Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Clin Chem ; 70(1): 339-349, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38175591

ABSTRACT

BACKGROUND: B-cell maturation antigen is a pivotal therapeutic target for multiple myeloma (MM). Membrane-bound BCMA can be cleaved by γ-secretase and shed as soluble BCMA (sBCMA). sBCMA can act as a neutralizing sink to compete with drug, as well as serve as a diagnostic/prognostic biomarker for MM. Antibody-capture based methods, such as enzyme-linked immunosorbent assay (ELISA) and immunoaffinity-liquid chromatography-multiple reaction monitoring (IA-LC-MRM), have been reported and well adopted to measure sBCMA in clinical samples. However, both methods are biased by capturing antibodies. METHODS: We have used various LC-MS workflows to characterize and quantify endogenous sBCMA in MM patient samples, including bottom-up peptide mapping, intact analysis, IA-based, and reagent-free (RF)-LC-MRM quantitation. RESULTS: We have confirmed that sBCMA contains a variable N-terminus and a C-terminus that extends to the transmembrane domain, ending at amino acid 61. Leveraging an in-house synthesized G-1-61 sBCMA recombinant standard, we developed a RF-LC-MRM method for unbiased sBCMA quantitation in MM patient samples. By comparing the results from RF-LC-MRM with ELISA and IA-LC-MRM, we demonstrated that RF-LC-MRM measures a more complete pool of endogenous sBCMA compared to the antibody-based methods. CONCLUSIONS: This work fills the knowledge gap of the exact sequence of endogenous sBCMA for the first time, which differs from the current commercially available standard. Additionally, this work highlights the necessity of identifying the actual sequence of an endogenous soluble target such as sBCMA, both for bioanalytical purposes and to underpin pharmacodynamic measurements.


Subject(s)
B-Cell Maturation Antigen , Multiple Myeloma , Humans , Chromatography, Liquid , Liquid Chromatography-Mass Spectrometry , Multiple Myeloma/diagnosis , Tandem Mass Spectrometry , Antibodies
2.
J Clin Oncol ; 41(3): 460-471, 2023 01 20.
Article in English | MEDLINE | ID: mdl-36351210

ABSTRACT

PURPOSE: Acquired resistance to anti-epidermal growth factor receptor (EGFR) inhibitor (EGFRi) therapy in colorectal cancer (CRC) has previously been explained by the model of acquiring new mutations in KRAS/NRAS/EGFR, among other MAPK-pathway members. However, this was primarily on the basis of single-agent EGFRi trials and little is known about the resistance mechanisms of EGFRi combined with effective cytotoxic chemotherapy in previously untreated patients. METHODS: We analyzed paired plasma samples from patients with RAS/BRAF/EGFR wild-type metastatic CRC enrolled in three large randomized trials evaluating EGFRi in the first line in combination with chemotherapy and as a single agent in third line. The mutational signature of the alterations acquired with therapy was evaluated. CRC cell lines with resistance to cetuximab, infusional fluorouracil, leucovorin, and oxaliplatin, and SN38 were developed, and transcriptional changes profiled. RESULTS: Patients whose tumors were treated with and responded to EGFRi alone were more likely to develop acquired mutations (46%) compared with those treated in combination with cytotoxic chemotherapy (9%). Furthermore, contrary to the generally accepted hypothesis of the clonal evolution of acquired resistance, we demonstrate that baseline resistant subclonal mutations rarely expanded to become clonal at progression, and most remained subclonal or disappeared. Consistent with this clinical finding, preclinical models with acquired resistance to either cetuximab or chemotherapy were cross-resistant to the alternate agents, with transcriptomic profiles consistent with epithelial-to-mesenchymal transition. By contrast, commonly acquired resistance alterations in the MAPK pathway do not affect sensitivity to cytotoxic chemotherapy. CONCLUSION: These findings support a model of resistance whereby transcriptomic mechanisms of resistance predominate in the presence of active cytotoxic chemotherapy combined with EGFRi, with a greater predominance of acquired MAPK mutations after single-agent EGFRi. The proposed model has implications for prospective studies evaluating EGFRi rechallenge strategies guided by acquired MAPK mutations, and highlights the need to address transcriptional mechanisms of resistance.


Subject(s)
Colorectal Neoplasms , ErbB Receptors , Humans , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cetuximab/pharmacology , Cetuximab/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , ErbB Receptors/antagonists & inhibitors , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Mutation , Prospective Studies , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Drug Resistance, Neoplasm
3.
Nature ; 599(7886): 679-683, 2021 11.
Article in English | MEDLINE | ID: mdl-34759319

ABSTRACT

Inactive state-selective KRAS(G12C) inhibitors1-8 demonstrate a 30-40% response rate and result in approximately 6-month median progression-free survival in patients with lung cancer9. The genetic basis for resistance to these first-in-class mutant GTPase inhibitors remains under investigation. Here we evaluated matched pre-treatment and post-treatment specimens from 43 patients treated with the KRAS(G12C) inhibitor sotorasib. Multiple treatment-emergent alterations were observed across 27 patients, including alterations in KRAS, NRAS, BRAF, EGFR, FGFR2, MYC and other genes. In preclinical patient-derived xenograft and cell line models, resistance to KRAS(G12C) inhibition was associated with low allele frequency hotspot mutations in KRAS(G12V or G13D), NRAS(Q61K or G13R), MRAS(Q71R) and/or BRAF(G596R), mirroring observations in patients. Single-cell sequencing in an isogenic lineage identified secondary RAS and/or BRAF mutations in the same cells as KRAS(G12C), where they bypassed inhibition without affecting target inactivation. Genetic or pharmacological targeting of ERK signalling intermediates enhanced the antiproliferative effect of G12C inhibitor treatment in models with acquired RAS or BRAF mutations. Our study thus suggests a heterogenous pattern of resistance with multiple subclonal events emerging during G12C inhibitor treatment. A subset of patients in our cohort acquired oncogenic KRAS, NRAS or BRAF mutations, and resistance in this setting may be delayed by co-targeting of ERK signalling intermediates. These findings merit broader evaluation in prospective clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Acetonitriles/pharmacology , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line , Cohort Studies , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Humans , MAP Kinase Signaling System/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Piperazines/pharmacology , Piperazines/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Pyridines/pharmacology , Pyridines/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Xenograft Model Antitumor Assays
4.
N Engl J Med ; 384(25): 2371-2381, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34096690

ABSTRACT

BACKGROUND: Sotorasib showed anticancer activity in patients with KRAS p.G12C-mutated advanced solid tumors in a phase 1 study, and particularly promising anticancer activity was observed in a subgroup of patients with non-small-cell lung cancer (NSCLC). METHODS: In a single-group, phase 2 trial, we investigated the activity of sotorasib, administered orally at a dose of 960 mg once daily, in patients with KRAS p.G12C-mutated advanced NSCLC previously treated with standard therapies. The primary end point was objective response (complete or partial response) according to independent central review. Key secondary end points included duration of response, disease control (defined as complete response, partial response, or stable disease), progression-free survival, overall survival, and safety. Exploratory biomarkers were evaluated for their association with response to sotorasib therapy. RESULTS: Among the 126 enrolled patients, the majority (81.0%) had previously received both platinum-based chemotherapy and inhibitors of programmed death 1 (PD-1) or programmed death ligand 1 (PD-L1). According to central review, 124 patients had measurable disease at baseline and were evaluated for response. An objective response was observed in 46 patients (37.1%; 95% confidence interval [CI], 28.6 to 46.2), including in 4 (3.2%) who had a complete response and in 42 (33.9%) who had a partial response. The median duration of response was 11.1 months (95% CI, 6.9 to could not be evaluated). Disease control occurred in 100 patients (80.6%; 95% CI, 72.6 to 87.2). The median progression-free survival was 6.8 months (95% CI, 5.1 to 8.2), and the median overall survival was 12.5 months (95% CI, 10.0 to could not be evaluated). Treatment-related adverse events occurred in 88 of 126 patients (69.8%), including grade 3 events in 25 patients (19.8%) and a grade 4 event in 1 (0.8%). Responses were observed in subgroups defined according to PD-L1 expression, tumor mutational burden, and co-occurring mutations in STK11, KEAP1, or TP53. CONCLUSIONS: In this phase 2 trial, sotorasib therapy led to a durable clinical benefit without new safety signals in patients with previously treated KRAS p.G12C-mutated NSCLC. (Funded by Amgen and the National Institutes of Health; CodeBreaK100 ClinicalTrials.gov number, NCT03600883.).


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Piperazines/therapeutic use , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Biomarkers/blood , Carcinoma, Non-Small-Cell Lung/genetics , Female , Humans , Lung Neoplasms/genetics , Male , Middle Aged , Mutation , Piperazines/adverse effects , Progression-Free Survival , Pyridines/adverse effects , Pyrimidines/adverse effects
5.
Cancer Biol Ther ; 21(10): 891-898, 2020 10 02.
Article in English | MEDLINE | ID: mdl-33026965

ABSTRACT

BACKGROUND: Antibodies against epidermal growth factor receptor (EGFR), panitumumab, a fully human monoclonal antibody, and cetuximab, a human/mouse chimeric monoclonal antibody, have shown clinical efficacy in metastatic colorectal cancer (mCRC). In the phase 3 noninferiority ASPECCT (ClinicalTrials.gov, NCT01001377) study, panitumumab was demonstrated to be noninferior to cetuximab and provided a similar overall survival benefit for patients with chemotherapy-refractory wild-type KRAS exon 2 mCRC. However, some patients eventually develop resistance to anti-EGFR therapy. EGFR p.S492R mutation was previously identified as conferring resistance to cetuximab, but not to panitumumab. METHODS: This biomarker study analyzed plasma samples from ASPECCT collected at both baseline and posttreatment. RESULTS: No EGFR p.S492R mutations were identified at baseline; however, after treatment the EGFR p.S492R mutation was detected in 1% of patients treated with panitumumab versus 16% of those treated with cetuximab, supporting that, in a large population, this mutation is more likely to be induced by cetuximab than by panitumumab. There were, however, no significant differences in progression-free survival or overall survival between patients who were wild-type compared with those with the S492R mutation within the cetuximab arm or the overall population. CONCLUSIONS: These results may support targeting treatment to small patient subgroups based on the presence of emerging EGFR mutations and provide a molecular rationale for rechallenging with a different anti-EGFR agent in patients who develop resistance. Prospective studies are needed to evaluate the efficacy of panitumumab in the EGFR p.S492R mutant population.


