Network


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

Hotspot


Dive into the research topics where Andrew D. Hughes is active.

Publication


Featured researches published by Andrew D. Hughes.


Journal of Controlled Release | 2012

E-selectin liposomal and nanotube-targeted delivery of doxorubicin to circulating tumor cells

Michael J. Mitchell; Christina S. Chen; Varun Ponmudi; Andrew D. Hughes; Michael R. King

The presence of circulating tumor cells (CTCs) is believed to lead to the formation of secondary tumors via an adhesion cascade involving interaction between adhesion receptors of endothelial cells and ligands on CTCs. Many CTCs express sialylated carbohydrate ligands on their surfaces that adhere to selectin protein found on inflamed endothelial cells. We have investigated the feasibility of using immobilized selectin proteins as a targeting mechanism for CTCs under flow. Herein, targeted liposomal doxorubicin (L-DXR) was functionalized with recombinant human E-selectin (ES) and polyethylene glycol (PEG) to target and kill cancer cells under shear flow, both when immobilized along a microtube device or sheared in a cone-and-plate viscometer in a dilute suspension. Healthy circulating cells such as red blood cells were not targeted by this mechanism and were left to freely circulate, and minimal leukocyte death was observed. Halloysite nanotube (HNT)-coated microtube devices immobilized with nanoscale liposomes significantly enhanced the targeting, capture, and killing of cancer cells. This work demonstrates that E-selectin functionalized L-DXR, sheared in suspension or immobilized onto microtube devices, provides a novel approach to selectively target and deliver chemotherapeutics to CTCs in the bloodstream.


Wiley Interdisciplinary Reviews-nanomedicine and Nanobiotechnology | 2012

Nanobiotechnology for the capture and manipulation of circulating tumor cells.

Andrew D. Hughes; Michael R. King

A necessary step in metastasis is the dissemination of malignant cells into the bloodstream, where cancer cells travel throughout the body as circulating tumor cells (CTC) in search of an opportunity to seed a secondary tumor. CTC represent a valuable diagnostic tool: evidence indicates that the quantity of CTC in the blood has been shown to relate to the severity of the illness, and samples are readily obtained through routine blood draws. As such, there has been a push toward developing technologies to reliably detect CTC using a variety of molecular and immunocytochemical techniques. In addition to their use in diagnostics, CTC detection systems that isolate CTC in such a way that the cells remain viable will allow for the performance of live-cell assays to facilitate the development of personalized cancer therapies. Moreover, techniques for the direct manipulation of CTC in circulation have been developed, intending to block metastasis in situ. We review a number of current and emerging micro- and nanobiotechnology approaches for the detection, capture, and manipulation of rare CTC aimed at advancing cancer treatment.


Journal of Visualized Experiments | 2012

Rapid Isolation of Viable Circulating Tumor Cells from Patient Blood Samples

Andrew D. Hughes; Jeff Mattison; John D. Powderly; Bryan T. Greene; Michael R. King

Circulating tumor cells (CTC) are cells that disseminate from a primary tumor throughout the circulatory system and that can ultimately form secondary tumors at distant sites. CTC count can be used to follow disease progression based on the correlation between CTC concentration in blood and disease severity1. As a treatment tool, CTC could be studied in the laboratory to develop personalized therapies. To this end, CTC isolation must cause no cellular damage, and contamination by other cell types, particularly leukocytes, must be avoided as much as possible2. Many of the current techniques, including the sole FDA-approved device for CTC enumeration, destroy CTC as part of the isolation process (for more information see Ref. 2). A microfluidic device to capture viable CTC is described, consisting of a surface functionalized with E-selectin glycoprotein in addition to antibodies against epithelial markers3. To enhance device performance a nanoparticle coating was applied consisting of halloysite nanotubes, an aluminosilicate nanoparticle harvested from clay4. The E-selectin molecules provide a means to capture fast moving CTC that are pumped through the device, lending an advantage over alternative microfluidic devices wherein longer processing times are necessary to provide target cells with sufficient time to interact with a surface. The antibodies to epithelial targets provide CTC-specificity to the device, as well as provide a readily adjustable parameter to tune isolation. Finally, the halloysite nanotube coating allows significantly enhanced isolation compared to other techniques by helping to capture fast moving cells, providing increased surface area for protein adsorption, and repelling contaminating leukocytes3,4. This device is produced by a straightforward technique using off-the-shelf materials, and has been successfully used to capture cancer cells from the blood of metastatic cancer patients. Captured cells are maintained for up to 15 days in culture following isolation, and these samples typically consist of >50% viable primary cancer cells from each patient. This device has been used to capture viable CTC from both diluted whole blood and buffy coat samples. Ultimately, we present a technique with functionality in a clinical setting to develop personalized cancer therapies.


Frontiers in Oncology | 2012

Circulating tumor cells: the substrate of personalized medicine?

Bryan T. Greene; Andrew D. Hughes; Michael R. King

Circulating tumor cells (CTCs) are believed to be responsible for the development of metastatic disease. Over the last several years there has been a great interest in understanding the biology of CTCs to understand metastasis, as well as for the development of companion diagnostics to predict patient response to anti-cancer targeted therapies. Understanding CTC biology requires innovative technologies for the isolation of these rare cells. Here we review several methods for the detection, capture, and analysis of CTCs and also provide insight on improvements for CTC capture amenable to cellular therapy applications.


Cancer Letters | 2014

Differential drug responses of circulating tumor cells within patient blood

Andrew D. Hughes; Jocelyn R. Marshall; Eric Keller; John D. Powderly; Bryan T. Greene; Michael R. King

Personalized medicine holds great promise for cancer treatment, with the potential to address challenges associated with drug sensitivity and interpatient variability. Circulating tumor cells (CTC) can be useful for screening cancer drugs as they may reflect the severity and heterogeneity of primary tumors. Here we present a platform for rapidly evaluating individualized drug susceptibility. Treatment efficacy is evaluated directly in blood, employing a relevant environment for drug administration, and assessed by comparison of CTC counts in treated and control samples. Multiple drugs at varying concentrations are evaluated simultaneously to predict an appropriate therapy for individual patients.


International Journal of Molecular Sciences | 2014

Modulation of Selectin-Mediated Adhesion of Flowing Lymphoma and Bone Marrow Cells by Immobilized SDF-1

Elizabeth A. Hedges; Andrew D. Hughes; Jane L. Liesveld; Michael R. King

The α-chemokine, stromal-derived factor-1 (SDF-1), has been linked to the homing of circulating tumor cells to bone. SDF-1 is expressed by bone microvascular cells and osteoblasts and normally functions to attract blood-borne hematopoietic stem and progenitor cells to marrow. It has been shown that treatment of cancer cells with soluble SDF-1 results in a more aggressive phenotype; however, the relevance of the administration of the soluble protein is unclear. As such, a flow device was functionalized with P-selectin and SDF-1 to mimic the bone marrow microvasculature and the initial steps of cell adhesion. The introduction of SDF-1 onto the adhesive surface was found to significantly enhance the adhesion of lymphoma cells, as well as low-density bone marrow cells (LDBMC), both in terms of the number of adherent cells and the strength of cell adhesion. Thus, SDF-1 has a synergistic effect with P-selectin on cancer cell adhesion and may be sufficient to promote preferential metastasis to bone.


Bellman Prize in Mathematical Biosciences | 2017

Effect of circulating tumor cell aggregate configuration on hemodynamic transport and wall contact

Kevin J. Anderson; Adelaide de Guillebon; Andrew D. Hughes; Weiwei Wang; Michael R. King

Selectin-mediated adhesion of circulating tumor cells (CTCs) to the endothelium is a critical step in cancer metastasis, a major factor contributing to the mortality of cancer. The formation of tethers between tumor cells and endothelial selectins initiates cell rolling, which can lead to firm adhesion, extravasation and the formation of secondary metastases. Tumor cells travel through the bloodstream as single cells, or as aggregates known as circulating tumor microemboli (CTM). CTM have increased survivability and metastatic potential relative to CTCs, and the presence of CTM is associated with worse patient prognosis. The motion of cells and cellular aggregates in flow is a function of their size and shape, and these differences influence the frequency and strength of their contact with the endothelium. In this study, a computational model consisting of the hydrodynamic component of the Multiparticle Adhesive Dynamics simulation analyzed the effects of model aggregate conformation and orientation on adhesive binding potential. Model aggregates of the Colo205 colorectal cancer cell line were created, consisting of two, three, and four cells in simple geometrical conformations. Contact time, contact area, and time integral of contact area were measured as a function of fluid shear rate, initial centroid height, and initial orientation for model aggregates that experienced hydrodynamic collisions with the plane wall. It was found that larger CTM conformations with intermediate nonsphericities had the highest adhesion potential. The results of this study shed light on the correlation between environmental conditions and extravasation efficiency, which could inform the development of new anti-metastatic drugs.


northeast bioengineering conference | 2011

Use of naturally-occurring halloysite nanotubes for enhanced capture of cells from flow

Andrew D. Hughes; Michael R. King

The development of individualized treatments for cancer can be facilitated by more efficient methods for separating circulating tumor cells (CTC) from patient blood in such a way that they remain viable for live cell assays. We have previously shown that immobilized P-selectin protein can be used on the inner surface of a microscale flow system to induce leukemic cells and leukocytes to roll at different velocities and relative fluxes, thereby creating a means for rapid cell fractionation without inflicting cellular damage [1]. In this study a method for more efficient capture was developed. This enhancement was achieved by altering the nanoscale topography of the inner surface of selectin-coated MicroRenathane (MRE) microtubes using naturally occurring halloysite nanotubes. Immobilized nanotubes on the MRE tube surface provide enhanced capture by two means: (1) The surface area of the MRE tube inner surface is increased, allowing for more protein adsorption, and (2) the characteristic dimensions of the nanotubes allow for selectin proteins to be presented away from the surface, spanning the hydrodynamic lubrication layer that opposes cell contact with the surface. Comprehensive theoretical and experimental analyses of the nanotube coating has confirmed this, as well as show that the macroscale and microscale fluid dynamics in the MRE tube are unaltered by the coating. Consequently, halloysite nanotube coatings provide a straightforward engineering solution to an inherent fluidics obstacle, potentiating our ability to capture viable CTC for study, diagnosis, and treatment of cancer on a patient-to-patient basis.


ASME 2010 8th International Conference on Nanochannels, Microchannels, and Minichannels collocated with 3rd Joint US-European Fluids Engineering Summer Meeting | 2010

Nanoparticle-Coated Microtubes for the Manipulation of Cancer Cells

Robin Zhou; Andrew D. Hughes; Jane L. Liesveld; Michael R. King

The development of novel methods for the isolation of primary stem and progenitor cells is important for the treatment of blood cancers, tissue engineering, and basic research in the biomedical sciences. Our lab has previously shown that microtubes coated with P-selectin protein can be used to capture and enrich hematopoietic stem and progenitor cells from a mixture of cells perfused through the tube at physiologically-relevant shear stresses[1][2], and that using a surface coating of colloidal silica nanoparticles (12 nm diameter, 30% by weight SiO2 ) increased cell capture and decreased rolling velocity[3]. Here we show that 50 nm colloidal silica nanoparticle coatings may similarly increase cell capture, and that these protocols are effective for enrichment of human adult CD34-positive HSCs from primary apheresis and bone marrow aspirate samples. Future research may include long-term colony-forming assays to confirm stem cell activity of enriched cells, and transplantation in immune-deficient mice.© 2010 ASME


Archive | 2010

Fluid Flow Device Containing Nanotubes and Method for Cell Trafficking Using Same

Michael R. King; Andrew D. Hughes

Collaboration


Dive into the Andrew D. Hughes'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