Network


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

Hotspot


Dive into the research topics where James Che is active.

Publication


Featured researches published by James Che.


Oncotarget | 2016

Classification of large circulating tumor cells isolated with ultra-high throughput microfluidic Vortex technology

James Che; Victor Yu; Manjima Dhar; Corinne Renier; Melissa Matsumoto; Kyra Heirich; Edward B. Garon; Jonathan W. Goldman; Jianyu Rao; George W. Sledge; Mark D. Pegram; Shruti Sheth; Stefanie S. Jeffrey; Rajan P. Kulkarni; Elodie Sollier; Dino Di Carlo

Circulating tumor cells (CTCs) are emerging as rare but clinically significant non-invasive cellular biomarkers for cancer patient prognosis, treatment selection, and treatment monitoring. Current CTC isolation approaches, such as immunoaffinity, filtration, or size-based techniques, are often limited by throughput, purity, large output volumes, or inability to obtain viable cells for downstream analysis. For all technologies, traditional immunofluorescent staining alone has been employed to distinguish and confirm the presence of isolated CTCs among contaminating blood cells, although cells isolated by size may express vastly different phenotypes. Consequently, CTC definitions have been non-trivial, researcher-dependent, and evolving. Here we describe a complete set of objective criteria, leveraging well-established cytomorphological features of malignancy, by which we identify large CTCs. We apply the criteria to CTCs enriched from stage IV lung and breast cancer patient blood samples using the High Throughput Vortex Chip (Vortex HT), an improved microfluidic technology for the label-free, size-based enrichment and concentration of rare cells. We achieve improved capture efficiency (up to 83%), high speed of processing (8 mL/min of 10x diluted blood, or 800 μL/min of whole blood), and high purity (avg. background of 28.8±23.6 white blood cells per mL of whole blood). We show markedly improved performance of CTC capture (84% positive test rate) in comparison to previous Vortex designs and the current FDA-approved gold standard CellSearch assay. The results demonstrate the ability to quickly collect viable and pure populations of abnormal large circulating cells unbiased by molecular characteristics, which helps uncover further heterogeneity in these cells.


Nature Communications | 2017

Profiling protein expression in circulating tumour cells using microfluidic western blotting.

Elly Sinkala; Elodie Sollier-Christen; Corinne Renier; Elisabet Rosàs-Canyelles; James Che; Kyra Heirich; Todd A. Duncombe; Julea Vlassakis; Kevin A. Yamauchi; Haiyan Huang; Stefanie S. Jeffrey; Amy E. Herr

Circulating tumour cells (CTCs) are rare tumour cells found in the circulatory system of certain cancer patients. The clinical and functional significance of CTCs is still under investigation. Protein profiling of CTCs would complement the recent advances in enumeration, transcriptomic and genomic characterization of these rare cells and help define their characteristics. Here we describe a microfluidic western blot for an eight-plex protein panel for individual CTCs derived from estrogen receptor-positive (ER+) breast cancer patients. The precision handling and analysis reveals a capacity to assay sparingly available patient-derived CTCs, a biophysical CTC phenotype more lysis-resistant than breast cancer cell lines, a capacity to report protein expression on a per CTC basis and two statistically distinct GAPDH subpopulations within the patient-derived CTCs. Targeted single-CTC proteomics with the capacity for archivable, multiplexed protein analysis offers a unique, complementary taxonomy for understanding CTC biology and ascertaining clinical impact.


PLOS ONE | 2013

Microfluidic Purification and Concentration of Malignant Pleural Effusions for Improved Molecular and Cytomorphological Diagnostics

James Che; Albert J. Mach; Derek E. Go; Ish Talati; Yong Ying; Jianyu Rao; Rajan P. Kulkarni; Dino Di Carlo

Evaluation of pleural fluids for metastatic cells is a key component of diagnostic cytopathology. However, a large background of smaller leukocytes and/or erythrocytes can make accurate diagnosis difficult and reduce specificity in identification of mutations of interest for targeted anti-cancer therapies. Here, we describe an automated microfluidic system (Centrifuge Chip) which employs microscale vortices for the size-based isolation and concentration of cancer cells and mesothelial cells from a background of blood cells. We are able to process non-diluted pleural fluids at 6 mL/min and enrich target cells significantly over the background; we achieved improved purity in all patient samples analyzed. The resulting isolated and viable cells are readily available for immunostaining, cytological analysis, and detection of gene mutations. To demonstrate the utility towards aiding companion diagnostics, we also show improved detection accuracy of KRAS gene mutations in lung cancer cells processed using the Centrifuge Chip, leading to an increase in the area under the curve (AUC) of the receiver operating characteristic from 0.90 to 0.99. The Centrifuge Chip allows for rapid concentration and processing of large volumes of bodily fluid samples for improved cytological diagnosis and purification of cells of interest for genetic testing, which will be helpful for enhancing diagnostic accuracy.


Biomicrofluidics | 2015

High efficiency vortex trapping of circulating tumor cells

Manjima Dhar; Jessica Wong; Armin Karimi; James Che; Corinne Renier; Melissa Matsumoto; Melanie Triboulet; Edward B. Garon; Jonathan W. Goldman; Matthew B. Rettig; Stefanie S. Jeffrey; Rajan P. Kulkarni; Elodie Sollier; Dino Di Carlo

Circulating tumor cells (CTCs) are important biomarkers for monitoring tumor dynamics and efficacy of cancer therapy. Several technologies have been demonstrated to isolate CTCs with high efficiency but achieve a low purity from a large background of blood cells. We have previously shown the ability to enrich CTCs with high purity from large volumes of blood through selective capture in microvortices using the Vortex Chip. The device consists of a narrow channel followed by a series of expansion regions called reservoirs. Fast flow in the narrow entry channel gives rise to inertial forces, which direct larger cells into trapping vortices in the reservoirs where they remain circulating in orbits. By studying the entry and stability of particles following entry into reservoirs, we discover that channel cross sectional area plays an important role in controlling the size of trapped particles, not just the orbital trajectories. Using these design modifications, we demonstrate a new device that is able to capture a wider size range of CTCs from clinical samples, uncovering further heterogeneity. This simple biophysical method opens doors for a range of downstream interventions, including genetic analysis, cell culture, and ultimately personalized cancer therapy.


Oncotarget | 2016

Enumeration and targeted analysis of KRAS , BRAF and PIK3CA mutations in CTCs captured by a label-free platform: Comparison to ctDNA and tissue in metastatic colorectal cancer

Evelyn Kidess-Sigal; Haiyan E. Liu; Melanie Triboulet; James Che; Vishnu C. Ramani; Brendan C. Visser; George A. Poultsides; Teri A. Longacre; Andre Marziali; Valentina Vysotskaia; Matthew Wiggin; Kyra Heirich; Violet R. Hanft; Ulrich Keilholz; Ingeborg Tinhofer; Jeffrey A. Norton; Mark Lee; Elodie Sollier-Christen; Stefanie S. Jeffrey

Treatment of advanced colorectal cancer (CRC) requires multimodal therapeutic approaches and need for monitoring tumor plasticity. Liquid biopsy biomarkers, including CTCs and ctDNA, hold promise for evaluating treatment response in real-time and guiding therapeutic modifications. From 15 patients with advanced CRC undergoing liver metastasectomy with curative intent, we collected 41 blood samples at different time points before and after surgery for CTC isolation and quantification using label-free Vortex technology. For mutational profiling, KRAS, BRAF, and PIK3CA hotspot mutations were analyzed in CTCs and ctDNA from 23 samples, nine matched liver metastases and three primary tumor samples. Mutational patterns were compared. 80% of patient blood samples were positive for CTCs, using a healthy baseline value as threshold (0.4 CTCs/mL), and 81.4% of captured cells were EpCAM+ CTCs. At least one mutation was detected in 78% of our blood samples. Among 23 matched CTC and ctDNA samples, we found a concordance of 78.2% for KRAS, 73.9% for BRAF and 91.3% for PIK3CA mutations. In several cases, CTCs exhibited a mutation that was not detected in ctDNA, and vice versa. Complementary assessment of both CTCs and ctDNA appears advantageous to assess dynamic tumor profiles.


npj Precision Oncology | 2017

Label-free isolation of prostate circulating tumor cells using Vortex microfluidic technology

Corinne Renier; Edward Pao; James Che; Haiyan E. Liu; Clementine A. Lemaire; Melissa Matsumoto; Melanie Triboulet; Sandy Srivinas; Stefanie S. Jeffrey; Matthew Rettig; Rajan P. Kulkarni; Dino Di Carlo; Elodie Sollier-Christen

There has been increased interest in utilizing non-invasive “liquid biopsies” to identify biomarkers for cancer prognosis and monitoring, and to isolate genetic material that can predict response to targeted therapies. Circulating tumor cells (CTCs) have emerged as such a biomarker providing both genetic and phenotypic information about tumor evolution, potentially from both primary and metastatic sites. Currently, available CTC isolation approaches, including immunoaffinity and size-based filtration, have focused on high capture efficiency but with lower purity and often long and manual sample preparation, which limits the use of captured CTCs for downstream analyses. Here, we describe the use of the microfluidic Vortex Chip for size-based isolation of CTCs from 22 patients with advanced prostate cancer and, from an enumeration study on 18 of these patients, find that we can capture CTCs with high purity (from 1.74 to 37.59%) and efficiency (from 1.88 to 93.75 CTCs/7.5 mL) in less than 1 h. Interestingly, more atypical large circulating cells were identified in five age-matched healthy donors (46–77 years old; 1.25–2.50 CTCs/7.5 mL) than in five healthy donors <30 years old (21–27 years old; 0.00 CTC/7.5 mL). Using a threshold calculated from the five age-matched healthy donors (3.37 CTCs/mL), we identified CTCs in 80% of the prostate cancer patients. We also found that a fraction of the cells collected (11.5%) did not express epithelial prostate markers (cytokeratin and/or prostate-specific antigen) and that some instead expressed markers of epithelial–mesenchymal transition, i.e., vimentin and N-cadherin. We also show that the purity and DNA yield of isolated cells is amenable to targeted amplification and next-generation sequencing, without whole genome amplification, identifying unique mutations in 10 of 15 samples and 0 of 4 healthy samples.Prostate cancer: a “liquid biopsy” test for circulating tumor cellsA microfluidic device can rapidly and efficiently isolate circulating tumor cells from the blood of prostate cancer patients. Elodie Sollier-Christen of Vortex Biosciences, Rajan Kulkarni of David Geffen School of Medicine at UCLA, Dino Di Carlo of UCLA and colleagues tested the company’s microfluidic technology on blood samples taken from 21 men with advanced prostate cancer and 10 healthy controls. They showed that, within an hour, the Vortex Chip could isolate circulating tumor cells in 80% of the cancer patients and that many of these cells did not display the usual surface markers that other approaches require to capture prostate cancer cells. The purities and DNA yields of the isolated cells were high enough to enable targeted genome sequencing, which revealed mutations potentially involved in tumor formation. The Vortex technology could help diagnose prostate cancer and inform therapeutic decision-making for those with the disease.


Scientific Reports | 2016

Label-free enumeration, collection and downstream cytological and cytogenetic analysis of circulating tumor cells

Manjima Dhar; Edward Pao; Corinne Renier; Derek E. Go; James Che; Rosita Montoya; Rachel Conrad; Melissa Matsumoto; Kyra Heirich; Melanie Triboulet; Jianyu Rao; Stefanie S. Jeffrey; Edward B. Garon; Jonathan W. Goldman; Nagesh Rao; Rajan P. Kulkarni; Elodie Sollier-Christen; Dino Di Carlo

Circulating tumor cells (CTCs) have a great potential as indicators of metastatic disease that may help physicians improve cancer prognostication, treatment and patient outcomes. Heterogeneous marker expression as well as the complexity of current antibody-based isolation and analysis systems highlights the need for alternative methods. In this work, we use a microfluidic Vortex device that can selectively isolate potential tumor cells from blood independent of cell surface expression. This system was adapted to interface with three protein-marker-free analysis techniques: (i) an in-flow automated image processing system to enumerate cells released, (ii) cytological analysis using Papanicolaou (Pap) staining and (iii) fluorescence in situ hybridization (FISH) targeting the ALK rearrangement. In-flow counting enables a rapid assessment of the cancer-associated large circulating cells in a sample within minutes to determine whether standard downstream assays such as cytological and cytogenetic analyses that are more time consuming and costly are warranted. Using our platform integrated with these workflows, we analyzed 32 non-small cell lung cancer (NSCLC) and 22 breast cancer patient samples, yielding 60 to 100% of the cancer patients with a cell count over the healthy threshold, depending on the detection method used: respectively 77.8% for automated, 60–100% for cytology, and 80% for immunostaining based enumeration.


Lab on a Chip | 2014

Research highlights: microfluidic single-cell analysis from nucleic acids to proteins to functions

Rajan P. Kulkarni; James Che; Manjima Dhar; Dino Di Carlo

We highlight recent reports using microfluidic systems to perform single-cell analysis. It has been demonstrated on numerous occasions that population averages are often not representative of single-cell behavior. These differences in behavior can be caused by stochastic fluctuations in temporal response, changes in the surrounding instructive environment, or hard-coded genetic changes. Because of the similar length scales, microfluidic approaches have been well-suited to isolating, analyzing, and culturing single-cells to better understand this heterogeneity. Here, we discuss recent works in which microfluidic researchers have extended single-cell characterization approaches, in order to improve analysis from nucleic acids to proteins to final functional behavior. Nucleic acid detection can be amplified beyond what is possible with fluorescence in situ hybridization using droplet-enabled PCR-activated cell sorting. Multiplexed protein detection that overcomes the problems with off-target antibody binding which are associated with traditional immunofluorescence is achieved with single-cell western blotting. Proliferation, migration, and secretion are analyzed in rare circulating tumor cells isolated in microwells. The next steps will be getting these new tools into the hands of a growing number of biologists and developing new tools to report on single-cell epigenetic modifications.


npj Genomic Medicine | 2017

Workflow optimization of whole genome amplification and targeted panel sequencing for CTC mutation detection

Haiyan E. Liu; Melanie Triboulet; Amin Zia; Meghah Vuppalapaty; Evelyn Kidess-Sigal; John A. Coller; Vanita Natu; Vida Shokoohi; James Che; Corinne Renier; Natalie H. Chan; Violet R. Hanft; Stefanie S. Jeffrey; Elodie Sollier-Christen

Genomic characterization of circulating tumor cells (CTCs) may prove useful as a surrogate for conventional tissue biopsies. This is particularly important as studies have shown different mutational profiles between CTCs and ctDNA in some tumor subtypes. However, isolating rare CTCs from whole blood has significant hurdles. Very limited DNA quantities often can’t meet NGS requirements without whole genome amplification (WGA). Moreover, white blood cells (WBC) germline contamination may confound CTC somatic mutation analyses. Thus, a good CTC enrichment platform with an efficient WGA and NGS workflow are needed. Here, Vortex label-free CTC enrichment platform was used to capture CTCs. DNA extraction was optimized, WGA evaluated and targeted NGS tested. We used metastatic colorectal cancer (CRC) as the clinical target, HCT116 as the corresponding cell line, GenomePlex® and REPLI-g as the WGA methods, GeneRead DNAseq Human CRC Panel as the 38 gene panel. The workflow was further validated on metastatic CRC patient samples, assaying both tumor and CTCs. WBCs from the same patients were included to eliminate germline contaminations. The described workflow performed well on samples with sufficient DNA, but showed bias for rare cells with limited DNA input. REPLI-g provided an unbiased amplification on fresh rare cells, enabling an accurate variant calling using the targeted NGS. Somatic variants were detected in patient CTCs and not found in age matched healthy donors. This demonstrates the feasibility of a simple workflow for clinically relevant monitoring of tumor genetics in real time and over the course of a patient’s therapy using CTCs.Liquid biopsy: Simple workflow allows DNA analysis of circulating tumor cellsA microfluidic device that isolates cancer cells circulating in a blood sample allows for real-time genetic monitoring. A team led by Elodie Sollier-Christen of Vortex Biosciences, a cancer diagnostics company in Menlo Park, California, USA, in collaboration with Professor Stefanie Jeffrey at Stanford University School of Medicine, developed a simple workflow for analyzing the genomes of rare circulating tumor cells (CTCs) found in the bloodstream after they’ve been collected through a proprietary microfluidic system. They optimized rare cell DNA extraction, compared different whole genome amplification methods, and then tested the workflow on blood samples from patients with metastatic colorectal cancer. The analysis also included white blood cells from the same patients to parse cancer-causing mutations from inherited ones. The method could aid in the translation of liquid biopsies for the clinical care of cancer patients.


Scientific Reports | 2018

Evaluation of PD-L1 expression on vortex-isolated circulating tumor cells in metastatic lung cancer

Manjima Dhar; Jessica Wong; James Che; Melissa Matsumoto; Tristan Grogan; David Elashoff; Edward B. Garon; Jonathan W. Goldman; Elodie Sollier Christen; Dino Di Carlo; Rajan P. Kulkarni

Metastatic non-small cell lung cancer (NSCLC) is a highly fatal and immunogenic malignancy. Although the immune system is known to recognize these tumor cells, one mechanism by which NSCLC can evade the immune system is via overexpression of programmed cell death ligand 1 (PD-L1). Recent clinical trials of PD-1 and PD-L1 inhibitors have returned promising clinical responses. Important for personalizing therapy, patients with higher intensity staining for PD-L1 on tumor biopsies responded better. Thus, there has been interest in using PD-L1 tumor expression as a criterion for patient selection. Currently available methods of screening involve invasive tumor biopsy, followed by histological grading of PD-L1 levels. Biopsies have a high risk of complications, and only allow sampling from limited tumor sections, which may not reflect overall tumor heterogeneity. Circulating tumor cell (CTC) PD-L1 levels could aid in screening patients, and could supplement tissue PD-L1 biopsy results by testing PD-L1 expression from disseminated tumor sites. Towards establishing CTCs as a screening tool, we developed a protocol to isolate CTCs at high purity and immunostain for PD-L1. Monitoring of PD-L1 expression on CTCs could be an additional biomarker for precision medicine that may help in determining response to immunotherapies.

Collaboration


Dive into the James Che's collaboration.

Top Co-Authors

Avatar

Dino Di Carlo

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elodie Sollier

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Manjima Dhar

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge