Quick chip assay using locked nucleic acid modified epithelial cell adhesion molecule and nucleolin aptamers for the capture of circulating tumor cells.

The role of circulating tumor cells (CTCs) in disease diagnosis, prognosis, monitoring of the therapeutic efficacy, and clinical decision making is immense and has attracted tremendous focus in the last decade. We designed and fabricated simple, flat channel microfluidic devices polydimethylsiloxane (PDMS based) functionalized with locked nucleic acid (LNA) modified aptamers (targeting epithelial cell adhesion molecule (EpCAM) and nucleolin expression) for quick and efficient capture of CTCs and cancer cells. With optimized flow rates (10 μl/min), it was revealed that the aptamer modified devices offered reusability for up to six times while retaining optimal capture efficiency (>90%) and specificity. High capture sensitivity (92%) and specificity (100%) was observed in whole blood samples spiked with Caco-2 cells (10-100 cells/ml). Analysis of blood samples obtained from 25 head and neck cancer patients on the EpCAM LNA aptamer functionalized chip revealed that an average count of 5 ± 3 CTCs/ml of blood were captured from 22/25 samples (88%). EpCAM intracellular domain (EpICD) immunohistochemistry on 9 oral squamous cell carcinomas showed the EpICD positivity in the tumor cells, confirming the EpCAM expression in CTCs from head and neck cancers. These microfluidic devices also maintained viability for in vitro culture and characterization. Use of LNA modified aptamers provided added benefits in terms of cost effectiveness due to increased reusability and sustainability of the devices. Our results present a robust, quick, and efficient CTC capture platform with the use of simple PDMS based devices that are easy to fabricate at low cost and have an immense potential in cancer diagnosis, prognosis, and therapeutic planning.

[1]  J. Wengel,et al.  Locked vs. unlocked nucleic acids (LNA vs. UNA): contrasting structures work towards common therapeutic goals. , 2011, Chemical Society reviews.

[2]  Ciprian Iliescu,et al.  Label-free isolation of circulating tumor cells in microfluidic devices: Current research and perspectives. , 2013, Biomicrofluidics.

[3]  Peng Li,et al.  Probing circulating tumor cells in microfluidics. , 2013, Lab on a chip.

[4]  M. Famulok,et al.  Nucleic acid aptamers-from selection in vitro to applications in vivo. , 2000, Accounts of chemical research.

[5]  Kyung-A Hyun,et al.  Advances and critical concerns with the microfluidic enrichments of circulating tumor cells. , 2014, Lab on a chip.

[6]  Weihong Tan,et al.  Enrichment of cancer cells using aptamers immobilized on a microfluidic channel. , 2009, Analytical chemistry.

[7]  Zhaohai Wang,et al.  Increased level of nucleolin confers to aggressive tumor progression and poor prognosis in patients with hepatocellular carcinoma after hepatectomy , 2014, Diagnostic Pathology.

[8]  S. Potrč,et al.  Generating Aptamers for Cancer Diagnosis and Therapy , 2012 .

[9]  M. Herlyn,et al.  Colorectal carcinoma-specific antigen: detection by means of monoclonal antibodies. , 1979, Proceedings of the National Academy of Sciences of the United States of America.

[10]  Brigitte Rack,et al.  Detection of Circulating Tumor Cells in Peripheral Blood of Patients with Metastatic Breast Cancer: A Validation Study of the CellSearch System , 2007, Clinical Cancer Research.

[11]  K. Pantel,et al.  Prognostic Relevance of Circulating Tumor Cells in Blood and Disseminated Tumor Cells in Bone Marrow of Patients with Squamous Cell Carcinoma of the Oral Cavity , 2013, Clinical Cancer Research.

[12]  R. Kanwar,et al.  Cancer Targeted Nanoparticles Specifically Induce Apoptosis in Cancer Cells and Spare Normal Cells , 2012 .

[13]  S. Warnakulasuriya Global epidemiology of oral and oropharyngeal cancer. , 2009, Oral oncology.