Subject(s)
Cetuximab/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Panitumumab/therapeutic use , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , ErbB Receptors/genetics , Female , Humans , Male , Mutation , Neoplasm Metastasis , Survival Analysis
6.
Clin Cancer Res ; 25(4): 1216-1225, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30487126

ABSTRACT

PURPOSE: Mutations in EGFR pathway genes are poor prognostic indicators in patients with metastatic colorectal cancer. Plasma analysis of cell-free DNA is a minimally invasive and highly sensitive method to detect somatic mutations in tumors. EXPERIMENTAL DESIGN: Plasma samples collected from panitumumab-treated patients in the ASPECCT study at baseline and safety follow-up (SFU) were analyzed by a next-generation sequencing-based approach for extended RAS mutant allele frequency as a continuous variable and their association with clinical outcomes and the mutational prevalence of 63 cancer-related genes. The correlation between patient outcome and baseline mutational status of EGFR pathway genes was also examined. RESULTS: Overall, 261 patients in the panitumumab arm had evaluable plasma samples. Patients with a higher RAS mutant allele frequency at baseline had worse clinical outcomes than those with a lower frequency (P < 0.001, Cox PH model); however, RAS mutations did not necessarily preclude patients from deriving benefits. The objective response rate (complete or partial response) was 10.8% for patients with baseline RAS mutations and 21.7% for those with BRAF mutations. The 63-gene panel analysis revealed an increase in tumor mutational burden from baseline to SFU (P < 0.001, Wilcoxon signed rank test). Baseline mutations in EGFR pathway genes, when analyzed both categorically and continuously, were associated with shorter survival. CONCLUSIONS: When mutations in EGFR pathway genes were analyzed continuously, higher mutant allele frequency correlated with poorer outcomes. However, extended RAS mutation, by itself, did not preclude clinical responses to panitumumab in a monotherapy setting.


Subject(s)
Cell-Free Nucleic Acids/blood , Colorectal Neoplasms/drug therapy , Panitumumab/administration & dosage , Adult , Biomarkers, Tumor/blood , Colorectal Neoplasms/blood , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease-Free Survival , ErbB Receptors/blood , ErbB Receptors/genetics , Female , Gene Frequency , Genetic Heterogeneity , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Mutant Proteins/blood , Mutant Proteins/genetics , Mutation , Neoplasm Metastasis , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction/drug effects , Treatment Outcome
7.
PLoS One ; 13(6): e0198329, 2018.
Article in English | MEDLINE | ID: mdl-29912960

ABSTRACT

BACKGROUND: Despite a more proactive approach to reducing new HIV infections in infants through lifelong treatment (Option B+ policy) for infected pregnant women, prevention of mother-to-child transmission of HIV (PMTCT) has not been fully effective in Papua, Indonesia. Mother-to-child transmission (MTCT) is the second greatest risk factor for HIV infection in the community, and an elimination target of <1% MTCT has not yet been achieved. The purpose of this study was to improve understanding of the implementation of Option B+ for PMTCT in Papua through investigation of facilitators and barriers to women's uptake and adherence to antiretroviral therapy (ART) in the program. This information is vital for improving program outcomes and success of program scale up in similar settings in Papua. METHODS: In-depth interviews were conducted with 20 women and 20 PMTCT health workers at two main referral hospitals for PMTCT in Papua. Development of interview guides was informed by the socio-ecological framework. Qualitative data were managed with NVivo11 software and themes were analysed using template analysis. Factors influencing women's uptake and adherence in Option B+ for PMTCT were identified through final analysis of key themes. RESULTS: Factors that motivated PMTCT uptake and adherence were good quality post-test HIV counselling, belief in the efficacy of antiretroviral (ARV) attained through personal or peer experiences, and a partner who did not prevent women from seeking PMTCT care. Key barriers for PMTCT participation included doubts about ARV efficacy, particularly for asymptomatic women, unsupportive partners who actively prevented women from seeking treatment, and women's concerns about community stigma and discrimination. CONCLUSIONS: Results suggest that PMTCT program success is determined by facilitators and barriers from across the spectrum of the socio-ecological model. While roll out of Option B+ as current national policy for pregnant women in Papua has improved detection and enrolment of HIV-positive women, health facilities need to address various existing and potential issues to ensure long-term adherence of women beyond the current PMTCT program, including during pregnancy, childbirth and breastfeeding.


Subject(s)
Anti-Retroviral Agents/therapeutic use , HIV Infections/drug therapy , HIV Infections/transmission , Infectious Disease Transmission, Vertical/prevention & control , Pregnancy Complications, Infectious/drug therapy , Adult , Child , Counseling , Female , Focus Groups , Health Personnel , Humans , Indonesia , Infant , Interviews as Topic , Patient Compliance , Peer Group , Pregnancy , Pregnant Women , Program Evaluation , Qualitative Research , Sexual Partners , Social Stigma , Young Adult
8.
Clin Cancer Res ; 24(22): 5602-5609, 2018 11 15.
Article in English | MEDLINE | ID: mdl-29898991

ABSTRACT

Purpose: The accumulation of emergent RAS mutations during anti-EGFR therapy is of interest as a mechanism for acquired resistance to anti-EGFR treatment. Plasma analysis of circulating tumor (ct) DNA is a minimally invasive and highly sensitive method to determine RAS mutational status.Experimental Design: This biomarker analysis of the global phase III ASPECCT study used next-generation sequencing to detect expanded RAS ctDNA mutations in panitumumab-treated patients. Plasma samples collected at baseline and posttreatment were analyzed categorically for the presence of RAS mutations by the PlasmaSelect-R 64-gene panel at 0.1% sensitivity.Results: Among panitumumab-treated patients with evaluable plasma samples at baseline (n = 238), 188 (79%) were wild-type (WT) RAS, and 50 (21%) were mutant RAS Of the 188 patients with baseline ctDNA WT RAS status, 164 had evaluable posttreatment results with a 32% rate of emergent RAS mutations. The median overall survival for WT and RAS mutant status by ctDNA at baseline was 13.7 (95% confidence interval, 11.5-15.4) and 7.9 months (6.4-9.6), respectively (P < 0.0001). Clinical outcomes were not significantly different between patients with and without emergent ctDNA RAS mutations.Conclusions: Although patients with baseline ctDNA RAS mutations had worse outcomes than patients who were WT RAS before initiating treatment, emergent ctDNA RAS mutations were not associated with less favorable patient outcomes in panitumumab-treated patients. Further research is needed to determine a clinically relevant threshold for baseline and emergent ctDNA RAS mutations. Clin Cancer Res; 24(22); 5602-9. ©2018 AACR.


Subject(s)
Biomarkers, Tumor , Circulating Tumor DNA , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/drug effects , Genes, ras , Mutation , Alleles , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Clinical Trials, Phase III as Topic , Colorectal Neoplasms/blood , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/mortality , DNA Mutational Analysis , Drug Resistance, Neoplasm/genetics , Gene Frequency , High-Throughput Nucleotide Sequencing , Humans , Liquid Biopsy , Prognosis
9.
Jpn J Clin Oncol ; 47(11): 1002-1009, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28973403

ABSTRACT

OBJECTIVE: To evaluate the safety (including adverse events and dose-limiting toxicities [DLTs]), tolerability, pharmacokinetics and antitumor activity of the investigational MET inhibitor rilotumumab alone in patients with advanced solid tumors (Part 1) or in combination with cisplatin plus capecitabine (CX) in patients with MET-positive advanced gastric or gastroesophageal junction cancer (Part 2). METHODS: Adult patients received 10 or 20 mg/kg intravenous (IV) rilotumumab every 2 weeks (Part 1) or 15 mg/kg IV rilotumumab every 3 weeks plus 80 mg/m2 cisplatin on Day 1 and 1000 mg/m2 capecitabine twice daily on Days 1-14 of every 21-day cycle (Part 2). RESULTS: Nine patients enrolled in Part 1; 12 patients enrolled in Part 2. One DLT occurred (Grade 3 decreased appetite and stomatitis [Part 2]). Adverse events related to any treatment occurred in 17 patients (81%) and were Grade ≥3 in nine patients (43%). Rilotumumab pharmacokinetics appeared linear, and exposure was unaffected by CX. No patient who received rilotumumab monotherapy in Part 1 had a response. In Part 2, five of eight patients (63%) with measureable disease at baseline had a partial response and two patients (25%) had stable disease; median (95% CI) progression-free survival was 7.0 (2.4-15.4) months; overall survival was 18.2 (5.6-20.4) months. CONCLUSIONS: In combination with CX, rilotumumab appeared tolerable and showed antitumor activity in Japanese patients with MET-positive gastric/gastroesophageal junction cancer. However, owing to the results of recent Phase 3 trials of MET inhibitors (including rilotumumab), further development of rilotumumab in this setting is not being pursued. ClinicalTrials.gov Identifier: NCT01791374.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Stomach Neoplasms/drug therapy , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Capecitabine/administration & dosage , Capecitabine/adverse effects , Cisplatin/administration & dosage , Cisplatin/adverse effects , Disease-Free Survival , Esophagogastric Junction/pathology , Female , Humans , Male , Middle Aged , Stomach Neoplasms/mortality
10.
Lancet Oncol ; 18(11): 1467-1482, 2017 11.
Article in English | MEDLINE | ID: mdl-28958504

ABSTRACT

BACKGROUND: Rilotumumab is a fully human monoclonal antibody that selectively targets the ligand of the MET receptor, hepatocyte growth factor (HGF). We aimed to assess the efficacy, safety, and pharmacokinetics of rilotumumab combined with epirubicin, cisplatin, and capecitabine, and to assess potential biomarkers, in patients with advanced MET-positive gastric or gastro-oesophageal junction adenocarcinoma. METHODS: This multicentre, randomised, double-blind, placebo-controlled, phase 3 study was done at 152 centres in 27 countries. We recruited adults (aged ≥18 years) with unresectable locally advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma, an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1, MET-positive tumours (≥25% of tumour cells with membrane staining of ≥1+ staining intensity), and evaluable disease, who had not received previous systemic therapy. Eligible patients were randomly assigned (1:1) via a computerised voice response system to receive rilotumumab 15 mg/kg intravenously or placebo in combination with open-label chemotherapy (epirubicin 50 mg/m2 intravenously; cisplatin 60 mg/m2 intravenously; capecitabine 625 mg/m2 orally twice daily) in 21-day cycles for up to ten cycles. After completion of chemotherapy, patients continued to receive rilotumumab or placebo monotherapy until disease progression, intolerability, withdrawal of consent, or study termination. Randomisation was stratified by disease extent and ECOG performance status. Both patients and physicians were masked to study treatment assignment. The primary endpoint was overall survival, analysed by intention to treat. We report the final analysis. This study is registered with ClinicalTrials.gov, number NCT01697072. FINDINGS: Between Nov 7, 2012, and Nov 21, 2014, 609 patients were randomly assigned to rilotumumab plus epirubicin, cisplatin, and capecitabine (rilotumumab group; n=304) or placebo plus epirubicin, cisplatin, and capecitabine (placebo group; n=305). Study treatment was stopped early after an independent data monitoring committee found a higher number of deaths in the rilotumumab group than in the placebo group; all patients in the rilotumumab group subsequently discontinued all study treatment. Median follow-up was 7·7 months (IQR 3·6-12·0) for patients in the rilotumumab group and 9·4 months (5·3-13·1) for patients in the placebo group. Median overall survival was 8·8 months (95% CI 7·7-10·2) in the rilotumumab group compared with 10·7 months (9·6-12·4) in the placebo group (stratified hazard ratio 1·34, 95% CI 1·10-1·63; p=0·003). The most common grade 3 or worse adverse events in the rilotumumab and placebo groups were neutropenia (86 [29%] of 298 patients vs 97 [32%] of 299 patients), anaemia (37 [12%] vs 43 [14%]), and fatigue (30 [10%] vs 35 [12%]). The frequency of serious adverse events was similar in the rilotumumab and placebo groups (142 [48%] vs 149 [50%]). More deaths due to adverse events occurred in the rilotumumab group than the placebo group (42 [14%] vs 31 [10%]). In the rilotumumab group, 33 (11%) of 298 patients had fatal adverse events due to disease progression, and nine (3%) had fatal events not due to disease progression. In the placebo group, 23 (8%) of 299 patients had fatal adverse events due to disease progression, and eight (3%) had fatal events not due to disease progression. INTERPRETATION: Ligand-blocking inhibition of the MET pathway with rilotumumab is not effective in improving clinical outcomes in patients with MET-positive gastric or gastro-oesophageal adenocarcinoma. FUNDING: Amgen.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/mortality , Stomach Neoplasms/drug therapy , Stomach Neoplasms/mortality , Adult , Aged , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Capecitabine/administration & dosage , Capecitabine/adverse effects , Cisplatin/administration & dosage , Cisplatin/adverse effects , Disease-Free Survival , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Epirubicin/administration & dosage , Epirubicin/adverse effects , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophagogastric Junction/pathology , Humans , Internationality , Kaplan-Meier Estimate , Middle Aged , Prognosis , Proportional Hazards Models , Proto-Oncogene Proteins c-met/drug effects , Proto-Oncogene Proteins c-met/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Survival Analysis , Treatment Outcome
11.
Cancer ; 123(6): 1061-1070, 2017 05 15.
Article in English | MEDLINE | ID: mdl-27926778

ABSTRACT

BACKGROUND: MET gene amplification and Met protein overexpression may be associated with a poor prognosis. The MET/Met status is typically determined with fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC), respectively. Targeted proteomics uses mass spectrometry-based selected reaction monitoring (SRM) to accurately quantitate Met expression. FISH, IHC, and SRM analyses were compared to characterize the prognostic value of MET/Met in gastroesophageal adenocarcinoma (GEC). METHODS: Samples from 447 GEC patients were analyzed for MET gene amplification (FISH) and Met protein expression (IHC and SRM). Cox proportional hazards models and Kaplan-Meier estimates were applied to explore relations between Met, overall survival (OS), and clinical/pathological characteristics. Spearman's rank coefficient was used to assess the correlation between parameters. RESULTS: Patients with MET-amplified tumors had worse OS when: the MET/centromere enumeration probe for chromosome 7 FISH ratio was ≥ 2 (hazard ratio [HR], 3.13; 95% confidence interval [CI], 1.84-5.33), the MET gene copy number was ≥5 (HR, 2.51; 95% CI, 1.45-4.34), or ≥ 10% of the cells had ≥15 copies (HR, 4.28; 95% CI, 2.18-8.39). Similar observations were made with Met protein overexpression by IHC (≥1 + intensity in ≥ 25% of the tumor cell membrane: HR, 1.39; 95% CI, 1.04-1.86) or SRM (≥400 amol/µg: HR, 1.76; 95% CI, 1.06-2.90). A significant correlation was observed between MET FISH/Met IHC, MET FISH/Met SRM, and Met IHC/Met SRM; only MET FISH and Met SRM were independent negative prognostic biomarkers in multivariate analyses. CONCLUSIONS: MET amplification and overexpression, assessed by multiple methods, were associated with a worse prognosis in univariate analyses. However, only MET amplification by FISH and Met expression by SRM were independent prognostic biomarkers. Compared with IHC, SRM may provide an added benefit for informed decisions about Met-targeted therapy. Cancer 2017;123:1061-70. © 2016 American Cancer Society.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/mortality , Esophageal Neoplasms/genetics , Esophageal Neoplasms/mortality , Gene Amplification , Gene Expression , Proto-Oncogene Proteins c-met/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Biomarkers, Tumor , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Mass Spectrometry , Prognosis , Proportional Hazards Models
12.
Clin Cancer Res ; 19(11): 3078-87, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23591447

ABSTRACT

PURPOSE: HER3 is a key dimerization partner for other HER family members, and its expression is associated with poor prognosis. This first-in-human study of U3-1287 (NCT00730470), a fully human anti-HER3 monoclonal antibody, evaluated its safety, tolerability, and pharmacokinetics in patients with advanced solid tumor. EXPERIMENTAL DESIGN: The study was conducted in 2 parts: part 1--sequential cohorts received escalating doses (0.3-20 mg/kg) of U3-1287 every 2 weeks, starting 3 weeks after the first dose; part 2--additional patients received 9, 14, or 20 mg/kg U3-1287 every 2 weeks, based on observed tolerability and pharmacokinetics from part 1. Recommended phase II dose, adverse event rates, pharmacokinetics, and tumor response were determined. RESULTS: Fifty-seven patients (part 1: 26; part 2: 31) received U3-1287. As no dose-limiting toxicities were reported, the maximum-tolerated dose was not reached. The maximum-administered dose was 20 mg/kg every 2 weeks. The most frequent adverse events related to U3-1287 were fatigue (21.1%), diarrhea (12.3%), nausea (10.5%), decreased appetite (7.0%), and dysgeusia (5.3%). No patient developed anti-U3-1287 antibodies. In these heavily pretreated patients, stable disease was maintained 9 weeks or more in 19.2% in part 1 and 10 weeks or more in 25.8% in part 2. CONCLUSION: U3-1287 treatment was well tolerated, and some evidence of disease stabilization was observed. Pharmacokinetic data support U3-1287 dosing of 9 to 20 mg/kg every 2 to 3 weeks. Combination studies of U3-1287 are ongoing.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Biomarkers/metabolism , Broadly Neutralizing Antibodies , Female , Humans , Male , Middle Aged , Neoplasm Staging , Neoplasms/metabolism , Prognosis , Receptor, ErbB-3/antagonists & inhibitors , Treatment Outcome
13.
Clin Cancer Res ; 16(18): 4654-65, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20670944

ABSTRACT

PURPOSE: This study aimed to identify molecular determinants of sensitivity of non-small cell lung cancer (NSCLC) to anti-insulin-like growth factor receptor (IGF-IR) therapy. EXPERIMENTAL DESIGN: A total of 216 tumor samples were investigated, of which 165 consisted of retrospective analyses of banked tissue and an additional 51 were from patients enrolled in a phase II study of figitumumab, a monoclonal antibody against IGF-IR, in stage IIIb/IV NSCLC. Biomarkers assessed included IGF-IR, epidermal growth factor receptor, IGF-II, IGF-IIR, insulin receptor substrate 1 (IRS-1), IRS-2, vimentin, and E-cadherin. Subcellular localization of IRS-1 and phosphorylation levels of mitogen-activated protein kinase and Akt1 were also analyzed. RESULTS: IGF-IR was differentially expressed across histologic subtypes (P = 0.04), with highest levels observed in squamous cell tumors. Elevated IGF-IR expression was also observed in a small number of squamous cell tumors responding to chemotherapy combined with figitumumab (P = 0.008). Because no other biomarker/response interaction was observed using classical histologic subtyping, a molecular approach was undertaken to segment NSCLC into mechanism-based subpopulations. Principal component analysis and unsupervised Bayesian clustering identified three NSCLC subsets that resembled the steps of the epithelial to mesenchymal transition: E-cadherin high/IRS-1 low (epithelial-like), E-cadherin intermediate/IRS-1 high (transitional), and E-cadherin low/IRS-1 low (mesenchymal-like). Several markers of the IGF-IR pathway were overexpressed in the transitional subset. Furthermore, a higher response rate to the combination of chemotherapy and figitumumab was observed in transitional tumors (71%) compared with those in the mesenchymal-like subset (32%; P = 0.03). Only one epithelial-like tumor was identified in the phase II study, suggesting that advanced NSCLC has undergone significant dedifferentiation at diagnosis. CONCLUSION: NSCLC comprises molecular subsets with differential sensitivity to IGF-IR inhibition.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm , Insulin-Like Growth Factor I/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/classification , Carcinoma, Non-Small-Cell Lung/metabolism , Clinical Trials, Phase II as Topic , Drug Resistance, Neoplasm/drug effects , Female , Hormone Antagonists/therapeutic use , Humans , Immunoglobulins, Intravenous , Insulin-Like Growth Factor I/immunology , Male , Mice , Molecular Diagnostic Techniques , NIH 3T3 Cells , Prognosis , Retrospective Studies , Tissue Array Analysis
14.
Am J Trop Med Hyg ; 80(3): 384-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19270286

ABSTRACT

Cysticercosis and taeniasis are known to be present in Papua, Indonesia. Several small studies have found a high prevalence of cysticercosis (23.5-56.9%) in the central highlands of Papua. A seroepidemiologic survey was carried out in four districts (Jayawijaya, Paniai, Pegunungan Bintang, and Puncak Jaya) of Papua. Anti-cysticercosis and anti-taeniasis antibodies were measured in 2,931 people using recombinant T24 and recombinant ES33 as a measure of cysticercosis and taeniasis exposures, respectively. Prevalence of cysticercosis-taeniasis is high in the Jayawijaya and Paniai districts (20.8% and 29.2% for cysticercosis and 7% and 9.6% for taeniasis, respectively) and lowest in the other two districts (Pegunungan Bintang and Puncak Jaya) (2% and 2% for cysticercosis and 1.7% and 10.7% for taeniasis, respectively). Our data show that the prevalence of cysticercosis and taeniasis are unchanged from that reported nearly 35 years ago at the beginning of cysticercosis-taeniasis epidemics in Papua, Indonesia.


Subject(s)
Antibodies, Helminth/blood , Taenia solium/immunology , Taeniasis/epidemiology , Adolescent , Adult , Animals , Child , Cysticercosis/epidemiology , Cysticercus/immunology , Female , Humans , Male , Middle Aged , Papua New Guinea/epidemiology , Seroepidemiologic Studies , Young Adult
15.
Virchows Arch ; 451(4): 823-34, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17674039

ABSTRACT

The pathogenesis of Fabry disease is poorly understood. We used a variety of immunohistological techniques to localize globotriaosylceramide, the main glycolipid that accumulates in Fabry disease. Globotriaosylceramide immunoreactivity in a heterogenous pattern was present in all organs examined of a patient on long-term enzyme replacement therapy. In the brain, immmunopositivity was found only in the parahippocampal region. Globotriaosylceramide immunostaining was present in the cell membrane and cytoplasm of endothelial cells, even in the absence of lysosomal inclusions. In kidney tissue, globotriaosylceramide colocalized with lysosomal, endoplasmic reticulum, and nuclear markers. Pre- and postembedding immunogold electron microscopy of skin biopsies and untreated patient cultured skin fibroblasts confirmed the presence of globotriaosylceramide in the cell membrane, in various cytoplasmic structures, and in the nucleus. Control organ tissues and cultured fibroblasts from five unaffected subjects were uniformly negative for globotriaosylceramide by immunohistochemistry and immunogold electron microscopy. We conclude that a substantial amount of lysosomal and extralysosomal globotriaosylceramide immunoreactivity remains in cells and tissues even after years of enzyme replacement therapy in Fabry disease. These findings are crucial for the understanding of the disease mechanism and suggest the usefulness of immunostaining for globotriaosylceramide as a means to assess response to novel, specific therapies.


Subject(s)
Cell Membrane/metabolism , Cell Nucleus/metabolism , Endoplasmic Reticulum/metabolism , Fabry Disease/metabolism , Lysosomes/metabolism , Trihexosylceramides/metabolism , Adult , Brain/metabolism , Brain/pathology , Cell Membrane/pathology , Cell Membrane/ultrastructure , Cell Nucleus/pathology , Cell Nucleus/ultrastructure , Cells, Cultured , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum/ultrastructure , Fabry Disease/etiology , Fabry Disease/pathology , Humans , Kidney/metabolism , Kidney/pathology , Lysosomes/pathology , Lysosomes/ultrastructure , Middle Aged , Skin/metabolism , Skin/pathology
16.
Traffic ; 8(1): 47-60, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17132146

ABSTRACT

The sorting of newly synthesized membrane proteins to the cell surface is an important mechanism of cell polarity. To identify more of the molecular machinery involved, we investigated the function of the small GTPase Rab10 in polarized epithelial Madin-Darby canine kidney cells. We find that GFP-tagged Rab10 localizes primarily to the Golgi during early cell polarization. Expression of an activated Rab10 mutant inhibits biosynthetic transport from the Golgi and missorts basolateral cargo to the apical membrane. Depletion of Rab10 by RNA interference has only mild effects on biosynthetic transport and epithelial polarization, but simultaneous inhibition of Rab10 and Rab8a more strongly impairs basolateral sorting. These results indicate that Rab10 functions in trafficking from the Golgi at early stages of epithelial polarization, is involved in biosynthetic transport to the basolateral membrane and may co-operate with Rab8.


Subject(s)
rab GTP-Binding Proteins/metabolism , Animals , Base Sequence , Biological Transport, Active , Cell Line , Cell Membrane/metabolism , Cell Polarity , DNA/genetics , Dogs , Epithelial Cells/metabolism , Golgi Apparatus/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Kidney/cytology , Kidney/metabolism , Membrane Glycoproteins/metabolism , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Viral Envelope Proteins/metabolism , rab GTP-Binding Proteins/antagonists & inhibitors , rab GTP-Binding Proteins/genetics , trans-Golgi Network/metabolism
17.
J Cell Biol ; 167(3): 531-43, 2004 Nov 08.
Article in English | MEDLINE | ID: mdl-15534004

ABSTRACT

The AP-1B clathrin adaptor complex is responsible for the polarized transport of many basolateral membrane proteins in epithelial cells. Localization of AP-1B to recycling endosomes (REs) along with other components (exocyst subunits and Rab8) involved in AP-1B-dependent transport suggested that RE might be an intermediate between the Golgi and the plasma membrane. Although the involvement of endosomes in the secretory pathway has long been suspected, we now present direct evidence using four independent methods that REs play a role in basolateral transport in MDCK cells. Newly synthesized AP-1B-dependent cargo, vesicular stomatitis virus glycoprotein G (VSV-G), was found by video microscopy, immunoelectron microscopy, and cell fractionation to enter transferrin-positive REs within a few minutes after exit from the trans-Golgi network. Although transient, RE entry appears essential because enzymatic inactivation of REs blocked VSV-G delivery to the cell surface. Because an apically targeted VSV-G mutant behaved similarly, these results suggest that REs not only serve as an intermediate but also as a common site for polarized sorting on the endocytic and secretory pathways.


Subject(s)
Cell Membrane/metabolism , Endocytosis , Endosomes/physiology , Golgi Apparatus/metabolism , Protein Transport , Adaptor Protein Complex 1/metabolism , Animals , Cell Line , Dogs , Membrane Glycoproteins/metabolism , Microscopy, Fluorescence , Microscopy, Video , Transferrin , Viral Envelope Proteins/metabolism
18.
Nat Cell Biol ; 6(2): 106-12, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14743217

ABSTRACT

Protein kinase D (PKD) binds to diacylglycerol (DAG) in the trans-Golgi network (TGN) and is activated by trimeric G-protein subunits beta gamma. This complex then regulates the formation of transport carriers in the TGN that traffic to the plasma membrane in non-polarized cells. Here we report specificity of different PKD isoforms in regulating protein trafficking from the TGN. Kinase-inactive forms of PKD1, PKD2 and PKD3 localize to the TGN in polarized and non-polarized cells. PKD activity is required only for the transport of proteins containing basolateral sorting information, and seems to be cargo specific.


Subject(s)
Protein Kinase C/metabolism , Protein Kinases/metabolism , trans-Golgi Network/metabolism , Animals , Cell Line , Cell Polarity , Diglycerides/metabolism , Dogs , Epithelial Cells/cytology , Epithelial Cells/metabolism , HeLa Cells , Humans , Isoenzymes/metabolism , Protein Kinase C/genetics , Protein Kinase D2 , Protein Kinases/genetics , Protein Transport/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
19.
J Cell Biol ; 163(2): 339-50, 2003 Oct 27.
Article in English | MEDLINE | ID: mdl-14581456

ABSTRACT

The AP-1B clathrin adaptor complex plays a key role in the recognition and intracellular transport of many membrane proteins destined for the basolateral surface of epithelial cells. However, little is known about other components that act in conjunction with AP-1B. We found that the Rab8 GTPase is one such component. Expression of a constitutively activated GTP hydrolysis mutant selectively inhibited basolateral (but not apical) transport of newly synthesized membrane proteins. Moreover, the effects were limited to AP-1B-dependent basolateral cargo; basolateral transport of proteins containing dileucine targeting motifs that do not interact with AP-1B were targeted normally despite overexpression of mutant Rab8. Similar results were obtained for a dominant-negative allele of the Rho GTPase Cdc42, previously implicated in basolateral transport but now shown to be selective for the AP-1B pathway. Rab8-GFP was localized to membranes in the TGN-recycling endosome, together with AP-1B complexes and the closely related but ubiquitously expressed AP-1A complex. However, expression of active Rab8 caused a selective dissociation of AP-1B complexes, reflecting the specificity of Rab8 for AP-1B-dependent transport.


Subject(s)
Cell Polarity/physiology , GTP Phosphohydrolases/metabolism , Kidney/cytology , rab GTP-Binding Proteins/metabolism , Adaptor Protein Complex 1/metabolism , Adaptor Protein Complex gamma Subunits/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Biological Transport , Biomarkers , Cell Line , Dogs , Endosomes/metabolism , Enzyme Activation , GTP Phosphohydrolases/ultrastructure , Gene Expression , Mutation , Transferrin/pharmacokinetics , cdc42 GTP-Binding Protein/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/ultrastructure , trans-Golgi Network/metabolism , trans-Golgi Network/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...