumor cells. The analysis of circulating tumor DNA (ctDNA), commonly known as liquid biopsy, can be exploited for a variety of clinical applications. ctDNA is being used to genotype solid cancers non-invasively, to track tumor dynamics and to detect the emergence of drug resistance. In a few settings, liquid biopsies have already entered clinical practice. For example, ctDNA is used to guide treatment in a subset of lung cancers. In this review, we discuss how recent improvements in the sensitivity and accuracy of ctDNA analyses have led to unprecedented advances in this research field. We further consider what is required for the routine deployment of liquid biopsies in the clinical diagnostic space. We pinpoint technical hurdles that liquid biopsies have yet to overcome, including preanalytical and analytical challenges. We foresee how liquid biopsies will transform clinical practice: by complementing (or replacing) imaging to monitor treatment response and by detecting minimal residual disease after surgery with curative intent.


Subject(s)
Biomarkers, Tumor/blood , Circulating Tumor DNA/blood , Clinical Decision-Making , DNA, Neoplasm/blood , Liquid Biopsy/methods , Neoplasms/diagnosis , Practice Patterns, Physicians'/statistics & numerical data , Biomarkers, Tumor/genetics , Circulating Tumor DNA/genetics , DNA, Neoplasm/genetics , Humans , Neoplasms/blood , Neoplasms/genetics , Precision Medicine , Prognosis
5.
Ann Oncol ; 30(2): 243-249, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30462160

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) has been shown to acquire RAS and EGFR ectodomain mutations as mechanisms of resistance to epidermal growth factor receptor (EGFR) inhibition (anti-EGFR). After anti-EGFR withdrawal, RAS and EGFR mutant clones lack a growth advantage relative to other clones and decay; however, the kinetics of decay remain unclear. We sought to determine the kinetics of acquired RAS/EGFR mutations after discontinuation of anti-EGFR therapy. PATIENTS AND METHODS: We present the post-progression circulating tumor DNA (ctDNA) profiles of 135 patients with RAS/BRAF wild-type metastatic CRC treated with anti-EGFR who acquired RAS and/or EGFR mutations during therapy. Our validation cohort consisted of an external dataset of 73 patients with a ctDNA profile suggestive of prior anti-EGFR exposure and serial sampling. A separate retrospective cohort of 80 patients was used to evaluate overall response rate and progression free survival during re-challenge therapies. RESULTS: Our analysis showed that RAS and EGFR relative mutant allele frequency decays exponentially (r2=0.93 for RAS; r2=0.94 for EGFR) with a cumulative half-life of 4.4 months. We validated our findings using an external dataset of 73 patients with a ctDNA profile suggestive of prior anti-EGFR exposure and serial sampling, confirming exponential decay with an estimated half-life of 4.3 months. A separate retrospective cohort of 80 patients showed that patients had a higher overall response rate during re-challenge therapies after increasing time intervals, as predicted by our model. CONCLUSION: These results provide scientific support for anti-EGFR re-challenge and guide the optimal timing of re-challenge initiation.


Subject(s)
Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Neoplastic Cells, Circulating/pathology , Protein Kinase Inhibitors/therapeutic use , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Progression , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Follow-Up Studies , Humans , Mutation , Neoplasm Metastasis , Prognosis , Retrospective Studies , Survival Rate , ras Proteins/genetics
6.
Ann Oncol ; 29(10): 2061-2067, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30412224

ABSTRACT

Background: Gene expression-based profiling of colorectal cancer (CRC) can be used to identify four molecularly homogeneous consensus molecular subtype (CMS) groups with unique biologic features. However, its applicability to colorectal premalignant lesions remains unknown. Patients and methods: We assembled the largest transcriptomic premalignancy dataset by integrating different public and proprietary cohorts of adenomatous and serrated polyps from sporadic (N = 311) and hereditary (N = 78) patient populations and carried out a comprehensive analysis of carcinogenesis pathways using the CMS random forest (RF) classifier. Results: Overall, transcriptomic subtyping of sporadic and hereditary polyps revealed CMS2 and CMS1 subgroups as the predominant molecular subtypes in premalignancy. Pathway enrichment analysis showed that adenomatous polyps from sporadic or hereditary cases (including Lynch syndrome) displayed a CMS2-like phenotype with WNT and MYC activation, whereas hyperplastic and serrated polyps with CMS1-like phenotype harbored prominent immune activation. Rare adenomas with CMS4-like phenotype showed significant enrichment for stromal signatures along with transforming growth factor-Ɵ activation. There was a strong association of CMS1-like polyps with serrated pathology, right-sided anatomic location and BRAF mutations. Conclusions: Based on our observations made in premalignancy, we propose a model of pathway activation associated with CMS classification in colorectal carcinogenesis. Specifically, while adenomatous polyps are largely CMS2, most hyperplastic and serrated polyps are CMS1 and may transition into other CMS groups during evolution into carcinomas. Our findings shed light on the transcriptional landscape of premalignant colonic polyps and may help guide the development of future biomarkers or preventive treatments for CRC.


Subject(s)
Adenoma/diagnosis , Biomarkers, Tumor/genetics , Colonic Polyps/diagnosis , Colorectal Neoplasms/classification , Colorectal Neoplasms/diagnosis , Mutation , Precancerous Conditions/diagnosis , Adenoma/genetics , Colonic Polyps/genetics , Colorectal Neoplasms/genetics , Female , Follow-Up Studies , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Neoplasm Staging , Phenotype , Precancerous Conditions/genetics , Predictive Value of Tests , Prognosis , Transcriptome
7.
Ann Oncol ; 29(1): 139-144, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29069279

ABSTRACT

Background: Hypermethylation of promoter CpG islands [CpG island methylator phenotype (CIMP)] represents a unique pathway for the development of colorectal cancer (CRC), characterized by lack of chromosomal instability and a low rate of adenomatous polyposis coli (APC) mutations, which have both been correlated with taxane resistance. Similarly, small bowel adenocarcinoma (SBA), a rare tumor, also has a low rate of APC mutations. This phase II study evaluated taxane sensitivity in SBA and CIMP-high CRC. Patients and methods: The primary objective was Response Evaluation Criteria in Solid Tumors version 1.1 response rate. Eligibility included Eastern Cooperative Oncology Group performance status 0/1, refractory disease, and SBA or CIMP-high metastatic CRC. Nab-paclitaxel was initially administered at a dose of 260 mg/m2 every 3 weeks but was reduced to 220 mg/m2 owing to toxicity. Results: A total of 21 patients with CIMP-high CRC and 13 with SBA were enrolled from November 2012 to October 2014. The efficacy-assessable population (patients who received at least three doses of the treatment) comprised 15 CIMP-high CRC patients and 10 SBA patients. Common grade 3 or 4 toxicities were fatigue (12%), neutropenia (9%), febrile neutropenia (9%), dehydration (6%), and thrombocytopenia (6%). No responses were seen in the CIMP-high CRC cohort and two partial responses were seen in the SBA cohort. Median progression-free survival was significantly greater in the SBA cohort than in the CIMP-high CRC cohort (3.2 months compared with 2.1 months, P = 0.03). Neither APC mutation status nor CHFR methylation status correlated with efficacy in the CIMP-high CRC cohort. In vivo testing of paclitaxel in an SBA patient-derived xenograft validated the activity of taxanes in this disease type. Conclusion: Although preclinical studies suggested taxane sensitivity was associated with chromosomal stability and wild-type APC, we found that nab-paclitaxel was inactive in CIMP-high metastatic CRC. Nab-paclitaxel may represent a novel therapeutic option for SBA.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Albumins/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Intestine, Small/pathology , Paclitaxel/therapeutic use , Adenocarcinoma/pathology , Adult , Aged , Albumins/adverse effects , Animals , Cell Cycle Proteins/genetics , Colorectal Neoplasms/pathology , CpG Islands , DNA Methylation , DNA Mutational Analysis , Female , Humans , Male , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Proteins/genetics , Paclitaxel/adverse effects , Phenotype , Poly-ADP-Ribose Binding Proteins/genetics , Promoter Regions, Genetic , Ubiquitin-Protein Ligases/genetics , Xenograft Model Antitumor Assays
8.
Ann Oncol ; 29(1): 44-70, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29155929