[14]  R. Kanwar,et al.  Locked nucleic acid modified bi-specific aptamer-targeted nanoparticles carrying survivin antagonist towards effective colon cancer therapy , 2015 .

[15]  S. Nisole,et al.  The cell-surface-expressed nucleolin is associated with the actin cytoskeleton. , 2000, Experimental cell research.

[16]  Qiao Lin,et al.  Specific capture and temperature-mediated release of cells in an aptamer-based microfluidic device. , 2012, Lab on a chip.

[17]  Weihong Tan,et al.  Multivalent DNA nanospheres for enhanced capture of cancer cells in microfluidic devices. , 2013, ACS nano.

[18]  J. R. Kanwar,et al.  The significance of free and immune-complexed hydatid-specific antigen(s) as an immunodiagnostic tool for human hydatidosis. , 1992, Journal of medical microbiology.

[19]  Tatsuro Watanabe,et al.  Nucleolin on the cell surface as a new molecular target for gastric cancer treatment. , 2010, Biological & pharmaceutical bulletin.

[20]  J. Wengel,et al.  Locked Nucleic Acids: Promising Nucleic Acid Analogs for Therapeutic Applications , 2010, Chemistry & biodiversity.

[21]  A. Norton,et al.  Brief, high‐temperature heat denaturation (pressure cooking): A simple and effective method of antigen retrieval for routinely processed tissues , 1994, The Journal of pathology.

[22]  Klaus Pantel,et al.  Circulating tumor cells: liquid biopsy of cancer. , 2013, Clinical chemistry.

[23]  Kemin Wang,et al.  Selection of aptamers for molecular recognition and characterization of cancer cells. , 2007, Analytical chemistry.

[24]  Kyung-A Hyun,et al.  Continual collection and re-separation of circulating tumor cells from blood using multi-stage multi-orifice flow fractionation. , 2013, Biomicrofluidics.

[25]  P. Bouvet,et al.  AS-1411, a guanosine-rich oligonucleotide aptamer targeting nucleolin for the potential treatment of cancer, including acute myeloid leukemia. , 2010, Current opinion in molecular therapeutics.

[26]  Paul I. Okagbare,et al.  Highly efficient capture and enumeration of low abundance prostate cancer cells using prostate‐specific membrane antigen aptamers immobilized to a polymeric microfluidic device , 2009, Electrophoresis.

[27]  Nam-Trung Nguyen,et al.  Rare cell isolation and analysis in microfluidics. , 2014, Lab on a chip.

[28]  Ronald R. Breaker,et al.  Natural and engineered nucleic acids as tools to explore biology , 2004, Nature.

[29]  P. Brown,et al.  Parallel human genome analysis: microarray-based expression monitoring of 1000 genes. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[30]  E. Lianidou,et al.  Circulating tumor cells as promising novel biomarkers in solid cancers , 2014, Critical reviews in clinical laboratory sciences.

[31]  P. Bates,et al.  Antiproliferative activity of G-quartet-forming oligonucleotides with backbone and sugar modifications. , 2002, Biochemistry.

[32]  Daniel F. Hayes,et al.  Sensitive capture of circulating tumour cells by functionalised graphene oxide nanosheets , 2013, Nature nanotechnology.

[33]  H. Pollard,et al.  Molecular dissection of nucleolin's role in growth and cell proliferation: new insights , 1999, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[34]  P. Bates,et al.  Antiproliferative Activity of G-rich Oligonucleotides Correlates with Protein Binding* , 1999, The Journal of Biological Chemistry.

[35]  Xingyu Jiang,et al.  Size-based hydrodynamic rare tumor cell separation in curved microfluidic channels. , 2013, Biomicrofluidics.

[36]  M F Lawrence,et al.  Impedance based DNA chip for direct T(m) measurement. , 2002, Talanta.

[37]  Weihong Tan,et al.  Aptamer-enabled efficient isolation of cancer cells from whole blood using a microfluidic device. , 2012, Analytical chemistry.

[38]  Thomas J George,et al.  Capture, release and culture of circulating tumor cells from pancreatic cancer patients using an enhanced mixing chip. , 2014, Lab on a chip.

[39]  Richard J. Lee,et al.  Circulating tumour cells—monitoring treatment response in prostate cancer , 2014, Nature Reviews Clinical Oncology.

[40]  Omid C. Farokhzad,et al.  Current Progress of Aptamer-Based Molecular Imaging , 2014, The Journal of Nuclear Medicine.

[41]  Alessandro Lugli,et al.  Frequent EpCam protein expression in human carcinomas. , 2004, Human pathology.

[42]  Ying Liu,et al.  Aptamer-containing surfaces for selective capture of CD4 expressing cells. , 2012, Langmuir : the ACS journal of surfaces and colloids.

[43]  K. Pantel,et al.  Technologies for detection of circulating tumor cells: facts and vision. , 2014, Lab on a chip.

[44]  J. Visvader,et al.  Cancer stem cells in solid tumours: accumulating evidence and unresolved questions , 2008, Nature Reviews Cancer.

[45]  W. Oberaigner,et al.  The expression of EGFR, HER2 and EpCam in Head and Neck squamous cell carcinomas , 2009 .

[46]  S. Kubota,et al.  5′-,3′-Inverted Thymidine-modified Antisense Oligodeoxynucleotide Targeting Midkine , 2002, The Journal of Biological Chemistry.

[47]  R. Kanwar,et al.  Chimeric aptamers in cancer cell-targeted drug delivery , 2011, Critical reviews in biochemistry and molecular biology.

[48]  Weihong Tan,et al.  Optimization and Modifications of Aptamers Selected from Live Cancer Cell Lines , 2007, Chembiochem : a European journal of chemical biology.

[49]  M Vasei,et al.  Frequent high-level expression of the immunotherapeutic target Ep-CAM in colon, stomach, prostate and lung cancers , 2006, British Journal of Cancer.

[50]  A. Armstrong,et al.  EpCAM: A New Therapeutic Target for an Old Cancer Antigen , 2003, Cancer biology & therapy.

[51]  P. Gascoyne,et al.  Antibody-independent isolation of circulating tumor cells by continuous-flow dielectrophoresis. , 2013, Biomicrofluidics.

[52]  Seonghwan Lee,et al.  Aptamers and Their Biological Applications , 2012, Sensors.

[53]  W. Duan,et al.  RNA aptamer against a cancer stem cell marker epithelial cell adhesion molecule , 2011, Cancer science.

[54]  D. Shangguan,et al.  Aptamers evolved from live cells as effective molecular probes for cancer study , 2006, Proceedings of the National Academy of Sciences.

[55]  S. Litvinov,et al.  The biology of the 17–1A antigen (Ep-CAM) , 1999, Journal of Molecular Medicine.

[56]  P. Bates,et al.  Oncogenic synergism between ErbB1, nucleolin, and mutant Ras. , 2011, Cancer research.

[57]  M. Rots,et al.  EpCAM in carcinogenesis: the good, the bad or the ugly. , 2010, Carcinogenesis.

[58]  I. Nagtegaal,et al.  The epithelial cell adhesion molecule (Ep-CAM) as a morphoregulatory molecule is a tool in surgical pathology. , 2003, The American journal of pathology.

[59]  Weihong Tan,et al.  Aptamer-based microfluidic device for enrichment, sorting, and detection of multiple cancer cells. , 2009, Analytical chemistry.

[60]  Brigitte Mack,et al.  Nuclear signalling by tumour-associated antigen EpCAM , 2009, Nature Cell Biology.

[61]  E. M. Reyes-Reyes,et al.  Cell-surface nucleolin is a signal transducing P-selectin binding protein for human colon carcinoma cells. , 2008, Experimental cell research.

[62]  L. Mcdonald,et al.  The pathology of oral cancer , 2003, Pathology.