Network


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

Hotspot


Dive into the research topics where Shannon L. Stott is active.

Publication


Featured researches published by Shannon L. Stott.


Science | 2013

Circulating Breast Tumor Cells Exhibit Dynamic Changes in Epithelial and Mesenchymal Composition

Min Yu; Aditya Bardia; Ben S. Wittner; Shannon L. Stott; Malgorzata E. Smas; David T. Ting; Steven J. Isakoff; Jordan C. Ciciliano; Mn Wells; Ajay M. Shah; Kyle Concannon; Maria C. Donaldson; Lecia V. Sequist; Elena F. Brachtel; Dennis C. Sgroi; José Baselga; Sridhar Ramaswamy; Mehmet Toner; Daniel A. Haber; Shyamala Maheswaran

Cells in Transit(ion) Epithelial-mesenchymal transition (EMT) is a developmental program that converts adherent epithelial cells to a migratory mesenchymal state. This cell-fate change has been linked to tumor metastasis in preclinical models. To investigate whether EMT occurs in human cancer, Yu et al. (p. 580) isolated circulating tumor cells (CTCs) from breast cancer patients and analyzed their expression of epithelial and mesenchymal markers by RNA–in situ hybridization and RNA sequencing. Biphenotypic cells expressing both types of markers were rare in primary breast tumors but were enriched among CTCs, as were cells expressing only mesenchymal markers. Serial blood samples from one patient revealed that CTCs in the mesenchymal state declined in number when the patient responded to therapy but rebounded when the disease began to progress—a pattern repeated when a different therapy was administered. Thus, EMT may facilitate tumor cell dissemination in humans. Tumor cells circulating in the blood of cancer patients undergo a phenotypic change that may facilitate their spread. Epithelial-mesenchymal transition (EMT) of adherent epithelial cells to a migratory mesenchymal state has been implicated in tumor metastasis in preclinical models. To investigate its role in human cancer, we characterized EMT in circulating tumor cells (CTCs) from breast cancer patients. Rare primary tumor cells simultaneously expressed mesenchymal and epithelial markers, but mesenchymal cells were highly enriched in CTCs. Serial CTC monitoring in 11 patients suggested an association of mesenchymal CTCs with disease progression. In an index patient, reversible shifts between these cell fates accompanied each cycle of response to therapy and disease progression. Mesenchymal CTCs occurred as both single cells and multicellular clusters, expressing known EMT regulators, including transforming growth factor (TGF)–β pathway components and the FOXC1 transcription factor. These data support a role for EMT in the blood-borne dissemination of human breast cancer.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Isolation of circulating tumor cells using a microvortex-generating herringbone-chip

Shannon L. Stott; Chia Hsien Hsu; Dina I. Tsukrov; Min Yu; David T. Miyamoto; Belinda A. Waltman; S. Michael Rothenberg; Ajay M. Shah; Malgorzata E. Smas; George K. Korir; Frederick P. Floyd; Anna J. Gilman; Jenna B. Lord; Daniel Winokur; Simeon Springer; Daniel Irimia; Sunitha Nagrath; Lecia V. Sequist; Richard J. Lee; Kurt J. Isselbacher; Shyamala Maheswaran; Daniel A. Haber; Mehmet Toner

Rare circulating tumor cells (CTCs) present in the bloodstream of patients with cancer provide a potentially accessible source for detection, characterization, and monitoring of nonhematological cancers. We previously demonstrated the effectiveness of a microfluidic device, the CTC-Chip, in capturing these epithelial cell adhesion molecule (EpCAM)-expressing cells using antibody-coated microposts. Here, we describe a high-throughput microfluidic mixing device, the herringbone-chip, or “HB-Chip,” which provides an enhanced platform for CTC isolation. The HB-Chip design applies passive mixing of blood cells through the generation of microvortices to significantly increase the number of interactions between target CTCs and the antibody-coated chip surface. Efficient cell capture was validated using defined numbers of cancer cells spiked into control blood, and clinical utility was demonstrated in specimens from patients with prostate cancer. CTCs were detected in 14 of 15 (93%) patients with metastatic disease (median = 63 CTCs/mL, mean = 386 ± 238 CTCs/mL), and the tumor-specific TMPRSS2-ERG translocation was readily identified following RNA isolation and RT-PCR analysis. The use of transparent materials allowed for imaging of the captured CTCs using standard clinical histopathological stains, in addition to immunofluorescence-conjugated antibodies. In a subset of patient samples, the low shear design of the HB-Chip revealed microclusters of CTCs, previously unappreciated tumor cell aggregates that may contribute to the hematogenous dissemination of cancer.


Journal of Cell Biology | 2011

Circulating tumor cells: approaches to isolation and characterization

Min Yu; Shannon L. Stott; Mehmet Toner; Shyamala Maheswaran; Daniel A. Haber

Circulating tumor cells (CTCs) shed from primary and metastatic cancers are admixed with blood components and are thus rare, making their isolation and characterization a major technological challenge. CTCs hold the key to understanding the biology of metastasis and provide a biomarker to noninvasively measure the evolution of tumor genotypes during treatment and disease progression. Improvements in technologies to yield purer CTC populations amenable to better cellular and molecular characterization will enable a broad range of clinical applications, including early detection of disease and the discovery of biomarkers to predict treatment responses and disease progression.


Cell | 2014

Circulating Tumor Cell Clusters Are Oligoclonal Precursors of Breast Cancer Metastasis

Nicola Aceto; Aditya Bardia; David T. Miyamoto; Maria C. Donaldson; Ben S. Wittner; Joel A. Spencer; Min Yu; Adam Pely; Amanda Engstrom; Huili Zhu; Brian W. Brannigan; Ravi Kapur; Shannon L. Stott; Toshi Shioda; Sridhar Ramaswamy; David T. Ting; Charles P. Lin; Mehmet Toner; Daniel A. Haber; Shyamala Maheswaran

Circulating tumor cell clusters (CTC clusters) are present in the blood of patients with cancer but their contribution to metastasis is not well defined. Using mouse models with tagged mammary tumors, we demonstrate that CTC clusters arise from oligoclonal tumor cell groupings and not from intravascular aggregation events. Although rare in the circulation compared with single CTCs, CTC clusters have 23- to 50-fold increased metastatic potential. In patients with breast cancer, single-cell resolution RNA sequencing of CTC clusters and single CTCs, matched within individual blood samples, identifies the cell junction component plakoglobin as highly differentially expressed. In mouse models, knockdown of plakoglobin abrogates CTC cluster formation and suppresses lung metastases. In breast cancer patients, both abundance of CTC clusters and high tumor plakoglobin levels denote adverse outcomes. Thus, CTC clusters are derived from multicellular groupings of primary tumor cells held together through plakoglobin-dependent intercellular adhesion, and though rare, they greatly contribute to the metastatic spread of cancer.


Science Translational Medicine | 2013

Inertial Focusing for Tumor Antigen–Dependent and –Independent Sorting of Rare Circulating Tumor Cells

Emre Özkumur; Ajay M. Shah; Jordan C. Ciciliano; Benjamin L. Emmink; David T. Miyamoto; Elena F. Brachtel; Min Yu; Pin-i Chen; Bailey Morgan; Julie Trautwein; Anya M. Kimura; Sudarshana Sengupta; Shannon L. Stott; Nezihi Murat Karabacak; Tom Barber; John Walsh; Kyle C. Smith; Philipp S. Spuhler; James P. Sullivan; Richard J. Lee; David T. Ting; Xi Luo; Alice T. Shaw; Aditya Bardia; Lecia V. Sequist; David N. Louis; Shyamala Maheswaran; Ravi Kapur; Daniel A. Haber; Mehmet Toner

A multistage microfluidic chip is capable of sorting rare EpCAM+ and EpCAM− CTCs from cancer patients’ whole blood. Positive and Negative Outcomes Usually people want the good news first, to help cope with the bad news that inevitably follows. However, patients will soon desire both the positive and the negative outcomes together, according to the latest study by Ozkumur and colleagues. These authors have developed a multistage microfluidic device that is capable of sorting rare circulating tumor cells (CTCs) that are either positive or negative for the surface antigen epithelial cell adhesion molecule (EpCAM). EpCAM+ cells found in the bloodstream have long defined the typical CTC. Many sorting technologies have been developed to enumerate EpCAM+ CTCs in cancer patient’s blood; however, these cells are not always detectable in cancers with low EpCAM expression, like triple-negative breast cancer or melanoma. Ozkumur et al. engineered an automated platform, called the “CTC-iChip,” that captured both EpCAM+ and EpCAM− cancer cells in clinical samples using a series of debulking, inertial focusing, and magnetic separation steps. The sorted CTCs could then be interrogated using standard clinical protocols, such as immunocytochemistry. The authors tested the “positive mode” of their device using whole blood from patients with prostate, lung, breast, pancreatic, and colorectal cancers. After successfully separating out the EpCAM+ CTCs, they confirmed that the cells were viable and had high-quality RNA for molecular analysis, in one example, detecting the EML4-ALK gene fusion in lung cancer. Using the “negative mode” of their device, the authors were able to capture EpCAM− CTCs from patients with metastatic breast cancer, pancreatic cancer, and melanoma. The isolated CTCs showed similar morphology when compared with primary tumor tissue from these patients, suggesting that the microfluidic device can be used for clinical diagnoses—delivering both positive and negative news at once. Ozkumur et al. also demonstrated that CTCs isolated using the iChip could be analyzed on the single-cell level. One such demonstration with 15 CTCs from a prostate cancer patient reveals marked heterogeneity in the expression of mesenchymal and stem cell markers as well as typical prostate cancer–related antigens. The CTC-iChip can therefore process large volumes of patient blood to obtain not just EpCAM+ CTCs but also the EpCAM− ones, thus giving a broader picture of an individual’s cancer status and also allowing the device to be used for more cancer types. With the ability to further analyze the molecular characteristics of CTCs, this CTC-iChip could be a promising addition to current diagnostic tools used in the clinic. Circulating tumor cells (CTCs) are shed into the bloodstream from primary and metastatic tumor deposits. Their isolation and analysis hold great promise for the early detection of invasive cancer and the management of advanced disease, but technological hurdles have limited their broad clinical utility. We describe an inertial focusing–enhanced microfluidic CTC capture platform, termed “CTC-iChip,” that is capable of sorting rare CTCs from whole blood at 107 cells/s. Most importantly, the iChip is capable of isolating CTCs using strategies that are either dependent or independent of tumor membrane epitopes, and thus applicable to virtually all cancers. We specifically demonstrate the use of the iChip in an expanded set of both epithelial and nonepithelial cancers including lung, prostate, pancreas, breast, and melanoma. The sorting of CTCs as unfixed cells in solution allows for the application of high-quality clinically standardized morphological and immunohistochemical analyses, as well as RNA-based single-cell molecular characterization. The combination of an unbiased, broadly applicable, high-throughput, and automatable rare cell sorting technology with generally accepted molecular assays and cytology standards will enable the integration of CTC-based diagnostics into the clinical management of cancer.


Science Translational Medicine | 2010

Isolation and Characterization of Circulating Tumor Cells from Patients with Localized and Metastatic Prostate Cancer

Shannon L. Stott; Richard J. Lee; Sunitha Nagrath; Min Yu; David T. Miyamoto; Lindsey Ulkus; Elizabeth J. Inserra; Matthew Ulman; Simeon Springer; Zev Nakamura; Alessandra L. Moore; Dina I. Tsukrov; Maria E. Kempner; Douglas M. Dahl; Chin-Lee Wu; A. John Iafrate; Matthew R. Smith; Ronald G. Tompkins; Lecia V. Sequist; Mehmet Toner; Daniel A. Haber; Shyamala Maheswaran

Automated imaging of prostate-specific cancer cells from the blood provides a measure of circulating tumor cell half-life after tumor resection. Circling Cancers Out Oftentimes a patient and his or her clinician learn together at the flip of a radiological imaging scan that a solid tumor, previously removed, has returned either at the original site or at new locations to which it has spread. A failure of cancer treatment—but could it have been predicted? Whether it is freely floating tumor DNA or tumor cells, the circulation of cancer-derived material in the blood holds great promise for the early detection and prevention of cancer metastases. The accurate identification, enumeration, and molecular classification of blood-borne cells—although postulated more than 140 years ago—remain the greatest challenge. Now, in a small cohort of individuals with and without prostate cancer, Stott and colleagues have used a silicon microfluidic cell-capture technology that, when coupled to an automated imaging system, enables the detection and enumeration of prostate cancer cells fished out from the blood. These cells express a surface protein that uniquely identifies epithelial cells in the circulation. Once efficiently captured by antibody to this protein, the cells are counterstained with antibodies that are prostate-specific and that indicate cell proliferation, suggesting that these circulating cells are ready to repopulate distant metastatic sites. In their study, tumor cells obtained from the blood of cancer patients were monitored before and after surgery; some circulating cells persisted months after surgery while others rapidly declined shortly thereafter. Whether the persistence or disappearance of lurking cancer cells reflects an intrinsic capacity for reseeding remains to be established, but the system used in the study offers great potential for oncologists to detect cancer-related changes earlier and to monitor responses to drug treatments. In the hope of ultimately applying personalized treatments to cancer patients, based on “real-time” monitoring of the tumor cell genetic makeup, this approach to identifying these circulating cells early on moves us beyond the finality of the films currently offered to patients in the clinic. Rare circulating tumor cells (CTCs) are present in the blood of patients with metastatic epithelial cancers but have been difficult to measure routinely. We report a quantitative automated imaging system for analysis of prostate CTCs, taking advantage of prostate-specific antigen (PSA), a unique prostate tumor–associated marker. The specificity of PSA staining enabled optimization of criteria for baseline image intensity, morphometric measurements, and integration of multiple signals in a three-dimensional microfluidic device. In a pilot analysis, we detected CTCs in prostate cancer patients with localized disease, before surgical tumor removal in 8 of 19 (42%) patients (range, 38 to 222 CTCs per milliliter). For 6 of the 8 patients with preoperative CTCs, a precipitous postoperative decline (<24 hours) suggests a short half-life for CTCs in the blood circulation. Other patients had persistent CTCs for up to 3 months after prostate removal, suggesting early but transient disseminated tumor deposits. In patients with metastatic prostate cancer, CTCs were detected in 23 of 36 (64%) cases (range, 14 to 5000 CTCs per milliliter). In previously untreated patients followed longitudinally, the numbers of CTCs declined after the initiation of effective therapy. The prostate cancer–specific TMPRSS2-ERG fusion was detectable in RNA extracted from CTCs from 9 of 20 (45%) patients with metastatic disease, and dual staining of captured CTCs for PSA and the cell division marker Ki67 indicated a broad range for the proportion of proliferating cells among CTCs. This method for analysis of CTCs will facilitate the application of noninvasive tumor sampling to direct targeted therapies in advanced prostate cancer and warrants the initiation of long-term clinical studies to test the importance of CTCs in invasive localized disease.


Nature Protocols | 2014

Microfluidic, marker-free isolation of circulating tumor cells from blood samples

Nezihi Murat Karabacak; Philipp S. Spuhler; Fabio Fachin; Eugene J. Lim; Vincent Pai; Emre Özkumur; Joseph M. Martel; Nikola Kojic; Kyle C. Smith; Pin-i Chen; Jennifer Yang; Henry Hwang; Bailey Morgan; Julie Trautwein; Tom Barber; Shannon L. Stott; Shyamala Maheswaran; Ravi Kapur; Daniel A. Haber; Mehmet Toner

The ability to isolate and analyze rare circulating tumor cells (CTCs) has the potential to further our understanding of cancer metastasis and enhance the care of cancer patients. In this protocol, we describe the procedure for isolating rare CTCs from blood samples by using tumor antigen–independent microfluidic CTC-iChip technology. The CTC-iChip uses deterministic lateral displacement, inertial focusing and magnetophoresis to sort up to 107 cells/s. By using two-stage magnetophoresis and depletion antibodies against leukocytes, we achieve 3.8-log depletion of white blood cells and a 97% yield of rare cells with a sample processing rate of 8 ml of whole blood/h. The CTC-iChip is compatible with standard cytopathological and RNA-based characterization methods. This protocol describes device production, assembly, blood sample preparation, system setup and the CTC isolation process. Sorting 8 ml of blood sample requires 2 h including setup time, and chip production requires 2–5 d.


Nature | 2012

RNA sequencing of pancreatic circulating tumour cells implicates WNT signalling in metastasis

Min Yu; David T. Ting; Shannon L. Stott; Ben S. Wittner; Fatih Ozsolak; Suchismita Paul; Jordan C. Ciciliano; Malgorzata E. Smas; Daniel Winokur; Anna J. Gilman; Matthew Ulman; Kristina Xega; Gianmarco Contino; Brinda Alagesan; Brian W. Brannigan; Patrice M. Milos; David P. Ryan; Lecia V. Sequist; Nabeel Bardeesy; Sridhar Ramaswamy; Mehmet Toner; Shyamala Maheswaran; Daniel A. Haber

Circulating tumour cells (CTCs) shed into blood from primary cancers include putative precursors that initiate distal metastases. Although these cells are extraordinarily rare, they may identify cellular pathways contributing to the blood-borne dissemination of cancer. Here, we adapted a microfluidic device for efficient capture of CTCs from an endogenous mouse pancreatic cancer model and subjected CTCs to single-molecule RNA sequencing, identifying Wnt2 as a candidate gene enriched in CTCs. Expression of WNT2 in pancreatic cancer cells suppresses anoikis, enhances anchorage-independent sphere formation, and increases metastatic propensity in vivo. This effect is correlated with fibronectin upregulation and suppressed by inhibition of MAP3K7 (also known as TAK1) kinase. In humans, formation of non-adherent tumour spheres by pancreatic cancer cells is associated with upregulation of multiple WNT genes, and pancreatic CTCs revealed enrichment for WNT signalling in 5 out of 11 cases. Thus, molecular analysis of CTCs may identify candidate therapeutic targets to prevent the distal spread of cancer.


Nature Methods | 2015

A microfluidic device for label-free, physical capture of circulating tumor cell clusters

A. Fatih Sarioglu; Nicola Aceto; Nikola Kojic; Maria C. Donaldson; Mahnaz Zeinali; Bashar Hamza; Amanda Engstrom; Huili Zhu; Tilak Sundaresan; David T. Miyamoto; Xi Luo; Aditya Bardia; Ben S. Wittner; Sridhar Ramaswamy; Toshi Shioda; David T. Ting; Shannon L. Stott; Ravi Kapur; Shyamala Maheswaran; Daniel A. Haber; Mehmet Toner

Cancer cells metastasize through the bloodstream either as single migratory circulating tumor cells (CTCs) or as multicellular groupings (CTC clusters). Existing technologies for CTC enrichment are designed to isolate single CTCs, and although CTC clusters are detectable in some cases, their true prevalence and significance remain to be determined. Here we developed a microchip technology (the Cluster-Chip) to capture CTC clusters independently of tumor-specific markers from unprocessed blood. CTC clusters are isolated through specialized bifurcating traps under low–shear stress conditions that preserve their integrity, and even two-cell clusters are captured efficiently. Using the Cluster-Chip, we identified CTC clusters in 30–40% of patients with metastatic breast or prostate cancer or with melanoma. RNA sequencing of CTC clusters confirmed their tumor origin and identified tissue-derived macrophages within the clusters. Efficient capture of CTC clusters will enable the detailed characterization of their biological properties and role in metastasis.


Clinical Cancer Research | 2016

Detection of T790M, the Acquired Resistance EGFR Mutation, by Tumor Biopsy versus Noninvasive Blood-Based Analyses

Tilak Sundaresan; Lecia V. Sequist; John V. Heymach; Gregory J. Riely; Pasi A. Jänne; Walter H. Koch; James P. Sullivan; Douglas Fox; Robert C. Maher; Alona Muzikansky; Andrew Webb; Hai T. Tran; Uma Giri; Martin Fleisher; Helena A. Yu; Wen Wei; Bruce E. Johnson; Tom Barber; John Walsh; Jeffrey A. Engelman; Shannon L. Stott; Ravi Kapur; Shyamala Maheswaran; Mehmet Toner; Daniel A. Haber

Purpose: The T790M gatekeeper mutation in the EGFR is acquired by some EGFR-mutant non–small cell lung cancers (NSCLC) as they become resistant to selective tyrosine kinase inhibitors (TKI). As third-generation EGFR TKIs that overcome T790M-associated resistance become available, noninvasive approaches to T790M detection will become critical to guide management. Experimental Design: As part of a multi-institutional Stand-Up-To-Cancer collaboration, we performed an exploratory analysis of 40 patients with EGFR-mutant tumors progressing on EGFR TKI therapy. We compared the T790M genotype from tumor biopsies with analysis of simultaneously collected circulating tumor cells (CTC) and circulating tumor DNA (ctDNA). Results: T790M genotypes were successfully obtained in 30 (75%) tumor biopsies, 28 (70%) CTC samples, and 32 (80%) ctDNA samples. The resistance-associated mutation was detected in 47% to 50% of patients using each of the genotyping assays, with concordance among them ranging from 57% to 74%. Although CTC- and ctDNA-based genotyping were each unsuccessful in 20% to 30% of cases, the two assays together enabled genotyping in all patients with an available blood sample, and they identified the T790M mutation in 14 (35%) patients in whom the concurrent biopsy was negative or indeterminate. Conclusions: Discordant genotypes between tumor biopsy and blood-based analyses may result from technological differences, as well as sampling different tumor cell populations. The use of complementary approaches may provide the most complete assessment of each patients cancer, which should be validated in predicting response to T790M-targeted inhibitors. Clin Cancer Res; 22(5); 1103–10. ©2015 AACR.

Collaboration


Dive into the Shannon L. Stott's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge