Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
PLoS One ; 14(5): e0216864, 2019.
Article in English | MEDLINE | ID: mdl-31125352

ABSTRACT

BACKGROUND: In non-small cell lung cancer (NSCLC), PD-L1 expression on either tumor cells (TC) or both TC and tumor-infiltrating immune cells (IC) is currently the most used biomarker in cancer immunotherapy. However, the mechanisms involved in PD-L1 regulation are not fully understood. To provide better insight in these mechanisms, a multiangular analysis approach was used to combine protein and mRNA expression with several clinicopathological characteristics. PATIENTS AND METHODS: Archival tissues from 640 early stage, resected NSCLC patients were analyzed with immunohistochemistry for expression of PD-L1 and CD8 infiltration. In addition, mutational status and expression of a selection of immune genes involved in the PD-L1/PD-1 axis and T-cell response was determined. RESULTS: Tumors with high PD-L1 expression on TC or on IC represent two subsets of NSCLC with minimal overlap. We observed that PD-L1 expression on IC irrespective of expression on TC is a good marker for inflammation within tumors. In the tumors with the highest IC expression and absent TC expression an association with reduced IFNγ downstream signaling in tumor cells was observed. CONCLUSIONS: These results show that PD-L1 expression on TC and IC are both independent hallmarks of the inflamed phenotype in NSCLC, and TC-negative/IC-high tumors can also be categorized as inflamed. The lack of correlation between PD-L1 TC and IC expression in this subgroup may be caused by impaired IFNγ signaling in tumor cells. These findings may bring a better understanding of the tumor-immune system interaction and the clinical relevance of PD-L1 expression on IC irrespective of PD-L1 expression on TC.


Subject(s)
B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Gene Expression Regulation, Neoplastic/immunology , Immunity, Cellular , Interferon-gamma/immunology , Lung Neoplasms/immunology , Neoplasm Proteins/immunology , Signal Transduction/immunology , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/genetics , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Interferon-gamma/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Staging , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Signal Transduction/genetics
2.
Breast Cancer Res ; 19(1): 16, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28183321

ABSTRACT

BACKGROUND: NeoSphere showed significantly higher pathologic complete response (pCR) with neoadjuvant pertuzumab, trastuzumab, and docetaxel compared with trastuzumab plus docetaxel, pertuzumab plus trastuzumab, or pertuzumab plus docetaxel. We assessed associations between human epidermal growth factor receptor 2 (HER2) pathway-related biomarkers and clinical outcome in response to these regimens. METHODS: Tumor, serum, and whole blood samples were collected at baseline and post neoadjuvant treatment before surgery. Associations between biomarkers and pCR, and between biomarkers and clinical variables were assessed in the overall and estrogen receptor (ER)-positive and ER-negative populations. Changes in serum marker levels between baseline and post-neoadjuvant treatment were examined. RESULTS: No markers were associated with pCR across all groups; however, significant associations were observed for two markers in individual groups. High HER2 was significantly associated with higher pCR rates (P = 0.001) and a significant treatment interaction (P = 0.0236) with pertuzumab, trastuzumab, and docetaxel (odds ratio 2.07, P = 0.01). Low serum transforming growth factor alpha (TGFα) was associated with higher pCR rates with pertuzumab plus trastuzumab (P = 0.04) without a significant treatment interaction. Presence of truncated HER2 did not affect pCR. A non-significant decreased pCR benefit was observed consistently across groups in patients with mutated PIK3CA while the treatment benefit from pertuzumab was maintained when comparing the trastuzumab plus docetaxel and pertuzumab, trastuzumab, and docetaxel groups. Notably, PIK3CA exon 9 mutations were associated with residual disease (pooled groups), which was not found for exon 20 mutations. Serum HER2 extracellular domain levels were significantly increased between baseline and post-neoadjuvant treatment in the non-trastuzumab-treated group, and decreased in the trastuzumab-containing groups (likely due to trastuzumab's mechanism of action). Differences in biomarker profiles according to ER status were observed. CONCLUSIONS: The observed associations of HER2 protein levels with sensitivity to pertuzumab, and of PIK3CA exon 9 mutation to lack of sensitivity to HER2-targeted monoclonal antibody treatment, warrant further investigation. Previously reported findings of truncated forms of HER2 as resistance markers to HER2-targeted treatment could not be confirmed in NeoSphere. Conventional HER2 assessment should continue and HER2 remains the only biomarker suitable for patient selection in this population. TRIAL REGISTRATION: Clinicaltrials.gov, NCT00545688 . Registered on 16 October 2007.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Antibodies, Monoclonal, Humanized/administration & dosage , Biomarkers, Tumor , Breast Neoplasms/genetics , Docetaxel , Female , Humans , Immunohistochemistry , Mutation , Neoadjuvant Therapy , Taxoids/administration & dosage , Trastuzumab/administration & dosage , Treatment Outcome
3.
PLoS One ; 11(11): e0165856, 2016.
Article in English | MEDLINE | ID: mdl-27846280

ABSTRACT

In the age of personalized medicine stratifying tumors into molecularly defined subtypes associated with distinctive clinical behaviors and predictable responses to therapies holds tremendous value. Towards this end, we developed a custom microfluidics-based bladder cancer gene expression panel for characterization of archival clinical samples. In silico analysis indicated that the content of our panel was capable of accurately segregating bladder cancers from several public datasets into the clinically relevant basal and luminal subtypes. On a technical level, our bladder cancer panel yielded robust and reproducible results when analyzing formalin-fixed, paraffin-embedded (FFPE) tissues. We applied our panel in the analysis of a novel set of 204 FFPE samples that included non-muscle invasive bladder cancers (NMIBCs), muscle invasive disease (MIBCs), and bladder cancer metastases (METs). We found NMIBCs to be mostly luminal-like, MIBCs to include both luminal- and basal-like types, and METs to be predominantly of a basal-like transcriptional profile. Mutational analysis confirmed the expected enrichment of FGFR3 mutations in luminal samples, and, consistently, FGFR3 IHC showed high protein expression levels of the receptor in these tumors. Our bladder cancer panel enables basal/luminal characterization of FFPE tissues and with further development could be used for stratification of bladder cancer samples in the clinic.


Subject(s)
Biological Specimen Banks , Gene Expression Regulation, Neoplastic , Microfluidics/methods , Transcription, Genetic , Urinary Bladder Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Cohort Studies , Computer Simulation , Female , Formaldehyde , Genes, Neoplasm , Humans , Male , Middle Aged , Paraffin Embedding , Reproducibility of Results , Tissue Fixation , Urinary Bladder Neoplasms/pathology
4.
Nat Commun ; 7: 11579, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27174596

ABSTRACT

Mutations in ESR1 have been associated with resistance to aromatase inhibitor (AI) therapy in patients with ER+ metastatic breast cancer. Little is known of the impact of these mutations in patients receiving selective oestrogen receptor degrader (SERD) therapy. In this study, hotspot mutations in ESR1 and PIK3CA from ctDNA were assayed in clinical trial samples from ER+ metastatic breast cancer patients randomized either to the SERD fulvestrant or fulvestrant plus a pan-PI3K inhibitor. ESR1 mutations are present in 37% of baseline samples and are enriched in patients with luminal A and PIK3CA-mutated tumours. ESR1 mutations are often polyclonal and longitudinal analysis shows distinct clones exhibiting divergent behaviour over time. ESR1 mutation allele frequency does not show a consistent pattern of increases during fulvestrant treatment, and progression-free survival is not different in patients with ESR1 mutations compared with wild-type patients. ESR1 mutations are not associated with clinical resistance to fulvestrant in this study.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Estradiol/analogs & derivatives , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor alpha/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast/pathology , Breast Neoplasms/blood , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/genetics , DNA Mutational Analysis , DNA, Neoplasm/genetics , Disease-Free Survival , Estradiol/pharmacology , Estradiol/therapeutic use , Estrogen Receptor Antagonists/therapeutic use , Estrogen Receptor alpha/antagonists & inhibitors , Estrogens/metabolism , Female , Fulvestrant , Humans , Indazoles/pharmacology , Indazoles/therapeutic use , Middle Aged , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Sulfonamides/pharmacology , Sulfonamides/therapeutic use
5.
Clin Cancer Res ; 21(11): 2462-70, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-26034219

ABSTRACT

PURPOSE: The novel dual-action humanized IgG1 antibody MEHD7945A targeting HER3 and EGFR inhibits ligand-dependent HER dimer signaling. This phase I study evaluated the safety, pharmacokinetics, pharmacodynamics, and antitumor activity of MEHD7945A. EXPERIMENTAL DESIGN: Patients with locally advanced or metastatic epithelial tumors received escalating doses of MEHD7945A (1-30 mg/kg) every 2 weeks (q2w) until disease progression or intolerable toxicity. An expansion cohort was enrolled at the recommended phase II dose (14 mg/kg, q2w). Plasma samples, tumor biopsies, FDG-PET were obtained for assessment of pharmacokinetics, and pharmacodynamic modulation downstream of EGFR and HER3. RESULTS: No dose-limiting toxicities or MEHD7945A-related grade ≥ 4 adverse events (AE) were reported in dose-escalation (n = 30) or expansion (n = 36) cohorts. Related grade 3 AEs were limited to diarrhea and nausea in the same patient (30 mg/kg). Related AEs in ≥20% of patients ≤24 hours after the first infusion included grade 1/2 headache, fever, and chills, which were managed with premedication and/or symptomatic treatment. Pharmacodynamic data indicated target inhibition in 25% of evaluable patients. Best response by RECIST included 2 confirmed partial responses in squamous cell carcinomas of head and neck (SCCHN) patients with high tumor tissue levels of the HER3 ligand heregulin; 14 patients had stable disease ≥8 weeks, including SCCHN (n = 3), colorectal cancer (n = 6), and non-small cell lung cancer (n = 3). CONCLUSIONS: MEHD7945A was well-tolerated as single agent with evidence of tumor pharmacodynamic modulation and antitumor activity in SCCHN. Phase II studies were initiated with flat (nonweight-based) dosing at 1,100 mg q2w in SCCHN and colorectal cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Squamous Cell/drug therapy , ErbB Receptors/immunology , Head and Neck Neoplasms/drug therapy , Immunoglobulin G/administration & dosage , Receptor, ErbB-3/immunology , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Cetuximab/administration & dosage , Dose-Response Relationship, Drug , Drug-Related Side Effects and Adverse Reactions/classification , Drug-Related Side Effects and Adverse Reactions/pathology , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/administration & dosage , Female , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Humans , Immunoglobulin G/adverse effects , Immunoglobulin G/immunology , Male , Middle Aged , Panitumumab , Receptor, ErbB-3/antagonists & inhibitors , Squamous Cell Carcinoma of Head and Neck
6.
Clin Cancer Res ; 21(13): 2941-51, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25838397

ABSTRACT

PURPOSE: Up to one third of ovarian cancer patients are intrinsically resistant to platinum-based treatment. However, predictive and therapeutic strategies are lacking due to a poor understanding of the underlying molecular mechanisms. This study aimed to identify key molecular characteristics that are associated with primary chemoresistance in epithelial ovarian cancers. EXPERIMENTAL DESIGN: Gene expression profiling was performed on a discovery set of 85 ovarian tumors with clinically well-defined response to chemotherapies as well as on an independent validation dataset containing 138 ovarian patients from the chemotreatment arm of the ICON7 trial. RESULTS: We identified a distinct "reactive stroma" gene signature that is specifically associated with primary chemoresistant tumors and was further upregulated in posttreatment recurrent tumors. Immunohistochemistry (IHC) and RNA in situ hybridization (RNA ISH) analyses on three of the highest-ranked signature genes (POSTN, LOX, and FAP) confirmed that modulation of the reactive stroma signature genes within the peritumoral stromal compartments was specifically associated with the clinical chemoresistance. Consistent with these findings, chemosensitive ovarian cells grown in the presence of recombinant POSTN promoted resistance to carboplatin and paclitaxel treatment in vitro. Finally, we validated the reactive stroma signature in an independent dataset and demonstrated that a high POSTN expression level predicts shorter progression-free survival following first-line chemotherapy. CONCLUSIONS: Our findings highlight the important interplay between cancer and the tumor microenvironment in ovarian cancer biology and treatment. The identified reactive stromal components in this study provide a molecular basis to the further development of novel diagnostic and therapeutic strategies for overcoming chemoresistance in ovarian cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Adhesion Molecules/metabolism , Drug Resistance, Neoplasm , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Adult , Aged , Carboplatin/pharmacology , Carcinoma, Ovarian Epithelial , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Disease-Free Survival , Female , Fibroblasts/metabolism , Humans , Middle Aged , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/mortality , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Paclitaxel/pharmacology , Transcriptome , Treatment Outcome , Tumor Microenvironment , Up-Regulation
7.
Breast Cancer Res ; 16(3): R50, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24887458

ABSTRACT

INTRODUCTION: The purpose of this study was to retrospectively explore the relationship between human epidermal growth factor receptor 2 (HER2) messenger RNA (mRNA) expression and efficacy in patients receiving trastuzumab plus docetaxel (HT) or trastuzumab emtansine (T-DM1). METHODS: Patients with HER2-positive, locally advanced or metastatic breast cancer (MBC) were randomly assigned to HT (n=70) or T-DM1 (n=67). HER2 status was assessed locally using immunohistochemistry or fluorescence in situ hybridization and confirmed retrospectively by central testing. HER2 mRNA expression was assessed using quantitative reverse transcriptase polymerase chain reaction. RESULTS: HER2 mRNA levels were obtained for 116/137 patients (HT=61; T-DM1=55). Median pretreatment HER2 mRNA was 8.9. The risk of disease progression in the overall population was lower with T-DM1 than with HT (hazard ratio (HR)=0.59; 95% confidence interval (CI) 0.36 to 0.97). This effect was more pronounced in patients with HER2 mRNA≥median (HR=0.39; 95% CI 0.18 to 0.85) versus

Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Breast Neoplasms/drug therapy , Maytansine/analogs & derivatives , Receptor, ErbB-2/genetics , Taxoids/therapeutic use , Ado-Trastuzumab Emtansine , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Disease-Free Survival , Docetaxel , Female , Humans , Maytansine/therapeutic use , Middle Aged , RNA, Messenger/biosynthesis , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/metabolism , Retrospective Studies , Trastuzumab , Treatment Outcome
8.
Epigenomics ; 6(1): 59-72, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24579947

ABSTRACT

Lung cancer remains the primary cause of cancer-related deaths worldwide. Improved tools for early detection and therapeutic stratification would be expected to increase the survival rate for this disease. Alterations in the molecular pathways that drive lung cancer, which include epigenetic modifications, may provide biomarkers to help address this major unmet clinical need. Epigenetic changes, which are defined as heritable changes in gene expression that do not alter the primary DNA sequence, are one of the hallmarks of cancer, and prevalent in all types of cancer. These modifications represent a rich source of biomarkers that have the potential to be implemented in clinical practice. This perspective describes recent advances in the discovery of epigenetic biomarkers in lung cancer, specifically those that result in the methylation of DNA at CpG sites. We discuss one approach for methylation-based biomarker assay development that describes the discovery at a genome-scale level, which addresses some of the practical considerations for design of assays that can be implemented in the clinic. We emphasize that an integrated technological approach will enable the development of clinically useful DNA methylation-based biomarker assays. While this article focuses on current literature and primary research findings in lung cancer, the principles we describe here apply to the discovery and development of epigenetic biomarkers for other types of cancer.


Subject(s)
Biomarkers, Tumor/genetics , DNA Methylation , Epigenomics/methods , Histones/physiology , Lung Neoplasms/genetics , RNA, Untranslated/genetics , CpG Islands , Epigenesis, Genetic , Epigenomics/instrumentation , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , High-Throughput Screening Assays , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Reproducibility of Results
9.
Clin Cancer Res ; 20(8): 2080-91, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24573554

ABSTRACT

PURPOSE: Tailoring cancer treatment to tumor molecular characteristics promises to make personalized medicine a reality. However, reliable genetic profiling of archived clinical specimens has been hindered by limited sensitivity and high false-positive rates. Here, we describe a novel methodology, MMP-seq, which enables sensitive and specific high-throughput, high-content genetic profiling in archived clinical samples. EXPERIMENTAL DESIGN: We first validated the technical performance of MMP-seq in 66 cancer cell lines and a Latin square cross-dilution of known somatic mutations. We next characterized the performance of MMP-seq in 17 formalin-fixed paraffin-embedded (FFPE) clinical samples using matched fresh-frozen tissue from the same tumors as benchmarks. To demonstrate the potential clinical utility of our methodology, we profiled FFPE tumor samples from 73 patients with endometrial cancer. RESULTS: We demonstrated that MMP-seq enabled rapid and simultaneous profiling of a panel of 88 cancer genes in 48 samples, and detected variants at frequencies as low as 0.4%. We identified DNA degradation and deamination as the main error sources and developed practical and robust strategies for mitigating these issues, and dramatically reduced the false-positive rate. Applying MMP-seq to a cohort of endometrial tumor samples identified extensive, potentially actionable alterations in the PI3K (phosphoinositide 3-kinase) and RAS pathways, including novel PIK3R1 hotspot mutations that may disrupt negative regulation of PIK3CA. CONCLUSIONS: MMP-seq provides a robust solution for comprehensive, reliable, and high-throughput genetic profiling of clinical tumor samples, paving the way for the incorporation of genomic-based testing into clinical investigation and practice.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Multiplex Polymerase Chain Reaction/methods , Mutation , Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Class Ia Phosphatidylinositol 3-Kinase , DNA Mutational Analysis/methods , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , ErbB Receptors/genetics , Genotyping Techniques , Humans , MCF-7 Cells , Neoplasms/metabolism , Neoplasms/pathology , PTEN Phosphohydrolase/genetics , Paraffin Embedding , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics , Reproducibility of Results , Tumor Suppressor Proteins/metabolism , ras Proteins/genetics
10.
PLoS One ; 8(2): e56765, 2013.
Article in English | MEDLINE | ID: mdl-23468880

ABSTRACT

PURPOSE: Tumors with oncogenic dependencies on the HER family of receptor tyrosine kinases (RTKs) often respond well to targeted inhibition. Our previous work suggested that many cell lines derived from squamous cell carcinomas of the head and neck (SCCHNs) depend on autocrine signaling driven by HER2/3 dimerization and high-level co-expression of HRG. Additionally, results from a Phase I trial of MEHD7495A, a dual-action antibody that blocks ligand binding to EGFR and HER3, suggest that high-level HRG expression was associated with clinical response in SCCHN patients. Here we explore the hypothesis that high-level HRG expression defines a subpopulation of SCCHNs with activated HER3. EXPERIMENTAL DESIGN: qRT-PCR expression profiling was performed on >750 tumors of diverse origin, including >150 therapy-naïve, primary, and recurrent SCCHNs. Activated HER3, defined by immunoprecipitation of phospho-HER3, was compared to HRG expression in SCCHN samples. Paracrine versus autocrine expression was evaluated using RNA-in situ hybridization. RESULTS: SCCHN tumors express the highest levels of HRG compared to a diverse collection of other tumor types. We show that high HRG expression is associated with activated HER3, whereas low HRG expression is associated with low HER3 activation in SCCHN tumors. Furthermore, HRG expression is higher in recurrent SCCHN compared to patient-matched therapy naïve specimens. CONCLUSIONS: HRG expression levels define a biologically distinct subset of SCCHN patients. We propose that high-level expression of HRG is associated with constitutive activation of HER3 in SCCHN and thus defines an actionable biomarker for interventions targeting HER3.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Neuregulin-1/metabolism , Receptor, ErbB-3/metabolism , Biomarkers, Tumor , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Gene Expression , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Humans , Immunohistochemistry , Neuregulin-1/genetics , Receptor, ErbB-3/genetics , Squamous Cell Carcinoma of Head and Neck
11.
Cancer Res ; 73(2): 824-33, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23172311

ABSTRACT

EGF receptor (EGFR) inhibition is efficacious in cancer therapy, but initially sensitive tumors often develop resistance. In this study, we investigated the potential to overcome acquired resistance to EGFR inhibitors with MEHD7945A, a monoclonal antibody that dually targets EGFR and HER3 (ErbB3). In cancer cells resistant to cetuximab and erlotinib, we found that MEHD7945A, but not single target EGFR inhibitors, could inhibit tumor growth and cell-cycle progression in parallel with EGFR/HER3 signaling pathway modulation. MEHD7945A was more effective than a combination of cetuximab and anti-HER3 antibody at inhibiting both EGFR/HER3 signaling and tumor growth. In human tumor xenograft models, we confirmed the greater antitumor potency of MEHD7945A than cetuximab or erlotinib. MEHD7945A retained potent activity in tumors refractory to EGFR inhibitor alone. Furthermore, MEHD7945A also limited cross-resistance to radiation in EGFR inhibitor-resistant cells by modulating cell-cycle progression and repair processes that control apoptotic cell death. Taken together, our findings confirm an important role of compensatory HER3 signaling in the development of acquired resistance to EGFR inhibitors and offer preclinical proof-of-concept that MEHD7945A can effectively overcome EGFR inhibitor resistance.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , Head and Neck Neoplasms/drug therapy , Immunoglobulin G/pharmacology , Lung Neoplasms/drug therapy , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Cetuximab , Erlotinib Hydrochloride , Humans , Lung Neoplasms/radiotherapy , Mice , Mice, Nude , Molecular Targeted Therapy , Quinazolines/therapeutic use , Xenograft Model Antitumor Assays
12.
Clin Cancer Res ; 18(24): 6771-83, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23136191

ABSTRACT

PURPOSE: Class 1 phosphatidylinositol 3-kinase (PI3K) plays a major role in cell proliferation and survival in a wide variety of human cancers. Here, we investigated biomarker strategies for PI3K pathway inhibitors in non-small-cell lung cancer (NSCLC). EXPERIMENTAL DESIGN: Molecular profiling for candidate PI3K predictive biomarkers was conducted on a collection of NSCLC tumor samples. Assays included comparative genomic hybridization, reverse-transcription polymerase chain reaction gene expression, mutation detection for PIK3CA and other oncogenes, PTEN immunohistochemistry, and FISH for PIK3CA copy number. In addition, a panel of NSCLC cell lines characterized for alterations in the PI3K pathway was screened with PI3K and dual PI3K/mTOR inhibitors to assess the preclinical predictive value of candidate biomarkers. RESULTS: PIK3CA amplification was detected in 37% of squamous tumors and 5% of adenocarcinomas, whereas PIK3CA mutations were found in 9% of squamous and 0% of adenocarcinomas. Total loss of PTEN immunostaining was found in 21% of squamous tumors and 4% of adenocarcinomas. Cell lines harboring pathway alterations (receptor tyrosine kinase activation, PI3K mutation or amplification, and PTEN loss) were exquisitely sensitive to the PI3K inhibitor GDC-0941. A dual PI3K/mTOR inhibitor had broader activity across the cell line panel and in tumor xenografts. The combination of GDC-0941 with paclitaxel, erlotinib, or a mitogen-activated protein-extracellular signal-regulated kinase inhibitor had greater effects on cell viability than PI3K inhibition alone. CONCLUSIONS: Candidate biomarkers for PI3K inhibitors have predictive value in preclinical models and show histology-specific alterations in primary tumors, suggesting that distinct biomarker strategies may be required in squamous compared with nonsquamous NSCLC patient populations.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Carcinoma, Non-Small-Cell Lung/enzymology , Indazoles/pharmacology , Lung Neoplasms/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Comparative Genomic Hybridization , DNA Copy Number Variations , DNA Mutational Analysis , Drug Synergism , Erlotinib Hydrochloride , Female , Gene Amplification , Humans , Inhibitory Concentration 50 , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Nude , Molecular Targeted Therapy , PTEN Phosphohydrolase/metabolism , Paclitaxel/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Quinazolines/pharmacology , Signal Transduction , Transcriptome , Xenograft Model Antitumor Assays
13.
Cancer Invest ; 30(10): 727-31, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23061802

ABSTRACT

In this multicenter phase Ib study, drozitumab was given in combination with the mFOLFOX6 regimen and bevacizumab in patients with previously untreated, locally advanced recurrent or metastatic colorectal cancer on day 1 of every 14-day cycle. Nine patients were treated at 2 different cohort dose levels of drozitumab. No dose-limiting toxicities occurred at either dose level and the maximum tolerated dose was not reached. Two patients had a partial response of 4.93 and 4.96 months duration. Cohort 2 dose level is the recommended starting dose level for future trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma/drug therapy , Colorectal Neoplasms/drug therapy , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Carcinoma/genetics , Carcinoma/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Disease-Free Survival , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Humans , Immunohistochemistry , Leucovorin/administration & dosage , Leucovorin/adverse effects , Male , Maximum Tolerated Dose , Middle Aged , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/adverse effects
14.
Clin Cancer Res ; 18(8): 2391-401, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22492982

ABSTRACT

PURPOSE: Elevated levels or increases in circulating tumor cells (CTC) portend poor prognosis in patients with epithelial cancers. Less is known about CTCs as surrogate endpoints or their use for predictive biomarker evaluation. This study investigated the utility of CTC enumeration and characterization using the CellSearch platform, as well as mutation detection in circulating tumor DNA (ctDNA), in patients with advanced non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN: Forty-one patients were enrolled in a single-arm phase II clinical trial of erlotinib and pertuzumab. Peripheral blood was analyzed for CTC enumeration, EGFR expression in CTCs, and detection of oncogenic mutations in CTCs and ctDNA. Changes in CTC levels were correlated with 2[18F]fluoro-2-deoxy-D-glucose-positron emission tomographic (FDG-PET) and computed tomographic (CT) imaging and survival endpoints. RESULTS: CTCs were detected (≥ 1 CTC) at baseline in 78% of patients. Greater sensitivity for mutation detection was observed in ctDNA than in CTCs and detected mutations were strongly concordant with mutation status in matched tumor. Higher baseline CTC counts were associated with response to treatment by Response Evaluation Criteria in Solid Tumors (RECIST, P = 0.009) and decreased CTC counts upon treatment were associated with FDG-PET and RECIST response (P = 0.014 and P = 0.019) and longer progression-free survival (P = 0.050). CONCLUSION: These data provide evidence of a correlation between decreases in CTC counts and radiographic response by either FDG-PET or RECIST in patients with advanced NSCLC. These findings require prospective validation but suggest a potential role for using CTC decreases as an early indication of response to therapy and ctDNA for real-time assessment of mutation status from blood.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , DNA, Neoplasm/blood , Lung Neoplasms/drug therapy , Neoplastic Cells, Circulating , Antibodies, Monoclonal, Humanized/administration & dosage , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Disease-Free Survival , Endpoint Determination , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Female , Genotype , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/genetics , Male , Mutation , Positron-Emission Tomography , Quinazolines/administration & dosage
15.
Mol Cancer Ther ; 11(5): 1143-54, 2012 May.
Article in English | MEDLINE | ID: mdl-22402123

ABSTRACT

The RAS/RAF/MEK pathway is activated in more than 30% of human cancers, most commonly via mutation in the K-ras oncogene and also via mutations in BRAF. Several allosteric mitogen-activated protein/extracellular signal-regulated kinase (MEK) inhibitors, aimed at treating tumors with RAS/RAF pathway alterations, are in clinical development. However, acquired resistance to these inhibitors has been documented both in preclinical and clinical samples. To identify strategies to overcome this resistance, we have derived three independent MEK inhibitor-resistant cell lines. Resistance to allosteric MEK inhibitors in these cell lines was consistently linked to acquired mutations in the allosteric binding pocket of MEK. In one cell line, concurrent amplification of mutant K-ras was observed in conjunction with MEK allosteric pocket mutations. Clonal analysis showed that both resistance mechanisms occur in the same cell and contribute to enhanced resistance. Importantly, in all cases the MEK-resistant cell lines retained their addiction to the mitogen-activated protein kinase (MAPK) pathway, as evidenced by their sensitivity to a selective inhibitor of the ERK1/2 kinases. These data suggest that tumors with acquired MEK inhibitor resistance remain dependent on the MAPK pathway and are therefore sensitive to inhibitors that act downstream of the mutated MEK target. Importantly, we show that dual inhibition of MEK and ERK by small molecule inhibitors was synergistic and acted to both inhibit the emergence of resistance, as well as to overcome acquired resistance to MEK inhibitors. Therefore, our data provide a rationale for cotargeting multiple nodes within the MAPK signaling cascade in K-ras mutant tumors to maximize therapeutic benefit for patients.


Subject(s)
Drug Resistance, Neoplasm , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Genes, ras , Humans , Inhibitory Concentration 50 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Protein Binding , Protein Kinase Inhibitors/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction/drug effects
16.
Clin Cancer Res ; 18(8): 2360-73, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22261801

ABSTRACT

PURPOSE: Non-small cell lung cancers (NSCLC) comprise multiple distinct biologic groups with different prognoses. For example, patients with epithelial-like tumors have a better prognosis and exhibit greater sensitivity to inhibitors of the epidermal growth factor receptor (EGFR) pathway than patients with mesenchymal-like tumors. Here, we test the hypothesis that epithelial-like NSCLCs can be distinguished from mesenchymal-like NSCLCs on the basis of global DNA methylation patterns. EXPERIMENTAL DESIGN: To determine whether phenotypic subsets of NSCLCs can be defined on the basis of their DNA methylation patterns, we combined microfluidics-based gene expression analysis and genome-wide methylation profiling. We derived robust classifiers for both gene expression and methylation in cell lines and tested these classifiers in surgically resected NSCLC tumors. We validate our approach using quantitative reverse transcriptase PCR and methylation-specific PCR in formalin-fixed biopsies from patients with NSCLC who went on to fail front-line chemotherapy. RESULTS: We show that patterns of methylation divide NSCLCs into epithelial-like and mesenchymal-like subsets as defined by gene expression and that these signatures are similarly correlated in NSCLC cell lines and tumors. We identify multiple differentially methylated regions, including one in ERBB2 and one in ZEB2, whose methylation status is strongly associated with an epithelial phenotype in NSCLC cell lines, surgically resected tumors, and formalin-fixed biopsies from patients with NSCLC who went on to fail front-line chemotherapy. CONCLUSIONS: Our data show that patterns of DNA methylation can divide NSCLCs into two phenotypically distinct subtypes of tumors and provide proof of principle that differences in DNA methylation can be used as a platform for predictive biomarker discovery and development.


Subject(s)
Carcinoma, Non-Small-Cell Lung/classification , Carcinoma, Non-Small-Cell Lung/genetics , DNA Methylation/genetics , Lung Neoplasms/classification , Lung Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , CpG Islands/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genome-Wide Association Study , Homeodomain Proteins/genetics , Humans , Lung/pathology , Lung Neoplasms/pathology , Phenotype , Prognosis , Receptor, ErbB-2/genetics , Repressor Proteins/genetics , Zinc Finger E-box Binding Homeobox 2
17.
Clin Cancer Res ; 17(10): 3304-15, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21364032

ABSTRACT

PURPOSE: Assessing clinical activity of molecularly targeted anticancer agents, especially in the absence of tumor shrinkage, is challenging. To evaluate on-treatment 18F-fluorodeoxyglucose (FDG) and/or 18F-fluorodeoxythymidine (FLT) positron emission tomography (PET) for this purpose, we conducted a prospective multicenter trial assessing PET response rates and associations with progression-free (PFS) and overall survival (OS) in 2nd/3rd-line non-small-cell lung cancer patients treated with erlotinib. EXPERIMENTAL DESIGN: PET/computed tomography (CT) scans were conducted at baseline, day (d)14 and d56 after the first daily erlotinib dose, with diagnostic CT at baseline and d56 (all scans centrally reviewed). PET partial metabolic response (PMR) was defined as a mean decrease (in ≤ 5 lesions/patient) of 15% or more maximum standardized uptake value. PFS was investigator-determined. RESULTS: Of 74 erlotinib-treated patients, 51 completed all imaging assessments through d56; 13 of 51 (26%) FDG-evaluable patients had PMR at d14, as did 9 of 50 (18%) FLT-evaluable patients. Four (7.8%) showed partial responses (PR) by d56 CT; all 4 had PMR by d14 FDG-PET with 3 PMRs by d14 FLT-PET. Three of the 4 patients with CT PR had evaluable archival tumor tissue; all 3 had epidermal growth factor receptor mutations. D14 and d56 PMRs by FDG or FLT were associated with improved PFS; HRs for PET responders versus nonresponders were 0.3 to 0.4. D14 FDG-PET PMR was associated with improved OS (P = 0.03) compared with FDG-PET nonresponders. CONCLUSION: Early (d14) FDG-PET PMR is associated with improved PFS and OS, even in the absence of subsequent Response Evaluation Criteria in Solid Tumors response. These data support inclusion of FDG-PET imaging in clinical trials testing novel targeted therapies, particularly those with anticipated cytostatic effects.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/drug therapy , Fluorodeoxyglucose F18 , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/drug therapy , Quinazolines/therapeutic use , Thymidine/analogs & derivatives , Aged , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Disease Progression , Erlotinib Hydrochloride , Female , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging/methods , Positron-Emission Tomography/methods , Radiopharmaceuticals , Survival Analysis
18.
PLoS One ; 5(9): e12517, 2010 Sep 08.
Article in English | MEDLINE | ID: mdl-20838621

ABSTRACT

BACKGROUND: Evaluation of cancer biomarkers from blood could significantly enable biomarker assessment by providing a relatively non-invasive source of representative tumor material. Circulating Tumor Cells (CTCs) isolated from blood of metastatic cancer patients hold significant promise in this regard. METHODOLOGY/PRINCIPAL FINDINGS: Using spiked tumor-cells we evaluated CTC capture on different CTC technology platforms, including CellSearch and two biochip platforms, and used the isolated CTCs to develop and optimize assays for molecular characterization of CTCs. We report similar performance for the various platforms tested in capturing CTCs, and find that capture efficiency is dependent on the level of EpCAM expression. We demonstrate that captured CTCs are amenable to biomarker analyses such as HER2 status, qRT-PCR for breast cancer subtype markers, KRAS mutation detection, and EGFR staining by immunofluorescence (IF). We quantify cell surface expression of EGFR in metastatic lung cancer patient samples. In addition, we determined HER2 status by IF and FISH in CTCs from metastatic breast cancer patients. In the majority of patients (89%) we found concordance with HER2 status from patient tumor tissue, though in a subset of patients (11%), HER2 status in CTCs differed from that observed in the primary tumor. Surprisingly, we found CTC counts to be higher in ER+ patients in comparison to HER2+ and triple negative patients, which could be explained by low EpCAM expression and a more mesenchymal phenotype of tumors belonging to the basal-like molecular subtype of breast cancer. CONCLUSIONS/SIGNIFICANCE: Our data suggests that molecular characterization from captured CTCs is possible and can potentially provide real-time information on biomarker status. In this regard, CTCs hold significant promise as a source of tumor material to facilitate clinical biomarker evaluation. However, limitations exist from a purely EpCAM based capture system and addition of antibodies to mesenchymal markers could further improve CTC capture efficiency to enable routine biomarker analysis from CTCs.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplastic Cells, Circulating/metabolism , Receptor, ErbB-2/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Receptor, ErbB-2/metabolism
19.
Expert Opin Biol Ther ; 10(9): 1343-55, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20695834

ABSTRACT

IMPORTANCE OF THE FIELD: Heterodimerization of human EGF receptor (HER) 2 and HER3, a co-receptor of HER2, plays an important and dominant role in the functionality and transformation of HER-mediated pathways. Understanding the role of HER3 in oncogenesis as well as its place as a target for anticancer therapy is an ongoing area of research. Determination of biomarkers for clinical benefit from agents targeting HER3 is an essential component of translating basic science into real-world effective anticancer therapies, with the aim of ensuring the patients most likely to benefit from such treatments can be identified. AREAS COVERED IN THIS REVIEW: This review focuses on the targeting of HER2 and HER3 by monoclonal antibodies and the potential for HER3 mRNA levels to predict treatment outcome in ovarian cancer. WHAT THE READER WILL GAIN: An understanding of the value of biomarkers for clinical benefit to anticancer therapy and the current status of HER3 mRNA as a biomarker for clinical benefit of the HER2-HER3 dimerization inhibitor pertuzumab. TAKE HOME MESSAGE: HER3 mRNA levels may be a biomarker for active ligand-induced HER2-HER3 signaling, with low HER3 mRNA levels correlated with clinical benefit from the HER2-HER3 dimerization inhibitor pertuzumab.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , RNA, Messenger/analysis , Receptor, ErbB-3/genetics , Antibodies, Monoclonal, Humanized , Female , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Precision Medicine , Protein Multimerization , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Treatment Outcome
20.
Clin Cancer Res ; 16(14): 3670-83, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20453058

ABSTRACT

PURPOSE: The class I phosphatidylinositol 3' kinase (PI3K) plays a major role in proliferation and survival in a wide variety of human cancers. A key factor in successful development of drugs targeting this pathway is likely to be the identification of responsive patient populations with predictive diagnostic biomarkers. This study sought to identify candidate biomarkers of response to the selective PI3K inhibitor GDC-0941. EXPERIMENTAL DESIGN: We used a large panel of breast cancer cell lines and in vivo xenograft models to identify candidate predictive biomarkers for a selective inhibitor of class I PI3K that is currently in clinical development. The approach involved pharmacogenomic profiling as well as analysis of gene expression data sets from cells profiled at baseline or after GDC-0941 treatment. RESULTS: We found that models harboring mutations in PIK3CA, amplification of human epidermal growth factor receptor 2, or dual alterations in two pathway components were exquisitely sensitive to the antitumor effects of GDC-0941. We found that several models that do not harbor these alterations also showed sensitivity, suggesting a need for additional diagnostic markers. Gene expression studies identified a collection of genes whose expression was associated with in vitro sensitivity to GDC-0941, and expression of a subset of these genes was found to be intimately linked to signaling through the pathway. CONCLUSION: Pathway focused biomarkers and the gene expression signature described in this study may have utility in the identification of patients likely to benefit from therapy with a selective PI3K inhibitor.


Subject(s)
Breast Neoplasms/drug therapy , Disease Models, Animal , Indazoles/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Sulfonamides/pharmacology , Animals , Apoptosis/drug effects , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Female , Gene Expression Profiling , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mutation , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Predictive Value of Tests , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Sensitivity and Specificity , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...