ABSTRACT

The most recent version of the European Society for Medical Oncology (ESMO) consensus guidelines for the treatment of patients with metastatic colorectal cancer (mCRC) was published in 2016, identifying both a more strategic approach to the administration of the available systemic therapy choices, and a greater emphasis on the use of ablative techniques, including surgery. At the 2016 ESMO Asia Meeting, in December 2016, it was decided by both ESMO and the Japanese Society of Medical Oncology (JSMO) to convene a special guidelines meeting, endorsed by both ESMO and JSMO, immediately after the JSMO 2017 Annual Meeting. The aim was to adapt the ESMO consensus guidelines to take into account the ethnic differences relating to the toxicity as well as other aspects of certain systemic treatments in patients of Asian ethnicity. These guidelines represent the consensus opinions reached by experts in the treatment of patients with mCRC identified by the Presidents of the oncological societies of Japan (JSMO), China (Chinese Society of Clinical Oncology), Korea (Korean Association for Clinical Oncology), Malaysia (Malaysian Oncological Society), Singapore (Singapore Society of Oncology) and Taiwan (Taiwan Oncology Society). The voting was based on scientific evidence and was independent of both the current treatment practices and the drug availability and reimbursement situations in the individual participating Asian countries.


Subject(s)
Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/therapy , Asian People , China , Colorectal Neoplasms/ethnology , Colorectal Neoplasms/pathology , Humans , Malaysia , Neoplasm Metastasis , Republic of Korea , Taiwan
9.
Br J Surg ; 105(6): 618-627, 2018 05.
Article in English | MEDLINE | ID: mdl-29579319

ABSTRACT

BACKGROUND: Despite a 5-year overall survival rate of 58 per cent after liver resection for colorectal liver metastases (CLMs), more than half of patients develop recurrence, highlighting the need for accurate risk stratification and prognostication. Traditional prognostic factors have been superseded by newer outcome predictors, including those defined by the molecular origin of the primary tumour. METHODS: This review synthesized findings in the literature using the PubMed database of articles in the English language published between 1998 and 2017 on prognostic and predictive biomarkers in patients undergoing resection of CLMs. RESULTS: Responses to preoperative chemotherapy define prognosis in patients undergoing CLM resection. There are differences by embryological origin too. Somatic mutations in the proto-oncogenes KRAS and NRAS are associated with positive surgical margins and tumour regrowth after ablation. Other mutations (such as BRAF) and co-occurring mutations in RAS/TP53 and APC/PIK3CA have emerged as important biomarkers that determine an individual patient's tumour biology and may be used to predict outcome after CLM resection. CONCLUSION: Knowledge of somatic mutations can guide the use of preoperative therapy, extent of surgical margin and selection for ablation alone.


Subject(s)
Colorectal Neoplasms/pathology , Liver Neoplasms/secondary , Biomarkers, Tumor , Colorectal Neoplasms/diagnosis , Genetic Markers , Humans , Liver Neoplasms/diagnosis , Liver Neoplasms/surgery , Microsatellite Instability , Prognosis , Recurrence , ras Proteins
10.
Br J Surg ; 105(9): 1200-1209, 2018 08.
Article in English | MEDLINE | ID: mdl-29664996

ABSTRACT

BACKGROUND: Although perihepatic lymph node metastases (PLNMs) are known to be a poor prognosticator for patients with colorectal liver metastases (CRLMs), optimal management remains unclear. This study aimed to determine the risk factors for PLNMs, and the survival impact of their number and location in patients with resectable CRLMs. METHODS: Data on patients with CRLM who underwent hepatectomy during 2003-2014 were analysed retrospectively. Recurrence-free (RFS) and overall (OS) survival were calculated according to presence, number and location of PLNMs. Risk factors for PLNM were evaluated by logistic regression analysis. RESULTS: Of 1485 patients, 174 underwent lymphadenectomy, and 54 (31Ā·0 per cent) had PLNM. Ten patients (5Ā·7 per cent) who had lymphadenectomy and 176 (13Ā·4 per cent) who did not underwent repeat hepatectomy. Survival of patients with PLNM was significantly poorer than that of patients without (RFS: 5Ā·3 versus 13Ā·8 months, P < 0Ā·001; OS: 20Ā·5 versus 71Ā·3 months; P < 0Ā·001). Median OS was significantly better in patients with para-aortic versus hepatoduodenal ligament PLNMs (58Ā·2 versus 15Ā·5 months; P = 0Ā·011). Patients with three or more PLNMs had significantly worse median OS than those with one or two (16Ā·3 versus 25Ā·4 months; P = 0Ā·039). The presence of primary tumour lymph node metastases (odds ratio 2Ā·35; P = 0Ā·037) and intrahepatic recurrence requiring repeat hepatectomy (odds ratio 5Ā·61; P = 0Ā·012) were significant risk factors for PLNM on multivariable analysis. CONCLUSION: Patients undergoing repeat hepatectomy and those with primary tumour lymph node metastases are at significant risk of PLNM. Although PLNM is a poor prognostic factor independent of perihepatic lymph node station, patients with one or two PLNMs have a more favourable outcome than those with more PLNMs.


Subject(s)
Colorectal Neoplasms/surgery , Hepatectomy , Liver Neoplasms/surgery , Neoplasm Staging , Aged , Biopsy , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Female , Follow-Up Studies , Humans , Liver Neoplasms/diagnosis , Liver Neoplasms/secondary , Lymphatic Metastasis , Male , Prognosis , Prospective Studies , Survival Rate/trends , Texas/epidemiology , Tomography, X-Ray Computed
11.
Ann Oncol ; 28(3): 468-477, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27998963

ABSTRACT

Current trajectory of clinical care is heading in the direction of personalized medicine. In an ideal scenario, clinicians can obtain extensive diagnostic and prognostic information via minimally-invasive assays. Information available in the peripheral blood has the potential to bring us closer to this goal. In this review we highlight the contributions of circulating tumor cells and circulating tumor DNA and RNA (ctDNA/ctRNA) towards cancer therapeutic field. We discuss clinical relevance, summarize available and upcoming technologies, and hypothesize how future care could be impacted by a combined study.


Subject(s)
Biomarkers, Tumor/blood , Circulating Tumor DNA/blood , Neoplasms/blood , Neoplastic Cells, Circulating/metabolism , DNA, Neoplasm/genetics , Humans , Mutation , Neoplasms/genetics , Precision Medicine
12.
Br J Surg ; 104(6): 760-768, 2017 May.
Article in English | MEDLINE | ID: mdl-28240361

ABSTRACT

BACKGROUND: Percutaneous ablation is a common treatment for colorectal liver metastasis (CLM). However, the effect of rat sarcoma viral oncogene homologue (RAS) mutation on outcome after ablation of CLMs is unclear. METHODS: Patients who underwent image-guided percutaneous ablation of CLMs from 2004 to 2015 and had known RAS mutation status were analysed. Patients were evaluated for local tumour progression as observed on imaging of CLMs treated with ablation. Multivariable Cox regression analysis was performed to determine factors associated with local tumour progression-free survival. RESULTS: The study included 92 patients who underwent ablation of 137 CLMs. Thirty-six patients (39 per cent) had mutant RAS. Rates of local tumour progression were 14 per cent (8 of 56) for patients with wild-type RAS and 39 per cent (14 of 36) for patients with mutant RAS (P = 0Ā·007). The actuarial 3-year local tumour progression-free survival rate after percutaneous ablation was worse in patients with mutant RAS than in those with wild-type RAS (35 versus 71 per cent respectively; P = 0Ā·001). In multivariable analysis, negative predictors of local tumour progression-free survival were a minimum ablation margin of less than 5 mm (hazard ratio (HR) 2Ā·48, 95 per cent c.i. 1Ā·31 to 4Ā·72; P = 0Ā·006) and mutant RAS (HR 3Ā·01, 1Ā·60 to 5Ā·77; P = 0Ā·001). CONCLUSION: Mutant RAS is associated with an earlier and higher rate of local tumour progression in patients undergoing ablation of CLMs.


Subject(s)
Catheter Ablation/methods , Colonic Neoplasms/genetics , Genes, ras/genetics , Liver Neoplasms/genetics , Mutation/genetics , Rectal Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Disease Progression , Disease-Free Survival , Female , Humans , Liver Neoplasms/secondary , Liver Neoplasms/surgery , Lymphatic Metastasis , Male , Margins of Excision , Middle Aged , Neoplasm Recurrence, Local/genetics , Retrospective Studies , Surgery, Computer-Assisted/methods , Treatment Outcome
13.
Br J Cancer ; 114(1): 103-9, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26679375

ABSTRACT

BACKGROUND: In contrast to the consistent evidence for obesity and colorectal cancer (CRC) risk, the impact of obesity in CRC patients is less clear. In a well-characterised cohort of CRC patients, we prospectively evaluated class I and class II obesity with survival outcomes. METHODS: The CRC patients (N=634) were followed from the date of diagnosis until disease progression/first recurrence (progression-free survival (PFS)) or death (overall survival (OS)). Body mass index (BMI) was calculated from reported usual weight prior to diagnosis. Hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated in models adjusted for clinicopathologic, treatment, and lifestyle factors. RESULTS: Over a median follow-up of 4 years, 208 (33%) patients died and 235 (37%) recurred or progressed. Class II obesity, as compared with either overweight or normal weight, was associated with an increased risk of death (HR and 95% CI: 1.55 (0.97-2.48) and 1.65 (1.02-2.68), respectively), but no clear association was observed with PFS. In analyses restricted to patients who presented as stages I-III, who reported stable weight, or who were aged <50 years, obesity was associated with a significant two- to five-fold increased risk of death. CONCLUSIONS: In CRC patients evaluated at a large cancer centre, severely obese patients experienced worse survival outcomes independent of many other factors.


Subject(s)
Colorectal Neoplasms/mortality , Obesity/complications , Adult , Aged , Body Mass Index , Female , Humans , Male , Middle Aged , Neoplasm Staging , Prospective Studies
14.
Ann Oncol ; 27(6): 1068-1074, 2016 06.
Article in English | MEDLINE | ID: mdl-27045102

ABSTRACT

BACKGROUND: Incorporation of multiple enrichment biomarkers into prospective clinical trials is an active area of investigation, but the factors that determine clinical trial enrollment following a molecular prescreening program have not been assessed. PATIENTS AND METHODS: Patients with 5-fluorouracil-refractory metastatic colorectal cancer at the MD Anderson Cancer Center were offered screening in the Assessment of Targeted Therapies Against Colorectal Cancer (ATTACC) program to identify eligibility for companion phase I or II clinical trials with a therapy targeted to an aberration detected in the patient, based on testing by immunohistochemistry, targeted gene sequencing panels, and CpG island methylation phenotype assays. RESULTS: Between August 2010 and December 2013, 484 patients were enrolled, 458 (95%) had a biomarker result, and 157 (32%) were enrolled on a clinical trial (92 on biomarker-selected and 65 on nonbiomarker selected). Of the 458 patients with a biomarker result, enrollment on biomarker-selected clinical trials was ninefold higher for predefined ATTACC-companion clinical trials as opposed to nonpredefined biomarker-selected clinical trials, 17.9% versus 2%, P < 0.001. Factors that correlated positively with trial enrollment in multivariate analysis were higher performance status, older age, lack of standard of care therapy, established patient at MD Anderson, and the presence of an eligible biomarker for an ATTACC-companion study. Early molecular screening did result in a higher rate of patients with remaining standard of care therapy enrolling on ATTACC-companion clinical trials, 45.1%, in contrast to nonpredefined clinical trials, 22.7%; odds ratio 3.1, P = 0.002. CONCLUSIONS: Though early molecular prescreening for predefined clinical trials resulted in an increase rate of trial enrollment of nonrefractory patients, the majority of patients enrolled on clinical trials were refractory to standard of care therapy. Within molecular prescreening programs, tailoring screening for preidentified and open clinical trials, temporally linking screening to treatment and optimizing both patient and physician engagement are efforts likely to improve enrollment on biomarker-selected clinical trials. CLINICAL TRIALS NUMBER: The study NCT number is NCT01196130.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , DNA Methylation/genetics , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/pathology , CpG Islands/genetics , Eligibility Determination , Female , Fluorouracil/therapeutic use , Humans , Male , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Staging , Patient Selection
15.
Ann Oncol ; 27(5): 795-800, 2016 05.
Article in English | MEDLINE | ID: mdl-26787237

ABSTRACT

BACKGROUND: Next-generation sequencing in cancer research may reveal germline variants of clinical significance. We report patient preferences for return of results and the prevalence of incidental pathogenic germline variants (PGVs). PATIENTS AND METHODS: Targeted exome sequencing of 202 genes was carried out in 1000 advanced cancers using tumor and normal DNA in a research laboratory. Pathogenic variants in 18 genes, recommended for return by The American College of Medical Genetics and Genomics, as well as PALB2, were considered actionable. Patient preferences of return of incidental germline results were collected. Return of results was initiated with genetic counseling and repeat CLIA testing. RESULTS: Of the 1000 patients who underwent sequencing, 43 had likely PGVs: APC (1), BRCA1 (11), BRCA2 (10), TP53 (10), MSH2 (1), MSH6 (4), PALB2 (2), PTEN (2), TSC2 (1), and RB1 (1). Twenty (47%) of 43 variants were previously known based on clinical genetic testing. Of the 1167 patients who consented for a germline testing protocol, 1157 (99%) desired to be informed of incidental results. Twenty-three previously unrecognized mutations identified in the research environment were confirmed with an orthogonal CLIA platform. All patients approached decided to proceed with formal genetic counseling; in all cases where formal genetic testing was carried out, the germline variant of concern validated with clinical genetic testing. CONCLUSIONS: In this series, 2.3% patients had previously unrecognized pathogenic germline mutations in 19 cancer-related genes. Thus, genomic sequencing must be accompanied by a plan for return of germline results, in partnership with genetic counseling.


Subject(s)
Germ-Line Mutation/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Exome/genetics , Genetic Counseling , High-Throughput Nucleotide Sequencing , Humans , Neoplasms/diagnosis , Neoplasms/pathology
16.
Br J Cancer ; 112(3): 424-8, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25535726

ABSTRACT

BACKGROUND: KRAS mutations have been associated with lung metastases at diagnosis of metastatic colorectal cancer (mCRC), but the impact of this mutation on subsequent development of lung metastasis is unknown. We investigated KRAS mutation as a predictor of lung metastasis development. METHODS: We retrospectively evaluated data from patients with mCRC whose tumour was tested for KRAS mutation from 2008 to 2010. The relationships of KRAS mutational status with time-to-lung metastasis (TTLM) and overall survival (OS) were analysed. RESULTS: Of the 494 patients identified, 202 (41%) had tumours with KRAS mutation. KRAS mutations were associated with a shorter TTLM (median 15.2 vs 22.4 months; hazard ratio=1.40; P=0.002) and a two-fold greater odds of developing lung metastases during the disease course in patients with liver-limited mCRC at diagnosis (72 vs 56%, P=0.007). Overall survival did not differ by KRAS status. CONCLUSIONS: Lung metastasis was more likely to develop during the disease course in patients whose tumour had a KRAS mutation than in those whose tumour did not have a KRAS mutation. This finding may have an impact on decision making for surgical resection of metastatic disease.


Subject(s)
Colorectal Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Mutation , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Disease Progression , Female , Genetic Association Studies , Humans , Lung Neoplasms/mortality , Male , Middle Aged , Proto-Oncogene Proteins p21(ras) , Retrospective Studies
17.
Br J Cancer ; 112(3): 539-46, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25535733

ABSTRACT

BACKGROUND: Isolation of colorectal cancer (CRC) cell populations enriched for cancer stem cells (CSCs) may facilitate target identification. There is no consensus regarding the best methods for isolating CRC stem cells (CRC-SCs). We determined the suitability of various cellular models and various stem cell markers for the isolation of CRC-SCs. METHODS: Established human CRC cell lines, established CRC cell lines passaged through mice, patient-derived xenograft (PDX)-derived cells, early passage/newly established cell lines, and cells directly from clinical specimens were studied. Cells were FAC-sorted for the CRC-SC markers CD44, CD133, and aldehyde dehydrogenase (ALDH). Sphere formation and in vivo tumorigenicity studies were used to validate CRC-SC enrichment. RESULTS: None of the markers studied in established cell lines, grown either in vitro or in vivo, consistently enriched for CRC-SCs. In the three other cellular models, CD44 and CD133 did not reliably enrich for stemness. In contrast, freshly isolated PDX-derived cells or early passage/newly established CRC cell lines with high ALDH activity formed spheres in vitro and enhanced tumorigenicity in vivo, whereas cells with low ALDH activity did not. CONCLUSIONS: PDX-derived cells, early passages/newly established CRC cell lines and cells from clinical specimen with high ALDH activity can be used to identify CRC-SC-enriched populations. Established CRC cell lines should not be used to isolate CSCs.


Subject(s)
Colorectal Neoplasms/pathology , Neoplastic Stem Cells/pathology , Primary Cell Culture/methods , Animals , Cell Proliferation , Cell Separation/methods , Coculture Techniques , HCT116 Cells , HT29 Cells , Humans , Hyaluronan Receptors/metabolism , Mice , Mice, Nude , Neoplastic Stem Cells/physiology , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Tumor Cells, Cultured
18.
Br J Cancer ; 112(6): 1088-97, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25688736

ABSTRACT

BACKGROUND: High circulating neutrophil-lymphocyte ratio (NLR) appears to be prognostic in metastatic colorectal cancer (mCRC). We investigated the relationship of NLR with circulating cytokines and molecular alterations. METHODS: We performed retrospective analyses on multiple cohorts of CRC patients (metastatic untreated (n=166), refractory metastatic (n=161), hepatectomy (n=198), stage 2/3 (n=274), and molecularly screened (n=342)). High NLR (ratio of absolute neutrophil-to-lymphocyte counts in peripheral blood) was defined as NLR>5. Plasma cytokines were evaluated using multiplex-bead assays. Kaplan-Meier estimates, non-parametric correlation analysis, and hierarchical cluster analyses were used. RESULTS: High NLR was associated with poor prognosis in mCRC (hazard ratio (HR) 1.73; 95% confidence interval (CI):1.03-2.89; P=0.039) independent of known prognostic factors and molecular alterations (KRAS/NRAS/BRAF/PIK3CA/CIMP). High NLR correlated with increased expression of interleukin 6 (IL-6), IL-8, IL-2Rα, hepatocyte growth factor, macrophage-colony stimulating factor, and vascular epidermal growth factor in exploratory (n=39) and validation (n=166) cohorts. Fourteen additional cytokines correlated with high NLR in the validation cohort. All 20 cytokines fell into three major clusters: inflammatory cytokines, angiogenic cytokines, and epidermal growth factor ligands. In mCRC, composite stratification based on NLR-cytokine score provided enhanced prognostic information (HR 2.09; 95% CI: 1.59-2.76; P<0.001) over and above NLR. CONCLUSIONS: High NLR is an independent poor prognostic marker in CRC and correlates with a distinct cytokine profile related to key biological processes involved in carcinogenesis. A composite NLR-cytokine stratification has enhanced prognostic value in mCRC.


Subject(s)
Colorectal Neoplasms/immunology , Cytokines/blood , Lymphocytes/pathology , Neutrophils/pathology , Adult , Aged , Cohort Studies , Colorectal Neoplasms/blood , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Cytokines/immunology , Female , Humans , Kaplan-Meier Estimate , Leukocyte Count/methods , Lymphocytes/immunology , Male , Middle Aged , Neoplasm Metastasis , Neutrophils/immunology , Prognosis , Retrospective Studies
19.
Ann Oncol ; 26(4): 731-736, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25628445

ABSTRACT

INTRODUCTION: KRAS and EGFR ectodomain-acquired mutations in patients with metastatic colorectal cancer (mCRC) have been correlated with acquired resistance to anti-EGFR monoclonal antibodies (mAbs). We investigated the frequency, co-occurrence, and distribution of acquired KRAS and EGFR mutations in patients with mCRC refractory to anti-EGFR mAbs using circulating tumor DNA (ctDNA). PATIENTS AND METHODS: Sixty-two post-treatment plasma and 20 matching pretreatment archival tissue samples from KRAS (wt) mCRC patients refractory to anti-EGFR mAbs were evaluated by high-sensitivity emulsion polymerase chain reaction for KRAS codon 12, 13, 61, and 146 and EGFR 492 mutations. RESULTS: Plasma analyses showed newly detectable EGFR and KRAS mutations in 5/62 [8%; 95% confidence interval (CI) 0.02-0.18] and 27/62 (44%; 95% CI 0.3-0.56) samples, respectively. KRAS codon 61 and 146 mutations were predominant (33% and 11%, respectively), and multiple EGFR and/or KRAS mutations were detected in 11/27 (41%) cases. The percentage of mutant allele reads was inversely correlated with time since last treatment with EGFR mAbs (P = 0.038). In the matching archival tissue, these mutations were detectable as low-allele-frequency clones in 35% of patients with plasma mutations after treatment with anti-EGFR mAbs and correlated with shorter progression-free survival (PFS) compared with the cases with no new mutations (3.0 versus 8.0 months, P = 0.0004). CONCLUSION: Newly detected KRAS and/or EGFR mutations in plasma ctDNA from patients refractory to anti-EGFR treatment appear to derive from rare, pre-existing clones in the primary tumors. These rare clones were associated with shorter PFS in patients receiving anti-EGFR treatment. Multiple simultaneous mutations in KRAS and EGFR in the ctDNA and the decline in allele frequency after discontinuation of anti-EGFR therapy in a subset of patients suggest that several resistance mechanisms can co-exist and that relative clonal burdens may change over time. Monitoring treatment-induced genetic alterations by sequencing ctDNA could identify biomarkers for treatment screening in anti-EGFR-refractory patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , Mutation/genetics , Neoplastic Cells, Circulating/pathology , Clone Cells , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , DNA, Neoplasm/blood , DNA, Neoplasm/genetics , ErbB Receptors/blood , ErbB Receptors/genetics , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , Proto-Oncogene Proteins/blood , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/blood , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras) , Retrospective Studies , Survival Rate , ras Proteins/blood , ras Proteins/genetics
20.
Invest New Drugs ; 33(4): 977-84, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26062928

ABSTRACT

BACKGROUND: Src has a critical role in tumor cell migration and invasion. Increased Src activity has been shown to correlate with disease progression and poor prognosis, suggesting Src could serve as a therapeutic target for kinase inhibition. Saracatinib (AZD0530) is a novel selective oral Src kinase inhibitor. METHODS: Metastatic colorectal cancer patients who had received one prior treatment and had measurable disease were enrolled in this phase 2 study. Saracatinib was administered at 175 mg by mouth daily for 28 day cycles until dose-limiting toxicity or progression as determined by staging every 2 cycles. The primary endpoint was improvement in 4 month progression-free survival. Design of Thall, Simon, and Estey was used to monitor proportion of patients that were progression free at 4 months. The trial was opened with plan to enroll maximum of 35 patients, with futility assessment every 10 patients. RESULTS: A total of 10 patients were enrolled between January and November 2007. Further enrollment was stopped due to futility. Median progression-free survival was 7.9 weeks, with all 10 patients showing disease progression following radiographic imaging. Median overall survival was 13.5 months. All patients were deceased by time of analysis. Observed adverse events were notable for a higher than expected number of patients with grade 3 hypophosphatemia (n = 5). CONCLUSION: Saracatinib is a novel oral Src kinase inhibitor that was well tolerated but failed to meet its primary endpoint of improvement in 4 month progression-free survival as a single agent in previously treated metastatic colorectal cancer patients.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Benzodioxoles/therapeutic use , Colorectal Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Adenocarcinoma/blood , Adenocarcinoma/pathology , Aged , Antineoplastic Agents/adverse effects , Benzodioxoles/adverse effects , Colorectal Neoplasms/blood , Colorectal Neoplasms/pathology , Disease-Free Survival , Female , Humans , Male , Middle Aged , Protein Kinase Inhibitors/adverse effects , Quinazolines/adverse effects , Vascular Endothelial Growth Factor A/blood , src-Family Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL