Network


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

Hotspot


Dive into the research topics where Jian-Mei Hou is active.

Publication


Featured researches published by Jian-Mei Hou.


Journal of Clinical Oncology | 2011

Evaluation and Prognostic Significance of Circulating Tumor Cells in Patients With Non–Small-Cell Lung Cancer

Matthew Krebs; Robert Sloane; Lynsey Priest; Lee Lancashire; Jian-Mei Hou; Alastair Greystoke; Timothy H Ward; Roberta Ferraldeschi; Andrew Hughes; Glen Clack; Malcolm R Ranson; Caroline Dive; Fiona Blackhall

PURPOSE Lung cancer is the leading cause of cancer-related death worldwide. Non-small-cell lung cancer (NSCLC) lacks validated biomarkers to predict treatment response. This study investigated whether circulating tumor cells (CTCs) are detectable in patients with NSCLC and what their ability might be to provide prognostic information and/or early indication of patient response to conventional therapy. PATIENTS AND METHODS In this single-center prospective study, blood samples for CTC analysis were obtained from 101 patients with previously untreated, stage III or IV NSCLC both before and after administration of one cycle of standard chemotherapy. CTCs were measured using a semiautomated, epithelial cell adhesion molecule-based immunomagnetic technique. RESULTS The number of CTCs in 7.5 mL of blood was higher in patients with stage IV NSCLC (n = 60; range, 0 to 146) compared with patients with stage IIIB (n = 27; range, 0 to 3) or IIIA disease (n = 14; no CTCs detected). In univariate analysis, progression-free survival was 6.8 v 2.4 months with P < .001, and overall survival (OS) was 8.1 v 4.3 months with P < .001 for patients with fewer than five CTCs compared with five or more CTCs before chemotherapy, respectively. In multivariate analysis, CTC number was the strongest predictor of OS (hazard ratio [HR], 7.92; 95% CI, 2.85 to 22.01; P < .001), and the point estimate of the HR was increased with incorporation of a second CTC sample that was taken after one cycle of chemotherapy (HR, 15.65; 95% CI, 3.63 to 67.53; P < .001). CONCLUSION CTCs are detectable in patients with stage IV NSCLC and are a novel prognostic factor for this disease. Further validation is warranted before routine clinical application.


American Journal of Pathology | 2011

Circulating tumor cells as a window on metastasis biology in lung cancer.

Jian-Mei Hou; Matthew Krebs; Timothy H Ward; Robert Sloane; Lynsey Priest; Andrew Hughes; Glen Clack; Malcolm R Ranson; Fiona Blackhall; Caroline Dive

Circulating tumor cell (CTC) number in metastatic cancer patients yields prognostic information consistent with enhanced cell migration and invasion via loss of adhesion, a feature of epithelial-to-mesenchymal transition (EMT). Tumor cells also invade via collective migration with maintained cell-cell contacts and consistent with this is the circulating tumor microemboli (CTM; contiguous groups of tumor cells) that are observed in metastatic cancer patients. Using a blood filtration approach, we examined markers of EMT (cytokeratins, E-cadherin, vimentin, neural cadherin) and prevalence of apoptosis in CTCs and CTM to explore likely mechanism(s) of invasion in lung cancer patients and address the hypothesis that cells within CTM have a survival advantage. Intra-patient and inter-patient heterogeneity was observed for EMT markers in CTCs and CTM. Vimentin was only expressed in some CTCs, but in the majority of cells within CTM; E-cadherin expression was lost, cytoplasmic or nuclear, and rarely expressed at the surface of the cells within CTM. A subpopulation of CTCs was apoptotic, but apoptosis was absent within CTM. This pilot study suggests that EMT is not prosecuted homogeneously in tumor cells within the circulation of lung cancer patients and that collective migration and enhanced survival of cells within CTM might contribute to lung cancer metastasis. Multiplex analysis and further detailed exploration of metastatic potential and EMT in CTCs/CTM is now warranted in a larger patient cohort.


Journal of Thoracic Oncology | 2012

Analysis of circulating tumor cells in patients with non-small cell lung cancer using epithelial marker-dependent and -independent approaches.

Matthew Krebs; Jian-Mei Hou; Robert Sloane; Lee Lancashire; Lynsey Priest; Daisuke Nonaka; Timothy H Ward; Alison C Backen; Glen Clack; Andrew Hughes; Malcolm R Ranson; Fiona Blackhall; Caroline Dive

Introduction: Epithelial circulating tumor cells (CTCs) are detectable in patients with non-small cell lung cancer (NSCLC). However, epithelial to mesenchymal transition, a widely reported prerequisite for metastasis, may lead to an underestimation of CTC number. We compared directly an epithelial marker-dependent (CellSearch) and a marker-independent (isolation by size of epithelial tumor cells [ISET]) technology platform for the ability to identify CTCs. Molecular characteristics of CTCs were also explored. Methods: Paired peripheral blood samples were collected from 40 chemonäive, stages IIIA to IV NSCLC patients. CTCs were enumerated by Epithelial Cell Adhesion Molecule-based immunomagnetic capture (CellSearch, Veridex) and by filtration (ISET, RareCell Diagnostics). CTCs isolated by filtration were assessed by immunohistochemistry for epithelial marker expression (cytokeratins, Epithelial Cell Adhesion Molecule, epidermal growth factor receptor) and for proliferation status (Ki67). Results: CTCs were detected using ISET in 32 of 40 (80%) patients compared with 9 of 40 (23%) patients using CellSearch. A subpopulation of CTCs isolated by ISET did not express epithelial markers. Circulating tumor microemboli (CTM, clusters of ≥3 CTCs) were observed in 43% patients using ISET but were undetectable by CellSearch. Up to 62% of single CTCs were positive for the proliferation marker Ki67, whereas cells within CTM were nonproliferative. Conclusions: Both technology platforms detected NSCLC CTCs. ISET detected higher numbers of CTCs including epithelial marker negative tumor cells. ISET also isolated CTM and permitted molecular characterization. Combined with our previous CellSearch data confirming CTC number as an independent prognostic biomarker for NSCLC, we propose that this complementary dual technology approach to CTC analysis allows more complete exploration of CTCs in patients with NSCLC.


American Journal of Pathology | 2009

Evaluation of Circulating Tumor Cells and Serological Cell Death Biomarkers in Small Cell Lung Cancer Patients Undergoing Chemotherapy

Jian-Mei Hou; Alastair Greystoke; Lee Lancashire; Jeff Cummings; Tim Ward; Ruth Board; Eitan Amir; Sarah J. Hughes; Matthew Krebs; Andrew Hughes; Malcolm R Ranson; Paul Lorigan; Caroline Dive; Fiona Blackhall

Serological cell death biomarkers and circulating tumor cells (CTCs) have potential uses as tools for pharmacodynamic blood-based assays and their subsequent application to early clinical trials. In this study, we evaluated both the expression and clinical significance of CTCs and serological cell death biomarkers in patients with small cell lung cancer. Blood samples from 88 patients were assayed using enzyme-linked immunosorbent assays for various cytokeratin 18 products (eg, M65, cell death, M30, and apoptosis) as well as nucleosomal DNA. CTCs (per 7.5 ml of blood) were quantified using Veridex CellSearch technology. Before therapeutic treatment, cell death biomarkers were elevated in patients compared with controls. CTCs were detected in 86% of patients; additionally, CD56 was detectable in CTCs, confirming their neoplastic origin. M30 levels correlated with the percentage of apoptotic CTCs. M30, M65, lactate dehydrogenase, and CTC number were prognostic for patient survival as determined by univariate analysis. Using multivariate analysis, both lactate dehydrogenase and M65 levels remained significant. CTC number fell following chemotherapy, whereas levels of serological cell death biomarkers peaked at 48 hours and fell by day 22, mirroring the tumor response. A 48-hour rise in nucleosomal DNA and M30 levels was associated with early response and severe toxicity, respectively. Our results provide a rationale to include the use of serological biomarkers and CTCs in early clinical trials of new agents for small cell lung cancer.


British Journal of Cancer | 2008

Biomarkers of apoptosis.

Timothy H Ward; Jeffrey Cummings; Emma Dean; Alastair Greystoke; Jian-Mei Hou; Alison C Backen; Malcolm R Ranson; Caroline Dive

Within the era of molecularly targeted anticancer agents, it has become increasingly important to provide proof of mechanism as early on as possible in the drug development cycle, especially in the clinic. Selective activation of apoptosis is often cited as one of the major goals of cancer chemotherapy. Thus, the present minireview focuses on a discussion of the pros and cons of a variety of methodological approaches to detect different components of the apoptotic cascade as potential biomarkers of programmed cell death. The bulk of the discussion centres on serological assays utilising the technique of ELISA, since here there is an obvious advantage of sampling multiple time points. Potential biomarkers of apoptosis including circulating tumour cells, cytokeratins and DNA nucleosomes are discussed at length. However, accepting that a single biomarker may not have the power to predict proof of concept and patient outcome, it is clear that in the future more emphasis will be placed on technologies that can analyse panels of biomarkers in small volumes of samples. To this end the increased throughput afforded by multiplex ELISA technologies is discussed.


Lung Cancer | 2012

Biomarkers for small cell lung cancer: neuroendocrine, epithelial and circulating tumour cells.

Rachel Stovold; Fiona Blackhall; Suzanne Meredith; Jian-Mei Hou; Caroline Dive; Anne White

Small cell lung cancer (SCLC) is characterised by an aggressive clinical course with invariable resistance to chemotherapy despite initially high response rates. There has been little improvement in outcome over the past few decades, with no breakthrough yet in targeted therapies. Recent preclinical data and studies of circulating tumour cells (CTCs) highlight distinct cellular heterogeneity within SCLC. Better understanding of how these phenotypes contribute to metastasis and tumour progression might pave the way for development of more successful targeted therapies. Here we review these studies, their implications for future research and for the incorporation of biomarkers reflecting neuroendocrine, epithelial and mesenchymal phenotypes in clinical studies.


Journal of Thoracic Oncology | 2012

Original ArticleAnalysis of Circulating Tumor Cells in Patients with Non-small Cell Lung Cancer Using Epithelial Marker-Dependent and -Independent Approaches

Matthew Krebs; Jian-Mei Hou; Robert Sloane; Lee Lancashire; Lynsey Priest; Daisuke Nonaka; Timothy H Ward; Alison C Backen; Glen Clack; Andrew Hughes; Malcolm R Ranson; Fiona Blackhall; Caroline Dive

Introduction: Epithelial circulating tumor cells (CTCs) are detectable in patients with non-small cell lung cancer (NSCLC). However, epithelial to mesenchymal transition, a widely reported prerequisite for metastasis, may lead to an underestimation of CTC number. We compared directly an epithelial marker-dependent (CellSearch) and a marker-independent (isolation by size of epithelial tumor cells [ISET]) technology platform for the ability to identify CTCs. Molecular characteristics of CTCs were also explored. Methods: Paired peripheral blood samples were collected from 40 chemonäive, stages IIIA to IV NSCLC patients. CTCs were enumerated by Epithelial Cell Adhesion Molecule-based immunomagnetic capture (CellSearch, Veridex) and by filtration (ISET, RareCell Diagnostics). CTCs isolated by filtration were assessed by immunohistochemistry for epithelial marker expression (cytokeratins, Epithelial Cell Adhesion Molecule, epidermal growth factor receptor) and for proliferation status (Ki67). Results: CTCs were detected using ISET in 32 of 40 (80%) patients compared with 9 of 40 (23%) patients using CellSearch. A subpopulation of CTCs isolated by ISET did not express epithelial markers. Circulating tumor microemboli (CTM, clusters of ≥3 CTCs) were observed in 43% patients using ISET but were undetectable by CellSearch. Up to 62% of single CTCs were positive for the proliferation marker Ki67, whereas cells within CTM were nonproliferative. Conclusions: Both technology platforms detected NSCLC CTCs. ISET detected higher numbers of CTCs including epithelial marker negative tumor cells. ISET also isolated CTM and permitted molecular characterization. Combined with our previous CellSearch data confirming CTC number as an independent prognostic biomarker for NSCLC, we propose that this complementary dual technology approach to CTC analysis allows more complete exploration of CTCs in patients with NSCLC.


Cancer Research | 2010

Abstract 1022: PI3-kinase inhibition enhances ABT-737-induced apoptosis in colorectal cancer cell lines

Christopher J. Morrow; Jian-Mei Hou; Caroline Dive

The PI3-kinase signalling pathway is widely reported to promote survival, proliferation and migration. This pathway is up-regulated in several solid tumours by a host of different mechanisms, including PTEN deletion, RAS or PIK3CA mutation and aberrant activation of receptor tyrosine kinases. As such, inhibitors of PI3-kinase and its down-stream effectors are of great interest as tractable anti-cancer agents. The effect of PI3-kinase inhibition was investigated in colorectal cancer cell lines and demonstrated that, while PI3-kinase inhibition by itself did not cause cell death, it enhanced apoptosis induced by the BH3 mimetic ABT-737 (assessed by nuclear fragmentation, caspase 3 cleavage and phosphatidylserine exposure). Furthermore, ABT-737 and PI3-kinase inhibition caused increased level of Bak activating conformational change compared to ABT-737 alone, suggesting that the effect of PI3-kinase inhibition is up-stream of cytochrome C release. PI3-kinase inhibition did not alter the expression level of Bcl-2 family members, but co-immunoprecipitation revealed that it increased the amount of Bim bound to Bcl-x L . No change was seen in the binding of other BH3 only proteins or of Bax or Bak to ABT-737 targets Bcl-2 or Bcl-xL. ABT-737-induced apoptosis was independent of AKT and mTOR signalling as it was unaffected by the inhibitors AKTi1/2 and rapamycin respectively. A model is proposed whereby ABT-737 interrupts PI-3K inhibition-induced Bim/Bcl-x L complexes to promote apoptosis via Bax/Bak activation. Function testing experiments are underway to interrogate this model. Note: This abstract was not presented at the AACR 101st Annual Meeting 2010 because the presenter was unable to attend. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 1022.


Journal of Clinical Oncology | 2011

Phase III randomized, open-label, multicenter trial (BRIM3) comparing BRAF inhibitor vemurafenib with dacarbazine (DTIC) in patients with V600E BRAF-mutated melanoma

Phil Chapman; Axel Hauschild; Caroline Robert; James Larkin; John B. A. G. Haanen; A. Ribas; David W. Hogg; Steven O'Day; Paolo Antonio Ascierto; Alessandro Testori; Paul Lorigan; Reinhard Dummer; J. A. Sosman; Claus Garbe; Rebecca Lee; Keith Nolop; Betty Nelson; Jian-Mei Hou; Keith T. Flaherty; Grant A. McArthur


Lung Cancer | 2012

182 Do circulating tumour cell (CTC) counts correlate with tumour volume in limited disease small cell lung cancer (LD-SCLC)? An exploratory clinical study with survival outcomes

P. Koh; Jian-Mei Hou; T. Westwood; J. Cain; G. Webster; Lynsey Priest; R. Sloane; M. Krebs; Corinne Faivre-Finn; Caroline Dive; Fiona Blackhall

Collaboration


Dive into the Jian-Mei Hou's collaboration.

Top Co-Authors

Avatar

Caroline Dive

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Matthew Krebs

University of Manchester

View shared research outputs
Top Co-Authors

Avatar

Lynsey Priest

University of Manchester

View shared research outputs
Top Co-Authors

Avatar

Timothy H Ward

University of Manchester

View shared research outputs
Top Co-Authors

Avatar

Lee Lancashire

Nottingham Trent University

View shared research outputs
Top Co-Authors

Avatar

Robert Sloane

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge